Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 297(2): 100982, 2021 08.
Article in English | MEDLINE | ID: mdl-34293347

ABSTRACT

NADPH oxidase 2 (NOX2) produces the superoxide anion radical (O2-), which has functions in both cell signaling and immune defense. NOX2 is a multimeric-protein complex consisting of several protein subunits including the GTPase Rac. NOX2 uniquely facilitates an oxidative burst, which is described by initially slow O2- production, which increases over time. The NOX2 oxidative burst is considered critical to immune defense because it enables expedited O2- production in response to infections. However, the mechanism of the initiation and progression of this oxidative burst and its implications for regulation of NOX2 have not been clarified. In this study, we show that the NOX2 oxidative burst is a result of autoactivation of NOX2 coupled with the redox function of Rac. NOX2 autoactivation begins when active Rac triggers NOX2 activation and the subsequent production of O2-, which in turn activates redox-sensitive Rac. This activated Rac further activates NOX2, amplifying the feedforward cycle and resulting in a NOX2-mediated oxidative burst. Using mutagenesis-based kinetic and cell analyses, we show that enzymatic activation of Rac is exclusively responsible for production of the active Rac trigger that initiates NOX2 autoactivation, whereas redox-mediated Rac activation is the main driving force of NOX2 autoactivation and contributes to generation of ∼98% of the active NOX2 in cells. The results of this study provide insight into the regulation of NOX2 function, which could be used to develop therapeutics to control immune responses associated with dysregulated NOX2 oxidative bursts.


Subject(s)
NADPH Oxidase 2/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism , rac GTP-Binding Proteins/metabolism , Cell Line , Cell Line, Tumor , Enzyme Activation , Humans , NADPH Oxidase 2/immunology , Oxidation-Reduction , Signal Transduction
2.
Small GTPases ; 12(1): 44-59, 2021 01.
Article in English | MEDLINE | ID: mdl-30983499

ABSTRACT

Son of Sevenless (SOS), one of guanine nucleotide exchange factors (GEFs), activates Ras. We discovered that the allosteric domain of SOS yields SOS to proceed a previously unrecognized autoactivation kinetics. Its essential feature is a time-dependent acceleration of SOS feedback activation with a reaction initiator or with the priming of active Ras. Thus, this mechanistic autoactivation feature explains the notion, previously only conjectured, of accelerative SOS activation followed by the priming of active Ras, an action produced by another GEF Ras guanyl nucleotide-releasing protein (RasGRP). Intriguingly, the kinetic transition from gradual RasGRP activation to accelerative SOS activation has been interpreted as an analog to digital conversion; however, from the perspective of autoactivation kinetics, it is a process of straightforward RasGRP-mediated SOS autoactivation. From the viewpoint of allosteric protein cooperativity, SOS autoactivation is a unique time-dependent cooperative SOS activation because it enables an active SOS to accelerate activation of other SOS as a function of time. This time-dependent SOS cooperativity does not belong to the classic steady-state protein cooperativity, which depends on ligand concentration. Although its hysteretic or sigmoid-like saturation curvature is a classic hallmark of steady-state protein cooperativity, its hyperbolic saturation figure typically represents protein noncooperativity. We also discovered that SOS autoactivation perturbs the previously predicted hysteresis of SOS activation in a steady state to produce a hyperbolic saturation curve. We interpret this as showing that SOS allostery elicits, through SOS autoactivation, cooperativity uniquely time-dependent but not ligand concentration dependent.


Subject(s)
Son of Sevenless Proteins
3.
J Biol Chem ; 295(39): 13651-13663, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32753483

ABSTRACT

Ras family proteins play an essential role in several cellular functions, including growth, differentiation, and survival. The mechanism of action of Ras mutants in Costello syndrome and cancers has been identified, but the contribution of Ras mutants to Noonan syndrome, a genetic disorder that prevents normal development in various parts of the body, is unknown. Son of Sevenless (SOS) is a Ras guanine nucleotide exchange factor. In response to Ras-activating cell signaling, SOS autoinhibition is released and is followed by accelerative allosteric feedback autoactivation. Here, using mutagenesis-based kinetic and pulldown analyses, we show that Noonan syndrome Ras mutants I24N, T50I, V152G, and D153V deregulate the autoactivation of SOS to populate their active form. This previously unknown process has been linked so far only to the development of Noonan syndrome. In contrast, other Noonan syndrome Ras mutants-V14I, T58I, and G60E-populate their active form by deregulation of the previously documented Ras GTPase activities. We propose a novel mechanism responsible for the deregulation of SOS autoactivation, where I24N, T50I, V152G, and D153V Ras mutants evade SOS autoinhibition. Consequently, they are capable of forming a complex with the SOS allosteric site, thus aberrantly promoting SOS autoactivation, resulting in the population of active Ras mutants in cells. The results of this study elucidate the molecular mechanism of the Ras mutant-mediated development of Noonan syndrome.


