Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
2.
J Immunol ; 164(10): 5369-74, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10799900

ABSTRACT

The RhoA GTPase is involved in regulating actin cytoskeletal organization, gene expression, cell proliferation, and survival. We report here that p115-RhoGEF, a specific guanine nucleotide exchange factor (GEF) and activator of RhoA, modulates HIV-1 replication. Ectopic expression of p115-RhoGEF or Galpha13, which activates p115-RhoGEF activity, leads to inhibition of HIV-1 replication. RhoA activation is required and the inhibition affects HIV-1 gene expression. The RhoA effector activity in inhibiting HIV-1 replication is genetically separable from its activities in transformation of NIH3T3 cells, activation of serum response factor, and actin stress fiber formation. These findings reveal that the RhoA signal transduction pathway regulates HIV-1 replication and suggest that RhoA inhibits HIV-1 replication via a novel effector activity.


Subject(s)
HIV-1/physiology , Virus Replication/physiology , rhoA GTP-Binding Protein/physiology , Amino Acid Sequence , Antiviral Agents/metabolism , Antiviral Agents/physiology , Cell Line , Gene Expression Regulation/physiology , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/physiology , HIV-1/genetics , HeLa Cells , Humans , Molecular Sequence Data , Rho Guanine Nucleotide Exchange Factors , Signal Transduction/genetics , Signal Transduction/physiology , Virus Replication/genetics , rhoA GTP-Binding Protein/genetics
3.
Oncogene ; 18(26): 3831-45, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10445846

ABSTRACT

The small Ras-related GTPase, TC10, has been classified on the basis of sequence homology to be a member of the Rho family. This family, which includes the Rho, Rac and CDC42 subfamilies, has been shown to regulate a variety of apparently diverse cellular processes such as actin cytoskeletal organization, mitogen-activated protein kinase (MAPK) cascades, cell cycle progression and transformation. In order to begin a study of TC10 biological function, we expressed wild type and various mutant forms of this protein in mammalian cells and investigated both the intracellular localization of the expressed proteins and their abilities to stimulate known Rho family-associated processes. Wild type TC10 was located predominantly in the cell membrane (apparently in the same regions as actin filaments), GTPase defective (75L) and GTP-binding defective (31N) mutants were located predominantly in cytoplasmic perinuclear regions, and a deletion mutant lacking the carboxyl terminal residues required for post-translational prenylation was located predominantly in the nucleus. The GTPase defective (constitutively active) TC10 mutant: (1) stimulated the formation of long filopodia; (2) activated c-Jun amino terminal kinase (JNK); (3) activated serum response factor (SRF)-dependent transcription; (4) activated NF-kappaB-dependent transcription; and (5) synergized with an activated Raf-kinase (Raf-CAAX) to transform NIH3T3 cells. In addition, wild type TC10 function is required for full H-Ras transforming potential. We demonstrate that an intact effector domain and carboxyl terminal prenylation signal are required for proper TC10 function and that TC10 signals to at least two separable downstream target pathways. In addition, TC10 interacted with the actin-binding and filament-forming protein, profilin, in both a two-hybrid cDNA library screen, and an in vitro binding assay. Taken together, these data support a classification of TC10 as a member of the Rho family, and in particular, suggest that TC10 functions to regulate cellular signaling to the actin cytoskeleton and processes associated with cell growth.


Subject(s)
Contractile Proteins , GTP Phosphohydrolases/physiology , Mitogen-Activated Protein Kinases , Signal Transduction/physiology , rho GTP-Binding Proteins , 3T3 Cells , Amino Acid Sequence , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins/chemistry , Cell Division , Cell Size , Cell Transformation, Neoplastic , Chlorocebus aethiops , DNA, Complementary/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation , GTP Phosphohydrolases/classification , GTP Phosphohydrolases/deficiency , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/chemistry , JNK Mitogen-Activated Protein Kinases , Mice , Microfilament Proteins/metabolism , Molecular Sequence Data , Multigene Family , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Profilins , Protein Binding , Recombinant Fusion Proteins/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Serum Response Factor , Transcription, Genetic , cdc42 GTP-Binding Protein , rac GTP-Binding Proteins , rhoA GTP-Binding Protein
4.
J Biol Chem ; 274(3): 1423-31, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9880516

