Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 146(29): 19782-19791, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39001846

ABSTRACT

RAP1 proteins belong to the RAS family of small GTPases that operate as molecular switches by cycling between GDP-bound inactive and GTP-bound active states. The C-terminal anchors of RAP1 proteins are known to direct membrane localization, but how these anchors organize RAP1 on the plasma membrane (PM) has not been investigated. Using high-resolution imaging, we show that RAP1A and RAP1B form spatially segregated nanoclusters on the inner leaflet of the PM, with further lateral segregation between GDP-bound and GTP-bound proteins. The C-terminal polybasic anchors of RAP1A and RAP1B differ in their amino acid sequences and exhibit different lipid binding specificities, which can be modified by single-point mutations in the respective polybasic domains (PBD). Molecular dynamics simulations reveal that single PBD mutations substantially reduce the interactions of the membrane anchors with the PM lipid phosphatidylserine. In summary, we show that aggregate lipid binding specificity encoded within the C-terminal anchor determines PM association and nanoclustering of RAP1A and RAP1B. Taken together with previous observations on RAC1 and KRAS, the study reveals that the PBD sequences of small GTPase membrane anchors can encode distinct lipid binding specificities that govern PM interactions.


Subject(s)
Amino Acid Sequence , Cell Membrane , Molecular Dynamics Simulation , rap GTP-Binding Proteins , rap GTP-Binding Proteins/metabolism , rap GTP-Binding Proteins/chemistry , rap GTP-Binding Proteins/genetics , Cell Membrane/metabolism , Cell Membrane/chemistry , Humans , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics , Protein Binding , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Binding Sites
2.
J Biol Chem ; 295(49): 16562-16571, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32948655

ABSTRACT

Phospholipase Cε (PLCε) is activated downstream of G protein-coupled receptors and receptor tyrosine kinases through direct interactions with small GTPases, including Rap1A and Ras. Although Ras has been reported to allosterically activate the lipase, it is not known whether Rap1A has the same ability or what its molecular mechanism might be. Rap1A activates PLCε in response to the stimulation of ß-adrenergic receptors, translocating the complex to the perinuclear membrane. Because the C-terminal Ras association (RA2) domain of PLCε was proposed to the primary binding site for Rap1A, we first confirmed using purified proteins that the RA2 domain is indeed essential for activation by Rap1A. However, we also showed that the PLCε pleckstrin homology (PH) domain and first two EF hands (EF1/2) are required for Rap1A activation and identified hydrophobic residues on the surface of the RA2 domain that are also necessary. Small-angle X-ray scattering showed that Rap1A binding induces and stabilizes discrete conformational states in PLCε variants that can be activated by the GTPase. These data, together with the recent structure of a catalytically active fragment of PLCε, provide the first evidence that Rap1A, and by extension Ras, allosterically activate the lipase by promoting and stabilizing interactions between the RA2 domain and the PLCε core.


Subject(s)
Phosphoinositide Phospholipase C/metabolism , rap1 GTP-Binding Proteins/metabolism , Allosteric Regulation , GTP Phosphohydrolases/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Phosphoinositide Phospholipase C/chemistry , Phosphoinositide Phospholipase C/genetics , Pleckstrin Homology Domains , Protein Binding , Protein Domains , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Scattering, Small Angle , X-Ray Diffraction , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics
3.
J Biol Chem ; 293(47): 18110-18122, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30282804

