Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(4): 1686-92, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245484

ABSTRACT

We identified TMPRSS2 as a gene that is down-regulated in androgen-independent prostate cancer xenograft tissue derived from a bone metastasis. Using specific monoclonal antibodies, we show that the TMPRSS2-encoded serine protease is expressed as a Mr 70,000 full-length form and a cleaved Mr 32,000 protease domain. Mutation of Ser-441 in the catalytic triad shows that the proteolytic cleavage is dependent on catalytic activity, suggesting that it occurs as a result of autocleavage. Mutational analysis reveals the cleavage site to be at Arg-255. A consequence of autocatalytic cleavage is the secretion of the protease domain into the media by TMPRSS2-expressing prostate cancer cells and into the sera of prostate tumor-bearing mice. Immunohistochemical analysis of clinical specimens demonstrates the highest expression of TMPRSS2 at the apical side of prostate and prostate cancer secretory epithelia and within the lumen of the glands. Similar luminal staining was detected in colon cancer samples. Expression was also seen in colon and pancreas, with little to no expression detected in seven additional normal tissues. These data demonstrate that TMPRSS2 is a secreted protease that is highly expressed in prostate and prostate cancer, making it a potential target for cancer therapy and diagnosis.


Subject(s)
Androgens/physiology , Prostate/metabolism , Prostatic Neoplasms/metabolism , Serine Endopeptidases/metabolism , Animals , Catalysis , Culture Media , Down-Regulation , Epithelium/enzymology , Epithelium/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Prostate/enzymology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Receptors, Androgen/physiology , Serine Endopeptidases/genetics , Signal Transduction/physiology , Transplantation, Heterologous , Tumor Cells, Cultured
2.
Proc Natl Acad Sci U S A ; 96(25): 14523-8, 1999 Dec 07.
Article in English | MEDLINE | ID: mdl-10588738

ABSTRACT

In search of novel genes expressed in metastatic prostate cancer, we subtracted cDNA isolated from benign prostatic hypertrophic tissue from cDNA isolated from a prostate cancer xenograft model that mimics advanced disease. One novel gene that is highly expressed in advanced prostate cancer encodes a 339-amino acid protein with six potential membrane-spanning regions flanked by hydrophilic amino- and carboxyl-terminal domains. This structure suggests a potential function as a channel or transporter protein. This gene, named STEAP for six-transmembrane epithelial antigen of the prostate, is expressed predominantly in human prostate tissue and is up-regulated in multiple cancer cell lines, including prostate, bladder, colon, ovarian, and Ewing sarcoma. Immunohistochemical analysis of clinical specimens demonstrates significant STEAP expression at the cell-cell junctions of the secretory epithelium of prostate and prostate cancer cells. Little to no staining was detected at the plasma membranes of normal, nonprostate human tissues, except for bladder tissue, which expressed low levels of STEAP at the cell membrane. Protein analysis located STEAP at the cell surface of prostate-cancer cell lines. Our results support STEAP as a cell-surface tumor-antigen target for prostate cancer therapy and diagnostic imaging.


Subject(s)
Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Prostatic Neoplasms/chemistry , Amino Acid Sequence , Antigens, Neoplasm/genetics , Antigens, Surface/analysis , Antigens, Surface/genetics , Chromosomes/chemistry , Cloning, Molecular , Humans , Immunohistochemistry , Male , Molecular Sequence Data , Prostatic Neoplasms/metabolism , RNA, Messenger/analysis , Telomere , Tumor Cells, Cultured
3.
Leukemia ; 12(12): 1858-65, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9844916

