Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Cells ; 10(11)2021 10 20.
Article in English | MEDLINE | ID: mdl-34831028

ABSTRACT

Coronary artery disease caused by atherosclerosis is a major cause of morbidity and mortality around the world. Data from preclinical and clinical studies support the belief that atherosclerosis is an inflammatory disease that is mediated by innate and adaptive immune signaling mechanisms. This review sought to highlight the role of Rac-mediated inflammatory signaling in the mechanisms driving atherosclerotic calcification. In addition, current clinical treatment strategies that are related to targeting hypercholesterolemia as a critical risk factor for atherosclerotic vascular disease are addressed in relation to the effects on Rac immune signaling and the implications for the future of targeting immune responses in the treatment of calcific atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Atherosclerosis/immunology , Signal Transduction , rac GTP-Binding Proteins/metabolism , Amino Acid Sequence , Atherosclerosis/drug therapy , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/complications , Inflammation/pathology , Models, Biological , rac GTP-Binding Proteins/chemistry
2.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article in English | MEDLINE | ID: mdl-33723071

ABSTRACT

Small GTPases of the Ras-homology (Rho) family are conserved molecular switches that control fundamental cellular activities in eukaryotic cells. As such, they are targeted by numerous bacterial toxins and effector proteins, which have been intensively investigated regarding their biochemical activities and discrete target spectra; however, the molecular mechanism of target selectivity has remained largely elusive. Here we report a bacterial effector protein that selectively targets members of the Rac subfamily in the Rho family of small GTPases but none in the closely related Cdc42 or RhoA subfamilies. This exquisite target selectivity of the FIC domain AMP-transferase Bep1 from Bartonella rochalimae is based on electrostatic interactions with a subfamily-specific pair of residues in the nucleotide-binding G4 motif and the Rho insert helix. Residue substitutions at the identified positions in Cdc42 enable modification by Bep1, while corresponding Cdc42-like substitutions in Rac1 greatly diminish modification. Our study establishes a structural understanding of target selectivity toward Rac-subfamily GTPases and provides a highly selective tool for their functional analysis.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Protein Processing, Post-Translational , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/metabolism , Amino Acid Sequence , Bartonella , Binding Sites , Models, Molecular , Multigene Family , Protein Binding , Protein Conformation , Structure-Activity Relationship , rac GTP-Binding Proteins/genetics
3.
J Biol Chem ; 295(34): 12130-12142, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32636302

ABSTRACT

The RAS-related C3 botulinum toxin substrate 2 (RAC2) is a member of the RHO subclass of RAS superfamily GTPases required for proper immune function. An activating mutation in a key switch II region of RAC2 (RAC2E62K) involved in recognizing modulatory factors and effectors has been identified in patients with common variable immune deficiency. To better understand how the mutation dysregulates RAC2 function, we evaluated the structure and stability, guanine nucleotide exchange factor (GEF) and GTPase-activating protein (GAP) activity, and effector binding of RAC2E62K Our findings indicate the E62K mutation does not alter RAC2 structure or stability. However, it does alter GEF specificity, as RAC2E62K is activated by the DOCK GEF, DOCK2, but not by the Dbl homology GEF, TIAM1, both of which activate the parent protein. Our previous data further showed that the E62K mutation impairs GAP activity for RAC2E62K As this disease mutation is also found in RAS GTPases, we assessed GAP-stimulated GTP hydrolysis for KRAS and observed a similar impairment, suggesting that the mutation plays a conserved role in GAP activation. We also investigated whether the E62K mutation alters effector binding, as activated RAC2 binds effectors to transmit signaling through effector pathways. We find that RAC2E62K retains binding to an NADPH oxidase (NOX2) subunit, p67phox, and to the RAC-binding domain of p21-activated kinase, consistent with our earlier findings. Taken together, our findings indicate that the RAC2E62K mutation promotes immune dysfunction by promoting RAC2 hyperactivation, altering GEF specificity, and impairing GAP function yet retaining key effector interactions.


