Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Virus Genes ; 52(3): 365-71, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27108997

ABSTRACT

Transduction of oncogenes by ALVs and generation of acute transforming viruses is common in natural viral infections. In order to understand the molecular basis for the rapid oncogenicity of Fu-J, an acutely transforming avian leukosis virus isolated from fibrosarcomas in crossbreed broilers infected with subgroup J avian leukosis virus (ALV-J) in China, complete genomic structure of Fu-J virus was determined by PCR amplification and compared with those of Fu-J1, Fu-J2, Fu-J3, Fu-J4, and Fu-J5 reported previously. The results showed that the genome of Fu-J was defective, with parts of gag gene replaced by the complete v-fps oncogene and encoded a 137 kDa Gag-fps fusion protein. Sequence analysis revealed that Fu-J and Fu-J1 to Fu-J5 were related quasi-species variants carrying different lengths of v-fps oncogenes generated from recombination between helper virus and c-fps gene. Comparison of virus carrying v-fps oncogene also gave us a glimpse of the molecular characterization and evolution process of the acutely transforming ALV.


Subject(s)
Avian Leukosis Virus/genetics , Avian Leukosis/virology , Fusion Proteins, gag-onc/genetics , Oncogene Proteins/genetics , Oncogenic Viruses/genetics , Poultry Diseases/virology , Protein-Tyrosine Kinases/genetics , Animals , Avian Leukosis Virus/isolation & purification , Avian Leukosis Virus/pathogenicity , Avian Sarcoma Viruses/genetics , Base Sequence , Chick Embryo , Chickens/virology , DNA, Viral , Fibrosarcoma/virology , Gene Products, gag/genetics , Genes, Viral , Helper Viruses/genetics , Retroviridae/genetics , Virus Replication
2.
Blood Cells Mol Dis ; 32(2): 302-8, 2004.
Article in English | MEDLINE | ID: mdl-15003822

ABSTRACT

The c-fps/fes protooncogene encodes a 92-kDa protein tyrosine kinase that is involved in myeloid cell development and immune responses of granulocytes and macrophages. To help define its biological role and mechanism of action, we have developed a gain of function allele of Fes that has potent biological activity in myeloid cells. Introduction of constitutively active Fes into myeloid progenitors induced the appearance of fully differentiated macrophages or granulocytes depending on the lineage commitment of the transduced cells. We found that Fes-induced macrophage differentiation correlated with activation of the ets family transcription factor PU.1, which is essential for macrophage development. On the other hand, granulocyte differentiation by Fes was mediated through activation of CCAAT/enhancer-binding protein alpha (C/EBP-alpha) and STAT3, two transcription factors that are critical for granulocytic differentiation. We postulate that Fes transduces inductive signals for terminal macrophage and granulocyte differentiation, and that this biological activity is mediated through the activation of lineage-specific transcription factors.


Subject(s)
Fusion Proteins, gag-onc/physiology , Myeloid Cells/metabolism , Protein-Tyrosine Kinases/physiology , Signal Transduction , Transcriptional Activation , Enzyme Activation , Fusion Proteins, gag-onc/genetics , Gene Expression Regulation , Granulocytes/cytology , Humans , Molecular Mimicry , Monocytes/cytology , Myeloid Cells/enzymology , Myelopoiesis , Protein-Tyrosine Kinases/genetics , Transcription Factors/metabolism , Transfection , U937 Cells
3.
Blood ; 103(3): 912-20, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14525765

ABSTRACT

Relatively little is known about the modulators of the vascular endothelial growth factor A (VEGF-A)/Flk1 signaling cascade. To functionally characterize this pathway, VEGF-A stimulation of endothelial cells was performed. VEGF-A-mediated Flk1 activation resulted in increased translocation of the endogenous Fps/Fes cytoplasmic tyrosine kinase to the plasma membrane and increased tyrosine phosphorylation, suggesting a role for Fps/Fes in VEGF-A/Flk1 signaling events. Addition of a myristoylation consensus sequence to Fps/Fes resulted in VEGF-A-independent membrane localization of Fps/Fes in endothelial cells. Expression of the activated Fps/Fes protein in Flk1-deficient embryonic stem (ES) cells rescued their contribution to the developing vascular endothelium in vivo by using ES cell-derived chimeras. Activated Fps/Fes contributed to this rescue event by restoring the migratory potential to Flk1 null progenitors, which is required for movement of hemangioblasts from the primitive streak region into the yolk sac proper. Activated Fps/Fes in the presence of Flk1 increased the number of hemangioblast colonies in vitro and increased the number of mesodermal progenitors in vivo. These results suggest that Fps/Fes may act synergistically with Flk1 to modulate hemangioblast differentiation into the endothelium. We have also demonstrated that activated Fps/Fes causes hemangioma formation in vivo, independently of Flk1, as a result of increasing vascular progenitor density.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fusion Proteins, gag-onc/metabolism , Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor Receptor-2/deficiency , Animals , Cell Differentiation , Cell Movement , Enzyme Activation , Fusion Proteins, gag-onc/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
Exp Hematol ; 31(12): 1259-67, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14662333

