Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
Oncogene ; 33(31): 4021-35, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24037532

ABSTRACT

The aberrant activity of Ras homologous (Rho) family small GTPases (20 human members) has been implicated in cancer and other human diseases. However, in contrast to the direct mutational activation of Ras found in cancer and developmental disorders, Rho GTPases are activated most commonly in disease by indirect mechanisms. One prevalent mechanism involves aberrant Rho activation via the deregulated expression and/or activity of Rho family guanine nucleotide exchange factors (RhoGEFs). RhoGEFs promote formation of the active GTP-bound state of Rho GTPases. The largest family of RhoGEFs is comprised of the Dbl family RhoGEFs with 70 human members. The multitude of RhoGEFs that activate a single Rho GTPase reflects the very specific role of each RhoGEF in controlling distinct signaling mechanisms involved in Rho activation. In this review, we summarize the role of Dbl RhoGEFs in development and disease, with a focus on Ect2 (epithelial cell transforming squence 2), Tiam1 (T-cell lymphoma invasion and metastasis 1), Vav and P-Rex1/2 (PtdIns(3,4,5)P3 (phosphatidylinositol (3,4,5)-triphosphate)-dependent Rac exchanger).


Subject(s)
Neoplasms/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Division , Congenital Abnormalities/metabolism , Gene Expression Regulation, Neoplastic , Genetic Diseases, Inborn/metabolism , Humans , Proto-Oncogene Proteins c-vav/physiology
2.
Oncogene ; 29(3): 380-91, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19838215

ABSTRACT

The Rheb1 and Rheb2 small GTPases and their effector mTOR are aberrantly activated in human cancer and are attractive targets for anti-cancer drug discovery. Rheb is targeted to endomembranes via its C-terminal CAAX (C=cysteine, A=aliphatic, X=terminal amino acid) motif, a substrate for posttranslational modification by a farnesyl isoprenoid. After farnesylation, Rheb undergoes two additional CAAX-signaled processing steps, Ras converting enzyme 1 (Rce1)-catalyzed cleavage of the AAX residues and isoprenylcysteine carboxyl methyltransferase (Icmt)-mediated carboxylmethylation of the farnesylated cysteine. However, whether these postprenylation processing steps are required for Rheb signaling through mTOR is not known. We found that Rheb1 and Rheb2 localize primarily to the endoplasmic reticulum and Golgi apparatus. We determined that Icmt and Rce1 processing is required for Rheb localization, but is dispensable for Rheb-induced activation of the mTOR substrate p70 S6 kinase (S6K). Finally, we evaluated whether farnesylthiosalicylic acid (FTS) blocks Rheb localization and function. Surprisingly, FTS prevented S6K activation induced by a constitutively active mTOR mutant, indicating that FTS inhibits mTOR at a level downstream of Rheb. We conclude that inhibitors of Icmt and Rce1 will not block Rheb function, but FTS could be a promising treatment for Rheb- and mTOR-dependent cancers.


Subject(s)
Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Protein Processing, Post-Translational , Signal Transduction , Animals , Binding Sites/genetics , Blotting, Western , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Farnesol/analogs & derivatives , Farnesol/pharmacology , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microscopy, Fluorescence , Monomeric GTP-Binding Proteins/genetics , Mutation , NIH 3T3 Cells , Neuropeptides/genetics , Phosphorylation/drug effects , Prenylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Ras Homolog Enriched in Brain Protein , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Salicylates/pharmacology , TOR Serine-Threonine Kinases , Transfection
3.
Cancer Metastasis Rev ; 28(1-2): 77-83, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19221866

ABSTRACT

DLC-1 was originally identified as a potential tumor suppressor. One of the key biochemical functions of DLC-1 is to serve as a GTPase activating protein (GAP) for members of the Rho family of GTPases, particularly Rho A-C and Cdc 42. Since these GTPases are critically involved in regulation of the cytoskeleton and cell migration, it seems clear that DLC-1 will also influence these processes. In this review we examine basic aspects of the actin cyoskeleton and how it relates to cell motility. We then delineate the characteristics of DLC-1 and other members of its family, and describe how they may have multiple effects on the regulation of cell polarity, actin organization, and cell migration.