ABSTRACT

We used genetic strategies which have been proven valuable to decipher signaling pathways in comparatively simple organisms such as Drosophila and Caenorhabditis elegans, to dissect signaling network activated by tyrosine kinases in mammals. The strategy was developed further towards a generally applicable expression cloning system to identify signal transducers in tyrosine kinase pathways. This system is based on the ability of downstream acting genes to rescue the transformation phenotype of partial loss-of-function mutants of BCR-ABL which still retain tyrosine kinase activity. Using this strategy we have previously shown that overexpression of c-Myc and Cyclin D1 can rescue a signaling defective SH2 mutant of BCR-ABL for transformation. In an unbiased approach to identify new compensating genes, a cDNA library was introduced by retroviral infection into fibroblasts which express the BCR-ABL SH2 mutant. CDNA clones, capable of rescuing the SH2 mutant for transformation should result in colony formation in soft agar. A PCR approach was used to recover these compensating genes from the genomic DNA of the transformed fibroblasts. Sequencing analysis of the initial cDNAs identified three known genes, the adapter molecule Shc, the kinases SPRK and p38 MAPK. These genes have been found to interact functionally with BCR-ABL for fibroblast and hematopoietic cell transformation. Currently, we are constructing and screening new libraries to identify novel genes which complement the BCR-ABL SH2 mutant. Our results demonstrate that this cloning approach is an effective means of identifying and characterizing signaling molecules that function in specific signaling pathways. This in turn may identify specific targets for mechanism-based therapeutic intervention to block altered signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Fusion Proteins, bcr-abl/genetics , Protein-Tyrosine Kinases/physiology , Signal Transduction/genetics , Animals , Biological Assay , Caenorhabditis elegans , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Transformation, Viral/genetics , Drosophila melanogaster , Fusion Proteins, bcr-abl/physiology , Mice , Oncogenes/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Proteins/genetics , Proteins/physiology , Retroviridae/genetics , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
4.
Hum Gene Ther ; 9(14): 2049-62, 1998 Sep 20.
Article in English | MEDLINE | ID: mdl-9759932

ABSTRACT

We examined the potential of generating an immune response against Philadelphia chromosome-positive acute lymphoblastic leukemia. The immunostimulatory molecules chosen for this study were the cytokines IL-2 and GM-CSF and the costimulatory ligand CD80 (B7.1). We used a murine model based on a BALB/c pre-B cell line, BM185wt, in which leukemia is induced by the p185 BCR-ABL oncogenic product, which reproduces Philadelphia chromosome-positive ALL. BM185wt cells were transduced with retroviral vectors and the transduced clones expressing mIL-2, mGM-CSF, or mCD80 were used for challenge. Expression of the immunomodulators by BM185 cells was correlated with delay in leukemia development in immunocompetent mice, but not in immunodeficient mice, indicating an immune response against the modified leukemia cells. Expression of CD80 caused leukemia rejection in 50% of the cohort, which was associated with the CD4+ and CD8+ T cell-dependent development of anti-leukemia cytotoxic T lymphocytes. Furthermore, mice surviving the BM185/CD80 challenge or preimmunized with irradiated BM185/CD80 cells developed an immune response against subsequent challenge with the parental leukemia. These studies provide evidence that immunotherapeutic approaches can be developed for the treatment of ALL.


Subject(s)
B7-1 Antigen/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-2/genetics , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Vaccines/immunology , Adjuvants, Immunologic/genetics , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , Cell Division/immunology , Cytotoxicity Tests, Immunologic , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Mice, SCID/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transduction, Genetic/genetics , Tumor Cells, Cultured
5.
Oncogene ; 16(11): 1383-90, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9525737

ABSTRACT

Bcr - Abl is the molecule responsible for both the transformation phenotype and the resistance to chemotherapeutic drugs found in chronic myelogenous leukemia (CML) cells. Wild-type HL-60, a transformed pro-myelocytic cell line, is very susceptible to apoptosis-inducing agents. We show here that expression of Bcr - Abl in HL-60 cells rendered them extremely resistant to apoptosis induced by a wide variety of agents. The anti-apoptotic effect of Bcr - Abl was found to be independent of the phase of the cell cycle. Treatment with antisense oligonucleotides directed to bcr decreased the expression of the ectopic bcr - abl and restored susceptibility to apoptosis. Double mutations affecting the autophosphorylation site and the phosphotyrosine-binding motif (FLVRES) have been previously shown to impair the transforming activity of Bcr - Abl in fibroblasts and hematopoietic cells, however HL-60 cells expressing this double mutant molecule exhibited the same level of resistance to apoptosis as those expressing the wild-type Bcr - Abl. Interestingly, wild type and mutant Bcr - Abl induced in HL-60 cells a dramatic down regulation of Bcl-2 and increased the levels of Bcl-xL. The level of Bax did not change in response to the presence of Bcr - Abl. Antisense oligonucleotides targeted to bcl-x downregulated the expression of Bcl-x, and increased the susceptibility of HL-60. Bcr - Abl cells to staurosporine. Importantly, HL-60 cells overexpressing Bcl-xL showed higher expression of Bcl-xL but lower resistance to apoptosis when compared to HL-60. Bcr - Abl cells. The results described here show that Bcr - Abl is a powerful mammalian anti-apoptotic molecule and can act independently of Bcl-2. Bcl-xL, however, seems to participate in part in Bcr - Abl-mediated resistance to apoptosis in HL-60 cells.