ABSTRACT

OBJECTIVE: The fps/fes proto-oncogene is abundantly expressed in myeloid cells, and the Fps/Fes cytoplasmic protein-tyrosine kinase is implicated in signaling downstream from hematopoietic cytokines, including interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), and erythropoietin (EPO). Studies using leukemic cell lines have previously suggested that Fps/Fes contributes to granulomonocytic differentiation, and that it might play a more selective role in promoting survival and differentiation along the monocytic pathway. In this study we have used a genetic approach to explore the role of Fps/Fes in hematopoiesis. METHODS: We used transgenic mice that tissue-specifically express a mutant human fps/fes transgene (fps(MF)) that was engineered to encode Fps/Fes kinase that is activated through N-terminal myristoylation (MFps). Hematopoietic function was assessed using lineage analysis, hematopoietic progenitor cell colony-forming assays, and biochemical approaches. RESULTS: fps(MF) transgenic mice displayed a skewed hematopoietic output reflected by increased numbers of circulating granulocytic and monocytic cells and a corresponding decrease in lymphoid cells. Bone marrow colony assays of progenitor cells revealed a significant increase in the number of both granulomonocytic and multi-lineage progenitors. A molecular analysis of signaling in mature monocytic cells showed that MFps promoted GM-CSF-induced STAT3, STAT5, and ERK1/2 activation. CONCLUSIONS: These observations support a role for Fps/Fes in signaling pathways that contribute to lineage determination at the level of multi-lineage hematopoietic progenitors as well as the more committed granulomonocytic progenitors.


Subject(s)
Fusion Proteins, gag-onc/physiology , Hematopoiesis , Protein-Tyrosine Kinases/physiology , Animals , Blood Cells/cytology , Cell Count , Cell Lineage , Colony-Forming Units Assay , Esterification , Fusion Proteins, gag-onc/genetics , Humans , Mice , Mice, Transgenic , Myelopoiesis , Protein Engineering , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Transgenes
5.
Exp Hematol ; 31(8): 673-81, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12901971

ABSTRACT

OBJECTIVE: The highly related protein-tyrosine kinases Fps (also called Fes) and Fer are sole members of a subfamily of kinases. In this study, knock-in mice harboring kinase-inactivating mutations in both fps and fer alleles were used to assess functional redundancy between Fps and Fer kinases in regulating hematopoiesis. METHODS: Mice harboring kinase-inactivating mutations in fps and fer alleles were generated previously. Compound homozygous mice were bred that lack both Fps and Fer kinase activities and progeny were analyzed for potential defects in viability and fertility. Potential differences in hematopoiesis were analyzed by lineage analysis of bone marrow cells, peripheral blood counts, and hematopoietic progenitor cell colony-forming assays. RESULTS: Mice devoid of both Fps and Fer kinase activities were viable and displayed reduced fertility. Circulating levels of neutrophils, erythrocytes, and platelets were elevated in compound mutant mice compared to wild-type controls, suggesting that hematopoiesis is deregulated in the absence of Fps and Fer kinases. Compound mutant mice also showed reduced overall bone marrow cellularity, and lineage analysis revealed elevated CD11b(hi)Ly-6G(lo) myeloid cells, which may reflect increased granulocyte progenitors. Although no differences in the overall number of granulocyte/monocyte colony-forming progenitors were observed, qualitative differences in myeloid colonies from compound mutant mice suggested a role for Fps and Fer kinases in regulating cell-cell adhesion or a skewing in cellularity of colonies. CONCLUSIONS: Mice lacking both Fps and Fer kinase activities develop normally, show reduced fertility, and display defects in hematopoiesis, thus providing evidence for functional redundancy between Fps and Fer kinases in regulating hematopoiesis.


