Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Blood Cancer J ; 3: e135, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23955590

ABSTRACT

Previous studies have demonstrated that p210 BCR/ABL1 interacts directly with the xeroderma pigmentosum group B (XPB) protein, and that XPB is phosphorylated on tyrosine in cells that express p210 BCR/ABL1. In the current study, we have constructed a p210 BCR/ABL1 mutant that can no longer bind to XPB. The mutant has normal kinase activity and interacts with GRB2, but can no longer phosphorylate XPB. Loss of XPB binding is associated with reduced expression of c-MYC and reduced transforming potential in ex-vivo clonogenicity assays, but does not affect nucleotide excision repair in lymphoid or myeloid cells. When examined in a bone marrow transplantation (BMT) model for chronic myelogenous leukemia, mice that express the mutant exhibit attenuated myeloproliferation and lymphoproliferation when compared with mice that express unmodified p210 BCR/ABL1. Thus, the mutant-transplanted mice show predominantly neutrophilic expansion and altered progenitor expansion, and have significantly extended lifespans. This was confirmed in a BMT model for B-cell acute lymphoblastic leukemia, wherein the majority of the mutant-transplanted mice remain disease free. These results suggest that the interaction between p210 BCR/ABL1 and XPB can contribute to disease progression by influencing the lineage commitment of lymphoid and myeloid progenitors.

2.
Leukemia ; 27(5): 1080-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23207522

ABSTRACT

We have previously identified a tyrosine kinase-independent, guanine nucleotide exchange factor (GEF) activity, which is contained within the region of p210 no expansion BCR/ABL that distinguishes it from p190 BCR/ABL. In the current study, we have compared the transforming activity of p190 BCR/ABL, p210 BCR/ABL and a mutant that lacks GEF activity (p210 BCR/ABL(S509A)). In cell-based, ex vivo, and murine bone marrow transplantation (BMT) assays the transforming activity of p210 BCR/ABL(S509A) mimics p190 BCR/ABL, and is distinct from p210 BCR/ABL. Thus, in the BMT assay, the p190 BCR/ABL- and p210 BCR/ABL(S509A)-transplanted mice exhibit a more rapid onset of disease than mice transplanted with p210 BCR/ABL. The reduced disease latency is associated with erythroid hyperplasia in the absence of anemia, and expansion of the megakaryocyte-erythrocyte progenitor (MEP), common myeloid progenitor (CMP) and granulocyte-macrophage progenitor (GMP) populations, producing a phenotype that is similar to acute myeloid leukemia (AML-M6). The disease phenotype is readily transplantable into secondary recipients. This is consistent with ex vivo clonogenicity assays, where p210 BCR/ABL preferentially supports the growth of colony forming unit (CFU)-granulocyte-macrophage (GM), whereas p190 BCR/ABL and the mutant preferentially support the growth of burst forming unit-erythroid (BFU-E). These results suggest that the GEF activity that distinguishes p210 BCR/ABL from p190 BCR/ABL actively regulates disease progression.


Subject(s)
Fusion Proteins, bcr-abl/physiology , Guanine Nucleotide Exchange Factors/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Animals , Bone Marrow Transplantation , Cells, Cultured , Disease Progression , Humans , Interleukin-3/pharmacology , Mice , Mice, Inbred BALB C , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , rhoA GTP-Binding Protein/physiology
3.
Oncogene ; 27(14): 2064-71, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-17922031

ABSTRACT

The BCR-ABL oncogene encodes an in-frame fusion protein containing N-terminal sequences derived from Bcr and C-terminal sequences derived from Abl. Bcr contains a centrally located Rho-specific guanine nucleotide exchange factor (RhoGEF) domain that is retained within p210 Bcr-Abl. Although this domain is subject to autoinhibition in the context of Bcr, here we show that it is constitutively activated in p210 Bcr-Abl. p210 Bcr-Abl can stimulate RhoA activation independently of its tyrosine kinase activity, and mutations within the RhoGEF domain that are predicted to eliminate RhoGEF activity inhibit RhoA activation. The RhoGEF mutant of p210 Bcr-Abl does not affect the tyrosine kinase activity of the molecule, nor the ability of p210 Bcr-Abl to interact with XPB through the RhoGEF domain. Despite retaining normal levels of tyrosine kinase activity, the RhoGEF mutant of p210 Bcr-Abl is impaired in transforming activity as measured by anchorage-independent growth. However, the mutant is still able to confer the phenotype of growth factor independence in myeloid cells, suggesting that some, but not all parameters of p210 Bcr-Abl transformation, are dependent upon a catalytically active RhoGEF domain. Collectively, these observations identify a gain-of-function activity attributable to the RhoGEF domain of p210 Bcr-Abl that is required to support the transformed phenotype.