Subject(s)
Guanosine Triphosphate/chemistry , Mutation, Missense , rac GTP-Binding Proteins/chemistry , Amino Acid Substitution , Enzyme Activation , Guanosine Triphosphate/genetics , Guanosine Triphosphate/immunology , Humans , Hydrolysis , NADPH Oxidase 2/chemistry , NADPH Oxidase 2/genetics , NADPH Oxidase 2/immunology , Protein Domains , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , p21-Activated Kinases/chemistry , p21-Activated Kinases/genetics , p21-Activated Kinases/immunology , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/immunology , RAC2 GTP-Binding Protein
4.
Fish Shellfish Immunol ; 84: 998-1006, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30399403

ABSTRACT

Rac1 and Rac2, belonging to the small Rho GTPase family, play an important role during the immune responses. In this study, a Rac1 homolog (CsRac1) and a Rac2 homolog (CsRac2) were cloned from the Cynoglossus semilaevis. The full-length of CsRac1 and CsRac2 cDNA was 1219 bp and 1047 bp, respectively. Both CsRac1 and CsRac2 contain a 579 bp open reading frame (ORF) which encoding a 192 amino acids putative protein. The predicted molecular weight of CsRac1 and CsRac2 was 21.41 kDa and 21.35 kDa, and their theoretical pI was 8.50 and 7.91, respectively. Sequence analysis showed that the conserved RHO domain was detected both from amino acid of CsRac1 and CsRac2. Homologous analysis showed that CsRac1 and CsRac2 share high conservation with other counterparts from different species. The CsRac1 and CsRac2 transcript showed wide tissue distribution, in which CsRac1 and CsRac2 exhibit the highest expression level in liver and gill, respectively. The expression level of CsRac1 and CsRac2 fluctuated in the liver and gill tissues at different time points after challenged by Vibrio harveyi. Specifically, CsRac1 and CsRac2 were significantly up-regulated at 48 h and 96 h post injection. Moreover, the knocking down of CsRac1 and CsRac2 in cell line (TSHKC) reduced the expression of CsPAK1, CsIL1-ß and CsTNF-α. The present data suggests that CsRac1 and CsRac2 might play important roles in the innate immunity of half-smooth tongue sole.


Subject(s)
Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Flatfishes/genetics , Flatfishes/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/chemistry , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary , Vibrio/physiology , Vibrio Infections/immunology , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/immunology , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology , RAC2 GTP-Binding Protein
5.
J Struct Biol ; 202(1): 13-24, 2018 04.
Article in English | MEDLINE | ID: mdl-29196061

ABSTRACT

Rho family GTPases regulate a wide range of cellular processes. This includes cellular dynamics where three subfamilies, Rho, Rac, and Cdc42, are known to regulate cell shape and migration though coordinate action. Activation of Rho proteins largely depends on Rho Guanine nucleotide Exchange Factors (RhoGEFs) through a catalytic Dbl homology (DH) domain linked to a pleckstrin homology (PH) domain that subserves various functions. The PH domains from Lbc RhoGEFs, which specifically activate RhoA, have been shown to bind to activated RhoA. Here, p190RhoGEF is shown to also bind Rac1·GTP. Crystal structures reveal that activated Rac1 and RhoA use their effector-binding surfaces to associate with the same hydrophobic surface on the PH domain. Both activated RhoA and Rac1 can stimulate exchange of nucleotide on RhoA by localization of p190RhoGEF to its substrate, RhoA·GDP, in vitro. The binding of activated RhoA provides a mechanism for positive feedback regulation as previously proposed for the family of Lbc RhoGEFs. In contrast, the novel interaction between activated Rac1 and p190RhoGEF reveals a potential mechanism for cross-talk regulation where Rac can directly effect stimulation of RhoA. The greater capacity of Rac1 to stimulate p190RhoGEF among the Lbc RhoGEFs suggests functional specialization.