Subject(s)
Apoptosis , Fusion Proteins, bcr-abl/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation , Drug Resistance, Multiple/genetics , Fusion Proteins, bcr-abl/genetics , HL-60 Cells , Humans , Oligonucleotides, Antisense/pharmacology , bcl-X Protein
6.
Immunity ; 6(6): 773-82, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9208849

ABSTRACT

RIN1 was originally identified by its ability to physically bind to and interfere with activated Ras in yeast. Paradoxically, RIN1 potentiates the oncogenic activity of the BCR-ABL tyrosine kinase in hematopoietic cells and dramatically accelerates BCR-ABL-induced leukemias in mice. RIN1 rescues BCR-ABL mutants for transformation in a manner distinguishable from the cell cycle regulators c-Myc and cyclin D1 and the Ras connector Shc. These biological effects require tyrosine phosphorylation of RIN1 and binding of RIN1 to the Abl-SH2 and SH3 domains. RIN1 is tyrosine phosphorylated and is associated with BCR-ABL in human and murine leukemic cells. RIN1 exemplifies a new class of effector molecules dependent on the concerted action of the SH3, SH2, and catalytic domains of a cytoplasmic tyrosine kinase.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/physiology , Fusion Proteins, bcr-abl/metabolism , Intracellular Signaling Peptides and Proteins , rab GTP-Binding Proteins , Animals , Cell Division , Cell Transformation, Neoplastic/genetics , GRB2 Adaptor Protein , Hematopoiesis , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Macromolecular Substances , Mice , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Proteins/metabolism , Signal Transduction , Structure-Activity Relationship , Survival Analysis , Tumor Cells, Cultured , src Homology Domains
7.
Proc Natl Acad Sci U S A ; 94(10): 4954-9, 1997 May 13.
Article in English | MEDLINE | ID: mdl-9144171

ABSTRACT

Human RIN1 was first characterized as a RAS binding protein based on the properties of its carboxyl-terminal domain. We now show that full-length RIN1 interacts with activated RAS in mammalian cells and defines a minimum region of 434 aa required for efficient RAS binding. RIN1 interacts with the "effector domain" of RAS and employs some RAS determinants that are common to, and others that are distinct from, those required for the binding of RAF1, a known RAS effector. The same domain of RIN1 that binds RAS also interacts with 14-3-3 proteins, extending the similarity between RIN1 and other RAS effectors. When expressed in mammalian cells, the RAS binding domain of RIN1 can act as a dominant negative signal transduction blocker. The amino-terminal domain of RIN1 contains a proline-rich sequence similar to consensus Src homology 3 (SH3) binding regions. This RIN1 sequence shows preferential binding to the ABL-SH3 domain in vitro. Moreover, the amino-terminal domain of RIN1 directly associates with, and is tyrosine phosphorylated by, c-ABL. In addition, RIN1 encodes a functional SH2 domain that has the potential to activate downstream signals. These data suggest that RIN1 is able to mediate multiple signals. A differential pattern of expression and alternate splicing indicate several levels of RIN1 regulation.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , rab GTP-Binding Proteins , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Genomic Library , Humans , Mice , Molecular Sequence Data , Open Reading Frames , Protein Binding , Rats , Sequence Deletion , Sequence Homology, Amino Acid , Signal Transduction , ras Proteins/metabolism
8.
Mol Cell Biol ; 16(8): 4107-16, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8754809