Subject(s)
Noonan Syndrome/metabolism , Son of Sevenless Proteins/metabolism , Allosteric Site , HEK293 Cells , Humans , Kinetics , Models, Molecular , Mutation , Noonan Syndrome/genetics , Son of Sevenless Proteins/chemistry
4.
J Biol Chem ; 291(26): 13699-714, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27189938

ABSTRACT

6-Thiopurine (6-TP) prodrugs include 6-thioguanine and azathioprine. Both are widely used to treat autoimmune disorders and certain cancers. This study showed that a 6-thioguanosine triphosphate (6-TGTP), converted in T-cells from 6-TP, targets Rac1 to form a disulfide adduct between 6-TGTP and the redox-sensitive GXXXXGK(S/T)C motif of Rac1. This study also showed that, despite the conservation of the catalytic activity of RhoGAP (Rho-specific GAP) on the 6-TGTP-Rac1 adduct to produce the biologically inactive 6-thioguanosine diphosphate (6-TGDP)-Rac1 adduct, RhoGEF (Rho-specific GEF) cannot exchange the 6-TGDP adducted on Rac1 with free guanine nucleotide. The biologically inactive 6-TGDP-Rac1 adduct accumulates in cells because of the ongoing combined actions of RhoGEF and RhoGAP. Because other Rho GTPases, such as RhoA and Cdc42, also possess the GXXXXGK(S/T)C motif, the proposed mechanism for the inactivation of Rac1 also applies to RhoA and Cdc42. However, previous studies have shown that CD3/CD28-stimulated T-cells contain more activated Rac1 than other Rho GTPases such as RhoA and Cdc42. Accordingly, Rac1 is the main target of 6-TP in activated T-cells. This explains the T-cell-specific Rac1-targeting therapeutic action of 6-TP that suppresses the immune response. This proposed mechanism for the action of 6-TP on Rac1 performs a critical role in demonstrating the capability to design a Rac1-targeting chemotherapeutic agent(s) for autoimmune disorders. Nevertheless, the results also suggest that the targeting action of other Rho GTPases in other organ cells, such as RhoA in vascular cells, may be linked to cytotoxicities because RhoA plays a key role in vasculature functions.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Immunosuppressive Agents/pharmacokinetics , Prodrugs/pharmacology , Thionucleosides/pharmacokinetics , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism , Amino Acid Motifs , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , GTPase-Activating Proteins/metabolism , Humans , Rho Guanine Nucleotide Exchange Factors/metabolism , cdc42 GTP-Binding Protein/metabolism
5.
Biochemistry ; 55(3): 543-59, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26765051

ABSTRACT

Embryonic Ras (ERas)--a new subset of Ras proteins--are characterized by a unique p-loop residue, unique Switch II residues, and an unusual extended N-terminus. When expressed, both murine and human ERas are highly populated in their GTP-bound forms. The expression of murine ERas is linked to the development of murine embryonic cells, and the expression of human ERas is correlated to certain human cancers. Mutation-based kinetic analyses, in combination with assessments of the kinetic parameter-based calculation of the fraction of the GTP-bound active form of ERas proteins, explain the kinetic mechanism that produces the unprecedented hyperactive ERas. The ERas-specific p-loop residue contributes ERas proteins to intrinsically populate their GTP-bound form in cells. Furthermore, the ERas-specific Switch II residues block the catalytic action of p120GAP on ERas proteins. This blockage sustains the previously mentioned GTP-bound ERas proteins. In essence, the combined work of the ERas-specific p-loop and Switch II residues populates the exceedingly high GTP-bound form of ERas in cells. This study also rules out any kinetic function of the unique ERas-specific N-terminus in the production of the hyperactive GTP-bound ERas in cells. The biological role of this N-terminus remains uninvestigated. Intriguingly, the ERas-specific p-loop residue matches the mutated Ser residue of the Costello Syndrome G12S HRas mutant that also intrinsically populates its GTP-bound form in cells. However, because the effector protein of ERas differs from that of G12S HRas, this kinetic similarity does not confer on ERas biological and/or pathophysiological similarity to G12S HRas.