Subject(s)
Cytoskeleton/metabolism , GTPase-Activating Proteins/metabolism , Tumor Suppressor Proteins/physiology , Animals , Cell Movement , Humans , Mice , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction , Tumor Suppressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
4.
Oncogene ; 28(11): 1401-9, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19151751

ABSTRACT

DLC1 (deleted in liver cancer 1), which encodes a Rho GTPase-activating protein (Rho-GAP), is a potent tumor suppressor gene that is frequently inactivated in several human cancers. DLC1 is a multidomain protein that has been shown previously to bind members of the tensin gene family. Here we show that p120Ras-GAP (Ras-GAP; also known as RASA1) interacts and extensively colocalizes with DLC1 in focal adhesions. The binding was mapped to the SH3 domain located in the N terminus of Ras-GAP and to the Rho-GAP catalytic domain located in the C terminus of the DLC1. In vitro analyses with purified proteins determined that the isolated Ras-GAP SH3 domain inhibits DLC1 Rho-GAP activity, suggesting that Ras-GAP is a negative regulator of DLC1 Rho-GAP activity. Consistent with this possibility, we found that ectopic overexpression of Ras-GAP in a Ras-GAP-insensitive tumor line impaired the growth-suppressing activity of DLC1 and increased RhoA activity in vivo. Our observations expand the complexity of proteins that regulate DLC1 function and define a novel mechanism of the cross talk between Ras and Rho GTPases.1R01CA129610


Subject(s)
Neoplasms/pathology , Tumor Suppressor Proteins/physiology , p120 GTPase Activating Protein/physiology , rhoA GTP-Binding Protein/metabolism , Catalytic Domain , Cell Line, Tumor , Cell Proliferation , GTPase-Activating Proteins , Humans , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , p120 GTPase Activating Protein/analysis , p120 GTPase Activating Protein/chemistry , src Homology Domains
5.
Oncogene ; 26(22): 3291-310, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17496923

ABSTRACT

Mitogen-activated protein kinase (MAPK) cascades are key signaling pathways involved in the regulation of normal cell proliferation, survival and differentiation. Aberrant regulation of MAPK cascades contribute to cancer and other human diseases. In particular, the extracellular signal-regulated kinase (ERK) MAPK pathway has been the subject of intense research scrutiny leading to the development of pharmacologic inhibitors for the treatment of cancer. ERK is a downstream component of an evolutionarily conserved signaling module that is activated by the Raf serine/threonine kinases. Raf activates the MAPK/ERK kinase (MEK)1/2 dual-specificity protein kinases, which then activate ERK1/2. The mutational activation of Raf in human cancers supports the important role of this pathway in human oncogenesis. Additionally, the Raf-MEK-ERK pathway is a key downstream effector of the Ras small GTPase, the most frequently mutated oncogene in human cancers. Finally, Ras is a key downstream effector of the epidermal growth factor receptor (EGFR), which is mutationally activated and/or overexpressed in a wide variety of human cancers. ERK activation also promotes upregulated expression of EGFR ligands, promoting an autocrine growth loop critical for tumor growth. Thus, the EGFR-Ras-Raf-MEK-ERK signaling network has been the subject of intense research and pharmaceutical scrutiny to identify novel target-based approaches for cancer treatment. In this review, we summarize the current status of the different approaches and targets that are under evaluation and development for the therapeutic intervention of this key signaling pathway in human disease.


Subject(s)
Drug Delivery Systems/methods , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , raf Kinases/antagonists & inhibitors , Animals , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , MAP Kinase Kinase Kinases/physiology , MAP Kinase Signaling System/physiology , Neoplasms/genetics , raf Kinases/genetics , raf Kinases/physiology
6.
Dev Cell ; 1(1): 63-72, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11703924

ABSTRACT

Mutations in the neurofibromatosis type II (NF2) tumor suppressor predispose humans and mice to tumor development. The study of Nf2+/- mice has demonstrated an additional effect of Nf2 loss on tumor metastasis. The NF2-encoded protein, merlin, belongs to the ERM (ezrin, radixin, and moesin) family of cytoskeleton:membrane linkers. However, the molecular basis for the tumor- and metastasis- suppressing activity of merlin is unknown. We have now placed merlin in a signaling pathway downstream of the small GTPase Rac. Expression of activated Rac induces phosphorylation and decreased association of merlin with the cytoskeleton. Furthermore, merlin overexpression inhibits Rac-induced signaling in a phosphorylation-dependent manner. Finally, Nf2-/- cells exhibit characteristics of cells expressing activated alleles of Rac. These studies provide insight into the normal cellular function of merlin and how Nf2 mutation contributes to tumor initiation and progression.