Subject(s)
Guanine Nucleotide Exchange Factors/chemistry , Protein Domains , rac GTP-Binding Proteins/chemistry , rhoA GTP-Binding Protein/chemistry , Amino Acid Sequence , Feedback, Physiological , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Models, Molecular , Protein Binding , Sequence Homology, Amino Acid , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
6.
J Biol Chem ; 292(35): 14334-14348, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28710284

ABSTRACT

Synaptic vesicles (SVs) form distinct pools at synaptic terminals, and this well-regulated separation is necessary for normal neurotransmission. However, how the SV cluster, in particular synaptic compartments, maintains normal neurotransmitter release remains a mystery. The presynaptic protein Neurexin (NRX) plays a significant role in synaptic architecture and function, and some evidence suggests that NRX is associated with neurological disorders, including autism spectrum disorders. However, the role of NRX in SV clustering is unclear. Here, using the neuromuscular junction at the 2-3 instar stages of Drosophila larvae as a model and biochemical imaging and electrophysiology techniques, we demonstrate that Drosophila NRX (DNRX) plays critical roles in regulating synaptic terminal clustering and release of SVs. We found that DNRX controls the terminal clustering and release of SVs by stimulating presynaptic F-actin. Furthermore, our results indicate that DNRX functions through the scaffold protein Scribble and the GEF protein DPix to activate the small GTPase Ras-related C3 Botulinum toxin substrate 1 (Rac1). We observed a direct interaction between the C-terminal PDZ-binding motif of DNRX and the PDZ domains of Scribble and that Scribble bridges DNRX to DPix, forming a DNRX-Scribble-DPix complex that activates Rac1 and subsequently stimulates presynaptic F-actin assembly and SV clustering. Taken together, our work provides important insights into the function of DNRX in regulating SV clustering, which could help inform further research into pathological neurexin-mediated mechanisms in neurological disorders such as autism.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Actin Cytoskeleton/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Neuromuscular Junction/metabolism , Synaptic Vesicles/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Animals , Animals, Genetically Modified , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Drosophila Proteins/agonists , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Electrophysiological Phenomena , Gene Deletion , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Larva/cytology , Larva/genetics , Larva/growth & development , Larva/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/cytology , Neuromuscular Junction/growth & development , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , rac GTP-Binding Proteins/agonists , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/metabolism
7.
J Biol Chem ; 292(29): 12178-12191, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28600358

ABSTRACT

Developmental angiogenesis and the maintenance of the blood-brain barrier involve endothelial cell adhesion, which is linked to cytoskeletal dynamics. GPR124 (also known as TEM5/ADGRA2) is an adhesion G protein-coupled receptor family member that plays a pivotal role in brain angiogenesis and in ensuring a tight blood-brain barrier. However, the signaling properties of GPR124 remain poorly defined. Here, we show that ectopic expression of GPR124 promotes cell adhesion, additive to extracellular matrix-dependent effect, coupled with filopodia and lamellipodia formation and an enrichment of a pool of the G protein-coupled receptor at actin-rich cellular protrusions containing VASP, a filopodial marker. Accordingly, GPR124-expressing cells also displayed increased activation of both Rac and Cdc42 GTPases. Mechanistically, we uncover novel direct interactions between endogenous GPR124 and the Rho guanine nucleotide exchange factors Elmo/Dock and intersectin (ITSN). Small fragments of either Elmo or ITSN1 that bind GPR124 blocked GPR124-induced cell adhesion. In addition, Gßγ interacts with the C-terminal tail of GPR124 and promotes the formation of a GPR124-Elmo complex. Furthermore, GPR124 also promotes the activation of the Elmo-Dock complex, as measured by Elmo phosphorylation on a conserved C-terminal tyrosine residue. Interestingly, Elmo and ITSN1 also interact with each other independently of their GPR124-recognition regions. Moreover, endogenous phospho-Elmo and ITSN1 co-localize with GPR124 at lamellipodia of adhering endothelial cells, where GPR124 expression contributes to polarity acquisition during wound healing. Collectively, our results indicate that GPR124 promotes cell adhesion via Elmo-Dock and ITSN. This constitutes a previously unrecognized complex formed of atypical and conventional Rho guanine nucleotide exchange factors for Rac and Cdc42 that is putatively involved in GPR124-dependent angiogenic responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Endothelium, Vascular/metabolism , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled/metabolism , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Vesicular Transport/chemistry , Animals , COS Cells , Cell Adhesion , Cells, Cultured , Chlorocebus aethiops , Endothelium, Vascular/cytology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Pseudopodia/metabolism , RNA Interference , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , rac GTP-Binding Proteins/chemistry
8.
Sci Rep ; 5: 14437, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26411260