Subject(s)
Fusion Proteins, gag-onc/physiology , Hematopoiesis/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Alleles , Animals , Blood Cell Count , Bone Marrow/pathology , Colony-Forming Units Assay , Female , Fusion Proteins, gag-onc/deficiency , Fusion Proteins, gag-onc/genetics , Male , Mice , Mice, Knockout , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics
7.
Nat Rev Mol Cell Biol ; 3(4): 278-89, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11994747

ABSTRACT

Fps/Fes and Fer are the only known members of a distinct subfamily of the non-receptor protein-tyrosine kinase family. Recent studies indicate that these kinases have roles in regulating cytoskeletal rearrangements and inside out signalling that accompany receptor ligand, cell matrix and cell cell interactions. Genetic analysis using transgenic mouse models also implicates these kinases in the regulation of inflammation and innate immunity.


Subject(s)
Fusion Proteins, gag-onc/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Biological Evolution , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 5/genetics , Fusion Proteins, gag-onc/chemistry , Fusion Proteins, gag-onc/genetics , Humans , Inflammation/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Models, Molecular , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Receptor Cross-Talk , Receptors, Platelet-Derived Growth Factor/physiology , Signal Transduction
8.
Mol Cell Biol ; 22(8): 2472-86, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11909942

ABSTRACT

The fps/fes proto-oncogene encodes a cytoplasmic protein tyrosine kinase implicated in growth factor and cytokine receptor signaling and thought to be essential for the survival and terminal differentiation of myeloid progenitors. Fps/Fes-null mice were healthy and fertile, displayed slightly reduced numbers of bone marrow myeloid progenitors and circulating mature myeloid cells, and were more sensitive to lipopolysaccharide (LPS). These phenotypes were rescued using a fps/fes transgene. This confirmed that Fps/Fes is involved in, but not required for, myelopoiesis and that it plays a role in regulating the innate immune response. Bone marrow-derived Fps/Fes-null macrophages showed no defects in granulocyte-macrophage colony-stimulating factor-, interleukin 6 (IL-6)-, or IL-3-induced activation of signal transducer and activator of transcription 3 (Stat3) and Stat5A or LPS-induced degradation of I kappa B or activation of p38, Jnk, Erk, or Akt.


Subject(s)
Fusion Proteins, gag-onc/deficiency , Fusion Proteins, gag-onc/genetics , Hematopoiesis/genetics , Lipopolysaccharides/toxicity , Milk Proteins , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Animals , B-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Female , Gene Targeting , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Homeostasis , Humans , Interleukin-3/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Proto-Oncogene Mas , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction , T-Lymphocytes/immunology , Trans-Activators/metabolism , Tumor Suppressor Proteins
9.
Mol Cell Biol ; 22(6): 1903-18, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11865067

ABSTRACT

The c-fps/fes proto-oncogene encodes a 92-kDa protein tyrosine kinase that is preferentially expressed in myeloid and endothelial cells. Fes is believed to play a role in vascular development and myelopoiesis and in the inflammatory responses of granulocytes and macrophages. To help define the biological role of this kinase and identify its downstream targets, we have developed a gain-of-function allele of Fes that has potent biological activity in myeloid cell progenitors. Introduction of constitutively active Fes into bipotential U937 cells induced the appearance of fully differentiated macrophages within 6 to 12 days. The Fes-expressing differentiated cells became adherent, had distinctive macrophage morphology, and exhibited increased expression of myelomonocytic differentiation markers, including CD11b, CD11c, CD18, CD14, and the macrophage colony-stimulating factor receptor. These cells acquired phagocytic properties and exhibited NADPH oxidase and nonspecific esterase activities, confirming that they were functionally active macrophages. Concomitantly, there was downregulation of the granulocytic marker granulocyte colony-stimulating factor receptor, indicating that the biological activity of Fes was coordinated in a lineage-specific manner. A constitutively active Src did not induce macrophage morphology or upregulation of myelomonocytic markers in U937 cells, suggesting that the biological activity we observed was not a general consequence of expression of an activated nonreceptor tyrosine kinase. Analysis of possible downstream targets of Fes revealed that this kinase activated the ets family transcription factor PU.1, which is essential for macrophage development. Our results strongly implicate Fes as a key regulator of terminal macrophage differentiation and identify PU.1 as a transcription factor that may mediate some of its biological activities in myeloid cells.