ABSTRACT

TEL is a member of the Ets family of transcription factors which are frequently rearranged in human leukemia. The mechanism of TEL-mediated transformation, however, is unknown. We report the cloning and characterization of a chromosomal translocation associated with acute myeloid leukemia which fuses TEL to the ABL tyrosine kinase. The TEL-ABL fusion confers growth factor-independent growth to the marine hematopoietic cell line Ba/F3 and transforms Rat-1 fibroblasts and primary murine bone marrow cells. TEL-ABL is constitutively tyrosine phosphorylated and localizes to the cytoskeleton. A TEL-ABL mutant containing an ABL kinase-inactivating mutation is not constitutively phosphorylated and is nontransforming but retains cytoskeletal localization. However, constitutive phosphorylation, cytoskeletal localization, and transformation are all dependent upon a highly conserved region of TEL termed the helix-loop-helix (HLH) domain. TEL-ABL formed HLH-dependent homo-oligomers in vitro, a process critical for tyrosine kinase activation. These experiments suggest that oligomerization of TEL-ABL mediated by the TEL HLH domain is required for tyrosine kinase activation, cytoskeletal localization, and transformation. These data also suggest that oligomerization of Ets proteins through the highly conserved HLH domain may represent a previously unrecognized phenomenon.


Subject(s)
DNA-Binding Proteins/metabolism , Leukemia, Myeloid/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Repressor Proteins , Transcription Factors/metabolism , Aged , Aged, 80 and over , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 12 , Cytoskeletal Proteins/metabolism , DNA Primers/chemistry , Gene Expression Regulation, Neoplastic , Helix-Loop-Helix Motifs , Humans , Male , Molecular Sequence Data , Phosphoproteins/metabolism , Protein Binding , Proto-Oncogene Proteins c-ets , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Recombinant Fusion Proteins , Sequence Alignment , Sequence Homology, Amino Acid , Translocation, Genetic , ETS Translocation Variant 6 Protein
9.
Mol Cell Biol ; 16(7): 3465-71, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668162

ABSTRACT

Loss of function of Bruton's tyrosine kinase (Btk) results in X-linked immunodeficiencies characterized by a broad spectrum of signaling defects, including those dependent on Src family kinase-linked cell surface receptors. A gain-of-function mutant, Btk*, induces the growth of fibroblasts in soft agar and relieves the interleukin-5 dependence of a pre-B-cell line. To genetically define Btk signaling pathways, we used a strategy to either activate or inactivate Src family kinases in fibroblasts that express Btk*. The transformation potential of Btk* was dramatically increased by coexpression with a partly activated c-Src mutant (E-378 --> G). This synergy was further potentiated by deletion of the Btk Src homology 3 domain. Downregulation of Src family kinases by the C-terminal Src kinase (Csk) suppressed Btk* activation and biological potency. In contrast, kinase-inactive Csk (K-222 --> R), which functioned as a dominant negative molecule, synergized with Btk* in biological transformation. Activation of Btk* correlated with increased phosphotyrosine on transphosphorylation and autophosphorylation sites. These findings suggest that the Src and Btk kinase families form specific signaling units in tissues in which both are expressed.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Blotting, Western , CSK Tyrosine-Protein Kinase , Cell Line , Humans , Peptide Mapping , Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Phosphorylation , Point Mutation , Protein-Tyrosine Kinases/isolation & purification , Rats , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection , src-Family Kinases
10.
J Biol Chem ; 271(26): 15353-7, 1996 Jun 28.
Article in English | MEDLINE | ID: mdl-8663064

