Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cells ; 13(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38334664

ABSTRACT

Small G proteins (e.g., Rac1) play critical regulatory roles in islet ß-cell function in health (physiological insulin secretion) and in metabolic stress (cell dysfunction and demise). Multiple regulatory factors for these G proteins, such as GDP dissociation inhibitors (GDIs), have been implicated in the functional regulation of these G proteins. The current set of investigations is aimed at understanding impact of chronic hyperglycemic stress on the expression and subcellular distribution of three known isoforms of RhoGDIs (RhoGDIα, RhoGDIß, and RhoGDIγ) in insulin-secreting ß-cells. The data accrued in these studies revealed that the expression of RhoGDIß, but not RhoGDIα or RhoGDIγ, is increased in INS-1 832/13 cells, rat islets, and human islets. Hyperglycemic stress also promoted the cleavage of RhoGDIß, leading to its translocation to the nuclear compartment. We also report that RhoGDIα, but not RhoGDIγ, is associated with the nuclear compartment. However, unlike RhoGDIß, hyperglycemic conditions exerted no effects on RhoGDIα's association with nuclear fraction. Based on these observations, and our earlier findings of the translocation of Rac1 to the nuclear compartment under the duress of metabolic stress, we conclude that the RhoGDIß-Rac1 signaling module promotes signals from the cytosolic to the nucleus, culminating in accelerated ß-cell dysfunction under metabolic stress.


Subject(s)
Insulin-Secreting Cells , rho Guanine Nucleotide Dissociation Inhibitor beta , Animals , Humans , Rats , GTP-Binding Proteins/metabolism , Insulin-Secreting Cells/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism
2.
Atherosclerosis ; 387: 117391, 2023 12.
Article in English | MEDLINE | ID: mdl-38029612

ABSTRACT

BACKGROUND AND AIMS: The pathological roles and mechanisms of Rho-specific guanine nucleotide dissociation inhibitor 3 (RhoGDI3) in vascular smooth muscle cell (VSMC) phenotypic modulation and neointima formation are currently unknown. This study aimed to investigate how RhoGDI3 regulates the Nod-like receptor protein 3 (NLRP3) inflammasome in platelet-derived growth factor-BB (PDGF-BB)-induced neointima formation. METHODS: For in vitro assays, human aortic VSMCs (HA-VSMCs) were transfected with pcDNA3.1-GDI3 and RhoGDI3 siRNA to overexpress and knockdown RhoGDI3, respectively. HA-VSMCs were also treated with an NLRP3 inhibitor (CY-09) or agonist (NSS). Protein transcription and expression, cell proliferation and migration, Golgi morphology, and protein binding and colocalization were measured. For the in vivo assays, balloon injury (BI) rats were injected with recombinant adenovirus carrying RhoGDI3 shRNA. Carotid arterial morphology, protein expression and colocalization, and activation of the NLRP3 inflammasome were measured. RESULTS: PDGF-BB treatment induced transcription and expression of RhoGDI3 through PDGF receptor αß (PDGFRαß) rather than PDGFRαα or PDGFRßß in HA-VSMCs. RhoGDI3 suppression blocked PDGF-BB-induced VSMC phenotypic transformation. In contrast, RhoGDI3 overexpression further promoted PDGF-BB-induced VSMC dedifferentiation. The in vivo results also confirmed that RhoGDI3 expressed in VSMCs participated in neointima formation and muscle fiber and collagen deposition caused by balloon injury. In addition, PDGF-BB increased binding of RhoGDI3 to NLRP3 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) at the trans-Golgi membrane, which depended on the normal Golgi network. However, recruitment of NLRP3 and ASC to the trans-Golgi network after PDGF-BB treatment was independent of RhoGDI3. Moreover, RhoGDI3 knockdown significantly inhibited ASC expression and NLRP3 inflammasome assembly and activation and reduced NLRP3 protein stability in PDGF-BB-treated HA-VSMCs. Inhibiting NLRP3 effectively prevented PDGF-BB-induced VSMC phenotypic modulation, and an NLRP3 agonist reversed the decline in VSMC phenotypic transformation caused by RhoGDI3 knockdown. Furthermore, RhoGDI3 suppression reduced the protein levels and assembly of NLRP3 and ASC, and the activation of the NLRP3 inflammasome in VSMCs in a rat balloon injury model. CONCLUSIONS: The results of this study reveal a novel mechanism through which RhoGDI3 regulates VSMC phenotypic modulation and neointima formation by activating the NLRP3 inflammasome.