ABSTRACT

Coronin7 (CRN7) stabilizes F-actin and is a regulator of processes associated with the actin cytoskeleton. Its loss leads to defects in phagocytosis, motility and development. It harbors a CRIB (Cdc42- and Rac-interactive binding) domain in each of its WD repeat domains which bind to Rac GTPases preferably in their GDP-loaded forms. Expression of wild type CRN7 in CRN7 deficient cells rescued these defects, whereas proteins with mutations in the CRIB motifs which were associated with altered Rac binding were effective to varying degrees. The presence of one functional CRIB was sufficient to reestablish phagocytosis, cell motility and development. Furthermore, by molecular modeling and mutational analysis we identified the contact regions between CRN7 and the GTPases. We also identified WASP, SCAR and PAKa as downstream effectors in phagocytosis, development and cell surface adhesion, respectively, since ectopic expression rescued these functions.


Subject(s)
Microfilament Proteins/metabolism , Protein Interaction Domains and Motifs , Protozoan Proteins/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , rac GTP-Binding Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Adhesion , Cyclic AMP-Dependent Protein Kinases/metabolism , Microfilament Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Mutation , Mycetozoa , Phagocytosis , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs/genetics , Sequence Alignment , Structure-Activity Relationship , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics
9.
Sci Rep ; 5: 12628, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26243281

ABSTRACT

Phosphoinositide-specific phospholipase C (PLC) is an important family of enzymes constituting a junction between phosphoinositide lipid signaling and the trans-membrane signal transduction processes that are crucial to many living cells. However, the regulatory mechanism of PLC is not yet understood in detail. To address this issue, activity studies were carried out using lipid vesicles in a model system that was specifically designed to study protein-protein and lipid-protein interactions in concert. Evidence was found for a direct interaction between PLC and the GTPases that mediate phospholipase activation. Furthermore, for the first time, the relationships between PLC activity and substrate presentation in lipid vesicles of various sizes, as well as lipid composition and membrane mechanical properties, were analyzed. PLC activity was found to depend upon the electrostatic potential and the stored curvature elastic stress of the lipid membranes.


Subject(s)
Lipid Bilayers/chemistry , Phospholipase C beta/chemistry , Dimyristoylphosphatidylcholine/chemistry , Elasticity , Electrochemistry , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Protein Prenylation , Scattering, Small Angle , X-Ray Diffraction , rac GTP-Binding Proteins/chemistry , RAC2 GTP-Binding Protein
10.
Small GTPases ; 6(2): 71-80, 2015.
Article in English | MEDLINE | ID: mdl-25942647

ABSTRACT

Rac and PI3Ks are intracellular signal transducers able to regulate multiple signaling pathways fundamental for cell behavior. PI3Ks are lipid kinases that produce phosphorylated lipids which, in turn, transduce extracellular cues within the cell, while Rac is a small G protein that impacts on actin organization. Compelling evidence indicates that in multiple circumstances the 2 signaling pathways appear intermingled. For instance, phosphorylated lipids produced by PI3Ks recruit and activate GEF and GAP proteins, key modulators of Rac function. Conversely, PI3Ks interact with activated Rac, leading to Rac signaling amplification. This review summarizes the molecular mechanisms underlying the cross-talk between Rac and PI3K signaling in 2 different processes, cell migration and ROS production.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Second Messenger Systems , rac GTP-Binding Proteins/metabolism , Animals , Humans , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics
11.
Small GTPases ; 6(2): 49-70, 2015.
Article in English | MEDLINE | ID: mdl-25961466

ABSTRACT

The P-Rex family are Dbl-type guanine-nucleotide exchange factors for Rac family small G proteins. They are distinguished from other Rac-GEFs through their synergistic mode of activation by the lipid second messenger phosphatidyl inositol (3,4,5) trisphosphate and the Gßγ subunits of heterotrimeric G proteins, thus acting as coincidence detectors for phosphoinositide 3-kinase and G protein coupled receptor signaling. Work in genetically-modified mice has shown that P-Rex1 has physiological importance in the inflammatory response and the migration of melanoblasts during development, whereas P-Rex2 controls the dendrite morphology of cerebellar Purkinje neurons as well as glucose homeostasis in liver and adipose tissue. Deregulation of P-Rex1 and P-Rex2 expression occurs in many types of cancer, and P-Rex2 is frequently mutated in melanoma. Both GEFs promote tumor growth or metastasis. This review critically evaluates the P-Rex literature and tools available and highlights exciting recent developments and open questions.