ABSTRACT

Bcr-Abl oncoproteins are responsible for the pathogenesis of human leukemias with a reciprocal chromosome translocation t(9;22). The amino-terminal Bcr sequence has a potential to form a homotetramer (tetramer domain), and destructions of the tetramer domain cause a complete loss of biological activities in Bcr-Abl. Here we show that Bcr-Abl in which the tetramer domain is replaced with glutathione S-transferase (GST) with a dimerizing ability (GST/Bcr-Abl-(Delta1-160)) can no longer induce an interleukin-3 (IL-3) independence in Ba/F3 cells or transform mouse bone marrow cells but still retains by 30-40% the ability to transform Rat1 cells. Compared with the wild type Bcr-Abl, autophosphorylation of GST/Bcr-Abl-(Delta1-160) in vivo was reduced by more than 50%. The Grb-2 binding to GST/Bcr-Abl-(Delta1-160) was 50% reduced in Rat1 cells and undetectable in Ba/F3 cells. In Rat1 cells expressing GST/Bcr-Abl-(Delta1-160), phosphotyrosine contents of p62 and Shc were 70% decreased.


Subject(s)
Adaptor Proteins, Signal Transducing , Fusion Proteins, bcr-abl/metabolism , Glutathione Transferase/metabolism , Animals , Bone Marrow Cells , Cell Transformation, Neoplastic , Cells, Cultured , GRB2 Adaptor Protein , Macromolecular Substances , Mice , Phosphotyrosine/metabolism , Protein Binding , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Rats , Recombinant Fusion Proteins , Structure-Activity Relationship
11.
Immunity ; 4(5): 515-25, 1996 May.
Article in English | MEDLINE | ID: mdl-8630736

ABSTRACT

Bruton's tyrosine kinase (Btk) plays a crucial role in B cell development. Overexpression of Btk with a Src family kinase increases tyrosine phosphorylation and catalytic activity of Btk. This occurs by transphosphorylation at Y551 in the Btk catalytic domain and the enhancement of Btk autophosphorylation at a second site. A gain-of-function mutant called Btk* containing E41 to K change within the pleckstrin homology domain induces fibroblast transformation. Btk* enhances the transphosphorylation of Y551 by endogenous Src family tyrosine kinases and autophosphorylation at the second site. We mapped the major Btk autophosphorylation site to Y223 within the SH3 domain. Mutation of Y223 to F blocks Btk autophosphorylation and dramatically potentiates the transforming activity of Btk* in fibroblasts. The location of Y223 in a potential ligand-binding pocket suggests that autophosphorylation regulates SH3-mediated signaling by Btk.


Subject(s)
Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism , src Homology Domains/genetics , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Sequence , Animals , Base Sequence , Humans , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein-Tyrosine Kinases/physiology , Transformation, Genetic , Tyrosine/genetics
12.
Proc Natl Acad Sci U S A ; 92(21): 9540-4, 1995 Oct 10.
Article in English | MEDLINE | ID: mdl-7568169

ABSTRACT

Oncogenic signals induce cellular proliferation by deregulating the cell division cycle. Cyclin D1, a regulator of G1-phase progression, acts synergistically with ABL oncogenes in transforming fibroblasts and hematopoietic cells in culture. Synergy with v-Abl depended on a motif in cyclin D1 that mediates its binding to the retinoblastoma protein, suggesting that ABL oncogenes in part mediate their mitogenic effects via a retinoblastoma protein-dependent pathway. Overexpression of cyclin D1, but not cyclin E, rescued a signaling-defective src-homology 2 (SH2) domain mutant of BCR-ABL for transformation of cells in culture and malignant tumor formation in vivo. These results demonstrate that cyclin D1 can provide essential signals for malignant transformation in concert with an activated tyrosine kinase.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclins/metabolism , Genes, abl , Oncogene Proteins/metabolism , Signal Transduction/genetics , Animals , Bone Marrow Cells , Cells, Cultured , Cyclin D1 , Cyclins/genetics , Fusion Proteins, bcr-abl/genetics , Leukemia, Experimental/genetics , Male , Mice , Mice, Inbred BALB C , Oncogene Proteins/genetics , Protein Binding , Recombinant Proteins , Retinoblastoma Protein/metabolism , src Homology Domains
13.
Cell ; 82(6): 981-8, 1995 Sep 22.
Article in English | MEDLINE | ID: mdl-7553858