Subject(s)
Neurofibromin 2/metabolism , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Gene Expression/physiology , Mice , Molecular Sequence Data , Neurofibromin 2/genetics , Phosphorylation , cdc42 GTP-Binding Protein/metabolism
7.
J Biol Chem ; 276(51): 48257-61, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11641393

ABSTRACT

PLC-epsilon was identified recently as a phosphoinositide-hydrolyzing phospholipase C (PLC) containing catalytic domains (X, Y, and C2) common to all PLC isozymes as well as unique CDC25- and Ras-associating domains. Novel regulation of this PLC isozyme by the Ras oncoprotein and alpha-subunits (Galpha(12)) of heterotrimeric G proteins was illustrated. Sequence analyses of PLC-epsilon revealed previously unrecognized PH and EF-hand domains in the amino terminus. The known interaction of Gbetagamma subunits with the PH domains of other proteins led us to examine the capacity of Gbetagamma to activate PLC-epsilon. Co-expression of Gbeta(1)gamma(2) with PLC-epsilon in COS-7 cells resulted in marked stimulation of phospholipase C activity. Gbeta(2) and Gbeta(4) in combination with Ggamma(1), Ggamma(2), Ggamma(3), or Ggamma(13) also activated PLC-epsilon to levels similar to those observed with Gbeta(1)-containing dimers of these Ggamma-subunits. Gbeta(3) in combination with the same Ggamma-subunits was less active, and Gbeta(5)-containing dimers were essentially inactive. Gbetagamma-promoted activation of PLC-epsilon was blocked by cotransfection with either of two Gbetagamma-interacting proteins, Galpha(i1) or the carboxyl terminus of G protein receptor kinase 2. Pharmacological inhibition of PI3-kinase-gamma had no effect on Gbeta(1)gamma(2)-promoted activation of PLC-epsilon. Similarly, activation of Ras in the action of Gbetagamma is unlikely, because a mutation in the second RA domain of PLC-epsilon that blocks Ras activation of PLC failed to alter the stimulatory activity of Gbeta(1)gamma(2). Taken together, these results reveal the presence of additional functional domains in PLC-epsilon and add a new level of complexity in the regulation of this novel enzyme by heterotrimeric G proteins.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Type C Phospholipases/metabolism , Amino Acid Sequence , Animals , Cell Line , Dimerization , Enzyme Activation , Heterotrimeric GTP-Binding Proteins/chemistry , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide Phospholipase C , Sequence Homology, Amino Acid , Signal Transduction , Type C Phospholipases/chemistry
8.
Nat Struct Biol ; 8(12): 1037-41, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11685227

ABSTRACT

Rho GTPases are activated by a family of guanine nucleotide exchange factors (GEFs) known as Dbl family proteins. The structural basis for how GEFs recognize and activate Rho GTPases is presently ill defined. Here, we utilized the crystal structure of the DH/PH domains of the Rac-specific GEF Tiam1 in complex with Rac1 to determine the structural elements of Rac1 that regulate the specificity of this interaction. We show that residues in the Rac1 beta2-beta3 region are critical for Tiam1 recognition. Additionally, we determined that a single Rac1-to-Cdc42 mutation (W56F) was sufficient to abolish Rac1 sensitivity to Tiam1 and allow recognition by the Cdc42-specific DH/PH domains of Intersectin while not impairing Rac1 downstream activities. Our findings identified unique GEF specificity determinants in Rac1 and provide important insights into the mechanism of DH/PH selection of GTPase targets.


Subject(s)
Adaptor Proteins, Vesicular Transport , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Protein Interaction Mapping , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/metabolism , 3T3 Cells , Amino Acid Substitution/genetics , Animals , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Transformation, Neoplastic , Enzyme Activation , Humans , Ligands , Mice , Models, Molecular , Mutagenesis, Site-Directed/genetics , Mutation/genetics , Protein Binding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Substrate Specificity , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics
9.
J Biol Chem ; 276(49): 45868-75, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11577097