ABSTRACT

Multifunctional autoprocessing repeats-in-toxin (MARTX) toxins are secreted by Gram-negative bacteria and function as primary virulence-promoting macromolecules that deliver multiple cytopathic and cytotoxic effector domains into the host cytoplasm. Among these effectors, Ras/Rap1-specific endopeptidase (RRSP) catalyzes the sequence-specific cleavage of the Switch I region of the cellular substrates Ras and Rap1 that are crucial for host innate immune defenses during infection. To dissect the molecular basis underpinning RRSP-mediated substrate inactivation, we determined the crystal structure of an RRSP from the sepsis-causing bacterial pathogen Vibrio vulnificus (VvRRSP). Structural and biochemical analyses revealed that VvRRSP is a metal-independent TIKI family endopeptidase composed of an N-terminal membrane-localization and substrate-recruitment domain (N lobe) connected via an inter-lobe linker to the C-terminal active site-coordinating core ß-sheet-containing domain (C lobe). Structure-based mutagenesis identified the 2His/2Glu catalytic residues in the core catalytic domain that are shared with other TIKI family enzymes and that are essential for Ras processing. In vitro KRas cleavage assays disclosed that deleting the N lobe in VvRRSP causes complete loss of enzymatic activity. Endogenous Ras cleavage assays combined with confocal microscopy analysis of HEK293T cells indicated that the N lobe functions both in membrane localization via the first α-helix and in substrate assimilation by altering the functional conformation of the C lobe to facilitate recruitment of cellular substrates. Collectively, these results indicate that RRSP is a critical virulence factor that robustly inactivates Ras and Rap1 and augments the pathogenicity of invading bacteria via the combined effects of its N and C lobes.


Subject(s)
Bacterial Proteins/metabolism , Endopeptidases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Sepsis/enzymology , Sepsis/microbiology , Vibrio vulnificus/enzymology , rap1 GTP-Binding Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins , Endopeptidases/chemistry , Endopeptidases/genetics , HEK293 Cells , Host-Pathogen Interactions , Humans , Protein Domains , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Sepsis/genetics , Vibrio vulnificus/chemistry , Vibrio vulnificus/genetics , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics
4.
Int J Mol Sci ; 19(10)2018 Sep 20.
Article in English | MEDLINE | ID: mdl-30241315

ABSTRACT

This review addresses the issue of the numerous roles played by Rap1 GTPase (guanosine triphosphatase) in different cell types, in terms of both physiology and pathology. It is one among a myriad of small G proteins with endogenous GTP-hydrolyzing activity that is considerably stimulated by posttranslational modifications (geranylgeranylation) or guanine nucleotide exchange factors (GEFs), and inhibited by GTPase-activating proteins (GAPs). Rap1 is a ubiquitous protein that plays an essential role in the control of metabolic processes, such as signal transduction from plasma membrane receptors, cytoskeleton rearrangements necessary for cell division, intracellular and substratum adhesion, as well as cell motility, which is needed for extravasation or fusion. We present several examples of how Rap1 affects cells and organs, pointing to possible molecular manipulations that could have application in the therapy of several diseases.


Subject(s)
rap1 GTP-Binding Proteins/physiology , Adaptive Immunity , Cell Differentiation , Cell Transformation, Neoplastic , Models, Molecular , Prenylation , Protein Processing, Post-Translational , Signal Transduction , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/metabolism
5.
Sci Rep ; 8(1): 12976, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154518

ABSTRACT

The effects of phosphorylation of a serine residue on the structural and dynamic properties of Ras-like protein, Rap, and its interactions with effector protein Ras binding domain (RBD) of Raf kinase, in the presence of GTP, are investigated via molecular dynamics simulations. The simulations show that phosphorylation significantly effects the dynamics of functional loops of Rap which participate in the stability of the complex with effector proteins. The effects of phosphorylation on Rap are significant and detailed conformational analysis suggest that the Rap protein, when phosphorylated and with GTP ligand, samples different conformational space as compared to non-phosphorylated protein. In addition, phosphorylation of SER11 opens up a new cavity in the Rap protein which can be further explored for possible drug interactions. Residue network analysis shows that the phosphorylation of Rap results in a community spanning both Rap and RBD and strongly suggests transmission of allosteric effects of local alterations in Rap to distal regions of RBD, potentially affecting the downstream signalling. Binding free energy calculations suggest that phosphorylation of SER11 residue increases the binding between Rap and Raf corroborating the network analysis results. The increased binding of the Rap-Raf complex can have cascading effects along the signalling pathways where availability of Raf can influence the oncogenic effects of Ras proteins. These simulations underscore the importance of post translational modifications like phosphorylation on the functional dynamics in proteins and can be an alternative to drug-targeting, especially in notoriously undruggable oncoproteins belonging to Ras-like GTPase family.