ABSTRACT

Biological function of the BCR-ABL oncogene is dependent on its activated tyrosine kinase. Mutations that inactivate the SRC homology 2 (SH2) domain, the GRB2-binding site in BCR, or the major autophosphorylation site of the kinase domain selectively disrupt downstream signaling but not tyrosine kinase activity. Despite a loss of fibroblast transformation activity, all three mutants retain the ability to render hematopoietic cell lines growth factor independent and transform primary bone marrow cells in vitro. In vivo tests of malignant potential reveal a most critical role for signals dependent on the BCR-ABL SH2 domain. The efficiency of both fibroblast and hematopoietic transformation by BCR-ABL is strongly affected by increased dosage of the SHC adapter protein, which can connect tyrosine kinase signals to RAS. The BCR-ABL oncogene activates multiple alternative pathways to RAS for hematopoietic transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Fusion Proteins, bcr-abl/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/physiology , Animals , Bone Marrow Cells , Cell Line, Transformed/physiology , Fibroblasts/physiology , Granulocytes/cytology , Hematopoietic Cell Growth Factors/physiology , Lymphocytes/cytology , Mice , Mice, Inbred BALB C , Mice, SCID , Mutation/physiology , Oncogenes/physiology , Protein-Tyrosine Kinases/physiology , ras Proteins/physiology , src Homology Domains/physiology
14.
Mol Cell Biol ; 15(2): 835-42, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7529874

ABSTRACT

The human bcr gene encodes a protein with serine/threonine kinase activity, CDC24/dbl homology, a GAP domain, and an SH2-binding region. However, the precise physiological functions of BCR are unknown. Coexpression of BCR with the cytoplasmic protein-tyrosine kinase encoded by the c-fes proto-oncogene in Sf-9 cells resulted in stable BCR-FES protein complex formation and tyrosine phosphorylation of BCR. Association involves the SH2 domain of FES and a novel binding domain localized to the first 347 amino acids of the FES N-terminal region. Deletion of the homologous N-terminal BCR-binding domain from v-fps, a fes-related transforming oncogene, abolished transforming activity and tyrosine phosphorylation of BCR in vivo. Tyrosine phosphorylation of BCR in v-fps-transformed cells induced its association with GRB-2/SOS, the RAS guanine nucleotide exchange factor complex. These data provide evidence that BCR couples the cytoplasmic protein-tyrosine kinase and RAS signaling pathways.


Subject(s)
Adaptor Proteins, Signal Transducing , ErbB Receptors/metabolism , Fusion Proteins, gag-onc/metabolism , Oncogene Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins , Animals , Cell Line , Cell Transformation, Neoplastic , GRB2 Adaptor Protein , Guanine Nucleotide Exchange Factors , Humans , Oncogene Proteins/biosynthesis , Oncogene Proteins/isolation & purification , Oncogenes , Phosphorylation , Phosphotyrosine , Protein Binding , Protein-Tyrosine Kinases/biosynthesis , Proteins/isolation & purification , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcr , Recombinant Fusion Proteins , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Restriction Mapping , Sequence Deletion , Spodoptera , Transfection , Tyrosine/analogs & derivatives , Tyrosine/metabolism , ras Guanine Nucleotide Exchange Factors
15.
Methods Enzymol ; 256: 125-9, 1995.
Article in English | MEDLINE | ID: mdl-7476425

ABSTRACT

BCR is an interesting signaling protein, whose cellular function is currently unknown. Its biochemical properties include serine kinase activity, SH2-binding activity, and a GTPase-activating activity. The SH2-binding activity is particularly interesting because it may link BCR to signaling pathways involving SH2-containing molecules. Since tyrosine phosphorylation of BCR has been detected in CML-derived cell lines and since tyrosine-phosphorylated BCR shows increased affinity toward certain SH2 domains, it seems particularly important to further characterize this activity. This chapter described a simple purification scheme for partial purification of BCR, which can be used to assess in vitro kinase and SH2-binding activities.