Subject(s)
Fusion Proteins, gag-onc/metabolism , Macrophages , Myeloid Progenitor Cells/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Trans-Activators/metabolism , Alleles , Antigens, CD/biosynthesis , Antigens, Differentiation/biosynthesis , Cell Differentiation/drug effects , Cytoplasm/metabolism , Enzyme Activation/physiology , Fusion Proteins, gag-onc/genetics , Fusion Proteins, gag-onc/pharmacology , Humans , Macrophages/cytology , Macrophages/drug effects , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Phenotype , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/pharmacology , Proto-Oncogene Mas , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Signal Transduction/physiology , Thromboplastin/metabolism , Transfection , U937 Cells
10.
Biochemistry ; 40(34): 10078-86, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11513586

ABSTRACT

Autophosphorylation of Tyr-1073 in the activation loop of the oncoprotein v-Fps enhances the phosphoryl transfer reaction without influencing substrate, ATP, or metal ion binding affinities [Saylor, P., et al. (1998) Biochemistry 37, 17875-17881]. A structural model of v-Fps, generated from the insulin receptor, indicates that pTyr-1073 chelates two arginines. Mutation of these residues to alanine (R1042A and R1066A) results in weakly phosphorylated enzymes, indicating that one electropositive center is insufficient for attaining maximum loop phosphorylation and concomitant high catalytic activity. While the turnover rate for R1066A is similar to that for a mutant lacking a phosphorylatable residue in the activation loop, the rate for R1042A is 50-fold slower. While solvent perturbation studies suggest that the former is due to a slow phosphoryl transfer step, the latter effect results from a slow conformational change in the mutant, potentially linked to motions in the catalytic loop. Binding of a stoichiometric quantity of Mg(2+) is essential for ATP binding and catalysis, while binding of an additional Mg(2+) ion activates further the wild-type enzyme. The affinity of the R1066A enzyme for the second Mg(2+) ion is 23-fold higher than that of the phosphorylated or unphosphorylated form of wild-type v-Fps, with substrate binding unaffected. Conversely, the affinity of R1066A for a substrate mimic lacking a phosphorylation site is 12-fold higher than that for the phosphorylated or unphosphorylated form of wild-type v-Fps, with binding of the second Mg(2+) ion unaffected. A comparison of these enzyme-independent parameters indicates that Arg-1042 and Arg-1066 induce strain in the active site in the repressed form of the enzyme. While this strain is not relieved in the phosphorylated form, the improvements in catalysis in activated v-Fps compensate for reduced metal and substrate binding affinities.


Subject(s)
Fusion Proteins, gag-onc/chemistry , Fusion Proteins, gag-onc/metabolism , Phosphotyrosine , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Amino Acid Substitution , Arginine , Cloning, Molecular , Computer Simulation , Enzyme Activation , Escherichia coli , Fusion Proteins, gag-onc/genetics , Kinetics , Magnesium/pharmacology , Models, Molecular , Mutagenesis, Site-Directed , Phosphorylation , Polymerase Chain Reaction , Protein Structure, Secondary , Static Electricity , Viscosity
11.
Cell Growth Differ ; 11(11): 581-92, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11095247

ABSTRACT

The c-fes proto-oncogene encodes a Mr 93,000 protein-tyrosine kinase (Fes) that is strongly expressed in myeloid cells and has been implicated in myelomonocytic differentiation. Fes autophosphorylation and transforming activity are highly restrained after ectopic expression in fibroblasts, indicating tight negative regulation of Fes kinase activity in vivo. Here we investigated the regulatory role of the Fes Src homology 2 (SH2) domain by producing a series of chimeric constructs in which the Fes SH2 domain was replaced with those of the transforming oncogenes v-Fps and v-Src or by the NH2-terminal SH2 domain of the Ras GTPase-activating protein. Wild-type and chimeric Fes proteins readily underwent tyrosine autophosphorylation in vitro and produced identical cyanogen bromide phosphopeptide cleavage patterns, indicating that the SH2 substitutions did not influence overall kinase activity or autophosphorylation site selection. However, metabolic labeling of Rat-2 fibroblasts expressing each construct showed that only the Fes/Src SH2 chimera was active in vivo. Consistent with this result, the Fes/Src SH2 domain chimera exhibited potent transforming activity in fibroblasts and enhanced differentiation-inducing activity in K-562 myeloid leukemia cells. In addition, the Fes/Src SH2 chimera exhibited constitutive localization to focal adhesions in Rat-2 fibroblasts and induced the attachment and spreading of TF-1 myeloid cells. These data demonstrate a central role for the SH2 domain in the regulation of Fes kinase activity and biological function in vivo.