Subject(s)
Diabetes Mellitus/metabolism , Neoplasms/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Humans , Protein Binding , Rho Guanine Nucleotide Exchange Factors/chemistry , Rho Guanine Nucleotide Exchange Factors/genetics , Second Messenger Systems , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics
12.
J Biol Chem ; 290(28): 17056-72, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25903139

ABSTRACT

The Rho GTPase Rac is crucially involved in controlling multiple B cell functions, including those regulated by the B cell receptor (BCR) through increased cytosolic Ca(2+). The underlying molecular mechanisms and their relevance to the functions of intact B cells have thus far remained unknown. We have previously shown that the activity of phospholipase Cγ2 (PLCγ2), a key constituent of the BCR signalosome, is stimulated by activated Rac through direct protein-protein interaction. Here, we use a Rac-resistant mutant of PLCγ2 to functionally reconstitute cultured PLCγ2-deficient DT40 B cells and to examine the effects of the Rac-PLCγ2 interaction on BCR-mediated changes of intracellular Ca(2+) and regulation of Ca(2+)-regulated and nuclear-factor-of-activated-T-cell-regulated gene transcription at the level of single, intact B cells. The results show that the functional Rac-PLCγ2 interaction causes marked increases in the following: (i) sensitivity of B cells to BCR ligation; (ii) BCR-mediated Ca(2+) release from intracellular stores; (iii) Ca(2+) entry from the extracellular compartment; and (iv) nuclear translocation of the Ca(2+)-regulated nuclear factor of activated T cells. Hence, Rac-mediated stimulation of PLCγ2 activity serves to amplify B cell receptor-induced Ca(2+) signaling.


Subject(s)
Calcium Signaling/physiology , Phospholipase C gamma/metabolism , Receptors, Antigen, B-Cell/metabolism , rac GTP-Binding Proteins/metabolism , Active Transport, Cell Nucleus , Amino Acid Substitution , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Avian Proteins/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Chickens , Humans , Mice , Models, Molecular , Mutagenesis, Site-Directed , NFATC Transcription Factors/metabolism , Phospholipase C gamma/chemistry , Phospholipase C gamma/genetics , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics
13.
Small GTPases ; 5: e28579, 2014.
Article in English | MEDLINE | ID: mdl-24809833

ABSTRACT

While numerous studies support regulation of Ras GTPases by reactive oxygen and nitrogen species, the Rho subfamily has received considerably less attention. Over the last few years, increasing evidence is emerging that supports the redox sensitivity of Rho GTPases. Moreover, as Rho GTPases regulate the cellular redox state by controlling enzymes that generate and convert reactive oxygen and nitrogen species, redox feedback loops likely exist. Here, we provide an overview of cellular oxidants, Rho GTPases, and their inter-dependence.


Subject(s)
rho GTP-Binding Proteins/metabolism , Animals , Antioxidants/chemistry , Antioxidants/metabolism , Cysteine/chemistry , Cysteine/metabolism , Humans , Oxidation-Reduction , Protein Processing, Post-Translational , Reactive Nitrogen Species/chemistry , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , Second Messenger Systems , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/chemistry , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism
14.
FEBS Lett ; 588(11): 1997-2002, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24792722

ABSTRACT

Rac is an activating factor for Nox1, an O2(-)-generating NADPH oxidase, expressed in the colon and other tissues. Rac requires a GDP-GTP exchange factor for activation. Nox1 activation by ßPix has been demonstrated in cell lines. We examined the effects of ßPix and its phosphomimetic mutant on endogenous Nox1 in Caco-2 cells transfected with Noxo1 and Noxa1. ßPix expression enhanced O2(-) production in resting cells and cells stimulated with EGF or phorbol ester. ßPix(S340E) further enhanced O2(-) production, while ßPix(S340A) eliminated the ßPix effect. ßPix(S340E), but not ßPix(S340A), had higher affinity and GEF activity for Rac than wild-type ßPix. These results suggest that ßPix phosphorylation at Ser-340 upregulates Nox1 through Rac activation, confirming Rac as a trigger for acute Nox1-dependent ROS production.


Subject(s)
NADPH Oxidases/metabolism , Protein Processing, Post-Translational , Rho Guanine Nucleotide Exchange Factors/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Sequence , Amino Acid Substitution , Caco-2 Cells , Enzyme Activation , Epidermal Growth Factor/physiology , Guanosine Triphosphate/chemistry , Humans , Hydrolysis , Mutagenesis, Site-Directed , NADPH Oxidase 1 , Phosphorylation , Protein Binding , Rho Guanine Nucleotide Exchange Factors/chemistry , Rho Guanine Nucleotide Exchange Factors/genetics , Serine/metabolism , Superoxides/metabolism , rac GTP-Binding Proteins/chemistry
15.
Small GTPases ; 5: e27952, 2014.
Article in English | MEDLINE | ID: mdl-24598074

ABSTRACT

The superoxide-generating NADPH oxidase of phagocytes consists of the membrane-associated cytochrome b 558 (a heterodimer of Nox2 and p22(phox)) and 4 cytosolic components: p47(phox), p67(phox), p40(phox), and the small GTPase, Rac, in complex with RhoGDI. Superoxide is produced by the NADPH-driven reduction of molecular oxygen, via a redox gradient located in Nox2. Electron flow in Nox2 is initiated by interaction with cytosolic components, which translocate to the membrane, p67(phox) playing the central role. The participation of Rac is expressed in the following sequence: (1) Translocation of the RacGDP-RhoGDI complex to the membrane; (2) Dissociation of RacGDP from RhoGDI; (3) GDP to GTP exchange on Rac, mediated by a guanine nucleotide exchange factor; (4) Binding of RacGTP to p67(phox); (5) Induction of a conformational change in p67(phox), promoting interaction with Nox2. The particular involvement of Rac in NADPH oxidase assembly serves as a paradigm for signaling by Rho GTPases, in general.


Subject(s)
NADPH Oxidases/metabolism , rac GTP-Binding Proteins/metabolism , Cell Membrane/metabolism , Cytochrome b Group/chemistry , Cytochrome b Group/metabolism , Humans , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/chemistry , Phagocytes/cytology , Phagocytes/enzymology , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Reactive Oxygen Species/metabolism , rac GTP-Binding Proteins/chemistry
16.
Cell Rep ; 6(6): 1153-1164, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24630994

ABSTRACT

The small G protein family Rac has numerous regulators that integrate extracellular signals into tight spatiotemporal maps of its activity to promote specific cell morphologies and responses. Here, we have generated a mouse strain, Rac-FRET, which ubiquitously expresses the Raichu-Rac biosensor. It enables FRET imaging and quantification of Rac activity in live tissues and primary cells without affecting cell properties and responses. We assessed Rac activity in chemotaxing Rac-FRET neutrophils and found enrichment in leading-edge protrusions and unexpected longitudinal shifts and oscillations during protruding and stalling phases of migration. We monitored Rac activity in normal or disease states of intestinal, liver, mammary, pancreatic, and skin tissue, in response to stimulation or inhibition and upon genetic manipulation of upstream regulators, revealing unexpected insights into Rac signaling during disease development. The Rac-FRET strain is a resource that promises to fundamentally advance our understanding of Rac-dependent responses in primary cells and native environments.


Subject(s)
Neutrophils/enzymology , rac GTP-Binding Proteins/metabolism , Animals , Enzyme Activation , Fluorescence Resonance Energy Transfer/methods , Mice , Neutrophils/cytology , Signal Transduction , Spatio-Temporal Analysis , rac GTP-Binding Proteins/chemistry
17.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 1): 113-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24419631

ABSTRACT

Small GTPases regulate a large variety of key cellular processes. Plant small Rac/Rop GTPases have recently received broad attention as it is becoming clear that these enzymes regulate various plant cellular processes. OsRac1, a rice Rac/Rop protein, is a key regulator of reactive oxygen species (ROS) production and induces immune responses. Although four structures of plant small GTPases have been reported, all of these were of the inactive form. Here, OsRac1 was purified and co-crystallized with the GTP analogue 5'-guanylyl imidodiphosphate (GMPPNP). The crystal belonged to space group P2(1)2(1)2(1) and a complete data set was collected to 1.9 Šresolution.