Subject(s)
GTP Phosphohydrolases/analysis , GTP Phosphohydrolases/metabolism , Oncogene Proteins/analysis , Oncogene Proteins/metabolism , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cell Line , Chromatography, Affinity/methods , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 9 , Electrophoresis, Polyacrylamide Gel/methods , Exons , GTP Phosphohydrolases/isolation & purification , Humans , Kinetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Methionine/metabolism , Oncogene Proteins/genetics , Phosphorus Radioisotopes , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcr , Radioisotope Dilution Technique , Recombinant Proteins/analysis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spodoptera , Sulfur Radioisotopes , Transfection/methods , Translocation, Genetic
16.
Proc Natl Acad Sci U S A ; 91(26): 12448-52, 1994 Dec 20.
Article in English | MEDLINE | ID: mdl-7809057

ABSTRACT

We report the characterization of a member of the ras gene family that is overexpressed in cells transformed by abl tyrosine kinase oncogenes. The gene, named kir (for kinase-inducible ras-like), is induced at the transcriptional level. kir mRNA has a rapid turnover and encodes a protein of 33 kDa with guanine nucleotide-binding activity but undetectable intrinsic GTPase activity. kir was cloned by differential screening of genes present in fully malignant versus growth factor-independent cell lines expressing wild-type or mutant forms of BCR/ABL. BCR/ABL and v-Abl induce transcription of the kir gene via specific signaling pathway(s), but kir overexpression alone is not sufficient to mediate transformation.


Subject(s)
Cell Transformation, Neoplastic , Fusion Proteins, bcr-abl/metabolism , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Immediate-Early Proteins/genetics , Monomeric GTP-Binding Proteins , Oncogene Proteins v-abl/metabolism , Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Cloning, Molecular , DNA, Complementary/genetics , GTP-Binding Proteins/metabolism , Immediate-Early Proteins/metabolism , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Transcription, Genetic
17.
J Biol Chem ; 269(44): 27240-5, 1994 Nov 04.
Article in English | MEDLINE | ID: mdl-7525550

ABSTRACT

Myelin-associated glycoprotein (MAG) is a myelin-specific cell adhesion molecule of the immunoglobulin supergene family and is tyrosine-phosphorylated in the developing brain. To define the role of MAG in signal transduction, the tyrosine phosphorylation sites were analyzed. The major tyrosine phosphorylation residue was identified as Tyr-620, which was found to interact specifically with the SH2 domains of phospholipase C (PLC gamma). This domain may represent a novel protein binding motif that can be regulated by tyrosine phosphorylation. MAG also specifically bound the Fyn tyrosine kinase, suggesting that MAG serves as a docking protein that allows the interaction between different signaling molecules.


Subject(s)
Myelin Proteins/metabolism , Nerve Fibers, Myelinated/metabolism , Oligodendroglia/metabolism , Tyrosine/analogs & derivatives , Animals , Base Sequence , DNA Primers/chemistry , Mice , Molecular Sequence Data , Myelin-Associated Glycoprotein , Peptide Mapping , Phosphorylation , Phosphotyrosine , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn , Rats , Recombinant Proteins , Signal Transduction , Type C Phospholipases/metabolism , Tyrosine/metabolism
19.
Science ; 264(5157): 424-6, 1994 Apr 15.
Article in English | MEDLINE | ID: mdl-8153630

ABSTRACT

A complementation strategy was developed to define the signaling pathways activated by the Bcr-Abl tyrosine kinase. Transformation inactive point mutants of Bcr-Abl were tested for complementation with c-Myc. Single point mutations in the Src-homology 2 (SH2) domain, the major tyrosine autophosphorylation site of the kinase domain, and the Grb-2 binding site in the Bcr region impaired the transformation of fibroblasts by Bcr-Abl. Hyperexpression of c-Myc efficiently restored transformation activity only to the Bcr-Abl SH2 mutant. These data support a model in which Bcr-Abl activates at least two independent pathways for transformation. This strategy may be useful for discerning signaling pathways activated by other oncogenes.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Transformation, Neoplastic , Fusion Proteins, bcr-abl/genetics , Genes, abl , Genes, myc , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Fusion Proteins, bcr-abl/physiology , GRB2 Adaptor Protein , Gene Expression , Genetic Complementation Test , Molecular Sequence Data , Phosphorylation , Point Mutation , Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Rats , Retroviridae/physiology , Signal Transduction , Transfection , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...