Subject(s)
Cell Transformation, Neoplastic/genetics , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Proto-Oncogene Proteins c-bcr/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line , Cell Transformation, Neoplastic/metabolism , Enzyme Activation , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Guanine Nucleotide Exchange Factors/chemistry , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Mice , Myeloid Cells/metabolism , Point Mutation , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-bcr/chemistry , Proto-Oncogene Proteins c-bcr/genetics , Rho Guanine Nucleotide Exchange Factors , cdc42 GTP-Binding Protein/metabolism
4.
Oncogene ; 20(16): 1953-63, 2001 Apr 12.
Article in English | MEDLINE | ID: mdl-11360179

ABSTRACT

We utilized a cDNA expression library derived from the B6SutA(1) mouse myeloid progenitor cell line to search for novel oncogenes that promote growth transformation of NIH3T3 cells. A 2.2 kb transforming cDNA was recovered that encodes the wild type thrombin-stimulated G protein-coupled receptor PAR-1. In addition to its potent focus forming activity, constitutive overexpression of PAR-1 in NIH3T3 cells promoted the loss of anchorage- and serum-dependent growth. Although inhibitors of thrombin failed to block PAR-1 transforming activity, a PAR-1 mutant that cannot be cleaved by thrombin was nontransforming. Since the foci of transformed cells induced by PAR-1 bear a striking resemblance to those induced by activated RhoA, we determined if PAR-1 transformation was due to the aberrant activation of a specific Rho family member. Like RhoA, PAR-1 cooperated with activated Raf-1 and caused synergistic enhancement of transforming activity, induced stress fibers when microinjected into porcine aortic endothelial cells, stimulated the activity of the serum response factor and NF-kappaB transcription factors, and PAR-1 transformation was blocked by co-expression of dominant negative RhoA. Finally, PAR-1 transforming activity was blocked by pertussis toxin and by co-expression of the RGS domain of Lsc, implicating Galpha(i) and Galpha(12)/Galpha(13) subunits, respectively, as mediators of PAR-1 transformation. Taken together, these observations suggest that PAR-1 growth transformation is mediated, in part, by activation of RhoA.


Subject(s)
Cell Transformation, Neoplastic , Receptors, Thrombin/physiology , Signal Transduction/physiology , rhoA GTP-Binding Protein/physiology , 3T3 Cells/cytology , 3T3 Cells/metabolism , 3T3 Cells/physiology , Actins/metabolism , Animals , Cell Adhesion/physiology , Cell Division/physiology , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA, Complementary/genetics , DNA-Binding Proteins/physiology , GTP-Binding Protein alpha Subunits, G12-G13 , Heterotrimeric GTP-Binding Proteins/physiology , Mice , Myeloid Cells/physiology , Receptor, PAR-1 , Receptors, Thrombin/biosynthesis , Receptors, Thrombin/genetics , Transfection , rho GTP-Binding Proteins/metabolism
5.
Oncogene ; 20(13): 1547-55, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11313901

ABSTRACT

G protein coupled receptors (GPCRs) constitute the largest family of cell surface receptors, with more than 1000 members, and are responsible for converting a diverse array of extracellular stimuli into intracellular signaling events. Most members of the family have defined roles in intermediary metabolism and generally perform these functions in well-differentiated cells. However, there is an increasing awareness that some GPCRs can also regulate proliferative signaling pathways and that chronic stimulation or mutational activation of receptors can lead to oncogenic transformation. Activating mutations in GPCRs are associated with several types of human tumors and some receptors exhibit potent oncogenic activity due to agonist overexpression. Additionally, expression screening analyses for novel oncogenes identified GPCRs whose expression causes the oncogenic transformation of NIH3T3 mouse fibroblasts. These include Mas, G2A, and the PAR-1 thrombin receptor. In this review we summarize the signaling and transforming properties of these GPCR oncoproteins. What has emerged from these studies is the delineation of a GTPase cascade where transforming GPCRs cause aberrant growth regulation via activation of Rho family small GTPases.