Subject(s)
Molecular Dynamics Simulation , Proto-Oncogene Proteins c-raf/chemistry , rap1 GTP-Binding Proteins/chemistry , Allosteric Regulation , Humans , Phosphorylation , Protein Binding , Protein Structure, Secondary , Proto-Oncogene Proteins c-raf/metabolism , rap1 GTP-Binding Proteins/metabolism
6.
Biochem Biophys Res Commun ; 499(4): 783-789, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29614268

ABSTRACT

The small GTPase Ras proteins are involved in diverse cellular processes. We investigated the functions of RapC, one of 15 Ras subfamily GTPases in Dictyostelium. Loss of RapC resulted in a spread shape of cells; severe defects in cytokinesis leading to multinucleation; decrease of migration speed in chemoattractant-mediated cell migration, likely through increased cell adhesion; and aberrations in multicellular development producing abnormal multiple tips from one mound and multi-branched developmental structures. Defects in cells lacking RapC were rescued by expressing GFP-RapC in rapC null cells. Our results demonstrate that RapC, despite its high sequence homology with Rap1, plays a negative role in cell spreading and cell adhesion, in contrast to Rap1, which is a key regulator of cell adhesion and cytoskeleton rearrangement. In addition, RapC appears to have a unique function in multicellular development and is involved in tip formation from mounds. This study contributes to the understanding of Ras-mediated cellular processes.


Subject(s)
Cell Movement , Cytokinesis , Dictyostelium/cytology , Dictyostelium/growth & development , Protozoan Proteins/metabolism , Cell Adhesion , Cell Shape , Dictyostelium/metabolism , Phenotype , Phylogeny , Protozoan Proteins/chemistry , Sequence Homology, Amino Acid , rap1 GTP-Binding Proteins/chemistry
7.
Biochemistry ; 56(21): 2747-2757, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28459538

ABSTRACT

Ras/Rap1-specific endopeptidase (RRSP) is a cytotoxic effector domain of the multifunctional autoprocessing repeats-in-toxin (MARTX) toxin of highly virulent strains of Vibrio vulnificus. RRSP blocks RAS-MAPK kinase signaling by cleaving Ras and Rap1 within the switch I region between Y32 and D33. Although the RRSP processing site is highly conserved among small GTPases, only Ras and Rap1 have been identified as proteolytic substrates. Here we report that residues Y32 and D33 at the scissile bond play an important role in RRSP substrate recognition, while the nucleotide state of Ras has an only minimal effect. In addition, substrate specificity is generated by residues across the entire switch I region. Indeed, swapping the Ras switch I region into either RalA or RhoA, GTPases that are not recognized by RRSP, generated chimeras that are substrates of RRSP. However, a difference in the processing efficiency of Ras switch I in the context of Ras, RalA, or RhoA indicates that protein regions outside Ras switch I also contribute to efficient RRSP substrate recognition. Moreover, we show that synthetic peptides corresponding to the Ras and Rap1, but not RalA, switch I regions are cleaved by RRSP, demonstrating sequence-specific substrate recognition. In conclusion, this work demonstrates that the GTPase recognition of RRSP is independent of the nucleotide state and is mainly driven by the Ras and Rap1 switch I loop and also influenced by additional protein-protein interactions, increasing the substrate specificity of RRSP.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , Vibrio vulnificus/enzymology , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Humans , Models, Molecular , Substrate Specificity , rap1 GTP-Binding Proteins/chemistry , ras Proteins/chemistry
8.
J Microbiol ; 55(5): 366-372, 2017 May.
Article in English | MEDLINE | ID: mdl-28124777

ABSTRACT

Establishment of cell polarity is mediated by a series of signaling molecules that are asymmetrically activated or localized in the cell upon extracellular stimulation. To understand the mechanism that mediates anterior/posterior asymmetric localization of RapGAP3 during migration, we determined the minimally required amino acids in the I/LWEQ domain that cause posterior localization and found that the minimal region of the F-actin binding domain for posterior localization could, with some additional deletion at the C-terminal, localize to the anterior. Analysis of the localization and translocation kinetics to the cell cortex of the truncated proteins suggests that the required regions for anterior/posterior localization might have a preferential binding affinity to preexisting F-actins at the rear and lateral sides of the cell or newly formed F-actins at the front of the cell, leading to distinct differential sites of the cell.


Subject(s)
Amino Acids/chemistry , Dictyostelium/metabolism , rap1 GTP-Binding Proteins/metabolism , Actins/metabolism , Amino Acid Sequence , Amino Acids/genetics , Animals , Cell Movement , Cell Polarity , Conserved Sequence , Cytoskeleton/metabolism , Dictyostelium/chemistry , Dictyostelium/cytology , Dictyostelium/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics
9.
Structure ; 24(12): 2152-2162, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27839947

ABSTRACT

Ras-interacting protein 1 (Rasip1) is an endothelial-specific Rap1 and Ras effector, important for vascular development and angiogenesis. Here, we report the crystal structure of the Rasip1 RA domain (RRA) alone, revealing the basis of dimerization, and in complex with Rap1 at 2.8 Å resolution. In contrast to most RA domains, RRA formed a dimer that can bind two Rap1 (KD = 0.9 µM) or Ras (KD = 2.2 µM) molecules. We solved the Rap1-RRA complex and found that Rasip1 binds Rap1 in the Switch I region, and Rap1 binding induces few conformation changes to Rasip1 stabilizing a ß strand and an unstructured loop. Our data explain how Rasip1 can act as a Rap1 and Ras effector and show that Rasip1 defines a subgroup of dimeric RA domains that could mediate cooperative binding to membrane-associated Ras superfamily members.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Binding Sites , Dimerization , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Structure, Secondary , rap1 GTP-Binding Proteins/chemistry , ras Proteins/chemistry
10.
J Mol Biol ; 428(24 Pt B): 4929-4945, 2016 12 04.
Article in English | MEDLINE | ID: mdl-27760305

ABSTRACT

Two isoforms of the small GTPase Rap1, Rap1A and Rap1B, participate in cell adhesion; Rap1A promotes steady state adhesion, while Rap1B regulates dynamic changes in cell adhesion. These events depend on the prenylation of Rap1, which promotes its membrane localization. Here, we identify previously unsuspected differences in the regulation of prenylation of Rap1A versus Rap1B, due in part to their different phosphorylation-dependent interactions with the chaperone protein SmgGDS-607. Previous studies indicate that the activation of Gαs protein-coupled receptors (GPCRs) phosphorylates S-179 and S-180 in the polybasic region (PBR) of Rap1B, which inhibits Rap1B binding to SmgGDS-607 and diminishes Rap1B prenylation and membrane localization. In this study, we investigate how phosphorylation in the PBR of multiple small GTPases, including K-Ras4B, RhoA, Rap1A, and Rap1B, affects their binding to SmgGDS, with emphasis on differences between Rap1A and Rap1B. We identify the amino acids in SmgGDS-607 necessary for binding of Rap1A and Rap1B, and present homology models examining the binding between Rap1A or Rap1B and SmgGDS-607. Unlike Rap1B, phosphorylation in the PBR of Rap1A does not detectably inhibit its prenylation or its binding to SmgGDS-607. Activation of GPCRs suppresses Rap1A prenylation, but unlike this effect on Rap1B, the GPCR-mediated suppression of Rap1A prenylation can occur independently of Rap1A phosphorylation and does not detectably diminish Rap1A membrane localization. These data demonstrate unexpected evolutionarily conserved differences in the ability of GPCRs to regulate the prenylation of Rap1B compared to Rap1A.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Prenylation , Protein Processing, Post-Translational , rap GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Cell Line , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Conformation , Protein Interaction Mapping , Sequence Alignment , rap GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/chemistry
11.
Nat Commun ; 6: 7396, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26051945

ABSTRACT

Ras (Rat sarcoma) protein is a central regulator of cell growth and proliferation. Mutations in the RAS gene are known to occur in human cancers and have been shown to contribute to carcinogenesis. In this study, we show that the multifunctional-autoprocessing repeats-in-toxin (MARTX) toxin-effector domain DUF5(Vv) from Vibrio vulnificus to be a site-specific endopeptidase that cleaves within the Switch 1 region of Ras and Rap1. DUF5(Vv) processing of Ras, which occurs both biochemically and in mammalian cell culture, inactivates ERK1/2, thereby inhibiting cell proliferation. The ability to cleave Ras and Rap1 is shared by DUF5(Vv) homologues found in other bacteria. In addition, DUF5(Vv )can cleave all Ras isoforms and KRas with mutations commonly implicated in malignancies. Therefore, we speculate that this new family of Ras/Rap1-specific endopeptidases (RRSPs) has potential to inactivate both wild-type and mutant Ras proteins expressed in malignancies.


Subject(s)
Bacterial Toxins/metabolism , Vibrio vulnificus/metabolism , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Amino Acid Sequence , HeLa Cells , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , rap1 GTP-Binding Proteins/chemistry , ras Proteins/chemistry
12.
J Exp Clin Cancer Res ; 34: 8, 2015 Jan 31.
Article in English | MEDLINE | ID: mdl-25636908

ABSTRACT

OBJECTIVE: Evidence supports an important role for miR-203 in the regulation of the proliferation, migration and invasion of prostate cancer (PCa) cells. However, the exact mechanisms of miR-203 in PCa are not entirely clear. METHODS: We examined the expression of miR-203 in prostate cancer tissues, adjacent normal tissues, PCa cell lines and normal prostate epithelial cells by qRT-PCR. Then, the effects of miR-203 or Rap1A on proliferation, adhesion and invasion of PCa cells were assayed using CKK-8, adhesion analysis, and transwell invasion assays. Luciferase reporter assay was performed to assess miR-203 binding to Rap1A mRNA. Tumor growth was assessed by subcutaneous inoculation of cells into BALB/c nude mice. RESULTS: Here, we confirmed that the expression of miR-203 was significantly downregulated in prostate cancer specimens compared with matched adjacent normal prostate specimens. Mechanistic dissection revealed that miR-203 mediated cell proliferation, adhesion and invasion in vitro, and tumor growth in vivo, as evidenced by reduced RAC1, p-PAK1, and p-MEK1 expression. In addition, we identified Rap1A as a direct target suppressed by miR-203, and there was an inverse relationship between the expression of miR-203 and Rap1A in PCa. Knockdown of Rap1A phenocopied the effects of miR-203 on PCa cell growth and invasion. Furthermore, Rap1A over-expression in PCa cells partially reversed the effects of miR-203-expression on cell adhesion and invasion. CONCLUSIONS: These findings provide further evidence that a crucial role for miR-203 in inhibiting metastasis of PCa through the suppression of Rap1A expression.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , rap1 GTP-Binding Proteins/genetics , Animals , Base Sequence , Binding Sites , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Models, Animal , Down-Regulation , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Humans , Male , MicroRNAs/chemistry , RNA Interference , RNA, Messenger/genetics , Xenograft Model Antitumor Assays , rap1 GTP-Binding Proteins/chemistry
13.
J Biol Chem ; 290(8): 4908-4927, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25533468

ABSTRACT

synGAP is a neuron-specific Ras and Rap GTPase-activating protein (GAP) found in high concentrations in the postsynaptic density (PSD) fraction from the mammalian forebrain. We have previously shown that, in situ in the PSD fraction or in recombinant form in Sf9 cell membranes, synGAP is phosphorylated by Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), another prominent component of the PSD. Here, we show that recombinant synGAP (r-synGAP), lacking 102 residues at the N terminus, can be purified in soluble form and is phosphorylated by cyclin-dependent kinase 5 (CDK5) as well as by CaMKII. Phosphorylation of r-synGAP by CaMKII increases its HRas GAP activity by 25% and its Rap1 GAP activity by 76%. Conversely, phosphorylation by CDK5 increases r-synGAP's HRas GAP activity by 98% and its Rap1 GAP activity by 20%. Thus, phosphorylation by both kinases increases synGAP activity; CaMKII shifts the relative GAP activity toward inactivation of Rap1, and CDK5 shifts the relative activity toward inactivation of HRas. GAP activity toward Rap2 is not altered by phosphorylation by either kinase. CDK5 phosphorylates synGAP primarily at two sites, Ser-773 and Ser-802. Phosphorylation at Ser-773 inhibits r-synGAP activity, and phosphorylation at Ser-802 increases it. However, the net effect of concurrent phosphorylation of both sites, Ser-773 and Ser-802, is an increase in GAP activity. synGAP is phosphorylated at Ser-773 and Ser-802 in the PSD fraction, and its phosphorylation by CDK5 and CaMKII is differentially regulated by activation of NMDA-type glutamate receptors in cultured neurons.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cyclin-Dependent Kinase 5 , GTPase-Activating Proteins , Oncogene Proteins , Proto-Oncogene Proteins p21(ras) , Synapses/enzymology , rap GTP-Binding Proteins , rap1 GTP-Binding Proteins , ras GTPase-Activating Proteins , ras Proteins , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 5/chemistry , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Neurons/cytology , Neurons/enzymology , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism , ras GTPase-Activating Proteins/chemistry , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/metabolism
14.
J Biol Chem ; 288(39): 27712-23, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23946483

ABSTRACT

The small G protein Rap1 can mediate "inside-out signaling" by recruiting effectors to the plasma membrane that signal to pathways involved in cell adhesion and cell migration. This action relies on the membrane association of Rap1, which is dictated by post-translational prenylation as well as by a stretch of basic residues within its carboxyl terminus. One feature of this stretch of acidic residues is that it lies adjacent to a functional phosphorylation site for the cAMP-dependent protein kinase PKA. This phosphorylation has two effects on Rap1 action. One, it decreases the level of Rap1 activity as measured by GTP loading and the coupling of Rap1 to RapL, a Rap1 effector that couples Rap1 GTP loading to integrin activation. Two, it destabilizes the membrane localization of Rap1, promoting its translocation into the cytoplasm. These two actions, decreased GTP loading and decreased membrane localization, are related, as the translocation of Rap1-GTP into the cytoplasm is associated with its increased GTP hydrolysis and inactivation. The consequences of this phosphorylation in Rap1-dependent cell adhesion and cell migration were also examined. Active Rap1 mutants that lack this phosphorylation site had a minimal effect on cell adhesion but strongly reduced cell migration, when compared with an active Rap1 mutant that retained the phosphorylation site. This suggests that optimal cell migration is associated with cycles of Rap1 activation, membrane egress, and inactivation, and requires the regulated phosphorylation of Rap1 by PKA.


Subject(s)
Cell Movement , Cyclic AMP-Dependent Protein Kinases/chemistry , rap1 GTP-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Cattle , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Enzyme Activation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Microscopy, Confocal , Molecular Sequence Data , Mutation , Phosphorylation , Signal Transduction
15.
J Phys Chem B ; 116(31): 9326-36, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22738401

ABSTRACT

The human protein Rap1A (Rap) is a member of the Ras superfamily of GTPases that binds to the downstream effector Ral guanine nucleotide dissociation stimulator (RalGDS). Although Ras and Rap have nearly identical amino acid sequences and structures along the effector binding surface, the charge reversal mutation Rap K31E has previously been shown to increase the dissociation constant of Rap-RalGDS docking to values similar to that of Ras-RalGDS docking. This indicates that the difference in charge at position 31 could provide a mechanism for Ral to distinguish two structurally similar but functionally distinct GTPases, which would be of vital importance for appropriate biological function. In this report, vibrational Stark effect spectroscopy, dissociation constant measurements, and molecular dynamics simulations were used to investigate the role that electrostatic field differences caused by the charge reversal mutation Rap K31E play in determining the binding specificity of RalGDS to Rap versus Ras. To do this, six variants of RalGDS carrying a thiocyanate electrostatic probe were docked with three Rap mutants, E30D, K31E, and E30D/K31E. The change in absorption energy of the thiocyanate probe caused by RalGDS docking to these Rap variants was then compared to that observed with wild-type Ras. Three trends emerged: the expected reversion behavior, an additive behavior of the two single mutations, and cancelation of the effects of each single mutation in the double mutant. These observations are explained with a physical model of the position of the thiocyanate probe with respect to the mutated residue based on molecular dynamics simulations.


Subject(s)
Point Mutation , ral Guanine Nucleotide Exchange Factor/metabolism , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism , Humans , Molecular Dynamics Simulation , Protein Binding , Spectrophotometry, Infrared/methods , Static Electricity , Thiocyanates/chemistry , ral Guanine Nucleotide Exchange Factor/chemistry , rap1 GTP-Binding Proteins/chemistry
16.
Proc Natl Acad Sci U S A ; 109(10): 3814-9, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22343288

ABSTRACT

Epac1 is a cAMP-regulated guanine nucleotide exchange factor for the small G protein Rap. Upon cAMP binding, Epac1 undergoes a conformational change that results in its release from autoinhibition. In addition, cAMP induces the translocation of Epac1 from the cytosol to the plasma membrane. This relocalization of Epac1 is required for efficient activation of plasma membrane-located Rap and for cAMP-induced cell adhesion. This translocation requires the Dishevelled, Egl-10, Pleckstrin (DEP) domain, but the molecular entity that serves as the plasma membrane anchor and the possible mechanism of regulated binding remains elusive. Here we show that Epac1 binds directly to phosphatidic acid. Similar to the cAMP-induced Epac1 translocation, this binding is regulated by cAMP and requires the DEP domain. Furthermore, depletion of phosphatidic acid by inhibition of phospholipase D1 prevents cAMP-induced translocation of Epac1 as well as the subsequent activation of Rap at the plasma membrane. Finally, mutation of a single basic residue within a polybasic stretch of the DEP domain, which abolishes translocation, also prevents binding to phosphatidic acid. From these results we conclude that cAMP induces a conformational change in Epac1 that enables DEP domain-mediated binding to phosphatidic acid, resulting in the tethering of Epac1 at the plasma membrane and subsequent activation of Rap.


Subject(s)
Cell Membrane/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Phosphatidic Acids/chemistry , Guanine Nucleotide Exchange Factors/chemistry , HEK293 Cells , Humans , Lipids/chemistry , Liposomes/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/chemistry , rap1 GTP-Binding Proteins/chemistry
17.
Trends Cell Biol ; 21(10): 615-23, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21820312

ABSTRACT

Signaling by the small G-protein Rap is under tight regulation by its GEFs and GAPs. These are multi-domain proteins that are themselves controlled by distinct upstream pathways, and thus couple different extra- and intracellular cues to Rap. The individual RapGEFs and RapGAPs are, in addition, targeted to specific cellular locations by numerous anchoring mechanisms and, consequently, may control different pools of Rap. Here, we review the various activating signals and targeting mechanisms of these proteins and discuss their contribution to the spatiotemporal regulation and biological functions of the Rap proteins.


Subject(s)
Cyclic AMP/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Models, Molecular , Monomeric GTP-Binding Proteins/metabolism , rap GTP-Binding Proteins/metabolism , Cyclic AMP/chemistry , GTPase-Activating Proteins/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Monomeric GTP-Binding Proteins/chemistry , Signal Transduction , rap GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/metabolism
18.
Mol Biol Cell ; 22(14): 2509-19, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21633110

ABSTRACT

Activation of Rap1 small GTPases stabilizes cell--cell junctions, and this activity requires Krev Interaction Trapped gene 1 (KRIT1). Loss of KRIT1 disrupts cardiovascular development and causes autosomal dominant familial cerebral cavernous malformations. Here we report that native KRIT1 protein binds the effector loop of Rap1A but not H-Ras in a GTP-dependent manner, establishing that it is an authentic Rap1-specific effector. By modeling the KRIT1-Rap1 interface we designed a well-folded KRIT1 mutant that exhibited a ~40-fold-reduced affinity for Rap1A and maintained other KRIT1-binding functions. Direct binding of KRIT1 to Rap1 stabilized endothelial cell-cell junctions in vitro and was required for cardiovascular development in vivo. Mechanistically, Rap1 binding released KRIT1 from microtubules, enabling it to locate to cell--cell junctions, where it suppressed Rho kinase signaling and stabilized the junctions. These studies establish that the direct physical interaction of Rap1 with KRIT1 enables the translocation of microtubule-sequestered KRIT1 to junctions, thereby supporting junctional integrity and cardiovascular development.