Subject(s)
ras Proteins/metabolism , Animals , Embryo, Mammalian , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Kinetics , Mice , Mutation , NIH 3T3 Cells , Protein Binding , p120 GTPase Activating Protein/metabolism , ras Proteins/chemistry , ras Proteins/genetics
6.
Biochemistry ; 53(3): 518-32, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24422478

ABSTRACT

Ras and Rho GTPases are molecular switches for various vital cellular signaling pathways. Overactivation of these GTPases often causes development of cancer. Guanine nucleotide exchange factors (GEFs) and oxidants function to upregulate these GTPases through facilitation of guanine nucleotide exchange (GNE) of these GTPases. However, the effect of oxidants on GEF functions, or vice versa, has not been known. We show that, via targeting Ras Cys(51), an oxidant inhibits the catalytic action of Cdc25-the catalytic domain of RasGEFs-on Ras. However, the enhancement of Ras GNE by an oxidant continues regardless of the presence of Cdc25. Limiting RasGEF action by an oxidant may function to prevent the pathophysiological overactivation of Ras in the presence of both RasGEFs and oxidants. The continuous exposure of Ras to nitric oxide and its derivatives can form S-nitrosated Ras (Ras-SNO). This study also shows that an oxidant not only inhibits the catalytic action of Cdc25 on Ras-SNO but also fails to enhance Ras-SNO GNE. This lack of enhancement then populates the biologically inactive Ras-SNO in cells, which may function to prevent the continued redox signaling of the Ras pathophysiological response. Finally, this study also demonstrates that, unlike the case with RasGEFs, an oxidant does not inhibit the catalytic action of RhoGEF-Vav or Dbs-on Rho GTPases such as Rac1, RhoA, RhoC, and Cdc42. This result explains the results of the previous study in which, despite the presence of an oxidant, the catalytic action of Dbs in cells continued to enhance RhoC GNE.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Superoxides/pharmacology , ras Guanine Nucleotide Exchange Factors/metabolism , rho GTP-Binding Proteins/metabolism , Humans , Kinetics , Oxidants/pharmacology , ras-GRF1/antagonists & inhibitors
7.
Biochemistry ; 52(47): 8465-79, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24224811

ABSTRACT

Costello syndrome is linked to activating mutations of a residue in the p-loop or the NKCD/SAK motifs of Harvey Ras (HRas). More than 10 HRas mutants that induce Costello syndrome have been identified; G12S HRas is the most prevalent of these. However, certain HRas p-loop mutations also are linked to cancer formation that are exemplified with G12V HRas. Despite these relations, specific links between types of HRas mutations and diseases evade definition because some Costello syndrome HRas p-loop mutations, such as G12S HRas, also often cause cancer. This study established novel kinetic parameter-based equations that estimate the value of the cellular fractions of the GTP-bound active form of HRas mutant proteins. Such calculations differentiate between two basic kinetic mechanisms that populate the GTP-bound form of Ras in cells. (i) The increase in the level of GTP-bound Ras is caused by the HRas mutation-mediated perturbation of the intrinsic kinetic characteristics of Ras. This generates a broad spectrum of the population of the GTP-bound form of HRas that typically causes Costello syndrome. The upper end of this spectrum of HRas mutants, as exemplified by G12S HRas, can also cause cancer. (ii) The increase in the level of GTP-bound Ras occurs because the HRas mutations perturb the action of p120GAP on Ras. This causes production of a significantly high population of the only GTP-bound form of HRas linked merely to cancer formation. HRas mutant G12V belongs to this category.


Subject(s)
Costello Syndrome/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Biocatalysis , Costello Syndrome/genetics , Costello Syndrome/metabolism , Enzyme Activation , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Kinetics , Mice , Molecular Sequence Data , Mutant Proteins/metabolism , NIH 3T3 Cells , Protein Structure, Secondary , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Recombinant Proteins/metabolism , Signal Transduction , p120 GTPase Activating Protein/genetics , p120 GTPase Activating Protein/metabolism , ras-GRF1/genetics , ras-GRF1/metabolism
8.
Biochemistry ; 50(25): 5731-42, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21568348