Subject(s)
Cell Transformation, Neoplastic , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , rho GTP-Binding Proteins/metabolism , Cell Cycle Proteins , Heterotrimeric GTP-Binding Proteins/metabolism , Models, Biological , Oncogenes , Proto-Oncogene Mas , Proto-Oncogene Proteins , Receptor, PAR-1 , Receptors, Thrombin , Signal Transduction
7.
Curr Biol ; 9(21): 1271-4, 1999 Nov 04.
Article in English | MEDLINE | ID: mdl-10556093

ABSTRACT

The long cytoplasmic tail of the human immunodeficiency virus (HIV)-1 transmembrane protein gp41 (gp41C) is implicated in the replication and cytopathicity of HIV-1 [1]. Little is known about the specific functions of gp41C, however. HIV-1 or simian immunodeficiency virus (SIV) mutants with defective gp41C have cell-type- or species-dependent phenotypes [2] [3] [4] [5] [6]. Thus, host factors are implicated in mediating the functions of gp41C. We report here that gp41C interacted with the carboxy-terminal regulatory domain of p115-RhoGEF [7], a specific guanine nucleotide exchange factor (GEF) and activator of the RhoA GTPase, which regulates actin stress fiber formation, activation of serum response factor (SRF) and cell proliferation [8] [9]. We demonstrate that gp41C inhibited p115-mediated actin stress fiber formation and activation of SRF. An amphipathic helix region with a leucine-zipper motif in gp41C is involved in its interaction with p115. Mutations in gp41C leading to loss of interaction with p115 impaired HIV-1 replication in human T cells. These findings suggest that an important function of gp41C is to modulate the activity of p115-RhoGEF and they thus reveal a new potential anti-HIV-1 target.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , HIV Envelope Protein gp41/metabolism , Leucine Zippers , Actins/physiology , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HIV Envelope Protein gp41/chemistry , HIV-1/physiology , HeLa Cells , Humans , Nuclear Proteins/metabolism , Protein Conformation , Regulatory Sequences, Nucleic Acid/genetics , Rho Guanine Nucleotide Exchange Factors , Serum Response Factor , T-Lymphocytes , Virus Replication
8.
Mol Cell Biol ; 19(11): 7759-70, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10523665

ABSTRACT

Dbs was identified initially as a transforming protein and is a member of the Dbl family of proteins (>20 mammalian members). Here we show that Dbs, like its rat homolog Ost and the closely related Dbl, exhibited guanine nucleotide exchange activity for the Rho family members RhoA and Cdc42, but not Rac1, in vitro. Dbs transforming activity was blocked by specific inhibitors of RhoA and Cdc42 function, demonstrating the importance of these small GTPases in Dbs-mediated growth deregulation. Although Dbs transformation was dependent upon the structural integrity of its pleckstrin homology (PH) domain, replacement of the PH domain with a membrane localization signal restored transforming activity. Thus, the PH domain of Dbs (but not Dbl) may be important in modulating association with the plasma membrane, where its GTPase substrates reside. Both Dbs and Dbl activate multiple signaling pathways that include activation of the Elk-1, Jun, and NF-kappaB transcription factors and stimulation of transcription from the cyclin D1 promoter. We found that Elk-1 and NF-kappaB, but not Jun, activation was necessary for Dbl and Dbs transformation. Finally, we have observed that Dbl and Dbs regulated transcription from the cyclin D1 promoter in a NF-kappaB-dependent manner. Previous studies have dissociated actin cytoskeletal activity from the transforming potential of RhoA and Cdc42. These observations, when taken together with those of the present study, suggest that altered gene expression, and not actin reorganization, is the critical mediator of Dbl and Rho family protein transformation.


Subject(s)
Cell Transformation, Neoplastic , Guanine Nucleotide Exchange Factors/metabolism , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases , Retroviridae Proteins, Oncogenic/metabolism , 3T3 Cells , Animals , MAP Kinase Kinase 1 , Membrane Proteins , Mice , Protein Structure, Tertiary , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
9.
J Biol Chem ; 274(43): 30410-8, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10521418

ABSTRACT

Deletion of the NH(2)-terminal 65 amino acids of proto-Vav (to form onco-Vav) activates its transforming activity, suggesting that these sequences serve a negative regulatory role in Vav function. However, the precise role of these NH(2)-terminal sequences and whether additional NH(2)-terminal sequences are also involved in negative regulation have not been determined. Therefore, we generated additional NH(2)-terminal deletion mutants of proto-Vav that lack the NH(2)-terminal 127, 168, or 186 amino acids, and assessed their abilities to cause focus formation in NIH 3T3 cells and to activate different signaling pathways. Since Vav mutants lacking 168 or 186 NH(2)-terminal residues showed a several 100-fold greater focus forming activity than that seen with deletion of 65 residues, residues spanning 66 to 187 also contribute significantly to negative regulation of Vav transforming activity. The increase in Vav transforming activity correlated with the activation of the c-Jun, Elk-1, and NF-kappaB transcription factors, as well as increased transcription from the cyclin D1 promoter. Tyrosine 174 is a key site of phosphorylation by Lck in vitro and Lck-mediated phosphorylation has been shown to be essential for proto-Vav GEF function in vitro. However, we found that an NH(2)-terminal Vav deletion mutant lacking this tyrosine residue (DeltaN-186 Vav) retained the ability to be phosphorylated by Lck in vivo and Lck still caused enhancement of DeltaN-186 Vav signaling and transforming activity. Thus, Lck can stimulate Vav via a mechanism that does not involve Tyr(174) or removal of NH(2)-terminal regulatory activity. Finally, we found that NH(2)-terminal deletion enhanced the degree of Vav association with the membrane-containing particulate fraction and that an isolated NH(2)-terminal fragment (residues 1-186) could impair DeltaN-186 Vav signaling. Taken together, these observations suggest that the NH(2) terminus may serve as a negative regulator of Vav by intramolecular interaction with COOH-terminal sequences to modulate efficient membrane association.


Subject(s)
Cell Cycle Proteins , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , 3T3 Cells , Animals , Cell Line , Cell Transformation, Neoplastic , Genes, Reporter , Humans , Mice , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-vav , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Restriction Mapping , Sequence Deletion , Transfection
10.
Mol Cell Biol ; 18(12): 6995-7008, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9819387

ABSTRACT

As part of a cDNA library screen for clones that induce transformation of NIH 3T3 fibroblasts, we have isolated a cDNA encoding the murine homolog of the guanine nucleotide exchange factor RasGRP. A point mutation predicted to prevent interaction with Ras abolished the ability of murine RasGRP (mRasGRP) to transform fibroblasts and to activate mitogen-activated protein kinases (MAP kinases). MAP kinase activation via mRasGRP was enhanced by coexpression of H-, K-, and N-Ras and was partially suppressed by coexpression of dominant negative forms of H- and K-Ras. The C terminus of mRasGRP contains a pair of EF hands and a C1 domain which is very similar to the phorbol ester- and diacylglycerol-binding C1 domains of protein kinase Cs. The EF hands could be deleted without affecting the ability of mRasGRP to transform NIH 3T3 cells. In contrast, deletion of the C1 domain or an adjacent cluster of basic amino acids eliminated the transforming activity of mRasGRP. Transformation and MAP kinase activation via mRasGRP were restored if the deleted C1 domain was replaced either by a membrane-localizing prenylation signal or by a diacylglycerol- and phorbol ester-binding C1 domain of protein kinase C. The transforming activity of mRasGRP could be regulated by phorbol ester when serum concentrations were low, and this effect of phorbol ester was dependent on the C1 domain of mRasGRP. The C1 domain could also confer phorbol myristate acetate-regulated transforming activity on a prenylation-defective mutant of K-Ras. The C1 domain mediated the translocation of mRasGRP to cell membranes in response to either phorbol ester or serum stimulation. These results suggest that the primary mechanism of activation of mRasGRP in fibroblasts is through its recruitment to diacylglycerol-enriched membranes. mRasGRP is expressed in lymphoid tissues and the brain, as well as in some lymphoid cell lines. In these cells, RasGRP has the potential to serve as a direct link between receptors which stimulate diacylglycerol-generating phospholipase Cs and the activation of Ras.


Subject(s)
DNA-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors , Tetradecanoylphorbol Acetate/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , DNA Mutational Analysis , DNA, Complementary/genetics , Enzyme Activation/physiology , Gene Expression/genetics , Humans , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Point Mutation/genetics , RNA, Messenger/genetics , Retroviridae/genetics , Sequence Homology, Amino Acid , Signal Transduction/physiology , Transformation, Genetic/genetics
11.
Genome ; 41(4): 495-503, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9796098

ABSTRACT

Suppressors of position-effect variegation (Su(var)s) in Drosophila melanogaster are usually studied in the presence of chromosomal rearrangements, which exhibit variegated expression of euchromatic genes moved near to, or heterochromatic genes moved away from, centromeric heterochromatin. However, the effects of Su(var) mutations on heterochromatic gene expression in the absence of a variegating re-arrangement have not yet been defined. Here we present a number of results which suggest that Su(var) gene products can interact to affect the expression of the light gene in its normal heterochromatic location. We initially observed that eye pigment was reduced in several Su(var) double mutants; the phenotype resembled that of light mutations and was more severe when only one copy of the light gene was present. This reduced pigmentation could be alleviated by a duplication for the light gene or by a reduction in the amount of cellular heterochromatin. In addition, the viability of most Su(var) double mutant combinations tested was greatly reduced in a genetic background of reduced light gene dosage, when extra heterochromatin is present. We conclude that Su(var) gene products can affect expression of the heterochromatic light gene in the absence of any chromosomal rearrangements. However, it is noteworthy that mutations in any single Su(var) gene have little effect on light expression; we observe instead that different pairings of Su(var) mutations are required to show an effect on light expression. Interestingly, we have obtained evidence that at least two of the second chromosome Su(var) mutations are gain-of-function lesions, which also suggests that there may be different modes of interaction among these genes. It may therefore be possible to use this more sensitive assay of Su(var) effects on heterochromatic genes to infer functional relationships among the products of the 50 or more known Su(var) loci.


Subject(s)
Chromosomes/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation , Genes, Insect , Heterochromatin/genetics , Animals , Chromosomes/ultrastructure , Crosses, Genetic , Female , Genotype , Homozygote , Male , Mutation , Retinal Pigments/genetics
12.
Mol Cell Biol ; 18(8): 4689-97, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9671479

ABSTRACT

Activated forms of different Rho family members (CDC42, Rac1, RhoA, RhoB, and RhoG) have been shown to transform NIH 3T3 cells as well as contribute to Ras transformation. Rho family guanine nucleotide exchange factors (GEFs) (also known as Dbl family proteins) that activate CDC42, Rac1, and RhoA also demonstrate oncogenic potential. The faciogenital dysplasia gene product, FGD1, is a Dbl family member that has recently been shown to function as a CDC42-specific GEF. Mutations within the FGD1 locus cosegregate with faciogenital dysplasia, a multisystemic disorder resulting in extensive growth impairments throughout the skeletal and urogenital systems. Here we demonstrate that FGD1 expression is sufficient to cause tumorigenic transformation of NIH 3T3 fibroblasts. Although both FGD1 and constitutively activated CDC42 cooperated with Raf and showed synergistic focus-forming activity, both quantitative and qualitative differences in their functions were seen. FGD1 and CDC42 also activated common nuclear signaling pathways. However, whereas both showed comparable activation of c-Jun, CDC42 showed stronger activation of serum response factor and FGD1 was consistently a better activator of Elk-1. Although coexpression of FGD1 with specific inhibitors of CDC42 function demonstrated the dependence of FGD1 signaling activity on CDC42 function, FGD1 signaling activities were not always consistent with the direct or exclusive stimulation of CDC42 function. In summary, FGD1 and CDC42 signaling and transformation are distinct, thus suggesting that FGD1 may be mediating some of its biological activities through non-CDC42 targets.


Subject(s)
Cell Cycle Proteins/metabolism , GTP-Binding Proteins/metabolism , Proteins/metabolism , Signal Transduction , Transformation, Genetic , 3T3 Cells , Animals , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors , Mice , Nuclear Proteins/metabolism , Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-raf/genetics , Serum Response Factor , Transcription Factors/metabolism , cdc42 GTP-Binding Protein , ets-Domain Protein Elk-1
13.
J Biol Chem ; 273(27): 16739-47, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9642229

ABSTRACT

The dbl family of oncogenes encodes a large, structurally related, family of growth-regulatory molecules that possess guanine nucleotide exchange factor activity for specific members of the Rho family of Ras-related GTPases. We have evaluated matched sets of weakly and strongly transforming versions of five Dbl family proteins (Lfc, Lsc, Ect2, Dbl, and Dbs) to determine their ability to stimulate signaling pathways that are activated by Rho family proteins. We found that the transforming potential of this panel did not correlate directly with their ability to activate Jun NH2-terminal kinase, p38/Mpk2, serum response factor, or c-Jun. In contrast, transient stimulation of transcription from the cyclin D1 promoter provided a strong correlation with transforming potential, and we found constitutive up-regulation of cyclin D1 protein in Dbl family protein-transformed cells. In addition, we observed that at least two Dbl family members (Lfc and Ect2) induced changes in the actin cytoskeleton and exhibited nuclear signaling profiles that are consistent with a broader range of in vivo substrate utilization than is predicted from their in vitro exchange specificities. In summary, although Dbl family proteins exhibit signaling profiles that are consistent with their in vivo activation of Rho proteins, stimulation of cyclin D1 transcription is the only activity that correlates with transforming potential, thus suggesting that deregulated cell cycle progression may be important for Dbl family protein transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin D1/genetics , Mitogen-Activated Protein Kinases , Promoter Regions, Genetic , Retroviridae Proteins, Oncogenic/metabolism , Transcription, Genetic/genetics , 3T3 Cells , Actins/metabolism , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation , Guanine Nucleotide Exchange Factors , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Mice , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-jun/metabolism , Serum Response Factor , Signal Transduction
14.
Nature ; 390(6660): 632-6, 1997 Dec 11.
Article in English | MEDLINE | ID: mdl-9403696

ABSTRACT

Transformation of mammary epithelial cells into invasive carcinoma results in alterations in their integrin-mediated responses to the extracellular matrix, including a loss of normal epithelial polarization and differentiation, and a switch to a more motile, invasive phenotype. Changes in the actin cytoskeleton associated with this switch suggest that the small GTPases Cdc42 and Rac, which regulate actin organization, might modulate motility and invasion. However, the role of Cdc42 and Rac1 in epithelial cells, especially with respect to integrin-mediated events, has not been well characterized. Here we show that activation of Cdc42 and Rac1 disrupts the normal polarization of mammary epithelial cells in a collagenous matrix, and promotes motility and invasion. This motility does not require the activation of PAK, JNK, p70 S6 kinase, or Rho, but instead requires phosphatidylinositol-3-OH kinase (PI(3)K). Further, direct PI(3)K activation is sufficient to disrupt epithelial polarization and induce cell motility and invasion. PI(3)K inhibition also disrupts actin structures, suggesting that activation of PI(3)K by Cdc42 and Rac1 alters actin organization, leading to increased motility and invasiveness.