Subject(s)
Amino Acid Substitution/genetics , Cell Transformation, Neoplastic , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , src Homology Domains/genetics , Animals , Cell Adhesion , Cell Differentiation , Cell Division , Cell Line , Cytoskeletal Proteins/metabolism , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Focal Adhesions/chemistry , Focal Adhesions/metabolism , Fusion Proteins, gag-onc/chemistry , Fusion Proteins, gag-onc/genetics , Hematopoietic Stem Cells/cytology , Humans , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Oncogene Protein pp60(v-src)/chemistry , Oncogene Protein pp60(v-src)/genetics , Paxillin , Phosphoproteins/metabolism , Phosphorylation , Protein Transport , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fes , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , ras GTPase-Activating Proteins/chemistry , ras GTPase-Activating Proteins/genetics
12.
Biochemistry ; 39(43): 13276-84, 2000 Oct 31.
Article in English | MEDLINE | ID: mdl-11052681

ABSTRACT

The three glycine residues in the glycine-rich loop of the oncoprotein, v-Fps, were mutated to determine the function of these highly conserved residues in catalysis. The kinase domains of six mutants (G928A,S, G930A,S, and G933A,S) and the wild-type enzyme were expressed and purified as fusion proteins of glutathione-S-transferase in Escherichia coli, and their catalytic properties were assessed using steady-state kinetic, inhibition, viscosity and autophosphorylation studies. Although both G928A and G930A had no detectable activity toward the substrate peptide (EAEIYEAIE), the other mutants had apparent, but varying activities. G930S lowered the rate of phosphoryl transfer by 130-fold while G928S and G933S had smaller (6-9-fold) reductions in this step. These effects on catalytic function parallel the reductions in turnover and autophosphorylation but, for G933S and G933A, net product release is still rate limiting at saturating substrate and ATP concentrations. On the basis of K(I) measurements, the effects on turnover for these mutants may be due to improved ADP affinity. While ADP affinity is reduced 2- and 3-fold for G928S and G930S, the affinity of this product is increased by 22- and 7-fold for G933S and G933A. In contrast, ATP affinity is enhanced by 5-fold for G928S and G933S and is reduced by less than 2-fold for G930S. These complex, differential effects on nucleotide binding indicate that the glycines influence the relative affinities of ADP and ATP. On the basis of the results of serine replacements, Gly-928 and Gly-930 enhance ADP affinity by 9- and 2-fold compared to ATP affinity whereas Gly-933 diminishes ADP affinity by approximately 4-fold compared to ATP affinity. These findings demonstrate that the functions of the loop lie not only in modulating the rate of the phosphoryl transfer step but also in balancing the relative affinities of ATP and ADP. These effects on nucleotide specificity may be a contributing element for the stabilization of the phosphoryl transition state and may also facilitate quick release of bound products.


Subject(s)
Fusion Proteins, gag-onc/genetics , Fusion Proteins, gag-onc/metabolism , Glycine/genetics , Glycine/metabolism , Mutagenesis, Site-Directed , Adenosine Diphosphate/pharmacology , Amino Acid Motifs/genetics , Animals , Binding, Competitive , Catalysis , Cattle , Conserved Sequence/genetics , Enzyme Activation/genetics , Enzyme Inhibitors/metabolism , Fusion Proteins, gag-onc/antagonists & inhibitors , Kinetics , Models, Molecular , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Rabbits , Viscosity
14.
J Neurovirol ; 6(4): 329-40, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10951557

ABSTRACT

Our preliminary studies suggested that the novel gag-truncated mos (tmos) open reading frame (ORF) of R7, a spontaneous deletion mutant of Moloney murine sarcoma virus 124 (MoMuSV124), may be responsible for R7's unique ability to induce brain lesions in all R7-injected mice. However, when we replaced the gag-tmos ORF with either the MoMuSV124 or the homologous myeloproliferative sarcoma virus env-mos gene, we found that both recombinant viruses also induced brain lesions in all injected mice. Although these studies suggested that the critical determinants for brain lesion induction may reside in the tmos sequence common to all three viruses, they did not demonstrate if the N-terminus of Mos was dispensable for this activity. By inserting the FLAG sequence at the 3' end of the R7 gag-tmos ORF, we demonstrated that R7 does synthesize a Gag-tMos fusion protein. Using R7 gag deletion mutants with and without the FLAG sequence, we further demonstrated that (i) deletion of the entire gag sequence abolished R7's transforming activity; (ii) the ability of the virus to transform cultured NIH/3T3 cells was significantly reduced only when most of gag was deleted; (iii) the ability of the virus to induce brain lesions was inversely proportional to the extent of its gag deletions; and (iv) the insertion of FLAG at the Mos C-terminus did not reduce the in vitro transforming activity of the FLAG-tagged viruses but did reduce their ability to induce brain lesions. Thus, we have demonstrated that altering the N- or C-terminus of the R7 Gag-tMos fusion protein can affect disease manifestation.