Subject(s)
Inflammasomes , Neointima , Animals , Humans , Rats , Becaplermin/pharmacology , Becaplermin/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Inflammasomes/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Rats, Sprague-Dawley , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism , trans-Golgi Network
3.
Biochemistry ; 60(19): 1533-1551, 2021 05 18.
Article in English | MEDLINE | ID: mdl-33913706

ABSTRACT

There are three RhoGDIs in mammalian cells, which were initially defined as negative regulators of Rho family small GTPases. However, it is now accepted that RhoGDIs not only maintain small GTPases in their inactive GDP-bound form but also act as chaperones for small GTPases, targeting them to specific intracellular membranes and protecting them from degradation. Studies to date with RhoGDIs have usually focused on the interactions between the "typical" or "classical" small GTPases, such as the Rho, Rac, and Cdc42 subfamily members, and either the widely expressed RhoGDI-1 or the hematopoietic-specific RhoGDI-2. Less is known about the third member of the family, RhoGDI-3 and its interacting partners. RhoGDI-3 has a unique N-terminal extension and is found to localize in both the cytoplasm and the Golgi. RhoGDI-3 has been shown to target RhoB and RhoG to endomembranes. In order to facilitate a more thorough understanding of RhoGDI function, we undertook a systematic study to determine all possible Rho family small GTPases that interact with the RhoGDIs. RhoGDI-1 and RhoGDI-2 were found to have relatively restricted activity, mainly binding members of the Rho and Rac subfamilies. RhoGDI-3 displayed wider specificity, interacting with the members of Rho, Rac, and Cdc42 subfamilies but also forming complexes with "atypical" small Rho GTPases such as Wrch2/RhoV, Rnd2, Miro2, and RhoH. Levels of RhoA, RhoB, RhoC, Rac1, RhoH, and Wrch2/RhoV bound to GTP were found to decrease following coexpression with RhoGDI-3, confirming its role as a negative regulator of these small Rho GTPases.


Subject(s)
rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , HEK293 Cells , Humans , Monomeric GTP-Binding Proteins/metabolism , Protein Binding , rho GTP-Binding Proteins/chemistry , rho Guanine Nucleotide Dissociation Inhibitor alpha/physiology , rho Guanine Nucleotide Dissociation Inhibitor beta/physiology , rho Guanine Nucleotide Dissociation Inhibitor gamma/physiology , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/physiology
4.
PLoS One ; 11(11): e0166370, 2016.
Article in English | MEDLINE | ID: mdl-27832197