Subject(s)
Intercellular Junctions/genetics , Microtubule-Associated Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Muscle Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , rap1 GTP-Binding Proteins/metabolism , Animals , Cardiovascular System/growth & development , Cardiovascular System/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Gene Expression , Genetic Vectors , HEK293 Cells , Humans , Intercellular Junctions/metabolism , KRIT1 Protein , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/genetics , Muscle Proteins/genetics , Muscle Proteins/physiology , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , RNA, Small Interfering , Signal Transduction , Structure-Activity Relationship , Umbilical Veins/cytology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics
19.
Bioorg Med Chem Lett ; 20(22): 6443-6, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20943388

ABSTRACT

As is the case with other ladder-shaped polyether compounds, yessotoxin is produced by marine dinoflagellate, and possesses various biological activities beside potent toxicity. To gain a better understanding of the molecular mechanism for high affinity between these polyethers and their binding proteins, which accounts for their powerful biological activities, we searched for its binding proteins from human blood cells by using the biotin-conjugate of desulfated YTX as a ligand. By a protein pull-down protocol with use of streptavidin beads, a band of specifically binding proteins was detected in SDS-PAGE. HPLC-tandem mass spectrometry (MS/MS) indicated that Rap 1A, one of Ras superfamily proteins, binds to the YTX-linked resins. Western blotting and surface plasmon resonance experiments further confirmed that Rap1A specifically binds to YTX with the K(D) value around 4 µM.


Subject(s)
Erythrocyte Membrane/chemistry , Oxocins/metabolism , rap1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Blotting, Western , Chromatography, High Pressure Liquid , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Mollusk Venoms , Protein Binding , Surface Plasmon Resonance , Tandem Mass Spectrometry , rap1 GTP-Binding Proteins/chemistry
20.
J Phys Chem B ; 114(46): 15331-44, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20964430

ABSTRACT

Electrostatic fields at the interface of the Ras binding domain of the protein Ral guanine nucleotide dissociation stimulator (RalGDS) with the structurally analogous GTPases Ras and Rap1A were measured with vibrational Stark effect (VSE) spectroscopy. Eleven residues on the surface of RalGDS that participate in this protein-protein interaction were systematically mutated to cysteine and subsequently converted to cyanocysteine in order to introduce a nitrile VSE probe in the form of the thiocyanate (SCN) functional group. The measured SCN absorption energy on the monomeric protein was compared with solvent-accessible surface area (SASA) calculations and solutions to the Poisson-Boltzmann equation using Boltzmann-weighted structural snapshots from molecular dynamics simulations. We found a weak negative correlation between SASA and measured absorption energy, indicating that water exposure of protein surface amino acids can be estimated from experimental measurement of the magnitude of the thiocyanate absorption energy. We found no correlation between calculated field and measured absorption energy. These results highlight the complex structural and electrostatic nature of the protein-water interface. The SCN-labeled RalGDS was incubated with either wild-type Ras or wild-type Rap1A, and the formation of the docked complex was confirmed by measurement of the dissociation constant of the interaction. The change in absorption energy of the thiocyanate functional group due to complex formation was related to the change in electrostatic field experienced by the nitrile functional group when the protein-protein interface forms. At some locations, the nitrile experiences the same shift in field when bound to Ras and Rap1A, but at others, the change in field is dramatically different. These differences identify residues on the surface of RalGDS that direct the specificity of RalGDS binding to its in vivo binding partner, Rap1A, through an electrostatic mechanism.


Subject(s)
Spectrum Analysis/methods , ral Guanine Nucleotide Exchange Factor/chemistry , rap1 GTP-Binding Proteins/chemistry , ras Proteins/chemistry , Binding Sites , Models, Molecular , Molecular Dynamics Simulation , Molecular Probes/chemistry , Molecular Probes/metabolism , Molecular Structure , Protein Binding , Protein Conformation , Static Electricity , Thiocyanates/chemistry , Vibration , ral Guanine Nucleotide Exchange Factor/genetics , ral Guanine Nucleotide Exchange Factor/metabolism , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism , ras Proteins/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...