Subject(s)
Brain/virology , Fusion Proteins, gag-onc/genetics , Moloney murine sarcoma virus/physiology , 3T3 Cells , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Brain/pathology , Cell Line , Fusion Proteins, gag-onc/biosynthesis , Gene Deletion , Genes, gag , Mice , Mice, Inbred BALB C , Moloney murine sarcoma virus/genetics , Mutagenesis, Insertional , Mutation , Open Reading Frames , RNA/analysis , RNA, Viral/analysis
15.
Blood ; 95(12): 3959-63, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10845934

ABSTRACT

Fes is a nonreceptor tyrosine kinase expressed at the highest level in macrophages. We previously showed that the overexpression of c-fes in murine macrophages of the BAC-1.2F5 cell line renders these cells independent of macrophage colony-stimulating factor (MCSF) for survival and proliferation, although no direct relationship could be established between tyrosine-phosphorylated substrates of Fes- and MCSF receptor-dependent signaling and mitogenesis. In this study, we investigated whether the mitogen-activated protein kinase (MAPK) pathway is involved in the growth factor-independent growth of v-fes-overexpressing macrophages. We found a constitutively increased phosphorylation of extracellularly regulated kinase (ERK) in v-fes-overexpressing macrophages as compared with mock-infected cells. This finding was associated with activation of mitogen/extracellular signal-regulated kinase (MEK) and with constitutive localization of ERK in the nucleus. Treatment of v-fes-overexpressing cells with the MEK-specific inhibitor PD98059 markedly reduced cell growth, hyperphosphorylation, and nuclear localization of ERK, indicating that the MAPK pathway mediates the mitogenic effect of v-fes. (Blood. 2000;95:3959-3963)


Subject(s)
Fusion Proteins, gag-onc/physiology , Macrophages/cytology , Macrophages/physiology , Mitogen-Activated Protein Kinases/metabolism , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Cell Division/physiology , Cell Line , Fusion Proteins, gag-onc/genetics , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Recombinant Fusion Proteins/metabolism , Signal Transduction
16.
Biochemistry ; 39(1): 255-62, 2000 Jan 11.
Article in English | MEDLINE | ID: mdl-10625501

ABSTRACT

Based on the X-ray structure of the insulin receptor kinase [Hubbard, S. R. (1997) EMBO J. 16, 5572-5581], Arg-1130 in the oncoprotein v-Fps, a nonreceptor tyrosine protein kinase, is predicted to interact with the P+1 glutamate in substrate peptides. To determine whether this residue is an important recognition element in v-Fps, Arg-1130 was substituted with leucine (R1130L) and glutamic acid (R1130E). The ability of these mutants to phosphorylate the peptide EAEIYXAIE, where X is glutamic acid, alanine, or lysine, was assessed. A comparison of the rates of peptide phosphorylation under limiting substrate concentrations (i.e., k(cat)/K(m) conditions) indicates that substrate specificity is altered by the electrostatic environment of the P+1 pocket. When the pocket displays a positive charge (Arg-1130; wild type), no charge (R1130L), or a negative charge (R1130E), v-Fps prefers to phosphorylate the glutamate peptide over the lysine peptide by a 200:1, 9:1, or 1:1 margin. While k(cat)/K(m) for the glutamate peptide is 50-fold higher for wild type compared to R1130E, k(cat)/K(m) for the lysine peptide is 3-fold higher for R1130E compared to wild type, a 150-fold change in relative substrate specificity. Analysis of the individual steps in the kinetic mechanism using viscosometric techniques indicates that the wild-type enzyme binds the glutamate peptide 3-fold better than the alanine peptide and, at least, 10-fold better than the lysine peptide. For R1130L, this margin range is reduced substantially, and for R1130E, no binding preference is observed. Nonetheless, the lysine peptide binds, at least, 4-fold better to R1130E than to wild type, and the glutamate peptide binds 3-fold poorer to R1130E than to wild type. The mutants lower the phosphoryl transfer rate by 4-30-fold for the three peptides, suggesting that Arg-1130 helps to position the tyrosine for optimum catalysis. The data indicate that a single mutation in v-Fps can alter significantly the relative substrate specificity by about 2 orders of magnitude with, at least, 50% of this effect occurring through relative changes in peptide binding affinity.