Subject(s)
Cell Cycle Proteins/physiology , Cell Movement , Cell Transformation, Neoplastic , GTP Phosphohydrolases/physiology , GTP-Binding Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Actins/metabolism , Breast/cytology , Cell Line , Epithelium , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , Transfection , cdc42 GTP-Binding Protein, Saccharomyces cerevisiae , rac GTP-Binding Proteins
15.
Gene ; 198(1-2): 229-36, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9370286

ABSTRACT

The Ub80 gene in eukaryotes produces a ubiquitin fusion protein in which ubiquitin is fused in frame to a tail protein (Redman and Rechsteiner, 1988; Finley et al., 1989; Barrio et al., 1994). The tail protein is incorporated into the ribosome, and ubiquitin is thought to act as a chaperone. The DUb80 gene of Drosophila melanogaster was cloned by Barrio et al. (1994) and contains a 5'-untranslated exon, followed by a large intron and then the first coding exon. We report that the large intron of DUb80 contains an open reading frame, which produces a 259-aa protein (IP259) that is conserved in eukaryotes from yeast to mammals. Transcription of the DUb80 and IP259 mRNAs begins at the same start sites. However, alternate splicing of the primary transcript produces two structurally unrelated proteins. This is the second reported instance of two structurally unrelated proteins being produced via alternate splicing, suggesting that this form of genomic organization may be more common than previously thought.


Subject(s)
Drosophila melanogaster/genetics , Genes, Insect , Genes, Overlapping , Insect Proteins/genetics , Ubiquitins/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Immunologic Techniques , Introns , Molecular Sequence Data , RNA, Messenger/genetics , Sequence Alignment , Transcription, Genetic
17.
J Biol Chem ; 271(44): 27374-81, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8910315

ABSTRACT

Lfc and Lsc are two recently identified oncoproteins that contain a Dbl homology domain in tandem with a pleckstrin homology domain and thus share sequence similarity with a number of other growth regulatory proteins including Dbl, Tiam-1, and Lbc. We show here that Lfc and Lsc, like their closest relative Lbc, are highly specific guanine nucleotide exchange factors (GEFs) for Rho, causing a >10-fold stimulation of [3H]GDP dissociation from Rho and a marked stimulation of GDP-[35S]GTPgammas (guanosine 5'-O-(3-thiotriphosphate) exchange. All three proteins (Lbc, Lfc, and Lsc) are able to act catalytically in stimulating the guanine nucleotide exchange activity, such that a single molecule of each of these oncoproteins can activate a number of molecules of Rho. Neither Lfc nor Lsc shows any ability to stimulate GDP dissociation from other related GTP-binding proteins such as Rac, Cdc42, or Ras. Thus Lbc, Lfc, and Lsc appear to represent a subgroup of Dbl-related proteins that function as highly specific GEFs toward Rho and can be distinguished from Dbl, Ost, and Dbs which are less specific and show GEF activity toward both Rho and Cdc42. Consistent with these results, Lbc, Lfc, and Lsc each form tight complexes with the guanine nucleotide-depleted form of Rho and bind weakly to the GDP- and GTPgammaS-bound states. None of these oncoproteins are able to form complexes with Cdc42 or Ras. However, Lfc (but not Lbc nor Lsc) can bind to Rac, and this binding occurs equally well when Rac is nucleotide-depleted or is in the GDP- or GTPgammaS-bound state. These findings raise the possibility that in addition to acting directly as a GEF for Rho, Lfc may play other roles that influence the signaling activities of Rac and/or coordinate the activities of the Rac and Rho proteins.


Subject(s)
GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , Proto-Oncogene Proteins/metabolism , 3T3 Cells , A Kinase Anchor Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cell Line , Glutathione Transferase , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine Diphosphate/metabolism , Kinetics , Mice , Minor Histocompatibility Antigens , Molecular Sequence Data , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/chemistry , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Rho Guanine Nucleotide Exchange Factors , Sequence Homology, Amino Acid , Spodoptera , Substrate Specificity , Transfection
18.
J Biol Chem ; 271(31): 18643-50, 1996 Aug 02.
Article in English | MEDLINE | ID: mdl-8702517

ABSTRACT

In a screen for genes with oncogenic potential expressed by the murine B6SUtA1 myeloid progenitor cell line, we isolated a 2. 5-kilobase pair cDNA whose expression causes strong morphological transformation and deregulated proliferation of NIH 3T3 cells. The transforming cDNA encodes a truncated protein (designated Lsc) with a region of sequence similarity to the product of the lbc oncogene. This region includes the tandem Dbl homology and pleckstrin homology domains that are hallmarks of the Dbl-like proteins, a family of presumptive or demonstrated guanine nucleotide exchange factors that act on Rho family GTPases. Lsc requires intact Dbl homology and pleckstrin homology domains for its oncogenic activity. The transforming activity of Lsc in NIH 3T3 cells is reduced by cotransfection with p190 (a GTPase activating protein for Rho family GTPases) and the Rho family dominant-negative mutants RhoA(19N), CDC42(17N), and Rac1(17N). These results indicate a role for the Rho family of GTPases in mediating the transforming activity of Lsc and are consistent with the exchange specificities that have been attributed to Dbl family members. The lsc gene is expressed in a variety of tissues and is particularly abundant in hemopoietic tissues (thymus, spleen, and bone marrow). Lsc is a member of a growing family of proteins that may function as activators of Rho family GTPases in a developmental or tissue-specific manner.


Subject(s)
GTPase-Activating Proteins , Oncogenes , Proteins/genetics , Retroviridae Proteins, Oncogenic/genetics , 3T3 Cells , A Kinase Anchor Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , Mice , Minor Histocompatibility Antigens , Molecular Sequence Data , Molecular Structure , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Proteins/chemistry , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Retroviridae Proteins, Oncogenic/chemistry , Rho Guanine Nucleotide Exchange Factors , Sequence Homology, Amino Acid , Transformation, Genetic
19.
Genome ; 36(4): 676-85, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8405984

ABSTRACT

In fission yeast, the product of the cdc2 gene is required both for entry into S phase and mitosis. Homologs of cdc2 have been isolated from a number of metazoans, but in general they have not been amenable to genetic analysis. Here we describe P element transposon tagging of Cdc2 in Drosophila melanogaster and the analysis of 10 Cdc2 mutants. The recessive lethality of Cdc2P is associated with a P element located in the 5' untranslated region of the gene. Seven other alleles have unique single base pair substitutions in the coding region of Cdc2. One allele, Cdc2B47, is mutated in the splice donor site of exon 1. Most mutations in Cdc2, including the presumptive null allele Cdc2B47, die at the pupal stage, suggesting that the maternally supplied Cdc2 gene product drives earlier cell divisions. The phenotypes of our mutants are consistent with a role for Cdc2 in cell proliferation; however, we did not observe any perturbation of the endoreduplication cycle associated with the acquisition of polyteny.


Subject(s)
CDC2 Protein Kinase/genetics , Drosophila melanogaster/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Cell Division/genetics , Cloning, Molecular , DNA, Complementary/genetics , Drosophila melanogaster/enzymology , Drosophila melanogaster/growth & development , Female , Genes, Insect , Male , Molecular Sequence Data , Mutation
20.
Genetics ; 134(1): 221-30, 1993 May.
Article in English | MEDLINE | ID: mdl-8514131

ABSTRACT

Cytogenetic region 31 of the second chromosome of Drosophila melanogaster was screened for recessive lethal mutations. One hundred and thirty nine new recessive lethal alleles were isolated that fail to complement Df(2L)J2 (31A-32A). These new alleles, combined with preexisting mutations in the region, define 52 complementation groups, 35 of which have not previously been described. Among the new mutations were alleles of the cdc2 and mfs(2)31 genes. Six new deficiencies were also isolated and characterized identifying 16 deficiency subintervals within region 31. The new deficiencies were used to further localize three loci believed to encode non-histone chromosomal proteins. Suvar(2)1/Su(var)214, a dominant suppressor of position-effect variegation (PEV), maps to 31A-B, while the recessive suppressors of PEV mfs(2)31 and wdl were localized to regions 31E and 31F-32A, respectively. In addition, the cytological position of several mutations that interact with heterochromatin were more precisely defined.


Subject(s)
Chromosome Mapping , Drosophila melanogaster/genetics , Alleles , Animals , Cytogenetics , Female , Gene Rearrangement , Genes, Lethal , Genes, Recessive , Genes, Suppressor , Genetic Complementation Test , Male , Mutation , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...