ABSTRACT

Normally, Rho GTPases are activated by the removal of bound GDP and the concomitant loading of GTP catalyzed by members of the Dbl family of guanine nucleotide exchange factors (GEFs). This family of GEFs invariantly contain a Dbl homology (DH) domain adjacent to a pleckstrin homology (PH) domain, and while the DH domain usually is sufficient to catalyze nucleotide exchange, possible roles for the conserved PH domain remain ambiguous. Here we demonstrate that the conserved PH domains of three distinct Dbl family proteins, intersectin, Dbs, and Tiam1, selectively bind lipid vesicles only when phosphoinositides are present. While the PH domains of intersectin and Dbs promiscuously bind several multiphosphorylated phosphoinositides, Tiam1 selectively interacts with phosphatidylinositol 3-phosphate (K(D) approximately 5-10 microm). In addition, and in contrast to recent reports, catalysis of nucleotide exchange on nonprenylated Rac1 provided by various extended portions of Tiam1 is not influenced by (a) soluble phosphoinositide head groups, (b) dibutyl versions of phosphoinositides, or (c) lipid vesicles containing phosphoinositides. Likewise, GEF activity afforded by DH/PH fragments of intersectin and Dbs are also not altered by phosphoinositide interactions. These results strongly suggest that unless all relevant components are localized to a lipid membrane surface, Dbl family GEFs generally are not intrinsically modulated by binding phosphoinositides.


Subject(s)
Adaptor Proteins, Vesicular Transport , Carrier Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Phosphatidylinositols/metabolism , Proteins/metabolism , Immunoblotting , Protein Binding , Rho Guanine Nucleotide Exchange Factors , Surface Plasmon Resonance , T-Lymphoma Invasion and Metastasis-inducing Protein 1
10.
J Biol Chem ; 276(45): 42259-67, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11533059

ABSTRACT

Using microarray analysis, we identified a unique ras superfamily gene, termed RERG (ras-related and estrogen-regulated growth inhibitor), whose expression was decreased or lost in a significant percentage of primary human breast tumors that show a poor clinical prognosis. Importantly, high RERG expression correlated with expression of a set of genes that define a breast tumor subtype that is estrogen receptor-positive and associated with a slow rate of tumor cell proliferation and a favorable prognosis for these cancer patients. RERG mRNA expression was induced rapidly in MCF-7 cells stimulated by beta-estradiol and repressed by tamoxifen treatment. Like Ras, RERG protein exhibited intrinsic GDP/GTP binding and GTP hydrolysis activity. Unlike Ras proteins, RERG lacks a known recognition signal for COOH-terminal prenylation and was localized primarily in the cytoplasm. Expression of RERG protein in MCF-7 breast carcinoma cells resulted in a significant inhibition of both anchorage-dependent and anchorage-independent growth in vitro and inhibited tumor formation in nude mice. These features of RERG are strikingly different from most Ras superfamily GTP-binding pro-teins and suggest that the loss of RERG expression may contribute to breast tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Estrogens/pharmacology , GTP-Binding Proteins/genetics , Genes, ras , Growth Inhibitors/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Breast Neoplasms/pathology , Female , GTP Phosphohydrolases/metabolism , Humans , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Tumor Cells, Cultured
11.
Cancer Lett ; 171(1): 1-10, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11485822

ABSTRACT

The important contribution of aberrant Ras activation in oncogenesis is well established. Our knowledge of the signaling pathways that are regulated by Ras is considerable. However, the number of downstream effectors of Ras continues to increase and our understanding of the role of these effector signaling pathways in mediating oncogenesis is far from complete and continues to evolve. Similarly, our understanding of the components that control mitogen-stimulated cell cycle progression is also very advanced. Where our understanding has lagged has been the delineation of the mechanism by which Ras causes a deregulation of cell cycle progression to promote the uncontrolled proliferation of the cancer cell. In this review, we summarize our current knowledge of how deregulated Ras activation alters the function of cyclin D1, p21(Cip1), and p27(Kip1). The two themes that we have emphasized are the involvement of Rho small GTPases in cell cycle regulation and the cell-type differences in how Ras signaling interfaces with the cell cycle machinery.


Subject(s)
Cell Cycle/physiology , Cell Transformation, Neoplastic , Tumor Suppressor Proteins , ras Proteins/physiology , rho GTP-Binding Proteins/physiology , 3T3 Cells/cytology , 3T3 Cells/metabolism , Animals , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cell Division/physiology , Cyclin D1/genetics , Cyclin D1/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/physiology , Cyclins/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Humans , MAP Kinase Signaling System/physiology , Mice , Models, Biological , Retinoblastoma Protein/physiology , Signal Transduction/physiology
12.
Mol Cell Biol ; 21(16): 5488-99, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463831