Subject(s)
Oryza/enzymology , Plant Proteins/chemistry , Plant Proteins/isolation & purification , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/isolation & purification , Crystallization , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Guanylyl Imidodiphosphate/chemistry
18.
Cell Mol Life Sci ; 71(9): 1703-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24276852

ABSTRACT

Rho GTPases are a class of evolutionarily conserved proteins comprising 20 members, which are predominantly known for their role in regulating the actin cytoskeleton. They are primarily regulated by binding of GTP/GDP, which is again controlled by regulators like GEFs, GAPs, and RhoGDIs. Rho GTPases are thus far well known for their role in the regulation of actin cytoskeleton and migration. Here we present an overview on the role of Rho GTPases in regulating cell shape and plasticity of cell migration. Finally, we discuss the emerging roles of ubiquitination and sumoylation in regulating Rho GTPases and cell migration.


Subject(s)
rho GTP-Binding Proteins/metabolism , Cell Movement , Cell Shape , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Sumoylation , Ubiquitination , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/classification
19.
Nat Commun ; 4: 1849, 2013.
Article in English | MEDLINE | ID: mdl-23673634

ABSTRACT

Chimaerins, a family of GTPase activating proteins for the small G-protein Rac, have been implicated in development, neuritogenesis and cancer. These Rac-GTPase activating proteins are regulated by the lipid second messenger diacylglycerol generated by tyrosine kinases such as the epidermal growth factor receptor. Here we identify an atypical proline-rich motif in chimaerins that binds to the adaptor protein Nck1. Unlike most Nck1 partners, chimaerins bind to the third SH3 domain of Nck1. This association is mediated by electrostatic interactions of basic residues within the Pro-rich motif with acidic clusters in the SH3 domain. Epidermal growth factor promotes the binding of ß2-chimaerin to Nck1 in the cell periphery in a diacylglycerol-dependent manner. Moreover, ß2-chimaerin translocation to the plasma membrane and its peripheral association with Rac1 requires Nck1. Our studies underscore a coordinated mechanism for ß2-chimaerin activation that involves lipid interactions via the C1 domain and protein-protein interactions via the N-terminal proline-rich region.


Subject(s)
Diglycerides/metabolism , GTPase-Activating Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Proline-Rich Protein Domains , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , COS Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Chlorocebus aethiops , Epidermal Growth Factor/pharmacology , GTPase-Activating Proteins/chemistry , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Structure-Activity Relationship , Vanadates/pharmacology , rac GTP-Binding Proteins/chemistry , src Homology Domains
20.
Nat Commun ; 4: 1706, 2013.
Article in English | MEDLINE | ID: mdl-23591873

ABSTRACT

The chemokine CXCL12 and its G-protein-coupled receptor CXCR4 control the migration, invasiveness and metastasis of breast cancer cells. Binding of CXCL12 to CXCR4 triggers activation of heterotrimeric Gi proteins that regulate actin polymerization and migration. However, the pathways linking chemokine G-protein-coupled receptor/Gi signalling to actin polymerization and cancer cell migration are not known. Here we show that CXCL12 stimulation promotes interaction between Gαi2 and ELMO1. Gi signalling and ELMO1 are both required for CXCL12-mediated actin polymerization, migration and invasion of breast cancer cells. CXCL12 triggers a Gαi2-dependent membrane translocation of ELMO1, which associates with Dock180 to activate small G-proteins Rac1 and Rac2. In vivo, ELMO1 expression is associated with lymph node and distant metastasis, and knocking down ELMO1 impairs metastasis to the lung. Our findings indicate that a chemokine-controlled pathway, consisting of Gαi2, ELMO1/Dock180, Rac1 and Rac2, regulates the actin cytoskeleton during breast cancer metastasis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chemokine CXCL12/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemotaxis , Enzyme Activation , Female , GTP-Binding Protein alpha Subunit, Gi2/chemistry , Humans , Molecular Sequence Data , Neoplasm Invasiveness , rac GTP-Binding Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL