159 results on '"Guanosine Triphosphate genetics"'
Search Results
2. Mutations in the α4-α5 allosteric lobe of RAS do not significantly impair RAS signaling or self-association.
- Author
-
Whaby M, Wallon L, Mazzei M, Khan I, Teng KW, Koide S, and O'Bryan JP
- Subjects
- Humans, Protein Binding, Mutation, Guanosine Triphosphate genetics, Genes, ras, Signal Transduction genetics
- Abstract
Mutations in one of the three RAS genes (HRAS, KRAS, and NRAS) are present in nearly 20% of all human cancers. These mutations shift RAS to the GTP-loaded active state due to impairment in the intrinsic GTPase activity and disruption of GAP-mediated GTP hydrolysis, resulting in constitutive activation of effectors such as RAF. Because activation of RAF involves dimerization, RAS dimerization has been proposed as an important step in RAS-mediated activation of effectors. The α4-α5 allosteric lobe of RAS has been proposed as a RAS dimerization interface. Indeed, the NS1 monobody, which binds the α4-α5 region within the RAS G domain, inhibits RAS-dependent signaling and transformation as well as RAS nanoclustering at the plasma membrane. Although these results are consistent with a model in which the G domain dimerizes through the α4-α5 region, the isolated G domain of RAS lacks intrinsic dimerization capacity. Furthermore, prior studies analyzing α4-α5 point mutations have reported mixed effects on RAS function. Here, we evaluated the activity of a panel of single amino acid substitutions in the α4-α5 region implicated in RAS dimerization. We found that these proposed "dimerization-disrupting" mutations do not significantly impair self-association, signaling, or transformation of oncogenic RAS. These results are consistent with a model in which activated RAS protomers cluster in close proximity to promote the dimerization of their associated effector proteins (e.g., RAF) without physically associating into dimers mediated by specific molecular interactions. Our findings suggest the need for a nonconventional approach to developing therapeutics targeting the α4-α5 region., Competing Interests: Conflict of interest J. P. O., A. K., and S. K. are listed as inventors on a patent application on Monobodies targeting the nucleotide-free state of RAS files by the Medical University of South Carolina and New York University (No. 62/862924). K. W. T., A. K., and S. K. are listed as inventors on a patent application on mutant RAS targeting Monobodies filed by New York University (Application No. 63/121903). A. K. and S. K. are listed as inventors on issued and pending patents on Monobody technology filed by The University of Chicago (US Patent 9512199 B2 and related pending applications). S. K. was an SAB member and holds equity in and received consulting fees from Black Diamond Therapeutics and receives research funding from Black Diamond Therapeutics, Puretech Health, and Argenx BVBA. The other authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
3. The hypervariable region of atlastin-1 is a site for intrinsic and extrinsic regulation.
- Author
-
Kelly CM, Byrnes LJ, Neela N, Sondermann H, and O'Donnell JP
- Subjects
- Animals, Cell Line, Endoplasmic Reticulum genetics, GTP Phosphohydrolases genetics, Guanosine Triphosphate genetics, Humans, Hydrolysis, Membrane Fusion genetics, Mice, NIH 3T3 Cells, Protein Processing, Post-Translational genetics, GTP-Binding Proteins genetics, Membrane Proteins genetics
- Abstract
Atlastin (ATL) GTPases catalyze homotypic membrane fusion of the peripheral endoplasmic reticulum (ER). GTP-hydrolysis-driven conformational changes and membrane tethering are prerequisites for proper membrane fusion. However, the molecular basis for regulation of these processes is poorly understood. Here we establish intrinsic and extrinsic modes of ATL1 regulation that involve the N-terminal hypervariable region (HVR) of ATLs. Crystal structures of ATL1 and ATL3 exhibit the HVR as a distinct, isoform-specific structural feature. Characterizing the functional role of ATL1's HVR uncovered its positive effect on membrane tethering and on ATL1's cellular function. The HVR is post-translationally regulated through phosphorylation-dependent modification. A kinase screen identified candidates that modify the HVR site specifically, corresponding to the modifications on ATL1 detected in cells. This work reveals how the HVR contributes to efficient and potentially regulated activity of ATLs, laying the foundation for the identification of cellular effectors of ATL-mediated membrane processes., (© 2021 Kelly et al.)
- Published
- 2021
- Full Text
- View/download PDF
4. Heterotrimeric G Proteins in Plants: Canonical and Atypical Gα Subunits.
- Author
-
Maruta N, Trusov Y, Jones AM, and Botella JR
- Subjects
- Animals, GTP-Binding Protein alpha Subunits genetics, Guanosine Diphosphate genetics, Guanosine Triphosphate genetics, Plant Proteins genetics, Plants genetics, GTP-Binding Protein alpha Subunits metabolism, Guanosine Diphosphate metabolism, Guanosine Triphosphate metabolism, Plant Proteins metabolism, Plants metabolism, Signal Transduction
- Abstract
Heterotrimeric GTP-binding proteins (G proteins), consisting of Gα, Gβ and Gγ subunits, transduce signals from a diverse range of extracellular stimuli, resulting in the regulation of numerous cellular and physiological functions in Eukaryotes. According to the classic G protein paradigm established in animal models, the bound guanine nucleotide on a Gα subunit, either guanosine diphosphate (GDP) or guanosine triphosphate (GTP) determines the inactive or active mode, respectively. In plants, there are two types of Gα subunits: canonical Gα subunits structurally similar to their animal counterparts and unconventional extra-large Gα subunits (XLGs) containing a C-terminal domain homologous to the canonical Gα along with an extended N-terminal domain. Both Gα and XLG subunits interact with Gβγ dimers and regulator of G protein signalling (RGS) protein. Plant G proteins are implicated directly or indirectly in developmental processes, stress responses, and innate immunity. It is established that despite the substantial overall similarity between plant and animal Gα subunits, they convey signalling differently including the mechanism by which they are activated. This review emphasizes the unique characteristics of plant Gα subunits and speculates on their unique signalling mechanisms.
- Published
- 2021
- Full Text
- View/download PDF
5. Oncogenic mutations Q61L and Q61H confer active form-like structural features to the inactive state (state 1) conformation of H-Ras protein.
- Author
-
Matsumoto S, Taniguchi-Tamura H, Araki M, Kawamura T, Miyamoto R, Tsuda C, Shima F, Kumasaka T, Okuno Y, and Kataoka T
- Subjects
- Crystallography, X-Ray, Guanosine Triphosphate genetics, Humans, Molecular Conformation, Molecular Dynamics Simulation, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Guanosine Triphosphate metabolism, Proto-Oncogene Proteins p21(ras) metabolism
- Abstract
GTP-bound forms of Ras proteins (Ras•GTP) assume two interconverting conformations, "inactive" state 1 and "active" state 2. Our previous study on the crystal structure of the state 1 conformation of H-Ras in complex with guanosine 5'-(β, γ-imido)triphosphate (GppNHp) indicated that state 1 is stabilized by intramolecular hydrogen-bonding interactions formed by Gln61. Since Ras are constitutively activated by substitution mutations of Gln61, here we determine crystal structures of the state 1 conformation of H-Ras•GppNHp carrying representative mutations Q61L and Q61H to observe the effect of the mutations. The results show that these mutations alter the mode of hydrogen-bonding interactions of the residue 61 with Switch II residues and induce conformational destabilization of the neighboring regions. In particular, Q61L mutation results in acquirement of state 2-like structural features. Moreover, the mutations are likely to impair an intramolecular structural communication between Switch I and Switch II. Molecular dynamics simulations starting from these structures support the above observations. These findings may give a new insight into the molecular mechanism underlying the aberrant activation of the Gln61 mutants., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
6. GTP-dependent formation of straight tubulin oligomers leads to microtubule nucleation.
- Author
-
Ayukawa R, Iwata S, Imai H, Kamimura S, Hayashi M, Ngo KX, Minoura I, Uchimura S, Makino T, Shirouzu M, Shigematsu H, Sekimoto K, Gigant B, and Muto E
- Subjects
- Animals, Drosophila Proteins genetics, Drosophila Proteins metabolism, Drosophila melanogaster, Guanosine Diphosphate chemistry, Guanosine Diphosphate metabolism, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Microtubules genetics, Microtubules metabolism, Tubulin genetics, Tubulin metabolism, Drosophila Proteins chemistry, Guanosine Triphosphate chemistry, Microtubules chemistry, Protein Multimerization, Tubulin chemistry
- Abstract
Nucleation of microtubules (MTs) is essential for cellular activities, but its mechanism is unknown because of the difficulty involved in capturing rare stochastic events in the early stage of polymerization. Here, combining rapid flush negative stain electron microscopy (EM) and kinetic analysis, we demonstrate that the formation of straight oligomers of critical size is essential for nucleation. Both GDP and GTP tubulin form single-stranded oligomers with a broad range of curvatures, but upon nucleation, the curvature distribution of GTP oligomers is shifted to produce a minor population of straight oligomers. With tubulin having the Y222F mutation in the β subunit, the proportion of straight oligomers increases and nucleation accelerates. Our results support a model in which GTP binding generates a minor population of straight oligomers compatible with lateral association and further growth to MTs. This study suggests that cellular factors involved in nucleation promote it via stabilization of straight oligomers., (© 2021 Ayukawa et al.)
- Published
- 2021
- Full Text
- View/download PDF
7. Guanosine triphosphate links MYC-dependent metabolic and ribosome programs in small-cell lung cancer.
- Author
-
Huang F, Huffman KE, Wang Z, Wang X, Li K, Cai F, Yang C, Cai L, Shih TS, Zacharias LG, Chung A, Yang Q, Chalishazar MD, Ireland AS, Stewart CA, Cargill K, Girard L, Liu Y, Ni M, Xu J, Wu X, Zhu H, Drapkin B, Byers LA, Oliver TG, Gazdar AF, Minna JD, and DeBerardinis RJ
- Subjects
- Animals, Cell Line, Tumor, GTP Phosphohydrolases genetics, GTP Phosphohydrolases metabolism, GTP-Binding Proteins genetics, GTP-Binding Proteins metabolism, Guanosine Triphosphate genetics, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Mice, Mutation, Proto-Oncogene Proteins c-myc genetics, RNA Polymerase I genetics, RNA Polymerase I metabolism, Ribosomes genetics, Ribosomes pathology, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology, Guanosine Triphosphate metabolism, Lung Neoplasms metabolism, Proto-Oncogene Proteins c-myc metabolism, Ribosomes metabolism, Small Cell Lung Carcinoma metabolism
- Abstract
MYC stimulates both metabolism and protein synthesis, but how cells coordinate these complementary programs is unknown. Previous work reported that, in a subset of small-cell lung cancer (SCLC) cell lines, MYC activates guanosine triphosphate (GTP) synthesis and results in sensitivity to inhibitors of the GTP synthesis enzyme inosine monophosphate dehydrogenase (IMPDH). Here, we demonstrated that primary MYChi human SCLC tumors also contained abundant guanosine nucleotides. We also found that elevated MYC in SCLCs with acquired chemoresistance rendered these otherwise recalcitrant tumors dependent on IMPDH. Unexpectedly, our data indicated that IMPDH linked the metabolic and protein synthesis outputs of oncogenic MYC. Coexpression analysis placed IMPDH within the MYC-driven ribosome program, and GTP depletion prevented RNA polymerase I (Pol I) from localizing to ribosomal DNA. Furthermore, the GTPases GPN1 and GPN3 were upregulated by MYC and directed Pol I to ribosomal DNA. Constitutively GTP-bound GPN1/3 mutants mitigated the effect of GTP depletion on Pol I, protecting chemoresistant SCLC cells from IMPDH inhibition. GTP therefore functioned as a metabolic gate tethering MYC-dependent ribosome biogenesis to nucleotide sufficiency through GPN1 and GPN3. IMPDH dependence is a targetable vulnerability in chemoresistant MYChi SCLC.
- Published
- 2021
- Full Text
- View/download PDF
8. Identification of Rho GEF and RhoA Activation by Pull-Down Assays.
- Author
-
Sajib MS, Zahra FT, Akwii RG, and Mikelis CM
- Subjects
- GTPase-Activating Proteins isolation & purification, Guanosine Diphosphate genetics, Guanosine Triphosphate genetics, Humans, Protein Binding genetics, Rho Guanine Nucleotide Exchange Factors genetics, Rho Guanine Nucleotide Exchange Factors isolation & purification, rhoA GTP-Binding Protein isolation & purification, GTPase-Activating Proteins genetics, Molecular Biology methods, rhoA GTP-Binding Protein genetics
- Abstract
The small GTPase RhoA participates in actin and microtubule machinery, cell migration and invasion, gene expression, vesicular trafficking and cell cycle, and its dysregulation is a determining factor in many pathological conditions. Similar to other Rho GTPases, RhoA is a key component of the wound-healing process, regulating the activity of different participating cell types. RhoA gets activated upon binding to guanine nucleotide exchange factors (GEFs), which catalyze the exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP). GTPase-activating proteins (GAPs) mediate the exchange of GTP to GDP, inactivating RhoA, whereas guanine nucleotide dissociation inhibitors (GDIs) preserve the inactive pool of RhoA proteins in the cytosol. RhoA and Rho GEF activation is detected by protein pull-down assays, which use chimeric proteins with Rhotekin and G17A mutant RhoA as "bait" to pull down active RhoA and RhoA GEFs, respectively. In this chapter, we describe an optimized protocol for performing RhoA and GEF pull-down assays.
- Published
- 2021
- Full Text
- View/download PDF
9. Genetic investigation of purine nucleotide imbalance in Saccharomyces cerevisiae.
- Author
-
Saint-Marc C, Ceschin J, Almyre C, Pinson B, and Daignan-Fornier B
- Subjects
- Guanosine Triphosphate genetics, Humans, Nucleotides genetics, Phenotype, Saccharomyces cerevisiae genetics, AMP Deaminase genetics, Amino Acid Transport Systems genetics, Aminohydrolases genetics, Purine Nucleosides genetics, Saccharomyces cerevisiae Proteins genetics
- Abstract
Because metabolism is a complex balanced process involving multiple enzymes, understanding how organisms compensate for transient or permanent metabolic imbalance is a challenging task that can be more easily achieved in simpler unicellular organisms. The metabolic balance results not only from the combination of individual enzymatic properties, regulation of enzyme abundance, but also from the architecture of the metabolic network offering multiple interconversion alternatives. Although metabolic networks are generally highly resilient to perturbations, metabolic imbalance resulting from enzymatic defect and specific environmental conditions can be designed experimentally and studied. Starting with a double amd1 aah1 mutant that severely and conditionally affects yeast growth, we carefully characterized the metabolic shuffle associated with this defect. We established that the GTP decrease resulting in an adenylic/guanylic nucleotide imbalance was responsible for the growth defect. Identification of several gene dosage suppressors revealed that TAT1, encoding an amino acid transporter, is a robust suppressor of the amd1 aah1 growth defect. We show that TAT1 suppression occurs through replenishment of the GTP pool in a process requiring the histidine biosynthesis pathway. Importantly, we establish that a tat1 mutant exhibits synthetic sickness when combined with an amd1 mutant and that both components of this synthetic phenotype can be suppressed by specific gene dosage suppressors. Together our data point to a strong phenotypic connection between amino acid uptake and GTP synthesis, a connection that could open perspectives for future treatment of related human defects, previously reported as etiologically highly conserved.
- Published
- 2020
- Full Text
- View/download PDF
10. Peculiarities in Activation of Hydrolytic Activity of Elongation Factors.
- Author
-
Paleskava A, Kaiumov MY, Kirillov SV, and Konevega AL
- Subjects
- Guanosine Triphosphate genetics, Hydrolysis, Peptide Elongation Factors genetics, Ribosomes genetics, Guanosine Triphosphate metabolism, Peptide Elongation Factors metabolism, Protein Biosynthesis, Ribosomes metabolism
- Abstract
Translational GTPases (trGTPases) belong to the family of G proteins and play key roles at all stages of protein biosynthesis on the ribosome. Unidirectional and cyclic functioning of G proteins is ensured by their ability to switch between the active and inactive states due to GTP hydrolysis accelerated by the auxiliary GTPase-activating proteins. Although trGTPases interact with the ribosomes in different conformational states, they bind to the same conserved region, which, unlike in classical GTPase-activating proteins, is represented by ribosomal RNA. The resulting catalytic sites have almost identical structure in all elongation factors suggesting a common mechanism of GTP hydrolysis. However, fine details of the activated state formation and significantly different rates of GTP hydrolysis indicate the existence of distinctive features upon GTP hydrolysis catalyzed by the different factors. Here, we present a contemporary view on the mechanism of GTPase activation and GTP hydrolysis by the elongation factors EF-Tu, EF-G, and SelB based on the analysis of structural, biochemical, and bioinformatics data.
- Published
- 2020
- Full Text
- View/download PDF
11. A ribosomal RNA fragment with 2',3'-cyclic phosphate and GTP-binding activity acts as RIG-I ligand.
- Author
-
Jung S, von Thülen T, Yang I, Laukemper V, Rupf B, Janga H, Panagiotidis GD, Schoen A, Nicolai M, Schulte LN, Obermann HL, Weber F, Kaufmann A, and Bauer S
- Subjects
- Guanosine Triphosphate genetics, Humans, Ligands, Phosphates metabolism, RNA chemistry, RNA Helicases metabolism, Receptors, Immunologic, Ribonucleases genetics, DEAD Box Protein 58 genetics, RNA genetics, RNA Helicases genetics, RNA, Ribosomal genetics
- Abstract
The RNA helicase RIG-I plays a key role in sensing pathogen-derived RNA. Double-stranded RNA structures bearing 5'-tri- or diphosphates are commonly referred to as activating RIG-I ligands. However, endogenous RNA fragments generated during viral infection via RNase L also activate RIG-I. Of note, RNase-digested RNA fragments bear a 5'-hydroxyl group and a 2',3'-cyclic phosphate. How endogenous RNA fragments activate RIG-I despite the lack of 5'-phosphorylation has not been elucidated. Here we describe an endogenous RIG-I ligand (eRL) that is derived from the internal transcribed spacer 2 region (ITS2) of the 45S ribosomal RNA after partial RNase A digestion in vitro, RNase A protein transfection or RNase L activation. The immunostimulatory property of the eRL is dependent on 2',3'-cyclic phosphate and its sequence is characterized by a G-quadruplex containing sequence motif mediating guanosine-5'-triphosphate (GTP) binding. In summary, RNase generated self-RNA fragments with 2',3'-cyclic phosphate function as nucleotide-5'-triphosphate binding aptamers activating RIG-I., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
12. Requirement of GTP binding for TIF-90-regulated ribosomal RNA synthesis and oncogenic activities in human colon cancer cells.
- Author
-
Nguyen DQ, Hoang DH, Nelson M, Nigam L, Nguyen VTT, Zhang L, Pham TKT, Ho HD, Nguyen DDT, Lam TQ, Tat TT, Elhajmoussa Y, Ly QT, Pichiorri F, Pullarkat V, Zhang B, Kuo YH, Marcucci G, and Nguyen LXT
- Subjects
- Cell Line, Tumor, Cell Proliferation genetics, DNA, Ribosomal genetics, HCT116 Cells, Humans, RNA Polymerase I genetics, Signal Transduction genetics, Carcinogenesis genetics, Colonic Neoplasms genetics, Guanosine Triphosphate genetics, RNA, Ribosomal genetics, Ribosomes genetics, Transcription Factors genetics, Transcription, Genetic genetics
- Abstract
Transcription initiation factor 90 (TIF-90), an alternatively spliced variant of TIF-IA, differs by a 90 base pair deletion of exon 6. TIF-90 has been shown to regulate ribosomal RNA (rRNA) synthesis by interacting with polymerase I (Pol I) during the initiation of ribosomal DNA (rDNA) transcription in the nucleolus. Recently, we showed that TIF-90-mediated rRNA synthesis can play an important role in driving tumorigenesis in human colon cancer cells. Here we show that TIF-90 binds GTP at threonine 310, and that GTP binding is required for TIF-90-enhanced rRNA synthesis. Overexpression of activated AKT induces TIF-90 T310, but not a GTP-binding site (TIF-90 T310N) mutant, to translocate into the nucleolus and increase rRNA synthesis. Complementing this result, treatment with mycophenolic acid (MPA), an inhibitor of GTP production, dissociates TIF-90 from Pol I and hence abolishes AKT-increased rRNA synthesis by way of TIF-90 activation. Thus, TIF-90 requires bound GTP to fulfill its function as an enhancer of rRNA synthesis. Both TIF variants are highly expressed in colon cancer cells, and depletion of TIF-IA expression in these cells results in significant sensitivity to MPA-inhibited rRNA synthesis and reduced cell proliferation. Finally, a combination of MPA and AZD8055 (an inhibitor of both AKT and mTOR) synergistically inhibits rRNA synthesis, in vivo tumor growth, and other oncogenic activities of primary human colon cancer cells, suggesting a potential avenue for the development of therapeutic treatments by targeting the regulation of rRNA synthesis by TIF proteins., (© 2020 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
13. A revised mechanism for (p)ppGpp synthesis by Rel proteins: The critical role of the 2'-OH of GTP.
- Author
-
Patil PR, Vithani N, Singh V, Kumar A, and Prakash B
- Subjects
- Bacterial Proteins genetics, Guanosine Pentaphosphate genetics, Guanosine Triphosphate genetics, Ligases genetics, Magnesium metabolism, Streptococcus genetics, Bacterial Proteins metabolism, Gene Expression Regulation, Bacterial, Guanosine Pentaphosphate biosynthesis, Guanosine Triphosphate metabolism, Ligases metabolism, Streptococcus metabolism
- Abstract
Bacterial Rel proteins synthesize hyperphosphorylated guanosine nucleotides, denoted as (p)ppGpp, which by inhibiting energy requiring molecular pathways help bacteria to overcome the depletion of nutrients in its surroundings. (p)ppGpp synthesis by Rel involves transferring a pyrophosphate from ATP to the oxygen of 3'-OH of GTP/GDP. Initially, a conserved glutamate at the active site was believed to generate the nucleophile necessary to accomplish the reaction. Later this role was alluded to a Mg
2+ ion. However, no study has unequivocally established a catalytic mechanism for (p)ppGpp synthesis. Here we present a revised mechanism, wherein for the first time we explore a role for 2'-OH of GTP and show how it is important in generating the nucleophile. Through a careful comparison of substrate-bound structures of Rel, we illustrate that the active site does not discriminate GTP from dGTP, for a substrate. Using biochemical studies, we demonstrate that both GTP and dGTP bind to Rel, but only GTP (but not dGTP) can form the product. Reactions performed using GTP analogs substituted with different chemical moieties at the 2' position suggest a clear role for 2'-OH in catalysis by providing an indispensable hydrogen bond; preliminary computational analysis further supports this view. This study elucidating a catalytic role for 2'-OH of GTP in (p)ppGpp synthesis allows us to propose different mechanistic possibilities by which it generates the nucleophile for the synthesis reaction. This study underscores the selection of ribose nucleotides as second messengers and finds its roots in the old RNA world hypothesis., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Patil et al.)- Published
- 2020
- Full Text
- View/download PDF
14. The molecular basis for immune dysregulation by the hyperactivated E62K mutant of the GTPase RAC2.
- Author
-
Arrington ME, Temple B, Schaefer A, and Campbell SL
- Subjects
- 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, Guanosine Triphosphate chemistry, Mutation, Missense, rac GTP-Binding Proteins chemistry
- 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 (RAC2
E62K ) 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., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Arrington et al.)- Published
- 2020
- Full Text
- View/download PDF
15. Disparate Phenotypes Resulting from Mutations of a Single Histidine in Switch II of Geobacillus stearothermophilus Translation Initiation Factor IF2.
- Author
-
Tomsic J, Smorlesi A, Caserta E, Giuliodori AM, Pon CL, and Gualerzi CO
- Subjects
- Amino Acid Substitution genetics, GTP Phosphohydrolases genetics, Guanosine Triphosphate genetics, Phenotype, Protein Biosynthesis genetics, Protein Domains genetics, RNA, Transfer, Met genetics, Ribosomes genetics, Bacterial Proteins genetics, Geobacillus stearothermophilus genetics, Histidine genetics, Mutation genetics, Peptide Initiation Factors genetics
- Abstract
The conserved Histidine 301 in switch II of Geobacillus stearothermophilus IF2 G2 domain was substituted with Ser, Gln, Arg, Leu and Tyr to generate mutants displaying different phenotypes. Overexpression of IF2H301S, IF2H301L and IF2H301Y in cells expressing wtIF2, unlike IF2H301Q and IF2H301R, caused a dominant lethal phenotype, inhibiting in vivo translation and drastically reducing cell viability. All mutants bound GTP but, except for IF2H301Q, were inactive in ribosome-dependent GTPase for different reasons. All mutants promoted 30S initiation complex (30S IC) formation with wild type (wt) efficiency but upon 30S IC association with the 50S subunit, the fMet-tRNA reacted with puromycin to different extents depending upon the IF2 mutant present in the complex (wtIF2 to IF2H301Q > IF2H301R >>> IF2H301S, IF2H301L and IF2H301Y) whereas only fMet-tRNA 30S-bound with IF2H301Q retained some ability to form initiation dipeptide fMet-Phe. Unlike wtIF2, all mutants, regardless of their ability to hydrolyze GTP, displayed higher affinity for the ribosome and failed to dissociate from the ribosomes upon 50S docking to 30S IC. We conclude that different amino acids substitutions of His301 cause different structural alterations of the factor, resulting in disparate phenotypes with no direct correlation existing between GTPase inactivation and IF2 failure to dissociate from ribosomes., Competing Interests: The authors declare no conflict of interest
- Published
- 2020
- Full Text
- View/download PDF
16. Converting GTP hydrolysis into motion: versatile translational elongation factor G.
- Author
-
Rodnina MV, Peske F, Peng BZ, Belardinelli R, and Wintermeyer W
- Subjects
- GTP Phosphohydrolases genetics, Guanosine Triphosphate genetics, Hydrolysis, Peptide Elongation Factor G biosynthesis, RNA, Messenger genetics, RNA, Transfer genetics, Guanosine Triphosphate biosynthesis, Peptide Elongation Factor G genetics, Protein Biosynthesis genetics, Ribosomes genetics
- Abstract
Elongation factor G (EF-G) is a translational GTPase that acts at several stages of protein synthesis. Its canonical function is to catalyze tRNA movement during translation elongation, but it also acts at the last step of translation to promote ribosome recycling. Moreover, EF-G has additional functions, such as helping the ribosome to maintain the mRNA reading frame or to slide over non-coding stretches of the mRNA. EF-G has an unconventional GTPase cycle that couples the energy of GTP hydrolysis to movement. EF-G facilitates movement in the GDP-Pi form. To convert the energy of hydrolysis to movement, it requires various ligands in the A site, such as a tRNA in translocation, an mRNA secondary structure element in ribosome sliding, or ribosome recycling factor in post-termination complex disassembly. The ligand defines the direction and timing of EF-G-facilitated motion. In this review, we summarize recent advances in understanding the mechanism of EF-G action as a remarkable force-generating GTPase.
- Published
- 2019
- Full Text
- View/download PDF
17. Biosynthesis of the human milk oligosaccharide 3-fucosyllactose in metabolically engineered Escherichia coli via the salvage pathway through increasing GTP synthesis and β-galactosidase modification.
- Author
-
Choi YH, Park BS, Seo JH, and Kim BG
- Subjects
- Guanosine Diphosphate Fucose genetics, Guanosine Diphosphate Fucose metabolism, Humans, Trisaccharides genetics, Trisaccharides metabolism, Escherichia coli genetics, Escherichia coli metabolism, Escherichia coli Proteins genetics, Escherichia coli Proteins metabolism, Guanosine Triphosphate biosynthesis, Guanosine Triphosphate genetics, Metabolic Engineering, Milk, Human chemistry, Oligosaccharides biosynthesis, Oligosaccharides chemistry, Oligosaccharides genetics, beta-Galactosidase genetics, beta-Galactosidase metabolism
- Abstract
3-Fucosyllactose (3-FL) is one of the major fucosylated oligosaccharides in human milk. Along with 2'-fucosyllactose (2'-FL), it is known for its prebiotic, immunomodulator, neonatal brain development, and antimicrobial function. Whereas the biological production of 2'-FL has been widely studied and made significant progress over the years, the biological production of 3-FL has been hampered by the low activity and insoluble expression of α-1,3-fucosyltransferase (FutA), relatively low abundance in human milk oligosaccharides compared with 2'-FL, and lower digestibility of 3-FL than 2'-FL by bifidobacteria. In this study, we report the gram-scale production of 3-FL using E. coli BL21(DE3). We previously generated the FutA quadruple mutant (mFutA) with four site mutations at S46F, A128N, H129E, Y132I, and its specific activity was increased by nearly 15 times compared with that of wild-type FutA owing to the increase in k
cat and the decrease in Km . We overexpressed mFutA in its maximum expression level, which was achieved by the optimization of yeast extract concentration in culture media. We also overexpressed L-fucokinase/GDP- L-fucose pyrophosphorylase to increase the supply of GDP-fucose in the cytoplasm. To increase the mass of recombinant whole-cell catalysts, the host E. coli BW25113 was switched to E. coli BL21(DE3) because of the lower acetate accumulation of E. coli BL21(DE3) than that of E. coli BW25113. Finally, the lactose operon was modified by partially deleting the sequence of LacZ (lacZΔm15) for better utilization of D-lactose. Production using the lacZΔm15 mutant yielded 3-FL concentration of 4.6 g/L with the productivity of 0.076 g·L-1 ·hr-1 and the specific 3-FL yield of 0.5 g/g dry cell weight., (© 2019 Wiley Periodicals, Inc.)- Published
- 2019
- Full Text
- View/download PDF
18. Engineering the Substrate Transport and Cofactor Regeneration Systems for Enhancing 2'-Fucosyllactose Synthesis in Bacillus subtilis .
- Author
-
Deng J, Gu L, Chen T, Huang H, Yin X, Lv X, Liu Y, Li N, Liu Z, Li J, Du G, and Liu L
- Subjects
- Fermentation genetics, Fucose genetics, Fucosyltransferases genetics, Guanosine Diphosphate genetics, Guanosine Triphosphate genetics, Helicobacter pylori genetics, Lactose genetics, Metabolic Engineering methods, Milk, Human metabolism, Oligosaccharides genetics, Bacillus subtilis genetics, Regeneration genetics, Trisaccharides genetics
- Abstract
Human milk oligosaccharides (HMOs) have been proven to be beneficial to infants' intestinal health and immune systems. 2'-Fucosyllactose (2'-FL) is the most abundant and thoroughly studied HMO and has been approved to be an additive of infant formula. How to construct efficient and safe microbial cell factories for the production of 2'-FL attracts increasing attention. In this work, we engineered the Bacillus subtilis as an efficient 2'-FL producer by engineering the substrate transport and cofactor guanosine 5'-triphosphate (GTP) regeneration systems. First, we constructed a synthesis pathway for the 2'-FL precursor guanosine 5'-diphosphate-l-fucose (GDP-l-fucose) by introducing the salvage pathway gene fkp from Bacteriodes fragilis and improved the fucose importation by overexpressing the transporters. Then, the complete synthesis pathway of 2'-FL was constructed by introducing the heterologous fucosyltransferases from different sources, and it was found that the gene from Helicobacter pylori was the best one for 2'-FL synthesis. We also improved the substrate lactose importation by introducing heterologous lactose permeases and eliminated endogenous β-galactosidase ( yesZ ) to block the lactose degradation. Next, the production of 2'-FL and GDP-l-fucose was improved by fine-tuning the expression of cofactor guanosine 5'-triphosphate regeneration module genes gmd , ndk , guaA , guaC , ykfN , deoD , and xpt . Finally, a 3 L fed-batch fermentation was performed, and the highest 2'-FL titer reached 5.01 g/L with a yield up to 0.85 mol/mol fucose. We optimized the synthesis modules of 2'-FL in B. subtilis , and this provides a good starting point for metabolic engineering to further improve 2'-FL production in the future.
- Published
- 2019
- Full Text
- View/download PDF
19. The trafficking protein JFC1 regulates Rac1-GTP localization at the uropod controlling neutrophil chemotaxis and in vivo migration.
- Author
-
Ramadass M, Johnson JL, Marki A, Zhang J, Wolf D, Kiosses WB, Pestonjamasp K, Ley K, and Catz SD
- Subjects
- Animals, Chemotaxis genetics, Guanosine Triphosphate genetics, Inflammation genetics, Inflammation immunology, Inflammation pathology, Membrane Proteins genetics, Mice, Mice, Knockout, Neuropeptides genetics, Neutrophils pathology, Pseudopodia genetics, Pseudopodia pathology, Vesicular Transport Proteins genetics, rab27 GTP-Binding Proteins genetics, rab27 GTP-Binding Proteins immunology, rac1 GTP-Binding Protein genetics, Chemotaxis immunology, Guanosine Triphosphate immunology, Membrane Proteins immunology, Neuropeptides immunology, Neutrophils immunology, Pseudopodia immunology, Vesicular Transport Proteins immunology, rac1 GTP-Binding Protein immunology
- Abstract
Neutrophil chemotaxis is essential in responses to infection and underlies inflammation. In neutrophils, the small GTPase Rac1 has discrete functions at both the leading edge and in the retraction of the trailing structure at the cell's rear (uropod), but how Rac1 is regulated at the uropod is unknown. Here, we identified a mechanism mediated by the trafficking protein synaptotagmin-like 1 (SYTL1 or JFC1) that controls Rac1-GTP recycling from the uropod and promotes directional migration of neutrophils. JFC1-null neutrophils displayed defective polarization and impaired directional migration to N-formyl-methionine-leucyl-phenylalanine in vitro, but chemoattractant-induced actin remodeling, calcium signaling and Erk activation were normal in these cells. Defective chemotaxis was not explained by impaired azurophilic granule exocytosis associated with JFC1 deficiency. Mechanistically, we show that active Rac1 localizes at dynamic vesicles where endogenous JFC1 colocalizes with Rac1-GTP. Super-resolution microscopy (STORM) analysis shows adjacent distribution of JFC1 and Rac1-GTP, which increases upon activation. JFC1 interacts with Rac1-GTP in a Rab27a-independent manner to regulate Rac1-GTP trafficking. JFC1-null cells exhibited Rac1-GTP accumulation at the uropod and increased tail length, and Rac1-GTP uropod accumulation was recapitulated by inhibition of ROCK or by interference with microtubule remodeling. In vivo, neutrophil dynamic studies in mixed bone marrow chimeric mice show that JFC1
-/- neutrophils are unable to move directionally toward the source of the chemoattractant, supporting the notion that JFC1 deficiency results in defective neutrophil migration. Our results suggest that defective Rac1-GTP recycling from the uropod affects directionality and highlight JFC1-mediated Rac1 trafficking as a potential target to regulate chemotaxis in inflammation and immunity., (©2019 Society for Leukocyte Biology.)- Published
- 2019
- Full Text
- View/download PDF
20. Determining selection free energetics from nucleotide pre-insertion to insertion in viral T7 RNA polymerase transcription fidelity control.
- Author
-
Long C, E C, Da LT, and Yu J
- Subjects
- Adenosine Triphosphate genetics, Bacteriophage T7 enzymology, Guanosine Triphosphate genetics, Kinetics, Molecular Dynamics Simulation, Nucleotides genetics, Substrate Specificity, Bacteriophage T7 genetics, DNA-Directed RNA Polymerases genetics, Transcription, Genetic, Viral Proteins genetics, Virus Replication genetics
- Abstract
An elongation cycle of a transcribing RNA polymerase (RNAP) usually consists of multiple kinetics steps, so there exist multiple kinetic checkpoints where non-cognate nucleotides can be selected against. We conducted comprehensive free energy calculations on various nucleotide insertions for viral T7 RNAP employing all-atom molecular dynamics simulations. By comparing insertion free energy profiles between the non-cognate nucleotide species (rGTP and dATP) and a cognate one (rATP), we obtained selection free energetics from the nucleotide pre-insertion to the insertion checkpoints, and further inferred the selection energetics down to the catalytic stage. We find that the insertion of base mismatch rGTP proceeds mainly through an off-path along which both pre-insertion screening and insertion inhibition play significant roles. In comparison, the selection against dATP is found to go through an off-path pre-insertion screening along with an on-path insertion inhibition. Interestingly, we notice that two magnesium ions switch roles of leave and stay during the dATP on-path insertion. Finally, we infer that substantial selection energetic is still required to catalytically inhibit the mismatched rGTP to achieve an elongation error rate ∼10-4 or lower; while no catalytic selection seems to be further needed against dATP to obtain an error rate ∼10-2., (© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
21. Translation termination depends on the sequential ribosomal entry of eRF1 and eRF3.
- Author
-
Beißel C, Neumann B, Uhse S, Hampe I, Karki P, and Krebber H
- Subjects
- Codon, Terminator genetics, Guanosine Triphosphate genetics, Protein Binding genetics, Protein Biosynthesis genetics, RNA, Transfer genetics, Ribosomes genetics, Saccharomyces cerevisiae genetics, DEAD-box RNA Helicases genetics, Nucleocytoplasmic Transport Proteins genetics, Peptide Chain Termination, Translational, Peptide Termination Factors genetics, Saccharomyces cerevisiae Proteins genetics
- Abstract
Translation termination requires eRF1 and eRF3 for polypeptide- and tRNA-release on stop codons. Additionally, Dbp5/DDX19 and Rli1/ABCE1 are required; however, their function in this process is currently unknown. Using a combination of in vivo and in vitro experiments, we show that they regulate a stepwise assembly of the termination complex. Rli1 and eRF3-GDP associate with the ribosome first. Subsequently, Dbp5-ATP delivers eRF1 to the stop codon and in this way prevents a premature access of eRF3. Dbp5 dissociates upon placing eRF1 through ATP-hydrolysis. This in turn enables eRF1 to contact eRF3, as the binding of Dbp5 and eRF3 to eRF1 is mutually exclusive. Defects in the Dbp5-guided eRF1 delivery lead to premature contact and premature dissociation of eRF1 and eRF3 from the ribosome and to subsequent stop codon readthrough. Thus, the stepwise Dbp5-controlled termination complex assembly is essential for regular translation termination events. Our data furthermore suggest a possible role of Dbp5/DDX19 in alternative translation termination events, such as during stress response or in developmental processes, which classifies the helicase as a potential drug target for nonsense suppression therapy to treat cancer and neurodegenerative diseases., (© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
22. High-resolution structure of RGS17 suggests a role for Ca 2+ in promoting the GTPase-activating protein activity by RZ subfamily members.
- Author
-
Sieng M, Hayes MP, O'Brien JB, Andrew Fowler C, Houtman JC, Roman DL, and Lyon AM
- Subjects
- Calcium metabolism, Crystallography, X-Ray, GTP-Binding Protein alpha Subunits genetics, GTP-Binding Protein alpha Subunits metabolism, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Humans, Hydrolysis, Magnesium chemistry, Magnesium metabolism, RGS Proteins genetics, RGS Proteins metabolism, Calcium chemistry, Calcium Signaling, RGS Proteins chemistry
- Abstract
Regulator of G protein signaling (RGS) proteins are negative regulators of G protein-coupled receptor (GPCR) signaling through their ability to act as GTPase-activating proteins (GAPs) for activated Gα subunits. Members of the RZ subfamily of RGS proteins bind to activated Gα
o , Gαz , and Gαi1-3 proteins in the nervous system and thereby inhibit downstream pathways, including those involved in Ca2+ -dependent signaling. In contrast to other RGS proteins, little is known about RZ subfamily structure and regulation. Herein, we present the 1.5-Å crystal structure of RGS17, the most complete and highest-resolution structure of an RZ subfamily member to date. RGS17 cocrystallized with Ca2+ bound to conserved positions on the predicted Gα-binding surface of the protein. Using NMR chemical shift perturbations, we confirmed that Ca2+ binds in solution to the same site. Furthermore, RGS17 had greater than 55-fold higher affinity for Ca2+ than for Mg2+ Finally, we found that Ca2+ promotes interactions between RGS17 and activated Gα and decreases the Km for GTP hydrolysis, potentially by altering the binding mechanism between these proteins. Taken together, these findings suggest that Ca2+ positively regulates RGS17, which may represent a general mechanism by which increased Ca2+ concentration promotes the GAP activity of the RZ subfamily, leading to RZ-mediated inhibition of Ca2+ signaling., (© 2019 Sieng et al.)- Published
- 2019
- Full Text
- View/download PDF
23. The Nucleotide-Dependent Interactome of Rice Heterotrimeric G-Protein α -Subunit.
- Author
-
Biswal AK, McConnell EW, Werth EG, Lo SF, Yu SM, Hicks LM, and Jones AM
- Subjects
- Guanosine Diphosphate genetics, Guanosine Triphosphate genetics, Nucleotides genetics, Signal Transduction genetics, Heterotrimeric GTP-Binding Proteins genetics, Oryza genetics, Protein Subunits genetics
- Abstract
The rice heterotrimeric G-protein complex, a guanine-nucleotide-dependent on-off switch, mediates vital cellular processes and responses to biotic and abiotic stress. Exchange of bound GDP (resting state) for GTP (active state) is spontaneous in plants including rice and thus there is no need for promoting guanine nucleotide exchange in vivo as a mechanism for regulating the active state of signaling as it is well known for animal G signaling. As such, a master regulator controlling the G-protein activation state is unknown in plants. Therefore, an ab initio approach is taken to discover candidate regulators. The rice Gα subunit (RGA1) is used as bait to screen for nucleotide-dependent protein partners. A total of 264 proteins are identified by tandem mass spectrometry of which 32 were specific to the GDP-bound inactive state and 22 specific to the transition state. Approximately, 10% are validated as previously identified G-protein interactors., (© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
24. Re-introducing non-optimal synonymous codons into codon-optimized constructs enhances soluble recovery of recombinant proteins from Escherichia coli.
- Author
-
Konczal J, Bower J, and Gray CH
- Subjects
- Amino Acid Sequence genetics, Centrifugation, Guanosine Triphosphate genetics, Protein Biosynthesis genetics, RNA, Transfer genetics, Recombinant Proteins chemistry, Solubility, Codon genetics, Escherichia coli genetics, Recombinant Proteins genetics, Ribosomes genetics
- Abstract
Gene synthesis services have largely superseded traditional PCR methods for the generation of cDNAs destined for bacterial expression vectors. This, in turn, has increased the application of codon-optimized cDNAs where codons rarely used by Escherchia coli are replaced with common synonymous codons to accelerate translation of the target. A markedly accelerated rate of expression often results in a significant uplift in the levels of target protein but a substantial proportion of the enhanced yield can partition to the insoluble fraction rendering a significant portion of the gains unavailable for native purification. We have assessed several expression attenuation strategies for their utility in the manipulation of the soluble fraction towards higher levels of soluble target recovery from codon optimized systems. Using a set of human small GTPases as a case study, we compare the degeneration of the T7 promoter sequence, the use of alternative translational start codons and the manipulation of synonymous codon usage. Degeneration of both the T7 promoter and the translational start codon merely depressed overall expression and did not increase the percentage of product recovered in native purification of the soluble fraction. However, the selective introduction of rare non-optimal codons back into the codon-optimized sequence resulted in significantly elevated recovery of soluble targets. We propose that slowing the rate of extension during translation using a small number of rare codons allows more time for the co-translational folding of the nascent polypeptide. This increases the proportion of the target recovered in the soluble fraction by immobilized metal affinity chromatography (IMAC). Thus, a "de-optimization" of codon-optimized cDNAs, to attenuate or pause the translation process, may prove a useful strategy for improved recombinant protein production., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
25. Parkinson's disease-associated mutations in the GTPase domain of LRRK2 impair its nucleotide-dependent conformational dynamics.
- Author
-
Wu CX, Liao J, Park Y, Reed X, Engel VA, Hoang NC, Takagi Y, Johnson SM, Wang M, Federici M, Nichols RJ, Sanishvili R, Cookson MR, and Hoang QQ
- Subjects
- Amino Acid Substitution, Guanosine Diphosphate chemistry, Guanosine Diphosphate genetics, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, HEK293 Cells, Humans, Protein Domains, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 chemistry, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Mutation, Missense, Parkinson Disease enzymology, Parkinson Disease genetics, Protein Multimerization
- Abstract
Mutation in leucine-rich repeat kinase 2 (LRRK2) is a common cause of familial Parkinson's disease (PD). Recently, we showed that a disease-associated mutation R1441H rendered the GTPase domain of LRRK2 catalytically less active and thereby trapping it in a more persistently "on" conformation. However, the mechanism involved and characteristics of this on conformation remained unknown. Here, we report that the Ras of complex protein (ROC) domain of LRRK2 exists in a dynamic dimer-monomer equilibrium that is oppositely driven by GDP and GTP binding. We also observed that the PD-associated mutations at residue 1441 impair this dynamic and shift the conformation of ROC to a GTP-bound-like monomeric conformation. Moreover, we show that residue Arg-1441 is critical for regulating the conformational dynamics of ROC. In summary, our results reveal that the PD-associated substitutions at Arg-1441 of LRRK2 alter monomer-dimer dynamics and thereby trap its GTPase domain in an activated state.
- Published
- 2019
- Full Text
- View/download PDF
26. Arg-78 of Nprl2 catalyzes GATOR1-stimulated GTP hydrolysis by the Rag GTPases.
- Author
-
Shen K, Valenstein ML, Gu X, and Sabatini DM
- Subjects
- Amino Acid Substitution, Arginine chemistry, Arginine genetics, Arginine metabolism, GTPase-Activating Proteins chemistry, GTPase-Activating Proteins genetics, GTPase-Activating Proteins metabolism, Humans, Hydrolysis, Mutation, Missense, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Monomeric GTP-Binding Proteins chemistry, Monomeric GTP-Binding Proteins genetics, Monomeric GTP-Binding Proteins metabolism, Repressor Proteins chemistry, Repressor Proteins genetics, Repressor Proteins metabolism, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism
- Abstract
mTOR complex 1 (mTORC1) is a major regulator of cell growth and proliferation that coordinates nutrient inputs with anabolic and catabolic processes. Amino acid signals are transmitted to mTORC1 through the Rag GTPases, which directly recruit mTORC1 onto the lysosomal surface, its site of activation. The Rag GTPase heterodimer has a unique architecture that consists of two GTPase subunits, RagA or RagB bound to RagC or RagD. Their nucleotide-loading states are strictly controlled by several lysosomal or cytosolic protein complexes that directly detect and transmit the amino acid signals. GATOR1 (GTPase-activating protein (GAP) activity toward Rags-1), a negative regulator of the cytosolic branch of the nutrient-sensing pathway, comprises three subunits, Depdc5 (DEP domain-containing protein 5), Nprl2 (NPR2-like GATOR1 complex subunit), and Nprl3 (NPR3-like GATOR1 complex subunit), and is a GAP for RagA. GATOR1 binds the Rag GTPases via two modes: an inhibitory mode that holds the Rag GTPase heterodimer and has previously been captured by structural determination, and a GAP mode that stimulates GTP hydrolysis by RagA but remains structurally elusive. Here, using site-directed mutagenesis, GTP hydrolysis assays, coimmunoprecipitation experiments, and structural analysis, we probed the GAP mode and found that a critical residue on Nprl2, Arg-78, is the arginine finger that carries out GATOR1's GAP function. Substitutions of this arginine residue rendered mTORC1 signaling insensitive to amino acid starvation and are found frequently in cancers such as glioblastoma. Our results reveal the biochemical bases of mTORC1 inactivation through the GATOR1 complex., (© 2019 Shen et al.)
- Published
- 2019
- Full Text
- View/download PDF
27. Exposure of Candida albicans β (1,3)-glucan is promoted by activation of the Cek1 pathway.
- Author
-
Chen T, Jackson JW, Tams RN, Davis SE, Sparer TE, and Reynolds TB
- Subjects
- Cell Wall genetics, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Gene Expression Regulation, Fungal, Gene Knockout Techniques, Guanosine Triphosphate genetics, Humans, Lectins, C-Type genetics, MAP Kinase Signaling System genetics, Mitogen-Activated Protein Kinases genetics, beta-Glucans chemistry, beta-Glucans metabolism, cdc42 GTP-Binding Protein genetics, CDPdiacylglycerol-Serine O-Phosphatidyltransferase genetics, Candida albicans genetics, Fungal Proteins genetics, MAP Kinase Kinase Kinases genetics, Mitogen-Activated Protein Kinase 3 genetics
- Abstract
Candida albicans is among the most common causes of human fungal infections and is an important source of mortality. C. albicans is able to diminish its detection by innate immune cells through masking of β (1,3)-glucan in the inner cell wall with an outer layer of heavily glycosylated mannoproteins (mannan). However, mutations or drugs that disrupt the cell wall can lead to exposure of β (1,3)-glucan (unmasking) and enhanced detection by innate immune cells through receptors like Dectin-1, the C-type signaling lectin. Previously, our lab showed that the pathway for synthesizing the phospholipid phosphatidylserine (PS) plays a role in β (1,3)-glucan masking. The homozygous PS synthase knockout mutant, cho1Δ/Δ, exhibits increased exposure of β (1,3)-glucan. Several Mitogen Activated Protein Kinase (MAPK) pathways and their upstream Rho-type small GTPases are important for regulating cell wall biogenesis and remodeling. In the cho1Δ/Δ mutant, both the Cek1 and Mkc1 MAPKs are constitutively activated, and they act downstream of the small GTPases Cdc42 and Rho1, respectively. In addition, Cdc42 activity is up-regulated in cho1Δ/Δ. Thus, it was hypothesized that activation of Cdc42 or Rho1 and their downstream kinases cause unmasking. Disruption of MKC1 does not decrease unmasking in cho1Δ/Δ, and hyperactivation of Rho1 in wild-type cells increases unmasking and activation of both Cek1 and Mkc1. Moreover, independent hyperactivation of the MAP kinase kinase kinase Ste11 in wild-type cells leads to Cek1 activation and increased β (1,3)-glucan exposure. Thus, upregulation of the Cek1 MAPK pathway causes unmasking, and may be responsible for unmasking in cho1Δ/Δ., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
28. Differential GAP requirement for Cdc42-GTP polarization during proliferation and sexual reproduction.
- Author
-
Gallo Castro D and Martin SG
- Subjects
- GTPase-Activating Proteins genetics, Guanosine Triphosphate genetics, Schizosaccharomyces genetics, Schizosaccharomyces pombe Proteins genetics, cdc42 GTP-Binding Protein genetics, GTPase-Activating Proteins metabolism, Guanosine Triphosphate metabolism, Mitosis physiology, Schizosaccharomyces enzymology, Schizosaccharomyces pombe Proteins metabolism, cdc42 GTP-Binding Protein metabolism
- Abstract
The formation of a local zone of Cdc42 GTPase activity, which governs cell polarization in many cell types, requires not only local activation but also switch-off mechanisms. In this study, we identify Rga3, a paralog of Rga4, as a novel Cdc42 GTPase-activating protein (GAP) in the fission yeast Schizosaccharomyces pombe Contrary to Rga4, Rga3 localizes with Cdc42-GTP to sites of polarity. Rga3 is dispensable for cell polarization during mitotic growth, but it limits the lifetime of unstable Cdc42-GTP patches that underlie cell pairing during sexual reproduction, masking a partly compensatory patch-wandering motion. In consequence, cells lacking rga3 hyperpolarize and lose out in mating competition. Rga3 synergizes with the Cdc42 GAPs Rga4 and Rga6 to restrict Cdc42-GTP zone sizes during mitotic growth. Surprisingly, triple-mutant cells, which are almost fully round, retain pheromone-dependent dynamic polarization of Cdc42-GTP, extend a polarized projection, and mate. Thus, the requirement for Cdc42-GTP hydrolysis by GAPs is distinct during polarization by intrinsic or extrinsic cues., (© 2018 Gallo Castro and Martin.)
- Published
- 2018
- Full Text
- View/download PDF
29. GTP hydrolysis promotes disassembly of the atlastin crossover dimer during ER fusion.
- Author
-
Winsor J, Machi U, Han Q, Hackney DD, and Lee TH
- Subjects
- Endoplasmic Reticulum genetics, GTP-Binding Proteins genetics, Guanosine Triphosphate genetics, Humans, Hydrolysis, Membrane Proteins genetics, Endoplasmic Reticulum metabolism, GTP-Binding Proteins metabolism, Guanosine Triphosphate metabolism, Membrane Fusion, Membrane Proteins metabolism, Protein Multimerization
- Abstract
Membrane fusion of the ER is catalyzed when atlastin GTPases anchored in opposing membranes dimerize and undergo a crossed over conformational rearrangement that draws the bilayers together. Previous studies have suggested that GTP hydrolysis triggers crossover dimerization, thus directly driving fusion. In this study, we make the surprising observations that WT atlastin undergoes crossover dimerization before hydrolyzing GTP and that nucleotide hydrolysis and Pi release coincide more closely with dimer disassembly. These findings suggest that GTP binding, rather than its hydrolysis, triggers crossover dimerization for fusion. In support, a new hydrolysis-deficient atlastin variant undergoes rapid GTP-dependent crossover dimerization and catalyzes fusion at an initial rate similar to WT atlastin. However, the variant cannot sustain fusion activity over time, implying a defect in subunit recycling. We suggest that GTP binding induces an atlastin conformational change that favors crossover dimerization for fusion and that the input of energy from nucleotide hydrolysis promotes complex disassembly for subunit recycling., (© 2018 Winsor et al.)
- Published
- 2018
- Full Text
- View/download PDF
30. Human eIF5 and eIF1A Compete for Binding to eIF5B.
- Author
-
Lin KY, Nag N, Pestova TV, and Marintchev A
- Subjects
- Eukaryotic Initiation Factors genetics, Eukaryotic Initiation Factors metabolism, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Humans, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Protein Binding, RNA, Transfer, Amino Acyl chemistry, RNA, Transfer, Amino Acyl genetics, RNA, Transfer, Amino Acyl metabolism, Ribosomes chemistry, Ribosomes genetics, Ribosomes metabolism, Eukaryotic Initiation Factors chemistry, Neoplasm Proteins chemistry, Nerve Tissue Proteins chemistry
- Abstract
Eukaryotic translation initiation is a multistep process requiring a number of eukaryotic translation initiation factors (eIFs). Two GTPases play key roles in the process. eIF2 brings the initiator Met-tRNA
i to the preinitiation complex (PIC). Upon start codon selection and GTP hydrolysis promoted by the GTPase-activating protein (GAP) eIF5, eIF2-GDP is displaced from Met-tRNAi by eIF5B-GTP and is released in complex with eIF5. eIF5B promotes ribosomal subunit joining, with the help of eIF1A. Upon subunit joining, eIF5B hydrolyzes GTP and is released together with eIF1A. We found that human eIF5 interacts with eIF5B and may help recruit eIF5B to the PIC. An eIF5B-binding motif was identified at the C-terminus of eIF5, similar to that found in eIF1A. Indeed, eIF5 competes with eIF1A for binding and has an ∼100-fold higher affinity for eIF5B. Because eIF5 is the GAP of eIF2, the newly discovered interaction offers a possible mechanism for coordination between the two steps in translation initiation controlled by GTPases: start codon selection and ribosomal subunit joining. Our results indicate that in humans, eIF5B displacing eIF2 from Met-tRNAi upon subunit joining may be coupled to eIF1A displacing eIF5 from eIF5B, allowing the eIF5:eIF2-GDP complex to leave the ribosome.- Published
- 2018
- Full Text
- View/download PDF
31. The universally conserved GTPase HflX is an RNA helicase that restores heat-damaged Escherichia coli ribosomes.
- Author
-
Dey S, Biswas C, and Sengupta J
- Subjects
- Adenosine Triphosphatases chemistry, Adenosine Triphosphatases genetics, Escherichia coli enzymology, Escherichia coli genetics, Escherichia coli Proteins chemistry, GTP Phosphohydrolases chemistry, GTP-Binding Proteins chemistry, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Protein Binding, RNA chemistry, RNA genetics, RNA Helicases chemistry, Ribosome Subunits, Large, Bacterial enzymology, Ribosomes enzymology, Ribosomes genetics, Escherichia coli Proteins genetics, GTP Phosphohydrolases genetics, GTP-Binding Proteins genetics, Heat-Shock Response genetics, RNA Helicases genetics
- Abstract
The ribosome-associated GTPase HflX acts as an antiassociation factor upon binding to the 50S ribosomal subunit during heat stress in Escherichia coli Although HflX is recognized as a guanosine triphosphatase, several studies have shown that the N-terminal domain 1 of HflX is capable of hydrolyzing adenosine triphosphate (ATP), but the functional role of its adenosine triphosphatase (ATPase) activity remains unknown. We demonstrate that E. coli HflX possesses ATP-dependent RNA helicase activity and is capable of unwinding large subunit ribosomal RNA. A cryo-electron microscopy structure of the 50S-HflX complex in the presence of nonhydrolyzable analogues of ATP and guanosine triphosphate hints at a mode of action for the RNA helicase and suggests the linker helical domain may have a determinant role in RNA unwinding. Heat stress results in inactivation of the ribosome, and we show that HflX can restore heat-damaged ribosomes and improve cell survival., (© 2018 Dey et al.)
- Published
- 2018
- Full Text
- View/download PDF
32. CodY-Mediated c-di-GMP-Dependent Inhibition of Mammalian Cell Invasion in Listeria monocytogenes.
- Author
-
Elbakush AM, Miller KW, and Gomelsky M
- Subjects
- Amino Acids, Branched-Chain genetics, Amino Acids, Branched-Chain metabolism, Bacterial Proteins genetics, Bacterial Proteins metabolism, Bacterial Proteins pharmacology, Cyclic GMP analysis, Cyclic GMP genetics, Cyclic GMP metabolism, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, HT29 Cells, Host-Pathogen Interactions genetics, Humans, Listeriosis microbiology, Peptide Termination Factors genetics, Promoter Regions, Genetic, Regulon, Virulence genetics, Cyclic GMP analogs & derivatives, Gene Expression Regulation, Bacterial, Listeria monocytogenes genetics, Listeria monocytogenes pathogenicity, Transcription Factors genetics
- Abstract
Elevated levels of the second messenger c-di-GMP suppress virulence in diverse pathogenic bacteria, yet mechanisms are poorly characterized. In the foodborne pathogen Listeria monocytogenes , high c-di-GMP levels inhibit mammalian cell invasion. Here, we show that invasion is impaired because of the decreased expression levels of internalin genes whose products are involved in invasion. We further show that at high c-di-GMP levels, the expression of the entire virulence regulon is suppressed, and so is the expression of the prfA gene encoding the master activator of the virulence regulon. Analysis of mechanisms controlling prfA expression pointed to the transcription factor CodY as a c-di-GMP-sensitive component. In high-c-di-GMP strains, codY gene expression is decreased, apparently due to the lower activity of CodY, which functions as an activator of codY transcription. We found that listerial CodY does not bind c-di-GMP in vitro and therefore investigated whether c-di-GMP levels affect two known cofactors of listerial CodY, branched-chain amino acids and GTP. Our manipulation of branched-chain amino acid levels did not perturb the c-di-GMP effect; however, our replacement of listerial CodY with the streptococcal CodY homolog, whose activity is GTP independent, abolished the c-di-GMP effect. The results of this study suggest that elevated c-di-GMP levels decrease the activity of the coordinator of metabolism and virulence, CodY, possibly via lower GTP levels, and that decreased CodY activity suppresses L. monocytogenes virulence by the decreased expression of the PrfA virulence regulon. IMPORTANCE Listeria monocytogenes is a pathogen causing listeriosis, a disease responsible for the highest mortality rate among foodborne diseases. Understanding how the virulence of this pathogen is regulated is important for developing treatments to decrease the frequency of listerial infections in susceptible populations. In this study, we describe the mechanism through which elevated levels of the second messenger c-di-GMP inhibit listerial invasion in mammalian cells. Inhibition is caused by the decreased activity of the transcription factor CodY that coordinates metabolism and virulence., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
33. Internally ratiometric fluorescent sensors for evaluation of intracellular GTP levels and distribution.
- Author
-
Bianchi-Smiraglia A, Rana MS, Foley CE, Paul LM, Lipchick BC, Moparthy S, Moparthy K, Fink EE, Bagati A, Hurley E, Affronti HC, Bakin AV, Kandel ES, Smiraglia DJ, Feltri ML, Sousa R, and Nikiforov MA
- Subjects
- Animals, Bacterial Proteins genetics, Cell Line, Tumor, Guanosine Triphosphate genetics, Humans, Hydrogen-Ion Concentration, Luminescent Proteins genetics, Mutation, Bacterial Proteins metabolism, Biosensing Techniques, Guanosine Triphosphate metabolism, Luminescent Proteins metabolism
- Abstract
GTP is a major regulator of multiple cellular processes, but tools for quantitative evaluation of GTP levels in live cells have not been available. We report the development and characterization of genetically encoded GTP sensors, which we constructed by inserting a circularly permuted yellow fluorescent protein (cpYFP) into a region of the bacterial G protein FeoB that undergoes a GTP-driven conformational change. GTP binding to these sensors results in a ratiometric change in their fluorescence, thereby providing an internally normalized response to changes in GTP levels while minimally perturbing those levels. Mutations introduced into FeoB to alter its affinity for GTP created a series of sensors with a wide dynamic range. Critically, in mammalian cells the sensors showed consistent changes in ratiometric signal upon depletion or restoration of GTP pools. We show that these GTP evaluators (GEVALs) are suitable for detection of spatiotemporal changes in GTP levels in living cells and for high-throughput screening of molecules that modulate GTP levels.
- Published
- 2017
- Full Text
- View/download PDF
34. Native kinesin-1 does not bind preferentially to GTP-tubulin-rich microtubules in vitro.
- Author
-
Li Q, King SJ, and Xu J
- Subjects
- Animals, Cattle, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Kinesins genetics, Kinesins metabolism, Microtubules genetics, Microtubules metabolism, Protein Binding, Protein Processing, Post-Translational, Tubulin genetics, Tubulin metabolism, Guanosine Triphosphate chemistry, Kinesins chemistry, Microtubules chemistry, Tubulin chemistry
- Abstract
Molecular motors such as kinesin-1 work in small teams to actively shuttle cargos in cells, for example in polarized transport in axons. Here, we examined the potential regulatory role of the nucleotide state of tubulin on the run length of cargos carried by multiple kinesin motors, using an optical trapping-based in vitro assay. Based on a previous report that kinesin binds preferentially to GTP-tubulin-rich microtubules, we anticipated that multiple-kinesin cargos would run substantially greater distances along GMPCPP microtubules than along GDP microtubules. Surprisingly, we did not uncover any significant differences in run length between microtubule types. A combination of single-molecule experiments, comparison with previous theory, and classic microtubule affinity pulldown assays revealed that native kinesin-1 does not bind preferentially to GTP-tubulin-rich microtubules. The apparent discrepancy between our observations and the previous report likely reflects differences in post-translational modifications between the native motors used here and the recombinant motors examined previously. Future investigations will help shed light on the interplay between the motor's post-translational modification and the microtubule's nucleotide-binding state for transport regulation in vivo., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
35. GTP binding regulates cellular localization of Parkinson's disease-associated LRRK2.
- Author
-
Blanca Ramírez M, Lara Ordóñez AJ, Fdez E, Madero-Pérez J, Gonnelli A, Drouyer M, Chartier-Harlin MC, Taymans JM, Bubacco L, Greggio E, and Hilfiker S
- Subjects
- GTP Phosphohydrolases metabolism, GTP-Binding Proteins genetics, GTP-Binding Proteins metabolism, Genetic Variation, Guanosine Triphosphate genetics, HEK293 Cells, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 antagonists & inhibitors, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Microtubules genetics, Microtubules metabolism, Mutation, Parkinson Disease genetics, Phosphorylation, Protein Kinase Inhibitors pharmacology, Signal Transduction, Guanosine Triphosphate metabolism, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Parkinson Disease metabolism
- Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) comprise the most common cause of familial Parkinson's disease (PD), and sequence variants modify risk for sporadic PD. Previous studies indicate that LRRK2 interacts with microtubules (MTs) and alters MT-mediated vesicular transport processes. However, the molecular determinants within LRRK2 required for such interactions have remained unknown. Here, we report that most pathogenic LRRK2 mutants cause relocalization of LRRK2 to filamentous structures which colocalize with a subset of MTs, and an identical relocalization is seen upon pharmacological LRRK2 kinase inhibition. The pronounced colocalization with MTs does not correlate with alterations in LRRK2 kinase activity, but rather with increased GTP binding. Synthetic mutations which impair GTP binding, as well as LRRK2 GTP-binding inhibitors profoundly interfere with the abnormal localization of both pathogenic mutant as well as kinase-inhibited LRRK2. Conversely, addition of a non-hydrolyzable GTP analog to permeabilized cells enhances the association of pathogenic or kinase-inhibited LRRK2 with MTs. Our data elucidate the mechanism underlying the increased MT association of select pathogenic LRRK2 mutants or of pharmacologically kinase-inhibited LRRK2, with implications for downstream MT-mediated transport events., (© The Author 2017. Published by Oxford University Press.)
- Published
- 2017
- Full Text
- View/download PDF
36. Recurrent Mutations in the MTOR Regulator RRAGC in Follicular Lymphoma.
- Author
-
Ying ZX, Jin M, Peterson LF, Bernard D, Saiya-Cork K, Yildiz M, Wang S, Kaminski MS, Chang AE, Klionsky DJ, and Malek SN
- Subjects
- Amino Acids genetics, Binding Sites genetics, Cell Line, Genes, Tumor Suppressor physiology, Guanosine Diphosphate genetics, Guanosine Triphosphate genetics, HEK293 Cells, Humans, Intracellular Signaling Peptides and Proteins genetics, Phosphorylation genetics, Regulatory-Associated Protein of mTOR genetics, Signal Transduction genetics, Lymphoma, Follicular genetics, Monomeric GTP-Binding Proteins genetics, Mutation genetics, Neoplasm Recurrence, Local genetics, TOR Serine-Threonine Kinases genetics
- Abstract
Purpose: This study was performed to further our understanding of the biological and genetic basis of follicular lymphoma and to identify potential novel therapy targets., Experimental Design: We analyzed previously generated whole exome sequencing data of 23 follicular lymphoma cases and one transformed follicular lymphoma case and expanded findings to a combined total of 125 follicular lymphoma/3 transformed follicular lymphoma. We modeled the three-dimensional location of RRAGC-associated hotspot mutations. We performed functional studies on novel RRAGC mutants in stable retrovirally transduced HEK293T cells, stable lentivirally transduced lymphoma cell lines, and in Saccharomyces cerevisiae RESULTS: We report recurrent mutations, including multiple amino acid hotspots, in the small G-protein RRAGC, which is part of a protein complex that signals intracellular amino acid concentrations to MTOR, in 9.4% of follicular lymphoma cases. Mutations in RRAGC distinctly clustered on one protein surface area surrounding the GTP/GDP-binding sites. Mutated RRAGC proteins demonstrated increased binding to RPTOR (raptor) and substantially decreased interactions with the product of the tumor suppressor gene FLCN (folliculin). In stable retrovirally transfected 293T cells, cultured in the presence or absence of leucine, multiple RRAGC mutations demonstrated elevated MTOR activation as evidenced by increased RPS6KB/S6-kinase phosphorylation. Similar activation phenotypes were uncovered in yeast engineered to express mutations in the RRAGC homolog Gtr2 and in multiple lymphoma cell lines expressing HA-tagged RRAGC-mutant proteins., Conclusions: Our discovery of activating mutations in RRAGC in approximately 10% of follicular lymphoma provides the mechanistic rationale to study mutational MTOR activation and MTOR inhibition as a potential novel actionable therapeutic target in follicular lymphoma. Clin Cancer Res; 22(21); 5383-93. ©2016 AACR., Competing Interests: There is no conflict of interest to disclose., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
37. A Conserved Hydrophobic Core in Gαi1 Regulates G Protein Activation and Release from Activated Receptor.
- Author
-
Kaya AI, Lokits AD, Gilbert JA, Iverson TM, Meiler J, and Hamm HE
- Subjects
- Enzyme Activation, GTP-Binding Protein alpha Subunits, Gi-Go genetics, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Guanosine Diphosphate genetics, Guanosine Diphosphate metabolism, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Humans, Hydrophobic and Hydrophilic Interactions, Protein Structure, Secondary, GTP-Binding Protein alpha Subunits, Gi-Go chemistry, Guanosine Diphosphate chemistry, Guanosine Triphosphate chemistry
- Abstract
G protein-coupled receptor-mediated heterotrimeric G protein activation is a major mode of signal transduction in the cell. Previously, we and other groups reported that the α5 helix of Gαi1, especially the hydrophobic interactions in this region, plays a key role during nucleotide release and G protein activation. To further investigate the effect of this hydrophobic core, we disrupted it in Gαi1 by inserting 4 alanine amino acids into the α5 helix between residues Gln(333) and Phe(334) (Ins4A). This extends the length of the α5 helix without disturbing the β6-α5 loop interactions. This mutant has high basal nucleotide exchange activity yet no receptor-mediated activation of nucleotide exchange. By using structural approaches, we show that this mutant loses critical hydrophobic interactions, leading to significant rearrangements of side chain residues His(57), Phe(189), Phe(191), and Phe(336); it also disturbs the rotation of the α5 helix and the π-π interaction between His(57) and Phe(189) In addition, the insertion mutant abolishes G protein release from the activated receptor after nucleotide binding. Our biochemical and computational data indicate that the interactions between α5, α1, and β2-β3 are not only vital for GDP release during G protein activation, but they are also necessary for proper GTP binding (or GDP rebinding). Thus, our studies suggest that this hydrophobic interface is critical for accurate rearrangement of the α5 helix for G protein release from the receptor after GTP binding., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
38. The Globular Tail Domain of Myosin-5a Functions as a Dimer in Regulating the Motor Activity.
- Author
-
Zhang WB, Yao LL, and Li XD
- Subjects
- Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing genetics, Amino Acid Substitution, Guanosine Diphosphate chemistry, Guanosine Diphosphate genetics, Guanosine Diphosphate metabolism, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Mutation, Missense, Myosin Heavy Chains chemistry, Myosin Heavy Chains genetics, Myosin Type V chemistry, Myosin Type V genetics, Protein Domains, Protein Structure, Quaternary, Adaptor Proteins, Signal Transducing metabolism, Myosin Heavy Chains metabolism, Myosin Type V metabolism, Protein Folding, Protein Multimerization physiology
- Abstract
Myosin-5a contains two heavy chains, which are dimerized via the coiled-coil regions. Thus, myosin-5a comprises two heads and two globular tail domains (GTDs). The GTD is the inhibitory domain that binds to the head and inhibits its motor function. Although the two-headed structure is essential for the processive movement of myosin-5a along actin filaments, little is known about the role of GTD dimerization. Here, we investigated the effect of GTD dimerization on its inhibitory activity. We found that the potent inhibitory activity of the GTD is dependent on its dimerization by the preceding coiled-coil regions, indicating synergistic interactions between the two GTDs and the two heads of myosin-5a. Moreover, we found that alanine mutations of the two conserved basic residues at N-terminal extension of the GTD not only weaken the inhibitory activity of the GTD but also enhance the activation of myosin-5a by its cargo-binding protein melanophilin (Mlph). These results are consistent with the GTD forming a head to head dimer, in which the N-terminal extension of the GTD interacts with the Mlph-binding site in the counterpart GTD. The Mlph-binding site at the GTD-GTD interface must be exposed prior to the binding of Mlph. We therefore propose that the inhibited Myo5a is equilibrated between the folded state, in which the Mlph-binding site is buried, and the preactivated state, in which the Mlph-binding site is exposed, and that Mlph is able to bind to the Myo5a in preactivated state and activates its motor function., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
39. Enhancement of β-catenin activity by BIG1 plus BIG2 via Arf activation and cAMP signals.
- Author
-
Li CC, Le K, Kato J, Moss J, and Vaughan M
- Subjects
- A Kinase Anchor Proteins genetics, A Kinase Anchor Proteins metabolism, ADP-Ribosylation Factors genetics, Cyclic AMP genetics, Cyclic AMP-Dependent Protein Kinases genetics, Cyclic AMP-Dependent Protein Kinases metabolism, Guanine Nucleotide Exchange Factors genetics, Guanosine Diphosphate genetics, Guanosine Diphosphate metabolism, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, HeLa Cells, Humans, Phospholipase D genetics, Phospholipase D metabolism, Phosphorylation physiology, Protein Domains, beta Catenin genetics, ADP-Ribosylation Factors metabolism, Cyclic AMP metabolism, Guanine Nucleotide Exchange Factors metabolism, Second Messenger Systems physiology, beta Catenin metabolism
- Abstract
Multifunctional β-catenin, with critical roles in both cell-cell adhesion and Wnt-signaling pathways, was among HeLa cell proteins coimmunoprecipitated by antibodies against brefeldin A-inhibited guanine nucleotide-exchange factors 1 and 2 (BIG1 or BIG2) that activate ADP-ribosylation factors (Arfs) by accelerating the replacement of bound GDP with GTP. BIG proteins also contain A-kinase anchoring protein (AKAP) sequences that can act as scaffolds for multimolecular assemblies that facilitate and limit cAMP signaling temporally and spatially. Direct interaction of BIG1 N-terminal sequence with β-catenin was confirmed using yeast two-hybrid assays and in vitro synthesized proteins. Depletion of BIG1 and/or BIG2 or overexpression of guanine nucleotide-exchange factor inactive mutant, but not wild-type, proteins interfered with β-catenin trafficking, leading to accumulation at perinuclear Golgi structures. Both phospholipase D activity and vesicular trafficking were required for effects of BIG1 and BIG2 on β-catenin activation. Levels of PKA-phosphorylated β-catenin S675 and β-catenin association with PKA, BIG1, and BIG2 were also diminished after BIG1/BIG2 depletion. Inferring a requirement for BIG1 and/or BIG2 AKAP sequence in PKA modification of β-catenin and its effect on transcription activation, we confirmed dependence of S675 phosphorylation and transcription coactivator function on BIG2 AKAP-C sequence.
- Published
- 2016
- Full Text
- View/download PDF
40. Neutron Crystal Structure of RAS GTPase Puts in Question the Protonation State of the GTP γ-Phosphate.
- Author
-
Knihtila R, Holzapfel G, Weiss K, Meilleur F, and Mattos C
- Subjects
- Catalysis, Guanosine Triphosphate analogs & derivatives, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Humans, Hydrolysis, Protein Structure, Tertiary, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Guanosine Triphosphate chemistry, Neutrons, Proto-Oncogene Proteins p21(ras) chemistry
- Abstract
RAS GTPase is a prototype for nucleotide-binding proteins that function by cycling between GTP and GDP, with hydrogen atoms playing an important role in the GTP hydrolysis mechanism. It is one of the most well studied proteins in the superfamily of small GTPases, which has representatives in a wide range of cellular functions. These proteins share a GTP-binding pocket with highly conserved motifs that promote hydrolysis to GDP. The neutron crystal structure of RAS presented here strongly supports a protonated γ-phosphate at physiological pH. This counters the notion that the phosphate groups of GTP are fully deprotonated at the start of the hydrolysis reaction, which has colored the interpretation of experimental and computational data in studies of the hydrolysis mechanism. The neutron crystal structure presented here puts in question our understanding of the pre-catalytic state associated with the hydrolysis reaction central to the function of RAS and other GTPases., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
41. Distinct profiles of functional discrimination among G proteins determine the actions of G protein-coupled receptors.
- Author
-
Masuho I, Ostrovskaya O, Kramer GM, Jones CD, Xie K, and Martemyanov KA
- Subjects
- Animals, GTP-Binding Protein alpha Subunits genetics, Guanosine Triphosphate genetics, Humans, Mice, Receptors, G-Protein-Coupled genetics, GTP-Binding Protein alpha Subunits metabolism, Guanosine Triphosphate metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction physiology
- Abstract
Members of the heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptor (GPCR) family play key roles in many physiological functions and are extensively exploited pharmacologically to treat diseases. Many of the diverse effects of individual GPCRs on cellular physiology are transduced by heterotrimeric G proteins, which are composed of α, β, and γ subunits. GPCRs interact with and stimulate the binding of guanosine triphosphate (GTP) to the α subunit to initiate signaling. Mammalian genomes encode 16 different G protein α subunits, each one of which has distinct properties. We developed a single-platform, optical strategy to monitor G protein activation in live cells. With this system, we profiled the coupling ability of individual GPCRs for different α subunits, simultaneously quantifying the magnitude of the signal and the rates at which the receptors activated the G proteins. We found that individual receptors engaged multiple G proteins with varying efficacy and kinetics, generating fingerprint-like profiles. Different classes of GPCR ligands, including full and partial agonists, allosteric modulators, and antagonists, distinctly affected these fingerprints to functionally bias GPCR signaling. Finally, we showed that intracellular signaling modulators further altered the G protein-coupling profiles of GPCRs, which suggests that their differential abundance may alter signaling outcomes in a cell-specific manner. These observations suggest that the diversity of the effects of GPCRs on cellular physiology may be determined by their differential engagement of multiple G proteins, coupling to which produces signals with varying signal magnitudes and activation kinetics, properties that may be exploited pharmacologically., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
42. GTP Binding and Oncogenic Mutations May Attenuate Hypervariable Region (HVR)-Catalytic Domain Interactions in Small GTPase K-Ras4B, Exposing the Effector Binding Site.
- Author
-
Lu S, Banerjee A, Jang H, Zhang J, Gaponenko V, and Nussinov R
- Subjects
- Amino Acid Substitution, Guanosine Diphosphate genetics, Guanosine Diphosphate metabolism, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Humans, Protein Structure, Tertiary, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Guanosine Diphosphate chemistry, Guanosine Triphosphate chemistry, Mutation, Proto-Oncogene Proteins p21(ras) chemistry
- Abstract
K-Ras4B, a frequently mutated oncogene in cancer, plays an essential role in cell growth, differentiation, and survival. Its C-terminal membrane-associated hypervariable region (HVR) is required for full biological activity. In the active GTP-bound state, the HVR interacts with acidic plasma membrane (PM) headgroups, whereas the farnesyl anchors in the membrane; in the inactive GDP-bound state, the HVR may interact with both the PM and the catalytic domain at the effector binding region, obstructing signaling and nucleotide exchange. Here, using molecular dynamics simulations and NMR, we aim to figure out the effects of nucleotides (GTP and GDP) and frequent (G12C, G12D, G12V, G13D, and Q61H) and infrequent (E37K and R164Q) oncogenic mutations on full-length K-Ras4B. The mutations are away from or directly at the HVR switch I/effector binding site. Our results suggest that full-length wild-type GDP-bound K-Ras4B (K-Ras4B(WT)-GDP) is in an intrinsically autoinhibited state via tight HVR-catalytic domain interactions. The looser association in K-Ras4B(WT)-GTP may release the HVR. Some of the oncogenic mutations weaken the HVR-catalytic domain association in the K-Ras4B-GDP/-GTP bound states, which may facilitate the HVR disassociation in a nucleotide-independent manner, thereby up-regulating oncogenic Ras signaling. Thus, our results suggest that mutations can exert their effects in more than one way, abolishing GTP hydrolysis and facilitating effector binding., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
43. From GTP and G proteins to TRPC channels: a personal account.
- Author
-
Birnbaumer L
- Subjects
- Animals, GTP-Binding Proteins chemistry, GTP-Binding Proteins genetics, Guanosine Triphosphate genetics, Humans, Signal Transduction genetics, Signal Transduction physiology, TRPC Cation Channels chemistry, TRPC Cation Channels genetics, GTP-Binding Proteins metabolism, Guanosine Triphosphate metabolism, TRPC Cation Channels metabolism
- Abstract
By serendipity and good fortune, as a postdoctoral fellow in 1967, I landed at the right place at the right time, as I was allowed to investigate the mechanism by which hormones activate the enzyme adenylyl cyclase (then adenyl cyclase) in Martin Rodbell's Laboratory at the NIH in Bethesda, Maryland. The work uncovered first, the existence of receptors separate from the enzyme and then, the existence of transduction mechanisms requiring guanosine-5'-triphosphate (GTP) and Mg(2+). With my laboratory colleagues first and postdoctoral fellows after leaving NIH, I participated in the development of the field "signal transduction by G proteins," uncovered by molecular cloning several G-protein-coupled receptors (GPCRs) and became interested in both the molecular makeup of voltage-gated Ca channels and Ca2+ homeostasis downstream of activation of phospholipase C (PLC) by the Gq/11 signaling pathway. We were able to confirm the hypothesis that there would be mammalian homologues of the Drosophila "transient receptor potential" channel and discovered the existence of six of the seven mammalian genes, now called transient receptor potential canonical (TRPC) channels. In the present article, I summarize from a bird's eye view of what I feel were key findings along this path, not only from my laboratory but also from many others, that allowed for the present knowledge of cell signaling involving G proteins to evolve. Towards the end, I summarize roles of TRPC channels in health and disease.
- Published
- 2015
- Full Text
- View/download PDF
44. The motor function of Drosophila melanogaster myosin-5 is activated by calcium and cargo-binding protein dRab11.
- Author
-
Ji HH, Zhang HM, Shen M, Yao LL, and Li XD
- Subjects
- Animals, Drosophila Proteins genetics, Drosophila melanogaster, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Myosins genetics, Rhodopsin genetics, Sf9 Cells, Spodoptera, rab GTP-Binding Proteins genetics, Calcium metabolism, Calcium Signaling physiology, Drosophila Proteins metabolism, Myosins metabolism, Rhodopsin metabolism, rab GTP-Binding Proteins metabolism
- Abstract
In the Drosophila melanogaster compound eye, myosin-5 (DmM5) plays two distinct roles in response to light stimulation: transport of pigment granules to the rhabdomere base to decrease light exposure and transport of rhodopsin-bearing vesicles to the rhabdomere base to compensate for the rhodopsin loss during light exposure. However, little is known of how the motor function of DmM5 is regulated at the molecular level. In the present study, we overexpressed DmM5 in Sf9 insect cells and investigated its regulation using purified proteins. We found that the actin-activated ATPase activity of DmM5 is significantly lower than that of the truncated DmM5 having the C-terminal globular tail domain (GTD) deleted, indicating that the GTD is the inhibitory domain. The actin-activated ATPase activity of DmM5 is significantly activated by micromolar levels of calcium. DmM5 associates with pigment granules and rhodopsin-bearing vesicles through cargo-binding proteins Lightoid (Ltd) and dRab11 respectively. We found that GTP-bound dRab11, but not Ltd, significantly activates DmM5 actin-activated ATPase activity. Moreover, we identified Gln(1689) in the GTD as the critical residue for the interaction with dRab11 and activation of DmM5 motor function by dRab11. Based on those results, we propose that DmM5-dependent transport of pigment granules is directly activated by light-induced calcium influx and the DmM5-dependent transport of rhodopsin-bearing vesicle is activated by active GTP-bound dRab11, whose formation is stimulated by light-induced calcium influx., (© 2015 Authors; published by Portland Press Limited.)
- Published
- 2015
- Full Text
- View/download PDF
45. UreE-UreG complex facilitates nickel transfer and preactivates GTPase of UreG in Helicobacter pylori.
- Author
-
Yang X, Li H, Lai TP, and Sun H
- Subjects
- Bacterial Proteins genetics, Bacterial Proteins metabolism, Biological Transport, Active physiology, Carrier Proteins genetics, Carrier Proteins metabolism, GTP Phosphohydrolases genetics, GTP Phosphohydrolases metabolism, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Helicobacter pylori genetics, Helicobacter pylori metabolism, Metallochaperones, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Mutagenesis, Nickel metabolism, Phosphate-Binding Proteins, Protein Binding, Urease chemistry, Urease genetics, Urease metabolism, Bacterial Proteins chemistry, Carrier Proteins chemistry, GTP Phosphohydrolases chemistry, Helicobacter pylori chemistry, Multiprotein Complexes chemistry, Nickel chemistry
- Abstract
The pathogenicity of Helicobacter pylori relies heavily on urease, which converts urea to ammonia to neutralize the stomach acid. Incorporation of Ni(2+) into the active site of urease requires a battery of chaperones. Both metallochaperones UreE and UreG play important roles in the urease activation. In this study, we demonstrate that, in the presence of GTP and Mg(2+), UreG binds Ni(2+) with an affinity (Kd) of ∼0.36 μm. The GTPase activity of Ni(2+)-UreG is stimulated by both K(+) (or NH4 (+)) and HCO3 (-) to a biologically relevant level, suggesting that K(+)/NH4 (+) and HCO3 (-) might serve as GTPase elements of UreG. We show that complexation of UreE and UreG results in two protein complexes, i.e. 2E-2G and 2E-G, with the former being formed only in the presence of both GTP and Mg(2+). Mutagenesis studies reveal that Arg-101 on UreE and Cys-66 on UreG are critical for stabilization of 2E-2G complex. Combined biophysical and bioassay studies show that the formation of 2E-2G complex not only facilitates nickel transfer from UreE to UreG, but also enhances the binding of GTP. This suggests that UreE might also serve as a structural scaffold for recruitment of GTP to UreG. Importantly, we demonstrate for the first time that UreE serves as a bridge to grasp Ni(2+) from HypA, subsequently donating it to UreG. The study expands our horizons on the molecular details of nickel translocation among metallochaperones UreE, UreG, and HypA, which further extends our knowledge on the urease maturation process., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
46. Unlike catalyzing error-free bypass of 8-oxodGuo, DNA polymerase λ is responsible for a significant part of Fapy·dG-induced G → T mutations in human cells.
- Author
-
Pande P, Haraguchi K, Jiang YL, Greenberg MM, and Basu AK
- Subjects
- Catalysis, DNA biosynthesis, DNA genetics, DNA Polymerase beta metabolism, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Humans, Thymine Nucleotides genetics, Thymine Nucleotides metabolism, DNA chemistry, DNA Polymerase beta chemistry, Guanosine Triphosphate analogs & derivatives, Point Mutation, Thymine Nucleotides chemistry
- Abstract
8-OxodGuo and Fapy·dG induced 10-22% mutations, predominantly G → T transversions, in human embryonic kidney 293T cells in four TG*N sequence contexts, where N = C, G, A, or T. siRNA knockdown of pol λ resulted in 34 and 55% increases in the level of mutations in the progeny from the 8-oxodGuo construct in the TG*T and TG*G sequences, respectively, suggesting that pol λ is involved in error-free bypass of 8-oxodGuo. For Fapy·dG, in contrast, the level of G → T mutations was reduced by 27 and 46% in the TG*T and TG*G sequences, respectively, suggesting that pol λ is responsible for a significant fraction of Fapy·dG-induced G → T mutations.
- Published
- 2015
- Full Text
- View/download PDF
47. Distinct features of cap binding by eIF4E1b proteins.
- Author
-
Kubacka D, Miguel RN, Minshall N, Darzynkiewicz E, Standart N, and Zuberek J
- Subjects
- Animals, Binding Sites genetics, Carrier Proteins genetics, Cloning, Molecular, Eukaryotic Initiation Factor-4E genetics, Gene Expression Regulation, Guanosine Triphosphate analogs & derivatives, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Humans, Models, Molecular, Protein Conformation, RNA, Messenger genetics, RNA, Messenger metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Sepharose analogs & derivatives, Sepharose chemistry, Sepharose genetics, Sequence Alignment, Xenopus laevis, Carrier Proteins metabolism, Eukaryotic Initiation Factor-4E metabolism, Protein Binding
- Abstract
eIF4E1b, closely related to the canonical translation initiation factor 4E (eIF4E1a), cap-binding protein is highly expressed in mouse, Xenopus and zebrafish oocytes. We have previously characterized eIF4E1b as a component of the CPEB mRNP translation repressor complex along with the eIF4E-binding protein 4E-Transporter, the Xp54/DDX6 RNA helicase and additional RNA-binding proteins. eIF4E1b exhibited only very weak interactions with m(7)GTP-Sepharose and, rather than binding eIF4G, interacted with 4E-T. Here we undertook a detailed examination of both Xenopus and human eIF4E1b interactions with cap analogues using fluorescence titration and homology modeling. The predicted structure of eIF4E1b maintains the α+β fold characteristic of eIF4E proteins and its cap-binding pocket is similarly arranged by critical amino acids: Trp56, Trp102, Glu103, Trp166, Arg112, Arg157 and Lys162 and residues of the C-terminal loop. However, we demonstrate that eIF4E1b is 3-fold less well able to bind the cap than eIF4E1a, both proteins being highly stimulated by methylation at N(7) of guanine. Moreover, eIF4E1b proteins are distinguishable from eIF4E1a by a set of conserved amino acid substitutions, several of which are located near to cap-binding residues. Indeed, eIF4E1b possesses several distinct features, namely, enhancement of cap binding by a benzyl group at N(7) position of guanine, a reduced response to increasing length of the phosphate chain and increased binding to a cap separated by a linker from Sepharose, suggesting differences in the arrangement of the protein's core. In agreement, mutagenesis of the amino acids differentiating eIF4E1b from eIF4E1a reduces cap binding by eIF4E1a 2-fold, demonstrating their role in modulating cap binding., (Copyright © 2014. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
48. Modeling the role of G12V and G13V Ras mutations in the Ras-GAP-catalyzed hydrolysis reaction of guanosine triphosphate.
- Author
-
Khrenova MG, Mironov VA, Grigorenko BL, and Nemukhin AV
- Subjects
- Amino Acid Sequence, Catalysis, GTP-Binding Protein alpha Subunits, G12-G13 chemistry, Guanosine Triphosphate chemistry, Guanosine Triphosphate genetics, Hydrolysis, Molecular Sequence Data, ras GTPase-Activating Proteins chemistry, ras GTPase-Activating Proteins genetics, GTP-Binding Protein alpha Subunits, G12-G13 physiology, Guanosine Triphosphate metabolism, Models, Molecular, Mutation physiology, ras GTPase-Activating Proteins metabolism
- Abstract
Cancer-associated point mutations in Ras, in particular, at glycine 12 and glycine 13, affect the normal cycle between inactive GDP-bound and active GTP-bound states. In this work, the role of G12V and G13V replacements in the GAP-stimulated intrinsic GTP hydrolysis reaction in Ras is studied using molecular dynamics (MD) simulations with quantum mechanics/molecular mechanics (QM/MM) potentials. A model molecular system was constructed by motifs of the relevant crystal structure (Protein Data Bank entry 1WQ1 ). QM/MM optimization of geometry parameters in the Ras-GAP-GTP complex and QM/MM-MD simulations were performed with a quantum subsystem comprising a large fraction of the enzyme active site. For the system with wild-type Ras, the conformations fluctuated near the structure ready to be involved in the efficient chemical reaction leading to the cleavage of the phosphorus-oxygen bond in GTP upon approach of the properly aligned catalytic water molecule. Dynamics of the system with the G13V mutant is characterized by an enhanced flexibility in the area occupied by the γ-phosphate group of GTP, catalytic water, and the side chains of Arg789 and Gln61, which should somewhat hinder fast chemical steps. Conformational dynamics of the system with the G12V mutant shows considerable displacement of the Gln61 side chain and catalytic water from their favorable arrangement in the active site that may lead to a marked reduction in the reaction rate. The obtained computational results correlate well with the recent kinetic measurements of the Ras-GAP-catalyzed hydrolysis of GTP.
- Published
- 2014
- Full Text
- View/download PDF
49. Human CalDAG-GEFI gene (RASGRP2) mutation affects platelet function and causes severe bleeding.
- Author
-
Canault M, Ghalloussi D, Grosdidier C, Guinier M, Perret C, Chelghoum N, Germain M, Raslova H, Peiretti F, Morange PE, Saut N, Pillois X, Nurden AT, Cambien F, Pierres A, van den Berg TK, Kuijpers TW, Alessi MC, and Tregouet DA
- Subjects
- Adenosine Diphosphate genetics, Adenosine Diphosphate metabolism, Cell Line, Female, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Heterozygote, Homozygote, Humans, Male, Megakaryocytes metabolism, Megakaryocytes pathology, Membrane Proteins genetics, Membrane Proteins metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Platelet Glycoprotein GPIIb-IIIa Complex, Protein Kinase C genetics, Protein Kinase C metabolism, Shelterin Complex, Telomere-Binding Proteins genetics, Telomere-Binding Proteins metabolism, Blood Coagulation Disorders, Inherited genetics, Blood Coagulation Disorders, Inherited metabolism, Blood Coagulation Disorders, Inherited pathology, Blood Platelets metabolism, Blood Platelets pathology, Guanine Nucleotide Exchange Factors genetics, Guanine Nucleotide Exchange Factors metabolism, Hemorrhage genetics, Hemorrhage metabolism, Hemorrhage pathology, Mutation, Platelet Aggregation genetics
- Abstract
The nature of an inherited platelet disorder was investigated in three siblings affected by severe bleeding. Using whole-exome sequencing, we identified the culprit mutation (cG742T) in the RAS guanyl-releasing protein-2 (RASGRP2) gene coding for calcium- and DAG-regulated guanine exchange factor-1 (CalDAG-GEFI). Platelets from individuals carrying the mutation present a reduced ability to activate Rap1 and to perform proper αIIbβ3 integrin inside-out signaling. Expression of CalDAG-GEFI mutant in HEK293T cells abolished Rap1 activation upon stimulation. Nevertheless, the PKC- and ADP-dependent pathways allow residual platelet activation in the absence of functional CalDAG-GEFI. The mutation impairs the platelet's ability to form thrombi under flow and spread normally as a consequence of reduced Rac1 GTP-binding. Functional deficiencies were confined to platelets and megakaryocytes with no leukocyte alteration. This contrasts with the phenotype seen in type III leukocyte adhesion deficiency caused by the absence of kindlin-3. Heterozygous did not suffer from bleeding and have normal platelet aggregation; however, their platelets mimicked homozygous ones by failing to undergo normal adhesion under flow and spreading. Rescue experiments on cultured patient megakaryocytes corrected the functional deficiency after transfection with wild-type RASGRP2. Remarkably, the presence of a single normal allele is sufficient to prevent bleeding, making CalDAG-GEFI a novel and potentially safe therapeutic target to prevent thrombosis., (© 2014 Canault et al.)
- Published
- 2014
- Full Text
- View/download PDF
50. Anillin regulates cell-cell junction integrity by organizing junctional accumulation of Rho-GTP and actomyosin.
- Author
-
Reyes CC, Jin M, Breznau EB, Espino R, Delgado-Gonzalo R, Goryachev AB, and Miller AL
- Subjects
- Actins genetics, Actins metabolism, Actomyosin metabolism, Animals, Contractile Proteins metabolism, Embryo, Nonmammalian metabolism, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Myosin Type II genetics, Myosin Type II metabolism, Rho Factor metabolism, Xenopus laevis embryology, Xenopus laevis metabolism, Actomyosin genetics, Contractile Proteins genetics, Intercellular Junctions metabolism, Rho Factor genetics, Xenopus laevis genetics
- Abstract
Anillin is a scaffolding protein that organizes and stabilizes actomyosin contractile rings and was previously thought to function primarily in cytokinesis [1-10]. Using Xenopus laevis embryos as a model system to examine Anillin's role in the intact vertebrate epithelium, we find that a population of Anillin surprisingly localizes to epithelial cell-cell junctions throughout the cell cycle, whereas it was previously thought to be nuclear during interphase [5, 11]. Furthermore, we show that Anillin plays a critical role in regulating cell-cell junction integrity. Both tight junctions and adherens junctions are disrupted when Anillin is knocked down, leading to altered cell shape and increased intercellular spaces. Anillin interacts with Rho, F-actin, and myosin II [3, 8, 9], all of which regulate cell-cell junction structure and function. When Anillin is knocked down, active Rho (Rho-guanosine triphosphate [GTP]), F-actin, and myosin II are misregulated at junctions. Indeed, increased dynamic "flares" of Rho-GTP are observed at cell-cell junctions, whereas overall junctional F-actin and myosin II accumulation is reduced when Anillin is depleted. We propose that Anillin is required for proper Rho-GTP distribution at cell-cell junctions and for maintenance of a robust apical actomyosin belt, which is required for cell-cell junction integrity. These results reveal a novel role for Anillin in regulating epithelial cell-cell junctions., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.