ABSTRACT

Activated Ras, but not Raf, causes transformation of RIE-1 rat intestinal epithelial cells, demonstrating the importance of Raf-independent effector signaling in mediating Ras transformation. To further assess the contribution of Raf-dependent and Raf-independent function in oncogenic Ras transformation, we evaluated the mechanism by which oncogenic Ras blocks suspension-induced apoptosis, or anoikis, of RIE-1 cells. We determined that oncogenic versions of H-, K-, and N-Ras, as well as the Ras-related proteins TC21 and R-Ras, protected RIE-1 cells from anoikis. Surprisingly, our analyses of Ras effector domain mutants or constitutively activated effectors indicated that activation of Raf-1, phosphatidylinositol 3-kinase (PI3K), or RalGDS alone is not sufficient to promote Ras inhibition of anoikis. Treatment of Ras-transformed cells with the U0126 MEK inhibitor caused partial reversion to an anoikis-sensitive state, indicating that extracellular signal-regulated kinase activation contributes to inhibition of anoikis. Unexpectedly, oncogenic Ras failed to activate Akt, and treatment of Ras-transformed RIE-1 cells with the LY294002 PI3K inhibitor did not affect anoikis resistance or growth in soft agar. Thus, while important for Ras transformation of fibroblasts, PI3K may not be involved in Ras transformation of RIE-1 cells. Finally, inhibition of epidermal growth factor receptor kinase activity did not overcome Ras inhibition of anoikis, indicating that this autocrine loop essential for transformation is not involved in anoikis protection. We conclude that a PI3K- and RalGEF-independent Ras effector(s) likely cooperates with Raf to confer anoikis resistance upon RIE-1 cells, thus underscoring the complex nature by which Ras transforms cells.


Subject(s)
Anoikis/genetics , Cell Transformation, Neoplastic/genetics , Genes, ras/genetics , Phosphatidylinositol 3-Kinases/genetics , Animals , Cell Line , Enzyme Activation/genetics , Gene Expression Regulation , Rats , Signal Transduction/genetics
14.
J Biol Chem ; 276(29): 27145-51, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11373293

ABSTRACT

Leukemia-associated Rho guanine nucleotide exchange factor (LARG) was originally identified as a fusion partner with mixed-lineage leukemia in a patient with acute myeloid leukemia. LARG possesses a tandem Dbl homology and pleckstrin homology domain structure and, consequently, may function as an activator of Rho GTPases. In this study, we demonstrate that LARG is a functional Dbl protein. Expression of LARG in cells caused activation of the serum response factor, a known downstream target of Rho-mediated signaling pathways. Transient overexpression of LARG did not activate the extracellular signal-regulated kinase or c-Jun NH(2)-terminal kinase mitogen-activated protein kinase cascade, suggesting LARG is not an activator of Ras, Rac, or Cdc42. We performed in vitro exchange assays where the isolated Dbl homology (DH) or DH/pleckstrin homology domains of LARG functioned as a strong activator of RhoA, but exhibited no activity toward Rac1 or Cdc42. We found that LARG could complex with RhoA, but not Rac or Cdc42, in vitro, and that expression of LARG caused an increase in the levels of the activated GTP-bound form of RhoA, but not Rac1 or Cdc42, in vivo. Thus, we conclude that LARG is a RhoA-specific guanine nucleotide exchange factor. Finally, like activated RhoA, we determined that LARG cooperated with activated Raf-1 to transform NIH3T3 cells. These data demonstrate that LARG is the first functional Dbl protein mutated in cancer and indicate LARG-mediated activation of RhoA may play a role in the development of human leukemias.


Subject(s)
Cell Transformation, Neoplastic , Guanine Nucleotide Exchange Factors/physiology , Leukemia, Myeloid, Acute/physiopathology , rhoA GTP-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Humans , Male , Nuclear Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors , Serum Response Factor
15.
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
16.
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
17.
Mol Cell Biol ; 21(8): 2847-57, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283263

ABSTRACT

The Rho family of Ras-related proteins, which includes Rac1, RhoA, and Cdc42, is distinguished from other members of the Ras superfamily of small GTPases in that its members possess additional sequences positioned between beta-strand 5 and alpha-helix 4, designated the insert region. Previous studies have established the importance of an intact insert region for the transforming, but not actin cytoskeletal reorganization, activities of Cdc42 and RhoA. Similarly, the insert region was determined to be essential for Rac1-mediated mitogenesis. Additionally, an intact insert region was also determined to be required for the antiapoptotic activity of Rac1 as well as for Rac1 activation of reactive oxygen species and the NF-kappaB transcription factor. However, it has not been determined whether the insert region is important for Rac1-mediated growth transformation. In this study, we assessed the requirement for the insert region in Rac1 transformation and signaling in NIH 3T3 cells. Unexpectedly, we found that a mutant of constitutively activated Rac1 that lacked the insert region retained potent transforming activity. The insert region of Rac1 was dispensable for Rac1 stimulation of transcription from the cyclin D1 promoter and for activation of the c-Jun, NF-kappaB, and E2F-1 transcription factors but was essential for Rac1 induction of serum response factor activity. While an intact insert region was dispensable for inducing reactive oxygen species production in vivo, it was required for Rac1 induction of lamellipodia. When taken together, these results show that the insert region of Rac1 serves roles in regulating actin organization and cell growth that are distinct from those of the analogous regions of Cdc42 and RhoA and support its involvement in regulating specific downstream effector interactions.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Transformation, Neoplastic , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cyclin D1/genetics , DNA Primers/genetics , DNA-Binding Proteins/biosynthesis , E2F Transcription Factors , E2F1 Transcription Factor , Humans , Mice , Molecular Sequence Data , Mutagenesis, Insertional , NF-kappa B/metabolism , Nuclear Proteins/biosynthesis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/metabolism , Reactive Oxygen Species/metabolism , Retinoblastoma-Binding Protein 1 , Serum Response Factor , Transcription Factor DP1 , Transcription Factors/metabolism , Transcription, Genetic , rac1 GTP-Binding Protein/genetics
18.
Methods Enzymol ; 332: 221-32, 2001.
Article in English | MEDLINE | ID: mdl-11305099

ABSTRACT

In conclusion, RDA provides a fast, technically simple, and inexpensive way to characterize genes aberrantly expressed due to Ras transformation. The identification and characterization of these genes may provide insight not only into the mechanism by which Ras causes transformation, but also may identify novel targets for rational drug design and development of anticancer drugs.


Subject(s)
Genes, ras , Genetic Techniques , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Down-Regulation , Gene Expression Regulation , Nucleic Acid Hybridization , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Rats
20.
Mol Med ; 7(12): 816-30, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11844870

ABSTRACT

BACKGROUND: Relatively few genes have been shown to directly affect the metastatic phenotype of breast cancer epithelial cells in vivo. The Rho family of proteins, incluing the Rho, Rac and Cdc42 subfamilies, are related to the small GTP binding protein Ras and regulated diverse biological processes including gene transcription, cytoskeletal organization, cell proliferation and transformation. The effects of Cdc42, Rac and Rho on the actin cytoskeleton suggested a possible role for Rho proteins in cellular motility and metastasis; however, a formal analysis of the role of Rho proteins in breast cancer cellular growth and metastasis in vivo had not previously been performed. MATERIALS AND METHODS: We generated a panel of MTLn3 rat mammary adenocarcinoma cells that expressed similar levels of dominant inhibitory mutants of Cdc42-, Rac- and Rho-dependent signaling, to examine the contribution of these GTPases to cell spreading, guided chemotaxis, and metastasis in vivo. The ability of Rho proteins to regulate intravasation into the peripheral blood was determined by implanting MTLn3 cell stable dominant negative lines in nude mice and measuring the formation of breast cancer cell colonies grown from the peripheral blood. Serial sectioning of the lungs was performed to determine the presence of metastasis in mice in which mammary tumors expressing the dominant negative Rho family proteins had grown to a similar size. RESULTS: Cell spreading of MTLn3 cells was selectively abrogated by N17Rac1. N19RhoA and N17Cdc42 reduced the number of focal contacts (FCs) and disrupted the co-localization of vinculin with phosphotyrosine at FCs. While N17Rac1 and N17Cdc42 preferentially inhibited colony formation in soft agar, all three GTPases affected cell growth in vivo. To distinguish effects on tumorigenicity from intravasation into the bloodstream, implanted tumors were grown to the same size in nude mice. Each dominant inhibitory Rho protein reduced intravasation into the peripheral blood. Lung metastasis of MTLn3 cells was also abrogated by the dominant inhibitory Rho proteins, despite the presence of residual CFU. CONCLUSIONS: These studies demonstrate for the first time a critical role for the Rho GTPases involving independent signaling pathways to limit mammary tumor cellular growth and metastasis in vivo.


Subject(s)
Mammary Neoplasms, Experimental/physiopathology , rho GTP-Binding Proteins/metabolism , Adenocarcinoma , Animals , Cell Division , Chemotaxis , Epidermal Growth Factor/metabolism , Epithelium/physiopathology , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/enzymology , Multigene Family , Rats , Signal Transduction , Stress Fibers , Transfection , Tumor Cells, Cultured , Tyrosine/metabolism , cdc42 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...