ABSTRACT

Our study showed that a combination of 6-thiopurine (6-TP) drugs and a redox agent effectively inhibits the motility of SUM cells derived from human inflammatory breast cancer (IBC) cells and RhoC-overexpressed mammary epithelium cells. This 6-TP-mediated inhibition of cell motility occurs because the treated 6-TPs target and inactivate RhoC. A molecular mechanism for inactivation by the 6-TP-mediated RhoC is proposed by which treated TPs are converted in cells into 6-thioguanosine phosphate (6-TGNP). This 6-TGNP in turn reacts with the Cys(20) side chain of the redox-sensitive GXXXCGK(S/T)C motif of RhoC to produce a 6-TGNP-RhoC disulfide adduct. A redox agent synergistically enhances the formation process of this disulfide. The adduct that is formed impedes RhoC guanine nucleotide exchange, which populates an inactive RhoC. Our results suggest that 6-TGNP can also react with the redox-sensitive GXXXCGK(S/T)C and GXXXXGK(S/T)C motif of RhoA and Rac, respectively, to produce a 6-TGNP-RhoA and 6-TGNP-Rac disulfide adduct. However, given that RhoC has been shown to be overexpressed in ∼90% of IBC lesions, the populated RhoC but not other Rho proteins is likely to be a primary target for 6-TPs and a redox agent to terminate the metastasis of IBC.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Migration Inhibition/drug effects , Inflammation Mediators/chemistry , Inflammation Mediators/therapeutic use , Mercaptopurine/chemistry , Mercaptopurine/therapeutic use , Neoplasm Invasiveness/prevention & control , Breast Neoplasms/chemistry , Cell Line, Tumor , Cell Migration Inhibition/physiology , Crystallography, X-Ray , Drug Delivery Systems/methods , Drug Synergism , Female , Gene Silencing/drug effects , Guanine Nucleotides , Humans , Neoplasm Invasiveness/pathology , Oxidation-Reduction/drug effects , Prodrugs/chemistry , Prodrugs/therapeutic use , Thionucleotides , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/biosynthesis , rho GTP-Binding Proteins/genetics , rhoC GTP-Binding Protein
9.
Antioxid Redox Signal ; 14(4): 689-724, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20649471

ABSTRACT

Small GTPases, including the proto-oncoprotein Ras and Rho GTPases, are involved in various cellular signaling events. Some of these small GTPases are redox sensitive, including Ras, Rho, Ran, Dexras1, and Rhes GTPases. Thus, the redox-mediated regulation of these GTPases often determines the course of their cellular signaling cascades. This article takes into consideration the application of Marcus theory to potential redox-based molecular mechanisms in the regulation of these redox-sensitive GTPases and the relevance of such mechanisms to a specific redox-sensitive motif. The discussion also takes into account various diseases, including cancers, heart, and neuronal disorders, that are often linked with the dysregulation of the redox signaling cascades associated with these redox-sensitive GTPases.


Subject(s)
GTP Phosphohydrolases/metabolism , Animals , GTP Phosphohydrolases/genetics , Humans , Oxidation-Reduction/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
10.
Biochemistry ; 49(18): 3965-76, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20377193

ABSTRACT

Thiopurine drugs are commonly used in the treatment of certain cancers, autoimmune disorders, organ transplant rejection, and bowel disease. Because long-term treatment with thiopurines for certain diseases is common, the cytotoxic effects associated with chronic exposure to thiopurine drugs are inevitable. The results shown in this study indicate that the oncogenic Ras in model cancer cell lines forms a complex with thioguanine nucleotide that is derived from long-term treatment with thiopurines. This study also showed that the Ras thioguanine nucleotide binary complex is likely to be a direct target of a redox agent, resulting in downregulation of the oncogenic Ras. This study proposes a radical-based molecular mechanism for the path of Ras-targeting thiopurines used in conjunction with redox agents. Given that Ras plays a central role in cellular signaling pathways, any interference with Ras activity by thiopurines and redox agents has the potential for devastating cytotoxic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/metabolism , Reactive Nitrogen Species/metabolism , Thioguanine/pharmacology , ras Proteins/antagonists & inhibitors , Cell Line, Tumor , Humans , Neoplasms/drug therapy , Oxidation-Reduction , Protein Binding , ras Proteins/metabolism
11.
Toxicol Appl Pharmacol ; 242(3): 333-43, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19909765

ABSTRACT

Protein thiol modifications including cysteinylation (CSSP) and glutathionylation (GSSP) in erythrocytes of rat treated with diamide have been reported, but mechanism and origin of CSSP formation are unknown. Experiments were performed to relate CSSP formation to GSH hydrolysis via gamma-glutamyltranspeptidase (gamma-GT) and know whether cysteine may act as deglutathionylation factor. Time-dependent variations of redox forms of glutathione and cysteine were investigated in erythrocytes, plasma, liver and kidney of diamide-treated rats (0.4 mmol/kg by infusion for 45 min followed by 135 min of washout) in the presence and absence of acivicin (10 mg/kg administered twice 1 h before diamide) a known gamma-GT inhibitor. Diamide-treated rats showed decreased concentrations of erythrocyte GSH and increased levels of GSSP and CSSP. The rate of CSSP formation was slower than that of GSSP. Besides the entity of CSSP accumulation of erythrocytes was high and equivalent to approximately 3-fold of the normal plasma content of total cysteine. The result was paradoxically poorly related to gamma-GT activity because the gamma-GT inhibition only partially reduced erythrocyte CSSP. After gamma-GT inhibition, a large concentration fluctuation of glutathione (increased) and cysteine (decreased) was observed in plasma of diamide-treated rats, while little changes were seen in liver and kidney. There were indications from in vitro experiments that the CSSP accumulation in erythrocytes of diamide-treated rats derives from the coexistence of GSH hydrolysis via gamma-GT and production of reduced cysteine via plasma thiol exchanges. Moreover, reduced cysteine was found to be involved in deglutathionylation processes. Mechanisms of protein glutathionylation by diamide and deglutathionylation by cysteine were proposed.


Subject(s)
Cysteine/metabolism , Diamide/pharmacology , Sulfhydryl Compounds/metabolism , gamma-Glutamyltransferase/metabolism , Animals , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Erythrocytes/metabolism , Glutathione/metabolism , Isoxazoles/pharmacology , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Sulfhydryl Reagents/pharmacology , Time Factors , gamma-Glutamyltransferase/antagonists & inhibitors
12.
Biochem Biophys Res Commun ; 376(3): 568-72, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18796295

ABSTRACT

Ran, a small Ras-like GTP-binding nuclear protein, plays a key role in modulation of various cellular signaling events including the cell cycle. This study shows that a cellular redox agent (nitrogen dioxide) facilitates Ran guanine nucleotide dissociation, and identifies a unique Ran redox architecture involved in that process. Sequence analysis suggests that Dexras1 and Rhes GTPases also possess the Ran redox architecture. As Ran releases an intact nucleotide, the redox regulation mechanism of Ran is likely to differ from the radical-based guanine nucleotide modification mechanism suggested for Ras and Rho GTPases. These results provide a mechanistic reason for the previously observed oxidative stress-induced perturbation of the Ran-mediated nuclear import, and suggest that oxidative stress could be a factor in the regulation of cell signal transduction pathways associated with Ran.


Subject(s)
Nitrogen Dioxide/metabolism , ran GTP-Binding Protein/metabolism , Cysteine/chemistry , Cysteine/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Nitrogen Dioxide/chemistry , Oxidation-Reduction , Oxidative Stress , ran GTP-Binding Protein/chemistry , ran GTP-Binding Protein/genetics
13.
Biochemistry ; 45(48): 14481-9, 2006 Dec 05.
Article in English | MEDLINE | ID: mdl-17128987

ABSTRACT

We have previously shown that redox agents including superoxide anion radical and nitrogen dioxide can react with GXXXXGK(S/T)C motif-containing GTPases (i.e., Rac1, Cdc42, and RhoA) to stimulate guanine nucleotide release. We now show that the reaction of RhoA with redox agents leads to different functional consequences from that of Rac1 and Cdc42 due to the presence of an additional cysteine (GXXXCGK(S/T)C) in the RhoA redox-active motif. While reaction of redox agents with RhoA stimulates guanine nucleotide dissociation, RhoA is subsequently inactivated through formation of an intramolecular disulfide that prevents guanine nucleotide binding thereby causing RhoA inactivation. Thus, redox agents may function to downregulate RhoA activity under conditions that stimulate Rac1 and Cdc42 activity. The opposing functions of these GTPases may be due in part to their differential redox regulation. In addition, the results presented herein suggest that the platinated-chemotherapeutic agent, cisplatin, which is known for targeting nucleic acids, reacts with RhoA to produce a RhoA thiol-cisplatin-thiol adduct, leading to inactivation of RhoA. Similarly, certain arsenic complexes (i.e., arsenate and arsenic trioxide) may inactivate RhoA by bridging the cysteine residues in the GXXXCGK(S/T)C motif. Thus, in addition to redox agents, platinated-chemotherapeutic agents and arsenic complexes may modulate the activity of GTPases containing the GXXXCGK(S/T)C motif (i.e., RhoA and RhoB).


Subject(s)
rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Catalysis , Cisplatin/pharmacology , Guanine Nucleotides/metabolism , Kinetics , Mass Spectrometry , Molecular Sequence Data , Mutation/genetics , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Oxidation-Reduction/drug effects , Protein Binding , rhoA GTP-Binding Protein/genetics
14.
Biochemistry ; 45(7): 2200-10, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16475808

ABSTRACT

Ras GTPases cycle between inactive GDP-bound and active GTP-bound states to modulate a diverse array of processes involved in cellular growth control. We have previously shown that both NO/O(2) (via nitrogen dioxide, (*)NO(2)) and superoxide radical anion (O(2)(*)(-)) promote Ras guanine nucleotide dissociation. We now show that hydrogen peroxide in the presence of transition metals (i.e., H(2)O(2)/transition metals) and peroxynitrite also trigger radical-based Ras guanine nucleotide dissociation. The primary redox-active reaction species derived from H(2)O(2)/transition metals and peroxynitrite is O(2)(*)(-) and (*)NO(2), respectively. A small fraction of hydroxyl radical (OH(*)) is also present in both. We also show that both carbonate radical (CO(3)(*)(-)) and (*)NO(2), derived from the mixture of peroxynitrite and bicarbonate, facilitate Ras guanine nucleotide dissociation. We further demonstrate that NO/O(2) and O(2)(*)(-) promote Ras GDP exchange with GTP in the presence of a radical-quenching agent, ascorbate, or NO, and generation of Ras-GTP promotes high-affinity binding of the Ras-binding domain of Raf-1, a downstream effector of Ras. S-Nitrosylated Ras (Ras-SNO) can be formed when NO serves as a radical-quenching agent, and hydroxyl radical but not (*)NO(2) or O(2)(*)(-) can further react with Ras-SNO to modulate Ras activity in vitro. However, given the lack of redox specificity associated with the high redox potential of OH(*), it is unclear whether this reaction occurs under physiological conditions.


Subject(s)
Reactive Nitrogen Species/pharmacology , Reactive Oxygen Species/pharmacology , ras Proteins/metabolism , Copper/pharmacology , Ferrous Compounds/pharmacology , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Hydrogen Peroxide/pharmacology , Peroxynitrous Acid/pharmacology , Proto-Oncogene Proteins c-raf/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism
15.
J Biol Chem ; 280(35): 31003-10, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-15994296

ABSTRACT

Rho GTPases regulate multiple cellular processes including actin cytoskeletal rearrangements, transcriptional regulation, and oxidant production. The studies described herein demonstrate that small molecule redox agents, in addition to protein regulatory factors, can regulate the activity of redox-active Rho GTPases. A novel (GXXXXGK(S/T)C) motif, conserved in a number of Rho GTPases, appears critical for redox-mediated guanine nucleotide dissociation in vitro. A detailed molecular mechanism for redox regulation of GXXXXGK(S/T)C motif-containing Rho GTPases is proposed.


Subject(s)
Amino Acid Motifs , Guanine Nucleotides/metabolism , rho GTP-Binding Proteins/metabolism , Guanine Nucleotides/chemistry , Molecular Structure , Molecular Weight , Oxidation-Reduction , Reactive Nitrogen Species , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/genetics
16.
Biochemistry ; 44(17): 6573-85, 2005 May 03.
Article in English | MEDLINE | ID: mdl-15850391

ABSTRACT

Vav proteins are Rho GTPase-specific guanine nucleotide exchange factors (GEFs) that are distinguished by the tandem arrangement of Dbl homology (DH), Pleckstrin homology (PH), and cysteine rich domains (CRD). Whereas the tandem DH-PH arrangement is conserved among Rho GEFs, the presence of the CRD is unique to Vav family members and is required for efficient nucleotide exchange. We provide evidence that Vav2-mediated nucleotide exchange of Rho GTPases follows the Theorell-Chance mechanism in which the Vav2.Rho GTPase complex is the major species during the exchange process and the Vav2.GDP-Mg(2+).Rho GTPase ternary complex is present only transiently. The GTPase specificity for the DH-PH-CRD Vav2 in vitro follows this order: Rac1 > Cdc42 > RhoA. Results obtained from fluorescence anisotropy and NMR chemical shift mapping experiments indicate that the isolated Vav1 CRD is capable of directly associating with Rac1, and residues K116 and S83 that are in the proximity of the P-loop and the guanine base either are part of this binding interface or undergo a conformational change in response to CRD binding. The NMR studies are supported by kinetic measurements on Rac1 mutants S83A, K116A, and K116Q and Vav2 CRD mutant K533A in that these mutants affect both the initial binding event of Vav2 with Rac1 (k(on)) and the rate-limiting dissociation of Vav2 from the Vav2.Rac1 binary complex (thereby influencing the enzyme turnover number, k(cat)). The results suggest that the CRD domain in Vav proteins plays an active role, affecting both the k(on) and the k(cat) for Vav-mediated nucleotide exchange on Rho GTPases.


Subject(s)
Cell Cycle Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , Oncogene Proteins/physiology , Proto-Oncogene Proteins/physiology , rho GTP-Binding Proteins/metabolism , Amino Acid Sequence , Blood Proteins/metabolism , Catalysis , Cell Cycle Proteins/isolation & purification , Cysteine/metabolism , Enzyme Activation , Fluorescence Polarization , Guanine Nucleotide Exchange Factors/isolation & purification , Humans , Kinetics , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Oncogene Proteins/isolation & purification , Phosphoproteins/metabolism , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Proto-Oncogene Proteins/isolation & purification , Proto-Oncogene Proteins c-vav , Retroviridae Proteins, Oncogenic/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
17.
J Biol Chem ; 280(13): 12438-45, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15684418

ABSTRACT

Ras GTPases cycle between inactive GDP-bound and active GTP-bound states to modulate a diverse array of processes involved in cellular growth control. The activity of Ras is up-regulated by cellular agents, including both protein (guanine nucleotide exchange factors) and redox-active agents (nitric oxide (NO) and superoxide anion radical (O2*). We have recently elucidated the mechanism by which NO promotes guanine nucleotide dissociation of redox-active NKCD motif-containing Ras and Ras-related GTPases. In this study, we show that guanine nucleotide dissociation is enhanced upon exposure of the redox-active GTPases, Ras and Rap1A, to O2* and provide evidence for the efficient guanine nucleotide reassociation in the presence of the radical quenching agent ascorbate to complete guanine nucleotide exchange. In vivo, guanine nucleotide reassociation is necessary to populate Ras in its biologically active GTP-bound form after the dissociation of GDP. We further show that treatment of the redox-active GTPases with O2* releases GDP in form of an unstable the oxygenated GDP adduct, putatively assigned as 5-oxo-GDP. 5-Oxo-GDP was not produced from either the C118S or the F28L Ras variants upon the treatment of O2*, supporting the involvement of residues Cys118 and Phe28 in O2*-mediated Ras guanine nucleotide dissociation. These results indicate that the mechanism of O2*-mediated Ras guanine nucleotide dissociation is similar to that of NO/O2-mediated Ras guanine nucleotide dissociation.


Subject(s)
Nitric Oxide/metabolism , Superoxides/metabolism , rap1 GTP-Binding Proteins/chemistry , ras Proteins/chemistry , Amino Acid Motifs , Deoxyguanine Nucleotides/chemistry , Free Radicals , Guanine/metabolism , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Hydrogen-Ion Concentration , Kinetics , Mass Spectrometry , Models, Chemical , Nitric Oxide/chemistry , Oxidation-Reduction , Oxygen/metabolism , Regression Analysis , Spectrometry, Fluorescence , Time Factors , Up-Regulation , ras Proteins/metabolism
18.
J Mol Biol ; 346(5): 1423-40, 2005 Mar 11.
Article in English | MEDLINE | ID: mdl-15713491

ABSTRACT

Ras proteins cycle between GDP-bound and GTP-bound states to modulate a diverse array of cellular growth processes. In this study, we have elucidated a mechanism by which nitric oxide, in the presence of oxygen (NO/O2), regulates Ras activity. We show that treatment of Ras with NO/O2 causes conversion of Ras-bound GDP into a free 463.3 Da nucleotide-nitration product. Mass and UV/visible spectroscopic analyses suggest that this nitration product is 5-guanidino-4-nitroimidazole diphosphate (NIm-DP), a degradation product of 5-nitro-GDP. These results indicate that NO/O2 mediates Ras guanine nucleotide exchange (GNE) by conversion of Ras-bound GDP into an unstable 5-nitro-GDP. 5-Nitro-GDP can be produced by radical-based reaction of the GDP guanine base with nitrogen dioxide (*NO2). We also provide evidence that the Ras Phe28 side-chain plays a key role in the formation of a NO/O2-induced Ras 5-nitro-GDP product. We previously proposed a mechanism of NO/O2-mediated Ras GNE, in which *NO2, formed by the reaction of NO with O2, generates a Ras Cys118 thiyl radical (Ras-S118) intermediate. In the present study, we provide evidence for a radical-based mechanism of NO/O2-mediated Ras GNE. According to this mechanism, reaction of NO with O2 produces *NO2. *NO2 then reacts with Ras to produce Ras-S118, which withdraws an electron from the Ras-bound guanine nucleotide base to produce a guanine nucleotide diphosphate cation radical (G(+)-DP) via the Phe28 side-chain. G(+)-DP is subsequently converted to a neutral radical, and can react with another *NO2 to produce 5-nitro-GDP. This radical-based reaction process disrupts key binding interactions between Ras and the guanine base, resulting in release of GDP from Ras and its conversion to free 5-nitro-GDP. This mechanism is likely to be common to other NKCD motif-containing Ras superfamily GTPases, as NO/O2 also facilitates GNE on the redox-active Rap1A and Rab3A GTPases.


Subject(s)
Free Radicals/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Nitric Oxide/metabolism , Oxygen/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism , Binding Sites , Chromatography, High Pressure Liquid , Humans , Mass Spectrometry , Models, Molecular , Nitrogen Dioxide/metabolism , Oxidation-Reduction , Protein Binding , Protein Conformation , Proto-Oncogene Proteins p21(ras)/genetics
19.
Biochemistry ; 43(31): 10102-11, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15287738

ABSTRACT

p21Ras (Ras) proteins cycle between active GTP-bound and inactive GDP-bound states to mediate signal transduction pathways that promote cell growth, differentiation, and apoptosis. To better understand how cellular regulatory factors, such as guanine nucleotide exchange factors (GEFs) and nitric oxide (NO), modulate Ras-guanine nucleotide binding interactions, we have conducted NMR and kinetic studies to investigate the pH dependence of Ras-GDP interactions and Ras-guanine nucleotide exchange (GNE). pH-sensitive amide protons were identified and found to be associated with residues in the switch I (Phe28-Asp30) and switch II (Asp57 and Thr58) regions of Ras. Furthermore, most of the residues that interact with Mg2+ exhibit pH-sensitive amide proton chemical shifts which appear to be coupled to pH-dependent Ras Mg2+ binding and guanine nucleotide binding affinity. These results suggest that perturbation of Mg2+ interactions within the Ras-guanine nucleotide complex is critical for pH-dependent dissociation of guanine nucleotide ligands from Ras. Notably, these same regions undergo conformational changes upon association with the Ras GEF, SOS. In addition, although we have recently shown that addition of NO to Ras in the presence of oxygen produces a Ras thiyl radical intermediate that promotes Ras GNE, we have also postulated that another byproduct of this reaction, a H+, may contribute to NO-mediated GNE. However, the results presented herein suggest that the H+ byproduct of the reaction is unlikely to be involved in the NO-mediated Ras GNE.


Subject(s)
Guanine Nucleotides/metabolism , Hydrogen-Ion Concentration , Proto-Oncogene Proteins p21(ras)/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism , Amino Acid Sequence , Guanosine Diphosphate/metabolism , Humans , Hydrogen Bonding , Kinetics , Ligands , Magnesium/metabolism , Molecular Sequence Data , Nitric Oxide/metabolism , Nitric Oxide/physiology , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Secondary , Static Electricity , Thermodynamics
20.
Biochemistry ; 43(8): 2314-22, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14979728

ABSTRACT

Nitric oxide (NO), a highly reactive redox molecule, can react with protein thiols and protein metal centers to regulate a multitude of physiological processes. NO has been shown to promote guanine nucleotide exchange on the critical cellular signaling protein p21Ras (Ras) by S-nitrosylation of a redox-active thiol group (Cys(118)). This increases cellular Ras-GTP levels in vivo, leading to activation of downstream signaling pathways. Yet the process by which this occurs is not clear. Although several feasible mechanisms for protein S-nitrosylation with NO and NO donating have been proposed, results obtained from our studies suggest that Ras can be S-nitrosylated by direct reaction of Cys(118) with nitrogen dioxide (*NO(2)), a reaction product of NO with O(2), via a Ras thiyl-radical intermediate (Ras-S*). Results from our studies also indicate that Ras Cys(118) can be S-nitrosylated by direct reaction of Cys(118) with a glutathionyl radical (GS*), a reaction product derived from homolytic cleavage of S-nitrosoglutathione (GSNO). Moreover, we present evidence that reaction of GS* with Ras generates a Ras-S* intermediate during GSNO-mediated Ras S-nitrosylation. The Ras-S(*) radical intermediate formed from reaction of the Ras thiol with either *NO(2) or GS*, in turn, reacts with NO to complete Ras S-nitrosylation. NO and GSNO modulate Ras activity by promoting guanine nucleotide dissociation from Ras. Our results suggest that formation of the Ras radical intermediate, Ras-S*, may perturb interactions between Ras and its guanine nucleotide substrate, resulting in enhancement of guanine nucleotide dissociation from Ras.


Subject(s)
Guanine Nucleotides/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Guanine Nucleotides/chemistry , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Kinetics , Molecular Weight , Nitric Oxide/chemistry , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/metabolism , Nitroso Compounds/chemistry , Nitroso Compounds/metabolism , Oxidation-Reduction , Proteins/chemistry , Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/chemistry , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/metabolism , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...