ABSTRACT

RhoGDI proteins have been implicated in several human cancers; changes in their expression levels have shown pro- or anti-tumorigenic effects. Pancreatic Ductal Adenocarcinoma (PDAC) is a complex pathology, with poor prognosis, and most patients die shortly after diagnosis. Efforts have been focused on understanding the role of RhoGDI's in PDAC, specially, RhoGDI1 and RhoGDI2. However, the role of RhoGDI3 has not been studied in relation to cancer or to PDAC. Here, we characterized the expression and functionality of RhoGDI3 and its target GTPases, RhoG and RhoB in pancreatic cell lines from both normal pancreatic tissue and tissue in late stages of PDAC, and compared them to human biopsies. Through immunofluorescences, pulldown assays and subcellular fractionation, we found a reduction in RhoGDI3 expression in the late stages of PDAC, and this reduction correlates with tumor progression and aggressiveness. Despite the reduction in the expression of RhoGDI3 in PDAC, we found that RhoB was underexpressed while RhoG was overexpressed, suggesting that cancerous cells preserve their capacity to activate this pathway, thus these cells may be more eager to response to the stimuli needed to proliferate and become invasive unlike normal cells. Surprisingly, we found nuclear localization of RhoGDI3 in non-cancerous pancreatic cell line and normal pancreatic tissue biopsies, which could open the possibility of novel nuclear functions for this protein, impacting gene expression regulation and cellular homeostasis.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , rho GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism , rhoB GTP-Binding Protein/metabolism , Cell Line, Tumor , Enzyme Activation , Fluorescent Antibody Technique , Humans , rho GTP-Binding Proteins/analysis , rho Guanine Nucleotide Dissociation Inhibitor gamma/analysis , rhoB GTP-Binding Protein/analysis
5.
J Neurosci Res ; 91(11): 1394-401, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23996536

ABSTRACT

Neural stem cell (NSC) migration relies heavily on the regulation of actin and microtubule cytoskeletons by Rho GTPases, which are critical regulators of key steps during NSC migration. However, the migration mechanism remains unclear. Rho-GDP-dissociation inhibitor-γ (Rho-GDIγ) was identified as an important downregulator of the Rho family of GTPases, because of its ability to prevent nucleotide exchange and thus membrane association. This study investigates the role of Rho-GDIγ in neural stem cells migration. Our results indicate that the overexpression of Rho-GDIγ maintains NSCs in the stem cell state, meanwhile preventing NSC migration through inhibition of Rac1 expression, one of the Rho-family GTPases. This study provides the basis for further study of the molecular mechanism of NSC migration.


Subject(s)
Cell Movement/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/biosynthesis , Animals , Cell Differentiation/physiology , Cell Line , Fluorescent Antibody Technique , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
6.
Mol Neurobiol ; 47(1): 228-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22944911

ABSTRACT

Neural stem cells (NSCs) are promising candidates for a variety of neurological diseases due to their ability to differentiate into neurons, astrocytes, and oligodentrocytes. During this process, Rho GTPases are heavily involved in neuritogenesis, axon formation and dendritic development, due to their effects on the cytoskeleton through downstream effectors. The activities of Rho GTPases are controlled by Rho-GDP dissociation inhibitors (Rho-GDIs). As shown in our previous study, these are also involved in the differentiation of NSCs; however, little is known about the underlying regulatory mechanism. Here, we describe how the transcription factors hepatic nuclear factor (HNF4-1) and myc-associated zinc finger protein (MAZ-1) regulate the expression of Rho-GDIγ in the stimulation of NSC differentiation. Using a transfection of cis-element double-stranded oligodeoxynucleotides (ODNs) strategy, referred to as "decoy" ODNs, we examined the effects of HNF4-1 and MAZ-1 on NSC differentiation in the NSC line C17.2. Our results show that HNF4-1 and MAZ-1 decoy ODNs significantly knock down Rho-GDIγ gene transcription, leading to NSC differentiation towards neurons. We observed that HNF4-1 and MAZ-1 decoy ODNs are able enter to the cell nucleolus and specifically bind to their target transcription factors. Furthermore, the expression of Rho-GDIγ-mediated genes was identified, suggesting that the regulatory mechanism for the differentiation of NSCs is triggered by the transcription factors MAZ-1 and HNF4-1. These findings indicate that HNF4-1 and MAZ-1 regulate the expression of Rho-GDIγ and contribute to the differentiation of NSCs. Our findings provide a new perspective within regulatory mechanism research during differentiation of NSCs, especially the clinical application of transcription factor decoys in vivo, suggesting potential therapeutic strategies for neurodegenerative disease.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Neural Stem Cells/cytology , Transcription Factors/metabolism , Animals , Base Sequence , Cell Proliferation , Cell Shape , Gene Expression Regulation , Gene Knockdown Techniques , Immunohistochemistry , Mice , Models, Biological , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Oligodeoxyribonucleotides/metabolism , Phenotype , Protein Binding , Transcription, Genetic , rho Guanine Nucleotide Dissociation Inhibitor gamma/genetics , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism
7.
Mol Biosyst ; 8(11): 2916-23, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22892720

ABSTRACT

Understanding the molecular mechanism that underlies the differentiation of neural stem cells (NSCs) is vital to develop regenerative medicines for neurological disorders. In our previous work, Rho-GDI-γ was found to be able to prompt neuronal differentiation when it was down regulated. However, it is unclear how Rho-GDI-γ regulates this differentiation process. Therefore, a novel systems biology approach is presented here to identify putative signalling pathways regulated by Rho-GDI-γ during NSC differentiation, and these pathways can provide insights into the NSC differentiation mechanisms. In particular, our proposed approach combines the predictive power of computational biology and molecular experiments. With different biological experiments, the genes in the computationally identified signalling network were validated to be indeed regulated by Rho-GDI-γ during the differentiation of NSCs. In particular, one randomly selected pathway involving Vcp, Mapk8, Ywhae and Ywhah was experimentally verified to be regulated by Rho-GDI-γ. These promising results demonstrate the effectiveness of our proposed systems biology approach, indicating the potential predictive power of integrating computational and experimental approaches.


Subject(s)
Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Systems Biology/methods , rho Guanine Nucleotide Dissociation Inhibitor gamma/pharmacology , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Computational Biology , Fluorescent Antibody Technique , Mice , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Neural Stem Cells/drug effects , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Valosin Containing Protein
8.
J Proteome Res ; 8(8): 3824-33, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19505086

ABSTRACT

Leukocyte integrins are functionally regulated by "inside-out" signaling, meaning that stimulus-induced signaling pathways act on the intracellular integrin tail and induce activation of the receptor at the outside. Both a change in conformation (affinity) and in clustering (avidity/valency) of the receptors has been described to occur. This inside-out signaling is essential for adequate migration of leukocytes to inflammatory sites; however, the exact underlying mechanism is not known. We used two variants of a mouse acute lymphocytic leukemia cell line (L1210), a suspension (L1210-S) and an adherent (L1210-A) variant that were characterized by nonactivated and activated integrins (beta(1), beta(2) and beta(3)), respectively. L1210-S and L1210-A cells were compared on protein expression profiles by two-dimensional fluorescence difference in-gel electrophoresis (2D-DIGE). We found 86 protein spots that were more than 1.25-fold different between L1210-A and L1210-S. Only 4 protein spots were more than 2.5-fold different. We identified 29 proteins by mass spectrometry among which were gelsolin, L-plastin, and Rho GTPase dissociation inhibitor 2. These proteins were upregulated in the L1210-A cells versus L1210-S, which was verified by Western blot analysis. Overexpression of gelsolin in U937 resulted in increased high affinity integrin expression and cell adhesion. Comparison of functionally different cell lines from similar origin by 2D-DIGE might be a successful approach to identify regulatory proteins involved in integrin inside-out control.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Gelsolin/analysis , Guanine Nucleotide Dissociation Inhibitors/analysis , Integrins/analysis , Membrane Glycoproteins/analysis , Microfilament Proteins/analysis , Animals , Cell Adhesion , Cell Line, Tumor , Gelsolin/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Humans , Integrins/metabolism , Leukemia L1210 , Membrane Glycoproteins/metabolism , Mice , Microfilament Proteins/metabolism , Reproducibility of Results , Signal Transduction , U937 Cells , rho Guanine Nucleotide Dissociation Inhibitor gamma
9.
Mol Cell Biochem ; 311(1-2): 233-40, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18273563

ABSTRACT

Rho-GDIgamma belongs to the Rho-GDI protein family, which was observed to have high level expression in the entire brain. Although it exists in neuronal population, its physiological function is poorly understood. This study shows that Rho-GDIgamma is a key factor in the G13 signaling pathway based on an analysis of global gene expression. By using RNAi technology to downregulate expression of Rho-GDIgamma we found distinct morphological changes in neural stem cell line C17.2. More important, RT-PCR confirmed that RNAi-mediated downregulation of Rho-GDIgamma decreased expression of Rho-GDIgamma-regulated genes RhoA, Cdc42, Limk2, and N-WASP and slightly increased expression of Rac1. Further, immunochemical staining indicated that downregulation of Rho-GDIgamma increased the tendency of C17.2 cells to differentiate. These data strongly suggest that Rho-GDIgamma plays a key role in the differentiation of neural stem cells.


Subject(s)
Cell Differentiation/physiology , Guanine Nucleotide Dissociation Inhibitors/metabolism , Neurons/physiology , Stem Cells/physiology , Animals , Base Sequence , Biomarkers/metabolism , Cell Line , Computational Biology , Down-Regulation , Gene Expression Regulation , Molecular Sequence Data , Neurons/cytology , Oligonucleotide Array Sequence Analysis , RNA Interference , Signal Transduction/physiology , Stem Cells/cytology , rho Guanine Nucleotide Dissociation Inhibitor gamma
10.
Brain Res ; 1054(1): 9-21, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16054116

ABSTRACT

Rho-GDIs are a family of Rho GDP-dissociation inhibitors that are critical in modulating the activity of the small GTPases, Cdc42 and RhoA. Two Rho-GDI isoforms are expressed in the brain, Rho-GDIgamma and Rho-GDIalpha. Here, we describe the expression of both of these isoforms in the developing and mature brain. The mRNA expression patterns of Rho-GDIgamma and Rho-GDIalpha were almost identical in the brain with expression in the developing and mature cerebral cortex, striatum, and hippocampus. In addition, we generated mice with targeted deletions of Rho-GDIgamma that are viable and fertile and have no obvious phenotypic abnormalities. Mutant brains looked histologically normal and demonstrated normal patterns of dendritogenesis and neuronal layering as determined by Golgi staining. Mutant mice had normal sleep/wake patterns and sleep EEGs and showed normal hippocampal-dependent learning as assayed by the Morris water maze task. Based on the co-expression of Rho-GDIalpha and Rho-GDIgamma in identical populations of cells in the brain, the lack of phenotype caused by targeted deletion of Rho-GDIgamma may not be surprising given that Rho-GDIalpha may compensate for the loss of Rho-GDIgamma. Whether deletion of both Rho-GDIalpha and Rho-GDIgamma, thereby eliminating all GDI activity in the brain, would produce an observable phenotype remains to be determined.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental/physiology , Guanine Nucleotide Dissociation Inhibitors/deficiency , Guanine Nucleotide Dissociation Inhibitors/metabolism , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal , Blotting, Northern/methods , Brain/embryology , Brain/growth & development , Electroencephalography/methods , Embryo, Mammalian , Guanine Nucleotide Dissociation Inhibitors/genetics , In Situ Hybridization/methods , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout/metabolism , RNA, Messenger/physiology , Reaction Time/genetics , Silver Staining/methods , Sleep/physiology , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho Guanine Nucleotide Dissociation Inhibitor gamma , rho-Specific Guanine Nucleotide Dissociation Inhibitors
11.
Biochem Soc Trans ; 33(Pt 4): 623-6, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16042558

ABSTRACT

rhoGDIs (Rho GDP dissociation inhibitors) are postulated to regulate the activity and the localization of small G-proteins of the Rho family by a shuttling process involving extraction of Rho from donor membranes, formation of inhibitory cytosolic rhoGDI/Rho complexes, and delivery of Rho to target membranes. However, the role of rhoGDIs in site-specific membrane targeting or extraction of Rho is still poorly understood. We investigated here the in vivo functions of two mammalian rhoGDIs: the specific rhoGDI-3 and the well-studied rhoGDI-1 (rhoGDI) after structure-based mutagenesis. We identified two sites in rhoGDIs, forming conserved interactions with their Rho target, whose mutation results in the uncoupling of inhibitory and shuttling functions of rhoGDIs in vivo. Remarkably, these rhoGDI mutants were detected at Rho-induced membrane ruffles or protrusions, where they co-localized with RhoG or Cdc42, probably identifying for the first time the site of extraction of a Rho protein by a rhoGDI in vivo. We propose that these mutations act by modifying the steady-state kinetics of the shuttling process regulated by rhoGDIs, such that transient steps at the cell membranes now become detectable. They should provide valuable tools for future investigations of the dynamics of membrane extraction or delivery of Rho proteins and their regulation by cellular partners.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , rho GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/genetics , Models, Molecular , Mutagenesis , Protein Conformation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/genetics , rho Guanine Nucleotide Dissociation Inhibitor gamma
12.
Trends Cell Biol ; 15(7): 356-63, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15921909

ABSTRACT

The GDP dissociation inhibitors (GDIs) are pivotal regulators of Rho GTPase function. GDIs control the access of Rho GTPases to regulatory guanine nucleotide exchange factors and GTPase-activating proteins, to effector targets and to membranes where such effectors reside. We discuss here our current understanding of how Rho GTPase-GDI complexes are regulated by various proteins, lipids and enzymes that exert GDI displacement activity. We propose that phosphorylation mediated by diverse kinases might provide a means of controlling and coordinating Rho GTPase activation.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Guanine Nucleotide Dissociation Inhibitors/physiology , Protein Kinases/metabolism , rab GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Enzyme Activators/pharmacology , Guanine Nucleotide Exchange Factors/pharmacology , Humans , Lipids/pharmacology , Phosphorylation , Proteins/physiology , Tumor Suppressor Proteins , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/physiology , rho Guanine Nucleotide Dissociation Inhibitor gamma , rho-Specific Guanine Nucleotide Dissociation Inhibitors
13.
Exp Cell Res ; 301(2): 211-22, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15530857

ABSTRACT

Cdc42 is a member of the Rho family of GTPases and plays an important role in the regulation of actin cytoskeletal organization. Activation of Cdc42 and associated signal transduction cascades are dependent upon proper localization of this GTPase. The studies described herein address the hypothesis that Rho GDP-dissociation inhibitor, RhoGDI, plays an essential role in the translocation of Cdc42 to signaling complexes at the plasma membrane and is essential for Cdc42-mediated actin cytoskeletal rearrangements. An activating mutant of Cdc42 that is RhoGDI-binding defective (Cdc42(G12V/R66E)) is characterized and used as a tool to study the functional importance of the Cdc42-RhoGDI interaction. Overexpression of mycCdc42(G12V/R66E) in COS-7 cells results in actin cytoskeletal rearrangements that are indistinguishable from those stimulated by overexpression of mycCdc42(G12V). In addition, the G12V activating mutant of Cdc42 was overexpressed in mesangial cells that are null for RhoGDI expression. MycCdc42(G12V) stimulation of filopodia formation in these cells was indistinguishable from that observed in wild-type mesangial cells. Taken together, the results presented herein indicate that although RhoGDI is a critical regulator of guanine nucleotide binding, cycling of Cdc42 between membranes and the cytosol and cellular transformation, it is not essential for Cdc42-mediated organization of the actin cytoskeleton.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Mutation/physiology , cdc42 GTP-Binding Protein/physiology , Adherens Junctions/metabolism , Animals , Cell Line , Guanine Nucleotide Dissociation Inhibitors/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Transport , Pseudopodia/metabolism , Recombinant Proteins , Transfection , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma
14.
Eur J Biochem ; 262(3): 879-89, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10411652

ABSTRACT

Rho GTPases have two interconvertible forms and two cellular localizations. In their GTP-bound conformation, they bind to the cell membrane and are activated. In the inactive GDP-bound conformation, they associate with a cytosolic protein called GDP dissociation inhibitor (GDI). We previously reported that the RhoA component of the RhoA/Rho-GDI complex was not accessible to the Clostridium botulinum C3 ADP-ribosyl transferase, unless the complex had been incubated with phosphoinositides. We show here that PtdIns, PtdIns4P, PtdIns3,4P2, PtdIns4,5P2 and PtdInsP3 enhance not only the C3-dependent ADP-ribosylation, but also the GDP/GTP exchange in the RhoA component of the prenylated RhoA/Rho-GDI complex. In contrast, in the nonprenylated RhoA/Rho-GDI complex, the levels of ADP-ribosylation and GDP/GTP exchange are of the same order as those measured on free RhoA and are not modified by phosphoinositides. In both cases, phosphoinositides partially opened, but did not fully dissociate the complex. Upon treatment of the prenylated RhoA/Rho-GDI complex with phosphoinositides, a GTP-dependent transfer to neutrophil membranes was evidenced. Using an overlay assay with the prenylated RhoA/Rho-GDI complex pretreated with PtdIns4P and labeled with [alpha32P]GTP, three membrane proteins with molecular masses between 26 and 32 kDa were radiolabeled. We conclude that in the presence of phosphoinositides, the prenylated RhoA/Rho-GDI complex partially opens, which allows RhoA to exchange GDP for GTP. The opened GTP-RhoA/Rho-GDI complex acquires the capacity to target specific membrane proteins.


Subject(s)
GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors , Hemiterpenes , Pentanes , Phosphatidylinositols/metabolism , rho GTP-Binding Proteins , Adenosine Diphosphate/metabolism , Butadienes/metabolism , Enzyme Activation , GTP-Binding Proteins/chemistry , Guanosine Triphosphate/metabolism , Humans , Macromolecular Substances , Membrane Proteins/metabolism , Phosphorus Radioisotopes/metabolism , Protein Prenylation , Ribonucleosides/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma , rhoA GTP-Binding Protein
15.
Genomics ; 53(1): 104-9, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9787082

ABSTRACT

GDP-dissociation inhibitors (GDIs) play a primary role in modulating the activity of GTPases. We recently reported the identification of a new GDI for the Rho-related GTPases named RhoGDIgamma. This gene is now designated ARHGDIG by HUGO. Here, in a detailed analysis of tissue expression of ARHGDIG, we observe high levels in the entire brain, with regional variations. The mRNA is also present at high levels in kidney and pancreas and at moderate levels in spinal cord, stomach, and pituitary gland. In other tissues examined, the mRNA levels are very low (lung, trachea, small intestine, colon, placenta) or undetectable. RT-PCR analysis of total RNA isolated from exocrine pancreas and islets shows that the gene is expressed in both tissues. We also report the genomic structure of ARHGDIG. The gene spans over 4 kb and is organized into six exons and five introns. The upstream region lacks a canonical TATA box and contains several putative binding sites for ubiquitous and tissue-specific factors active in central nervous system development. Using FISH, we have mapped the gene to chromosome band 16p13.3. This band is rich in deletion mutants of genes involved in several human diseases, notably polycystic kidney disease, alpha-thalassemia, tuberous sclerosis, mental retardation, and cancer. The promoter structure and the chromosomal location of RhoGDIgamma suggest its importance and underscore the need for further investigation into its biology.


Subject(s)
Chromosomes, Human, Pair 16/genetics , GTP-Binding Proteins/genetics , Guanine Nucleotide Dissociation Inhibitors , rho GTP-Binding Proteins , Base Sequence , Chromosome Mapping , Cloning, Molecular , Gene Expression Regulation, Developmental/genetics , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , rho Guanine Nucleotide Dissociation Inhibitor gamma
16.
Proc Natl Acad Sci U S A ; 94(9): 4279-84, 1997 Apr 29.
Article in English | MEDLINE | ID: mdl-9113980

ABSTRACT

GDP-dissociation inhibitors (GDIs) play a primary role in modulating the activation of GTPases and may also be critical for the cellular compartmentalization of GTPases. RhoGDI and GDI/D4 are two currently known GDIs for the Rho-subfamily of GTPases. Using their cDNAs to screen a human brain cDNA library under low stringency, we have cloned a homologous cDNA preferentially expressed at high levels in brain and pancreas. The predicted protein, named RhoGDIgamma, is approximately 50% identical to GDI/D4 and RhoGDI. It binds to CDC42 and RhoA with less affinity compared with RhoGDI and does not bind with Rac1, Rac2, or Ras. RhoGDIgamma functions as a GDI for CDC42 but with approximately 20 times less efficiency than RhoGDI. Immunohistochemical studies showed a diffuse punctate distribution of the protein in the cytoplasm with concentration around the nucleus in cytoplasmic vesicles. Overexpression of the protein in baby hamster kidney cells caused the cells to round up with loss of stress fibers. A distinct hydrophobic amino terminus in RhoGDIgamma, not seen in the other two RhoGDIs, could provide a mechanism for localization of the GDI to specific membranous compartment thus determining function distinct from RhoGDI or GDI/D4. Our results provide evidence that there is a family of GDIs for the Rho-related GTPases and that they differ in binding affinity, target specificity, and tissue expression. We propose that RhoGDI be renamed RhoGDIalpha and GDID4 be renamed RhoGDIbeta. The new GDI should widen the scope of investigation of this important class of regulatory protein.


Subject(s)
Brain/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors , Guanosine Diphosphate/metabolism , Pancreas/metabolism , rho GTP-Binding Proteins , Amino Acid Sequence , Base Sequence , Cell Compartmentation , Cloning, Molecular , DNA, Complementary/genetics , Fluorescent Antibody Technique , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/genetics , GTP-Binding Proteins/isolation & purification , Humans , Molecular Sequence Data , Protein Binding , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , rho Guanine Nucleotide Dissociation Inhibitor gamma
17.
J Biol Chem ; 271(48): 30366-74, 1996 Nov 29.
Article in English | MEDLINE | ID: mdl-8939998

ABSTRACT

RhoB is a small GTP-binding protein highly homologous to the RhoA protein. While RhoA is known to regulate the assembly of focal adhesions and stress fibers in response to growth factors, the function of RhoB remains unknown. We have reported that the transient expression of the endogenous RhoB protein is regulated during the cell cycle, contrasting with the permanent RhoA protein expression (). Using the yeast two-hybrid system to characterize proteins interacting with RhoB, we identified a new mouse Rho GDP dissociation inhibitor, referenced as RhoGDI-3. The NH2-terminal alpha helix of RhoGDI-3 is strongly amphipatic and differs thus from that found in previously described bovine, human, and yeast RhoGDI proteins and mouse and human D4/Ly-GDIs. Contrary to the cytosolic localization of all known GDI proteins, acting on Rab or Rho, RhoGDI-3 is associated to a Triton X-100-insoluble membranous or cytoskeletal subcellular fraction. In the two-hybrid system, RhoGDI-3 interacts specifically with GDP- and GTP-bound forms of post-translationally processed RhoB and RhoG proteins, both of which show a growth-regulated expression in mammalian cells. No interaction is found with RhoA, RhoC, or Rac1 proteins. We show that GDI-3 is able to inhibit GDP/GTP exchange of RhoB and to release GDP-bound but not GTP-bound RhoB from cell membranes.


Subject(s)
GTP Phosphohydrolases , GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors , Membrane Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 16 , Cloning, Molecular , GTP-Binding Proteins/genetics , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Membranes/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , RNA, Messenger/genetics , Saccharomyces cerevisiae , Sequence Alignment , Tissue Distribution , rho GTP-Binding Proteins , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho Guanine Nucleotide Dissociation Inhibitor gamma , rho-Specific Guanine Nucleotide Dissociation Inhibitors , rhoB GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...