ABSTRACT

Ha-Ras undergoes post-translational modifications (including attachment of farnesyl and palmitate) that culminate in localization of the protein to the plasma membrane. Because palmitate is not attached without prior farnesyl addition, the distinct contributions of the two lipid modifications to membrane attachment or biological activity have been difficult to examine. To test if palmitate is able to support these crucial functions on its own, novel C-terminal mutants of Ha-Ras were constructed, retaining the natural sites for palmitoylation, but replacing the C-terminal residue of the CAAX signal for prenylation with six lysines. Both the Ext61L and ExtWT proteins were modified in a dynamic fashion by palmitate, without being farnesylated; bound to membranes modestly (40% as well as native Ha-Ras); and retained appropriate GTP binding properties. Ext61L caused potent transformation of NIH 3T3 cells and, unexpectedly, an exaggerated differentiation of PC12 cells. Ext61L with the six lysines but lacking palmitates was inactive. Thus, farnesyl is not needed as a signal for palmitate attachment or removal, and a combination of transient palmitate modification and basic residues can support Ha-Ras membrane binding and two quite different biological functions. The roles of palmitate can therefore be independent of and distinct from those of farnesyl. Reciprocally, if membrane association can be sustained largely through palmitates, farnesyl is freed to interact with other proteins.


Subject(s)
Palmitic Acid/metabolism , Protein Prenylation , ras Proteins/metabolism , 3T3 Cells , Animals , Cell Differentiation , Cell Membrane/metabolism , Cysteine/metabolism , DNA, Complementary/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Mice , PC12 Cells , Rats , Structure-Activity Relationship , Transfection , ras Proteins/genetics
5.
Oncogene ; 13(9): 1901-9, 1996 Nov 07.
Article in English | MEDLINE | ID: mdl-8934536

ABSTRACT

Although it is well-established that modification of Ras by farnesol is a critical step for its membrane association and transforming activity, the contribution of other C-terminal sequences and palmitate modification to Ras localization and function remains unclear. We have characterized H-Ras mutant proteins with alterations in the palmitoylated cysteines or in sequences flanking these residues. We found that non-palmitoylated proteins were impaired not only in membrane association but also in transforming activity. Mutations which drastically altered residues adjacent to the palmitoylated cysteine did not abolish palmitoylation. However, despite continued lipid modification the mutant proteins failed to bind to plasma membranes and instead accumulated on internal membranes and, importantly, were not transforming. Addition of an N-terminal myristoylation signal to these defective mutants, or to proteins entirely lacking the C-terminal 25 residues restored both plasma membrane association and transforming activity. Thus, H-Ras does not absolutely require prenylation or palmitoylation nor indeed its hypervariable domain in order to interact with effectors that ultimately cause transformation. However, in this native state, the C-terminus appears to provide a combination of lipids and a previously unrecognized signal for specific plasma membrane targeting that are essential for the correct localization and biological function of H-Ras.


Subject(s)
Cell Membrane/metabolism , Cell Transformation, Neoplastic/genetics , ras Proteins/genetics , ras Proteins/metabolism , Animals , Binding Sites , Cell Membrane/ultrastructure , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Farnesol/chemistry , Farnesol/metabolism , Genes, ras , Mice , Mutation , Myristic Acids/metabolism , Palmitates/chemistry , Palmitates/metabolism , Sensitivity and Specificity
6.
Mol Cell Biol ; 16(7): 3923-33, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668210

ABSTRACT

Substantial evidence supports a critical role for the activation of the Raf-1/MEK/mitogen-activated protein kinase pathway in oncogenic Ras-mediated transformation. For example, dominant negative mutants of Raf-1, MEK, and mitogen-activated protein kinase all inhibit Ras transformation. Furthermore, the observation that plasma membrane-localized Raf-1 exhibits the same transforming potency as oncogenic Ras suggests that Raf-1 activation alone is sufficient to mediate full Ras transforming activity. However, the recent identification of other candidate Ras effectors (e.g., RalGDS and phosphatidylinositol-3 kinase) suggests that activation of other downstream effector-mediated signaling pathways may also mediate Ras transforming activity. In support of this, two H-Ras effector domain mutants, H-Ras(12V, 37G) and H-Ras(12V, 40C), which are defective for Raf binding and activation, induced potent tumorigenic transformation of some strains of NIH 3T3 fibroblasts. These Raf-binding defective mutants of H-Ras induced a transformed morphology that was indistinguishable from that induced by activated members of Rho family proteins. Furthermore, the transforming activities of both of these mutants were synergistically enhanced by activated Raf-1 and inhibited by the dominant negative RhoA(19N) mutant, indicating that Ras may cause transformation that occurs via coordinate activation of Raf-dependent and -independent pathways that involves Rho family proteins. Finally, cotransfection of H-Ras(12V, 37G) and H-Ras(12V, 40C) resulted in synergistic cooperation of their focus-forming activities, indicating that Ras activates at least two Raf-independent, Ras effector-mediated signaling events.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Transformation, Neoplastic , Genes, ras , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3 Cells , Animals , GTP-Binding Proteins/metabolism , Humans , Kinetics , Luciferases/metabolism , MAP Kinase Kinase 4 , Mice , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , Recombinant Proteins/metabolism , Transfection
7.
J Biol Chem ; 271(14): 7992-8, 1996 Apr 05.
Article in English | MEDLINE | ID: mdl-8626480

ABSTRACT

Overexpression of Neu (ErbB-2/HER2) is found in approximately 20% of breast tumors. Activation of Neu by a point mutation (NeuT) causes constitutive tyrosine kinase activity of this transmembrane receptor and transforming activity in fibroblasts. To identify downstream targets of Neu, we have analyzed the ability of Neu to activate gene expression. Expression of NeuT, but not normal Neu, caused transcriptional activation of Ets, AP-1, or NF-kappaB-dependent reporter genes. Dominant inhibitory Ras or Raf mutants blocked the Neu-mediated transcriptional activation, confirming that Ras signaling pathways were required for this activation. Analysis with Ets2 mutants indicated that activation of Ets2 transcriptional activity mediated by NeuT or oncogenic Ras required phosphorylation of the same Ets2 residue, threonine 72. Cotransfection of dominant inhibitory Ets2 mutants specifically blocked NeuT-mediated activation of Ets-dependent reporter genes. Furthermore, in focus formation assays using NIH 3T3 cells, the transforming activity of NeuT was inhibited 5-fold when NeuT was cotransfected with a dominant negative Ets2 mutant. However, parallel colony formation assays showed that the Ets2 dominant negative mutant did not inhibit the growth of normal cells. Together, these data show that NeuT activates a variety of transcription factor families via the Ras signaling pathway and that Ets activation is required for NeuT-mediated cellular transformation. Thus, downstream targets of Neu, including Ets transcription factors, may be useful points for therapeutic intervention in Neu/ErbB-2-associated cancers.


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptor, ErbB-2/physiology , Repressor Proteins , Trans-Activators/physiology , Transcription Factors , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Regulation, Neoplastic , Mice , Molecular Sequence Data , NF-kappa B/physiology , Proteins/physiology , Proto-Oncogene Protein c-ets-2 , Proto-Oncogene Proteins p21(ras)/physiology , Signal Transduction , TNF Receptor-Associated Factor 3 , Transcription Factor AP-1/metabolism , Transcription, Genetic
8.
J Biol Chem ; 271(11): 6497-501, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8626452

ABSTRACT

Mitogen-activated protein (MAP) kinases can be grouped into three structural families, ERK, JNK, and p38, which are thought to carry out unique functions within cells. We demonstrate that ERK, JNK, and p38 are activated by distinct combinations of stimuli in T cells that simulate full or partial activation through the T cell receptor. These kinases are regulated by reversible phosphorylation on Tyr and Thr, and the dual specific phosphatases PAC1 and MKP-1 previously have been implicated in the in vivo inactivation of ERK or of ERK and JNK, respectively. Here we characterize a new MAP kinase phosphatase, MKP-2, that is induced in human peripheral blood T cells with phorbol 12-myristate 13-acetate and is expressed in a variety of nonhematopoietic tissues as well. We show that the in vivo substrate specificities of individual phosphatases are unique. PAC1, MKP-2, and MKP-1 recognize ERK and p38, ERK and JNK, and ERK, p38, and JNK, respectively. Thus, individual MAP kinase phosphatases can differentially regulate the potential for cross-talk between the various MAP kinase pathways. A hyperactive allele of ERK2 (D319N), analogous to the Drosophila sevenmaker gain-of-function mutation, has significantly reduced sensitivity to all three MAP kinase phosphatases in vivo.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins , Immediate-Early Proteins/metabolism , Mitogen-Activated Protein Kinases , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/metabolism , Animals , Cell Line , Drosophila Proteins , Dual Specificity Phosphatase 1 , Dual Specificity Phosphatase 2 , Dual-Specificity Phosphatases , Enzyme Induction , Female , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase Phosphatases , Mutation , Protein Phosphatase 1 , Protein Phosphatase 2 , Protein Tyrosine Phosphatases/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , Tetradecanoylphorbol Acetate/pharmacology , Tissue Distribution
9.
Mol Reprod Dev ; 42(4): 468-76, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8607978

ABSTRACT

Members of the Ras superfamily of proteins function as regulated GDP/GTP switches that cycle between active GTP-complexed and inactive GDP-complexed states. Guanine nucleotide exchange factors (GEFs) stimulate formation of the GTP-bound state, whereas GTPase activating proteins (GAPs) catalyze the formation of the GDP-bound state. We describe three studies that evaluate the mechanism of action of GEFs for Ras (SOS1 and RasGRF/CDC25) or Ras-related Rho (Dbl and Vav) proteins. Growth factor-mediated activation of Ras is believed to be mediated by activation of Ras GEFs (CDC25/GRF and SOS1/2). Although the mechanisms of Ras GEF regulation are unclear, recent studies suggest that translocation of SOS1 to the plasma membrane, where Ras is located, might be responsible for Ras activation. Our observation that the addition of the Ras plasma membrane-targeting sequence to the catalytic domains of CDC25 and SOS1 greatly enhanced their transforming and transactivation activities (10-50 fold and 5-10 fold, respectively) suggests that membrane translocation alone is sufficient to potentiate GEF activation of Ras. We have determined that two Ras-related proteins, designated R-Ras and R-Ras2/TC21, can trigger the malignant transformation of NIH 3T3 cells via activation of the Ras signal transduction pathway. Furthermore, like Ras and R-Ras, we observed that TC21 GTPase activity was stimulated by Ras GAPs. However, we observed that both SOS1 and CDC25 were activators of normal TC21, but not R-Ras, transforming activities. Therefore, TC21, but not R-Ras, may be activated by the same extracellular signaling events that activate Ras proteins. Dbl family proteins are believed to function as GEFs and activators of the Ras-related Rho family of proteins. However, one Dbl family oncogene, designated Vav, has been reported to be a GEF for Ras proteins. Therefore we were interested in determining whether Dbl family oncogenes cause transformation by triggering the constitutive activation of Rho or Ras proteins. Our results suggest that Dbl oncogenes cause transformation via a Ras-independent activation of MAP kinases and Rho family proteins.


Subject(s)
Proteins/metabolism , Signal Transduction , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Guanine Nucleotide Exchange Factors , Humans , Molecular Sequence Data , ras Guanine Nucleotide Exchange Factors , ras-GRF1
10.
Mol Cell Biol ; 15(11): 6443-53, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7565796

ABSTRACT

Although substantial evidence supports a critical role for the activation of Raf-1 and mitogen-activated protein kinases (MAPKs) in oncogenic Ras-mediated transformation, recent evidence suggests that Ras may activate a second signaling pathway which involves the Ras-related proteins Rac1 and RhoA. Consequently, we used three complementary approaches to determine the contribution of Rac1 and RhoA function to oncogenic Ras-mediated transformation. First, whereas constitutively activated mutants of Rac1 and RhoA showed very weak transforming activity when transfected alone, their coexpression with a weakly transforming Raf-1 mutant caused a greater than 35-fold enhancement of transforming activity. Second, we observed that coexpression of dominant negative mutants of Rac1 and RhoA reduced oncogenic Ras transforming activity. Third, activated Rac1 and RhoA further enhanced oncogenic Ras-triggered morphologic transformation, as well as growth in soft agar and cell motility. Finally, we also observed that kinase-deficient MAPKs inhibited Ras transformation. Taken together, these data support the possibility that oncogenic Ras activation of Rac1 and RhoA, coupled with activation of the Raf/MAPK pathway, is required to trigger the full morphogenic and mitogenic consequences of oncogenic Ras transformation.


Subject(s)
Cell Transformation, Neoplastic , GTP-Binding Proteins/physiology , Proto-Oncogene Proteins p21(ras)/physiology , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cell Movement , Cytoskeleton/ultrastructure , Enzyme Activation , Genes, Dominant , Humans , Mice , Point Mutation , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-raf , Signal Transduction , rac GTP-Binding Proteins , rhoA GTP-Binding Protein
11.
Oncogene ; 10(12): 2289-96, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7784077

ABSTRACT

We demonstrate that Rho, a regulator of cytoskeletal actin, is necessary for Ras transformation. A dominant inhibitory Rho gene (RhoBN19) specifically suppressed Rat1 cell focus formation induced by oncogenic Ras but not by Raf. An activated Rho gene (RhoBV14) lacked focus formation activity but augmented the focus formation activity of both oncogenes. NIH3T3 cell lines expressing RhoBV14 grew to higher saturation density and displayed reduced serum and anchorage requirements for growth. We concluded that Rho played a role in cell growth regulation and was required for transformation by oncogenic Ras but not Raf. A model for Ras signal transduction proposing separate Rho-dependent and Raf-dependent pathways is discussed.


Subject(s)
Cell Transformation, Neoplastic/genetics , GTP-Binding Proteins/physiology , Genes, ras/physiology , Signal Transduction , ras Proteins/physiology , 3T3 Cells , Animals , Base Sequence , Cell Line, Transformed , Cell Transformation, Neoplastic/pathology , Mice , Molecular Sequence Data , Oncogene Proteins/physiology , Rats
13.
Methods Enzymol ; 250: 435-54, 1995.
Article in English | MEDLINE | ID: mdl-7651170

ABSTRACT

Covalent attachment of lipids appears to be an important mechanism by which many proteins interact with membranes. As we learn more about how lipids and adjacent amino acids participate in addressing proteins to specific membranes within the cell, it should be possible to design more elegant and precise membrane targeting systems that can be used to guide proteins to functionally relevant destinations.


Subject(s)
Myristic Acids/metabolism , Palmitic Acids/metabolism , Protein Processing, Post-Translational , Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , GTP-Binding Proteins/metabolism , Genetic Vectors , Mammals , Molecular Sequence Data , Mutagenesis, Insertional , Myristic Acid , Oligodeoxyribonucleotides , Palmitic Acid , Polymerase Chain Reaction/methods , Protein Prenylation , Protein Sorting Signals/metabolism , Protein-Tyrosine Kinases/metabolism , Restriction Mapping , ras Proteins/biosynthesis
14.
Mol Cell Biol ; 14(10): 6848-57, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7935402

ABSTRACT

Vav and Dbl are members of a novel class of oncogene proteins that share significant sequence identity in a approximately 250-amino-acid domain, designated the Dbl homology domain. Although Dbl functions as a guanine nucleotide exchange factor (GEF) and activator of Rho family proteins, recent evidence has demonstrated that Vav functions as a GEF for Ras proteins. Thus, transformation by Vav and Dbl may be a consequence of constitutive activation of Ras and Rho proteins, respectively. To address this possibility, we have compared the transforming activities of Vav and Dbl with that of the Ras GEF, GRF/CDC25. As expected, GRF-transformed cells exhibited the same reduction in actin stress fibers and focal adhesions as Ras-transformed cells. In contrast, Vav- and Dbl-transformed cells showed the same well-developed stress fibers and focal adhesions observed in normal or RhoA(63L)-transformed NIH 3T3 cells. Furthermore, neither Vav- or Dbl-transformed cells exhibited the elevated levels of Ras-GTP (60%) observed with GRF-transformed cells. Finally, GRF, but not Vav or Dbl, induced transcriptional activation from Ras-responsive DNA elements (ets/AP-1, fos promoter, and kappa B). However, like Ras- and GRF-transformed cells, both Vav- and Dbl-transformed cells exhibited constitutively activated mitogen-activated protein kinases (MAPKs) (primarily p42MAPK/ERK2). Since kinase-deficient forms of p42MAPK/ERK2 and p44MAPK/ERK1 inhibited Dbl transformation, MAPK activation may be an important component of its transforming activity. Taken together, our observations indicate that Vav and Dbl transformation is not a consequence of Ras activation and instead may involve the constitutive activation of MAPKs.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Transformation, Neoplastic , Drosophila Proteins , Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Retroviridae Proteins, Oncogenic/metabolism , 3T3 Cells , Actins/metabolism , Actins/ultrastructure , Animals , Cell Adhesion , Guanine Nucleotide Exchange Factors , Guanosine Triphosphate/metabolism , Membrane Proteins/metabolism , Mice , Oncogene Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-vav , Retroviridae Proteins, Oncogenic/genetics , Signal Transduction , ras Guanine Nucleotide Exchange Factors , ras-GRF1
15.
J Biol Chem ; 268(16): 11548-52, 1993 Jun 05.
Article in English | MEDLINE | ID: mdl-8505289

ABSTRACT

Although the heterotrimeric Gi alpha subunit terminates in an apparent CXXX prenylation signal (CGLF), it is not modified by isoprenylation. To determine if the Gi alpha CXXX sequence can signal prenylation when placed at the carboxyl termini of normally prenylated proteins, we have characterized the processing and biological activity of chimeric oncogenic Ras proteins that terminate in the Gi alpha CXXX sequence (Ras/Gi alpha). Surprisingly, these chimeras were prenylated both in vivo and in vitro, demonstrated significant membrane association, exhibited transforming activity, and induced transcriptional transactivation from Ras-responsive elements. We then extended these studies to determine if, unlike the CC or CXC carboxyl-terminal sequences of other Rab proteins, the carboxyl-terminal CXXX sequences of the Ras-related Rab5 and Rab11 proteins represent conventional CXXX prenylation signals that can support Ras processing and transforming activity. Unexpectedly, these Ras/Rab chimeras were nonprenylated, were cytosolic, and lacked detectable transforming or transcriptional transactivation activity. Taken together, these results suggest that the context within which a CXXX sequence occurs may also critically control the modification of a protein by prenylation, and that the Rab5 and Rab11 carboxyl termini do not possess conventional CXXX sequences. Instead, their CCXX and CCXXX motifs may represent additional classes of protein prenylation signals.


Subject(s)
Cell Transformation, Neoplastic , GTP-Binding Proteins/metabolism , Genes, ras , Mevalonic Acid/analogs & derivatives , Protein Processing, Post-Translational , 3T3 Cells , Amino Acid Sequence , Animals , Cysteine/metabolism , GTP-Binding Proteins/genetics , Methionine/metabolism , Mevalonic Acid/metabolism , Mice , Molecular Sequence Data , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/isolation & purification , Proto-Oncogene Proteins p21(ras)/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Sulfur Radioisotopes , Transfection , Tritium , rab5 GTP-Binding Proteins
16.
Mol Cell Biol ; 11(3): 1523-30, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1899909

ABSTRACT

Although the Rap1A protein resembles the oncogenic Ras proteins both structurally and biochemically, Rap1A exhibits no oncogenic properties. Rather, overexpression of Rap1A can reverse Ras-induced transformation of NIH 3T3 cells. Because the greatest divergence in amino acid sequence between Ras and Rap1A occurs at the COOH terminus, the role of this domain in the opposing biological activities of these proteins was examined. COOH-terminal processing and membrane association of Rap1A were studied by constructing and expressing a chimeric protein (composed of residues 1 to 110 of an H-Ras activated by a Leu-61 mutation attached to residues 111 to 184 of Rap1A) in NIH 3T3 cells and a full-length human Rap1A protein in a baculovirus-Sf9 insect cell system. Both the chimeric protein and the full-length protein were synthesized as a 23-kDa cytosolic precursor that rapidly bound to membranes and was converted into a 22-kDa form that incorporated label derived from [3H]mevalonate. The mature 22-kDa form also contained a COOH-terminal methyl group. Full-length Rap1A, expressed in insect cells, was modified by a C20 (geranylgeranyl) isoprenoid. In contrast, H-Ras, expressed in either Sf9 insect or NIH 3T3 mouse cells contained a C15 (farnesyl) group. This suggests that the Rap1A COOH terminus is modified by a prenyl transferase that is distinct from the farnesyl transferase that modifies Ras proteins. Nevertheless, in NIH 3T3 cells the chimeric Ras:Rap1A protein retained the transforming activity conferred by the NH2-terminal Ras61L domain. This demonstrates that the modifications and localization signals of the COOH terminus of Rap1A can support the interactions between H-Ras and membranes that are required for transformation.


Subject(s)
GTP-Binding Proteins/physiology , Membrane Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/physiology , Animals , Cells, Cultured , Cloning, Molecular , DNA Mutational Analysis , In Vitro Techniques , Methylation , Moths , Palmitates/metabolism , Protein Processing, Post-Translational , Recombinant Fusion Proteins , Structure-Activity Relationship , Terpenes/metabolism , rap GTP-Binding Proteins
17.
Proc Natl Acad Sci U S A ; 87(21): 8511-5, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2236060

ABSTRACT

Covalent attachment of myristic acid (C14:0) to the NH2-terminal glycine residue of a number of cellular, viral, and oncogene-encoded proteins is essential for full expression of their biological function. Substitution of oxygen for methylene groups in this fatty acid does not produce a significant change in chain length or stereochemistry but does result in a reduction in hydrophobicity. These heteroatom-containing analogs serve as alternative substrates for mammalian myristoyl-CoA:protein N-myristoyltransferase (EC 2.3.1.97) and offer the opportunity to explore structure/function relationships of myristate in N-myristoyl proteins. We have synthesized three tritiated analogs of myristate with oxygen substituted for methylene groups at C6, C11, and C13. Metabolic labeling studies were performed with these compounds and (i) a murine myocyte cell line (BC3H1), (ii) a rat fibroblast cell that produces p60v-src (3Xsrc), or (iii) NIH 3T3 cells that have been engineered to express a fusion protein consisting of an 11-residue myristoylation signal from the Rasheed sarcoma virus (RaSV) gag protein linked to c-Ha-ras with a Cys----Ser-186 mutation. This latter mutation prevents isoprenylation and palmitoylation of ras. Two-dimensional gel electrophoresis of membrane and soluble fractions prepared from cell lysates revealed different patterns of incorporation of the analogs into cellular N-myristoyl proteins (i.e., protein-sequence-specific incorporation). In addition, proteins were identified that underwent redistribution from membrane to soluble fractions after incorporating one but not another analog (analog-specific redistribution). Comparable studies using the model RaSV-ras chimeric protein also demonstrated analog-specific differences in incorporation, varying from approximately 25% of the total RaSV-ras chimeric protein with 5-octyloxypentanoate to greater than 50% with 12-methoxydodecanoate. Modification by this latter compound was so extensive that the amount of membrane-associated N-myristoylated protein was decreased. Incorporation of each of the analogs caused a dramatic redistribution to the soluble fraction, comparable to that seen when myristoylation was completely blocked by mutating the protein's site of myristate attachment (glycine) to an alanine residue. The demonstration that these analogs differ in the extent to which they are incorporated and in their ability to cause redistribution of any single protein suggests that they may also have sufficient selectivity to be of potential therapeutic value.


Subject(s)
Myristic Acids/metabolism , Protein Biosynthesis , Animals , Cell Line , Electrophoresis, Gel, Two-Dimensional , Membrane Proteins/isolation & purification , Mice , Molecular Weight , Myristic Acids/chemical synthesis , Protein Processing, Post-Translational , Proteins/genetics , Proteins/isolation & purification , Sulfur Radioisotopes , Tritium
18.
Proc Natl Acad Sci U S A ; 87(8): 3042-6, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2183224

ABSTRACT

Oncogenic forms of ras proteins are synthesized in the cytosol and must become membrane associated to cause malignant transformation. Palmitic acid and an isoprenoid (farnesol) intermediate in cholesterol biosynthesis are attached to separate cysteine residues near the C termini of H-ras, N-ras, and Kirsten-ras (K-ras) exon 4A-encoded proteins. These lipid modifications have been suggested to promote or stabilize the association of ras proteins with membranes. Because preventing isoprenylation also prevents palmitoylation, examining the importance of isoprenylation alone has not been possible. However, the oncogenic human [Val12]K-ras 4B protein is not palmitoylated but is isoprenylated, membrane associated, and fully transforming. We therefore constructed mutant [Val12]K-ras 4B proteins that were not isoprenylated to examine the effects of isoprenylation in the absence of palmitoylation. The nonisoprenylated mutant proteins both failed to associate with membranes and did not transform NIH 3T3 cells. In addition, inhibition of isoprenoid and cholesterol synthesis with the drug compactin also decreased [Val12]K-ras 4B protein isoprenylation and membrane association. These results unequivocally demonstrate that isoprenylation, rather than palmitoylation, is essential for ras membrane binding and ras transforming activity. These findings clearly indicate the biological significance of ras protein modification by farnesol and suggest that this modification may be important for facilitating the processing, trafficking, and biological activity of other isoprenylated proteins. Because K-ras is the most frequently activated oncogene in a wide spectrum of human malignancies, study of this pathway could lead to important therapeutic treatments.


Subject(s)
Cell Transformation, Neoplastic , Exons , Farnesol/pharmacology , Proto-Oncogene Proteins/metabolism , Animals , Anticholesteremic Agents/pharmacology , Cell Line , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Genetic Vectors , Humans , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Mevalonic Acid/pharmacology , Mice , Mutation , Oligonucleotide Probes , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Transfection
19.
Proc Natl Acad Sci U S A ; 86(21): 8323-7, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2682646

ABSTRACT

Association of oncogenic ras proteins with cellular membranes appears to be a crucial step in transformation, ras is synthesized as a cytosolic precursor, which is processed to a mature form that localizes to the plasma membrane. This processing involves, in part, a conserved sequence, Cys-Ali-Ali-Xaa (in which Ali is an amino acid with an aliphatic side chain and Xaa is any amino acid), at the COOH terminus of ras proteins. Yeast a-factor mating hormone precursor also possesses a COOH-terminal Cys-Ali-Ali-Xaa sequence. However, while the COOH-terminal cysteine has been implicated as a site of palmitoylation of ras proteins, in mature a-type mating factor this residue is modified by an isoprenoid, a farnesyl moiety. We asked whether the Cys-Ali-Ali-Xaa sequence signaled different modifications for the yeast peptides (farnesylation) than for ras proteins (palmitoylation) or whether ras proteins were similar to the mating factors and contained a previously undiscovered isoprenoid. We report here that the processing of ras proteins involves addition of a farnesyl moiety, apparently at the COOH-terminal cysteine analogous to the cysteine modified in the yeast peptides, and that farnesylation may be important for membrane association and transforming activity of ras proteins.


Subject(s)
Farnesol/metabolism , Genes, ras , Mevalonic Acid/metabolism , Oncogene Protein p21(ras)/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Mice , Molecular Sequence Data , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/isolation & purification , Transfection
20.
Biochem Soc Trans ; 17(5): 867-9, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2695362

ABSTRACT

p21ras is palmitoylated on a cysteine residue near the C-terminus. Changing Cys-186 to Ser in oncogenic forms produces a non-palmitoylated protein that fails to associate with membranes and does not transform NIH 3T3 cells. To examine whether palmitate acts in a general way to increase ras protein hydrophobicity, or is involved in more specific interactions between p21ras and membranes, we constructed genes that encode non-palmitoylated ras proteins containing myristic acid at their N-termini. Myristoylated, activated ras, without palmitate (61Leu/186Ser) exhibited both efficient membrane association and full transforming activity. Unexpectedly, we found that myristoylated forms of normal cellular ras were also potently transforming. Myristoylated c-ras retained the high GTP binding and GTPase characteristic of the cellular protein and, moreover, bound predominantly GDP in vivo. This implied that it continued to interact with GAP (GTPase-activating protein). While the membrane binding induced by myristate permitted transformation, only palmitate produced a normal (non-transforming) association of ras with membranes and must therefore regulate ras function by some unique property that myristate does not mimic. Myristoylation thus represents a novel mechanism by which the ras proto-oncogene protein can become transforming.


Subject(s)
Cell Membrane/metabolism , Myristates/metabolism , Myristic Acids/metabolism , Oncogene Protein p21(ras)/physiology , Acylation , Animals , Cell Line , Mice , Palmitates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...