Subject(s)
Fusion Proteins, gag-onc/metabolism , Mutagenesis, Site-Directed , Peptides/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Fusion Proteins, gag-onc/genetics , Humans , Kinetics , Molecular Sequence Data , Peptides/genetics , Phosphorylation , Protein Binding/genetics , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/metabolism , Substrate Specificity/genetics , Viscosity
17.
Crit Rev Oncog ; 9(1): 43-62, 1998.
Article in English | MEDLINE | ID: mdl-9754447

ABSTRACT

The human c-fes protooncogene encodes a protein-tyrosine kinase (c-Fes) distinct from c-Src, c-Abl and other nonreceptor tyrosine kinases. Although originally identified as the cellular homolog of several transforming retroviral oncoproteins, Fes was later found to exhibit strong expression in myeloid hematopoietic cells and to play a direct role in their differentiation. Recent work has shown that Fes exhibits a more widespread expression pattern in both developing and adult tissues, suggesting a general physiological function for this kinase and its closely related homolog, Fer. This review highlights the unique aspects of Fes structure, regulation, and function that set it apart from other tyrosine kinase families.


Subject(s)
Protein-Tyrosine Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Animals , Cell Differentiation , Cell Division , Fusion Proteins, gag-onc/genetics , Fusion Proteins, gag-onc/metabolism , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/enzymology , Humans , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-bcr , Proto-Oncogene Proteins c-fes , src Homology Domains
18.
Biochemistry ; 37(51): 17875-81, 1998 Dec 22.
Article in English | MEDLINE | ID: mdl-9922154

ABSTRACT

Mutations were made in the activation loop tyrosine of the kinase domain of the oncoprotein v-Fps to assess the role of autophosphorylation in catalysis. Three mutant proteins, Y1073E, Y1073Q, and Y1073F, were expressed and purified as fusion proteins of glutathione-S-transferase from Escherichia coli and their catalytic properties were evaluated. Y1073E, Y1073Q, and Y1073F have k(cat) values that are reduced by 5-, 35-, and 40-fold relative to the wild-type enzyme, respectively. For all mutant enzymes, the Km values for ATP and a peptide substrate, EAEIYEAIE, are changed by 0.4-2-fold compared to the wild-type enzyme. The slopes for the plots of relative turnover versus solvent viscosity [(k(cat))eta] are 0.71 +/- 0.08, 0.10 +/- 0.06, and approximately 0 for wild type, Y1073Q, and Y1073E, respectively. These results imply that the phosphoryl transfer rate constant is reduced by 19- and 130-fold for Y1073E and Y1073Q compared to the wild-type enzyme. The dissociation constant of the substrate peptide is 1.5-2.5-fold lower for the mutants compared to wild type. The inhibition constant for EAEIFEAIE, a competitive inhibitor, is unaffected for Y1073E and raised 3-fold for Y1073Q compared to wild type. Y1073E and Y1073Q are strongly activated by free magnesium to the same extent and the apparent affinity constant for the metal is similar to that for the wild-type enzyme. The data indicate that the major role of autophosphorylation in the tyrosine kinase domain of v-Fps is to increase the rate of phosphoryl transfer without greatly affecting active-site accessibility or the local environment of the activating metal. Finally, the similar rate enhancements for phosphoryl transfer in v-Fps compared to protein kinase A [Adams et al. (1995) Biochemistry 34, 2447-2454] upon autophosphorylation suggest a conserved mechanism for communication between the activation loop and the catalytic residues of these two enzymes.


Subject(s)
Fusion Proteins, gag-onc/genetics , Fusion Proteins, gag-onc/metabolism , Mutagenesis, Site-Directed , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Animals , Binding Sites/genetics , Chickens , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fusion Proteins, gag-onc/antagonists & inhibitors , Kinetics , Magnesium/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Substrate Specificity/genetics , Viscosity
19.
Oncogene ; 14(13): 1571-80, 1997 Apr 03.
Article in English | MEDLINE | ID: mdl-9129148

ABSTRACT

Concentrations of the bioactive lipids, phosphatidate and diacylglycerol, increased with time in culture in ras- and tyrosine kinase (fps)-transformed fibroblasts but not in control fibroblasts. On Day 3, diacylglycerol and phosphatidate concentrations were about 3.3- and 5.5-fold higher respectively in the ras-transformed compared to control fibroblasts. These concentrations in fps-transformed fibroblasts were increased about twofold. The changes in phosphatidate and diacylglycerol resulted from enhanced phospholipid turnover rather than from synthesis de novo. The increased ratio of phosphatidate to diacylglycerol is explained by decreased activities of two distinct phosphatidate phosphohydrolases and increased diacylglycerol kinase in ras-transformed fibroblasts. Ceramide concentrations were about 2.5- and threefold higher in the fps- and ras-transformed cells respectively on Day 3 compared to the controls. Incubating control fibroblasts from Days 1 to 3 with phosphatidylcholine-specific phospholipase C increased diacylglycerol, phosphatidate and ceramide concentrations, and decreased Mg2+-independent-phosphatidate phosphohydrolase activity. 8-(4-chlorophenylthio)-cAMP had a cytostatic effect in ras-transformed cells, it decreased the concentrations of phosphatidate and diacylglycerol, but increased that of ceramide. The consequences of increased ceramide and phosphatidate concentrations in ras-transformed cells are discussed in relation to signal transduction, cell division and the transformed phenotype.


Subject(s)
Cell Transformation, Neoplastic , Ceramides/metabolism , Diglycerides/metabolism , Fusion Proteins, gag-onc/genetics , Genes, ras , Phosphatidic Acids/metabolism , Animals , Cell Division , Cell Line, Transformed , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Diacylglycerol Kinase , Fibroblasts , Glucose/metabolism , Phosphatidate Phosphatase/metabolism , Phospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein-Tyrosine Kinases/genetics , Signal Transduction , Thionucleotides/pharmacology , Type C Phospholipases/metabolism
20.
Biochemistry ; 35(5): 1533-9, 1996 Feb 06.
Article in English | MEDLINE | ID: mdl-8634284

ABSTRACT

The rate-determining steps in the phosphorylation of four tyrosine-containing peptides by the kinase domain of the nonreceptor tyrosine protein kinase v-fps were measured using viscosometric methods. The peptides were phosphorylated by a fusion protein of glutathione-S-transferase and the kinase domain of v-fps (GST-kin) and the initial velocities were determined by a coupled enzyme assay. Peptides I (EEEIYEEIE), II (EAEIYEAIE), and III (DADIYDAID) were phosphorylated by GST-kin with similar kinetic constants. The viscosogens, glycerol and sucrose, were found to have intermediate effects on kcat and no effect on kcat/Kpeptide for the phosphorylation of these three peptides. The data are interpreted according to the Stokes-Einstein equation and a simple three-step mechanism involving substrate binding, phosphoryl group transfer, and net product release. Two competitive inhibitors (EAEIFEAIE and DADIFDAID) exhibited K1 values that are 6-10-fold higher than the Kpeptide values for their analogous peptide substrates. The data imply that peptides I-III are in rapid equilibrium with the enzyme and that kcat is partially limited by both phosphoryl group transfer (40-100 s-1) and product release (17-22 s-1). GST-kin phosphorylates peptide IV (R5AENLEYamide) with a low Km (100 microM) and a kcat that is 40-fold lower than that for peptide I. No effect of solvent viscosity was observed for the phosphorylation of this peptide on either kcat or kcat/Kpeptide. This suggests that highly viscous solutions do not perturb structure and that the rate-determining step for this poor substrate is phosphoryl group transfer. The data indicate that the kinase domain of v-fps phosphorylates its best substrate with a chemical rate constant that is at least 5-fold lower than that for the serine-specific cAMP-dependent protein kinase and its best substrate LRRASLG (Adams & Taylor, 1992). Interestingly, both enzymes exhibit a similar affinity for their substrates and both enzymes release their products at a similar rate. This implies that the differences in catalytic efficiency between serine- and tyrosine-specific protein kinases lie exclusively in the rate constants for phosphoryl group transfer and not in substrate absorption or product desorption.


Subject(s)
Fusion Proteins, gag-onc/metabolism , Oligopeptides/metabolism , Peptide Fragments/metabolism , Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Fusion Proteins, gag-onc/genetics , Glutathione Transferase/genetics , Kinetics , Models, Chemical , Molecular Sequence Data , Peptide Fragments/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/metabolism , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL