Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 104
1.
Biophys J ; 123(1): 57-67, 2024 01 02.
Article En | MEDLINE | ID: mdl-37978802

Rho-specific guanine nucleotide dissociation inhibitors (RhoGDIs) play a crucial role in the regulation of Rho family GTPases. They act as negative regulators that prevent the activation of Rho GTPases by forming complexes with the inactive GDP-bound state of GTPase. Release of Rho GTPase from the RhoGDI-bound complex is necessary for Rho GTPase activation. Biochemical studies provide evidence of a "phosphorylation code," where phosphorylation of some specific residues of RhoGDI selectively releases its GTPase partner (RhoA, Rac1, Cdc42, etc.). This work attempts to understand the molecular mechanism behind this specific phosphorylation-induced reduction in binding affinity. Using several microseconds long atomistic molecular dynamics simulations of the wild-type and phosphorylated states of the RhoA-RhoGDI complex, we propose a molecular-interaction-based mechanistic model for the dissociation of the complex. Phosphorylation induces major structural changes, particularly in the positively charged polybasic region (PBR) of RhoA and the negatively charged N-terminal region of RhoGDI that contribute most to the binding affinity. Molecular mechanics Poisson-Boltzmann surface area binding energy calculations show a significant weakening of interaction on phosphorylation at the RhoA-specific site of RhoGDI. In contrast, phosphorylation at a Rac1-specific site does not affect the overall binding affinity significantly, which confirms the presence of a phosphorylation code. RhoA-specific phosphorylation leads to a reduction in the number of contacts between the PBR of RhoA and the N-terminal region of RhoGDI, which manifests a reduction of the binding affinity. Using hydrogen bond occupancy analysis and energetic perturbation network, we propose a mechanistic model for the allosteric response, i.e., long-range signal propagation from the site of phosphorylation to the PBR and buried geranylgeranyl group in the form of rearrangement and rewiring of hydrogen bonds and salt bridges. Our results highlight the crucial role of specific electrostatic interactions in manifestation of the phosphorylation code.


Guanine Nucleotide Dissociation Inhibitors , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , Phosphorylation , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , Protein Binding , rhoA GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
2.
FEBS Lett ; 597(6): 836-849, 2023 03.
Article En | MEDLINE | ID: mdl-36658753

RhoGTPases are well known for being controllers of cell cytoskeleton and share common features in the way they act and are controlled. These include their switch from GDP to GTP states, their regulations by different guanine exchange factors (GEFs), GTPase-activating proteins and guanosine dissociation inhibitors (GDIs), and their similar structure of active sites/membrane anchors. These very similar features often lead to the common consideration that the differences in their biological effects mainly arise from the different types of regulators and specific effectors associated with each GTPase. Focusing on data obtained through biosensors, live cell microscopy and recent optogenetic approaches, we highlight in this review that the regulation of RhoA appears to depart from Cdc42 and Rac1 modes of regulation through its enhanced lability at the plasma membrane. RhoA presents a high dynamic turnover at the membrane that is regulated not only by GDIs but also by GEFs, effectors and a possible soluble conformational state. This peculiarity of RhoA regulation may be important for the specificities of its functions, such as the existence of activity waves or its putative dual role in the initiation of protrusions and contractions.


Guanine Nucleotide Exchange Factors , rhoA GTP-Binding Protein , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Guanine Nucleotide Exchange Factors/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/metabolism
3.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article En | MEDLINE | ID: mdl-34830380

Three decades of research have documented the spatiotemporal dynamics of RHO family GTPase membrane extraction regulated by guanine nucleotide dissociation inhibitors (GDIs), but the interplay of the kinetic mechanism and structural specificity of these interactions is as yet unresolved. To address this, we reconstituted the GDI-controlled spatial segregation of geranylgeranylated RHO protein RAC1 in vitro. Various biochemical and biophysical measurements provided unprecedented mechanistic details for GDI function with respect to RHO protein dynamics. We determined that membrane extraction of RHO GTPases by GDI occurs via a 3-step mechanism: (1) GDI non-specifically associates with the switch regions of the RHO GTPases; (2) an electrostatic switch determines the interaction specificity between the C-terminal polybasic region of RHO GTPases and two distinct negatively-charged clusters of GDI1; (3) a non-specific displacement of geranylgeranyl moiety from the membrane sequesters it into a hydrophobic cleft, effectively shielding it from the aqueous milieu. This study substantially extends the model for the mechanism of GDI-regulated RHO GTPase extraction from the membrane, and could have implications for clinical studies and drug development.


Prenylation/drug effects , rac1 GTP-Binding Protein/chemistry , rho GTP-Binding Proteins/chemistry , rho-Specific Guanine Nucleotide Dissociation Inhibitors/chemistry , Amino Acid Sequence/genetics , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Kinetics , Static Electricity , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/genetics , rho-Specific Guanine Nucleotide Dissociation Inhibitors/genetics
4.
Biochemistry ; 60(19): 1533-1551, 2021 05 18.
Article En | MEDLINE | ID: mdl-33913706

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


rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , HEK293 Cells , Humans , Monomeric GTP-Binding Proteins/metabolism , Protein Binding , rho GTP-Binding Proteins/chemistry , rho Guanine Nucleotide Dissociation Inhibitor alpha/physiology , rho Guanine Nucleotide Dissociation Inhibitor beta/physiology , rho Guanine Nucleotide Dissociation Inhibitor gamma/physiology , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/physiology
5.
Small GTPases ; 10(3): 227-242, 2019 05.
Article En | MEDLINE | ID: mdl-29065764

The small GTPase Rab5 is the key regulator of early endosomal fusion. It is post-translationally modified by covalent attachment of two geranylgeranyl (GG) chains to adjacent cysteine residues of the C-terminal hypervariable region (HVR). The GDP dissociation inhibitor (GDI) recognizes membrane-associated Rab5(GDP) and serves to release it into the cytoplasm where it is kept in a soluble state. A detailed new structural and dynamic model for human Rab5(GDP) recognition and binding with human GDI at the early endosome membrane and in its dissociated state is presented. In the cytoplasm, the GDI protein accommodates the GG chains in a transient hydrophobic binding pocket. In solution, two different binding modes of the isoprenoid chains inserted into the hydrophobic pocket of the Rab5(GDP):GDI complex can be identified. This equilibrium between the two states helps to stabilize the protein-protein complex in solution. Interprotein contacts between the Rab5 switch regions and characteristic patches of GDI residues from the Rab binding platform (RBP) and the C-terminus coordinating region (CCR) reveal insight on the formation of such a stable complex. GDI binding to membrane-anchored Rab5(GDP) is initially mediated by the solvent accessible switch regions of the Rab-specific RBP. Formation of the membrane-associated Rab5(GDP):GDI complex induces a GDI reorientation to establish additional interactions with the Rab5 HVR. These results allow to devise a detailed structural model for the process of extraction of GG-Rab5(GDP) by GDI from the membrane and the dissociation from targeting factors and effector proteins prior to GDI binding.


Diterpenes/chemistry , Guanine Nucleotide Dissociation Inhibitors/chemistry , Molecular Dynamics Simulation , Multiprotein Complexes , Protein Prenylation , rab5 GTP-Binding Proteins/chemistry , Animals , Cattle , Diterpenes/metabolism , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanine Nucleotide Dissociation Inhibitors/metabolism , Humans , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
6.
Biochemistry ; 57(47): 6562-6569, 2018 11 27.
Article En | MEDLINE | ID: mdl-30406994

GoLoco motif-containing proteins regulate the nucleotide-binding state of Gα proteins in various signaling pathways. As guanine nucleotide dissociation inhibitors (GDIs), they bind Gα·GDP and inhibit GDP to GTP exchange. GoLoco proteins show binding selectivity toward different members of the Gα family. Although the Gαi1·GDP/RGS14 crystal structure explains the specific binding selectivity of the RGS14 GoLoco domain well, the mechanism of selective binding has not been understood for the more general features of short GoLoco domains found in tandem arrays in proteins like GPSM2/LGN/ dPins and GPSM1/AGS3. We explored the mechanism of differential interactions of GoLoco protein LGN with hGαi3 and hGαo. By combining mutagenesis experiments and molecular dynamics simulations, we identified a residue (Asp229 in hGαi3) away from the binding interface that remarkably affects the interaction between LGN and hGαi/o. A negatively charged residue at this position is required for high binding affinity. This affinity regulation mechanism was further verified by the cases of hGαi2 and dGαo, suggesting that this pathway is conserved among members of the Gα family.


Carrier Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Protein Interaction Domains and Motifs , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cell Cycle Proteins , Crystallography, X-Ray , Drosophila , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Guanine Nucleotide Dissociation Inhibitors/chemistry , Humans , Mice , Models, Molecular , Protein Binding , Protein Conformation , Sequence Homology
7.
Nat Commun ; 9(1): 4042, 2018 10 02.
Article En | MEDLINE | ID: mdl-30279442

Controlling protein activity with chemogenetics and optogenetics has proven to be powerful for testing hypotheses regarding protein function in rapid biological processes. Controlling proteins by splitting them and then rescuing their activity through inducible reassembly offers great potential to control diverse protein activities. Building split proteins has been difficult due to spontaneous assembly, difficulty in identifying appropriate split sites, and inefficient induction of effective reassembly. Here we present an automated approach to design effective split proteins regulated by a ligand or by light (SPELL). We develop a scoring function together with an engineered domain to enable reassembly of protein halves with high efficiency and with reduced spontaneous assembly. We demonstrate SPELL by applying it to proteins of various shapes and sizes in living cells. The SPELL server (spell.dokhlab.org) offers an automated prediction of split sites.


Optogenetics , Protein Engineering/methods , Algorithms , Automation , Guanine Nucleotide Dissociation Inhibitors/chemistry , Proto-Oncogene Proteins c-vav/chemistry , Tacrolimus Binding Protein 1A/chemistry , src-Family Kinases/chemistry
8.
Nat Commun ; 9(1): 1025, 2018 03 09.
Article En | MEDLINE | ID: mdl-29523789

Asymmetric cell divisions balance stem cell proliferation and differentiation to sustain tissue morphogenesis and homeostasis. During asymmetric divisions, fate determinants and niche contacts segregate unequally between daughters, but little is known on how this is achieved mechanistically. In Drosophila neuroblasts and murine mammary stem cells, the association of the spindle orientation protein LGN with the stem cell adaptor Inscuteable has been connected to asymmetry. Here we report the crystal structure of Drosophila LGN in complex with the asymmetric domain of Inscuteable, which reveals a tetrameric arrangement of intertwined molecules. We show that Insc:LGN tetramers constitute stable cores of Par3-Insc-LGN-GαiGDP complexes, which cannot be dissociated by NuMA. In mammary stem cells, the asymmetric domain of Insc bound to LGN:GαiGDP suffices to drive asymmetric fate, and reverts aberrant symmetric divisions induced by p53 loss. We suggest a novel role for the Insc-bound pool of LGN acting independently of microtubule motors to promote asymmetric fate specification.


Adaptor Proteins, Signal Transducing/metabolism , Asymmetric Cell Division , Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/cytology , Drosophila/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Stem Cells/cytology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Drosophila/chemistry , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Molecular , Protein Binding , Stem Cells/chemistry , Stem Cells/metabolism
9.
EMBO Rep ; 18(9): 1509-1520, 2017 09.
Article En | MEDLINE | ID: mdl-28684399

In many cell types, mitotic spindle orientation relies on the canonical "LGN complex" composed of Pins/LGN, Mud/NuMA, and Gαi subunits. Membrane localization of this complex recruits motor force generators that pull on astral microtubules to orient the spindle. Drosophila Pins shares highly conserved functional domains with its two vertebrate homologs LGN and AGS3. Whereas the role of Pins and LGN in oriented divisions is extensively documented, involvement of AGS3 remains controversial. Here, we show that AGS3 is not required for planar divisions of neural progenitors in the mouse neocortex. AGS3 is not recruited to the cell cortex and does not rescue LGN loss of function. Despite conserved interactions with NuMA and Gαiin vitro, comparison of LGN and AGS3 functional domains in vivo reveals unexpected differences in the ability of these interactions to mediate spindle orientation functions. Finally, we find that Drosophila Pins is unable to substitute for LGN loss of function in vertebrates, highlighting that species-specific modulations of the interactions between components of the Pins/LGN complex are crucial in vivo for spindle orientation.


Carrier Proteins/genetics , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Spindle Apparatus/metabolism , Animals , Carrier Proteins/chemistry , Cell Cycle Proteins , Cell Division , Cell Polarity , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/genetics , Mice , Microtubules/metabolism , Neocortex/physiology , Nuclear Proteins/metabolism , Protein Binding , Protein Domains , Spindle Apparatus/genetics
10.
Gene ; 599: 78-86, 2017 Jan 30.
Article En | MEDLINE | ID: mdl-27836664

Vesicle shuttling is critical for many cellular and organismal processes, including embryonic development. GDI proteins contribute to vesicle shuttling by regulating the activity of Rab GTPases, controlling their cycling between the inactive cytosol and active membrane bound states. While identifying genes controlled by A-form DNA sequences we discovered a previously unknown member of the GDI family, GDI3. The GDI3 gene is found only in amphibians and fish and is developmentally expressed in Xenopus from neurula stages onwards in the neural plate, and subsequently in both dorsal and anterior structures. Depletion or over-expression of the GDI3 protein in Xenopus embryos gives rise to very similar phenotypes, suggesting that strict control of GDI3 protein levels is required for correct embryonic development. Our analysis suggests the evolutionary origins of GDI3 and that it is functionally distinct from GDI1. Predicted structural analysis of GDI3 suggests that the key difference between GDI1 and GDI3 lies in their lipid binding pockets.


Guanine Nucleotide Dissociation Inhibitors/metabolism , Neurogenesis/physiology , Xenopus Proteins/metabolism , Xenopus/embryology , Xenopus/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Base Sequence , Cloning, Molecular , DNA/genetics , Gene Expression Regulation, Developmental , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/genetics , Models, Molecular , Neurogenesis/genetics , Phylogeny , Xenopus/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/genetics
11.
Nat Chem Biol ; 12(10): 802-809, 2016 10.
Article En | MEDLINE | ID: mdl-27501396

Guanine-nucleotide dissociation inhibitors (GDIs) are negative regulators of Rho family GTPases that sequester the GTPases away from the membrane. Here we ask how GDI-Cdc42 interaction regulates localized Cdc42 activation for cell motility. The sensitivity of cells to overexpression of Rho family pathway components led us to a new biosensor, GDI.Cdc42 FLARE, in which Cdc42 is modified with a fluorescence resonance energy transfer (FRET) 'binding antenna' that selectively reports Cdc42 binding to endogenous GDIs. Similar antennae could also report GDI-Rac1 and GDI-RhoA interaction. Through computational multiplexing and simultaneous imaging, we determined the spatiotemporal dynamics of GDI-Cdc42 interaction and Cdc42 activation during cell protrusion and retraction. This revealed remarkably tight coordination of GTPase release and activation on a time scale of 10 s, suggesting that GDI-Cdc42 interactions are a critical component of the spatiotemporal regulation of Cdc42 activity, and not merely a mechanism for global sequestration of an inactivated pool of signaling molecules.


Fluorescence Resonance Energy Transfer , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/metabolism , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism , Binding Sites , HEK293 Cells , Humans , Spatio-Temporal Analysis
12.
Nucleic Acids Res ; 44(20): 9698-9709, 2016 Nov 16.
Article En | MEDLINE | ID: mdl-27458202

In protein synthesis translation factor eIF2 binds initiator tRNA to ribosomes and facilitates start codon selection. eIF2 GDP/GTP status is regulated by eIF5 (GAP and GDI functions) and eIF2B (GEF and GDF activities), while eIF2α phosphorylation in response to diverse signals is a major point of translational control. Here we characterize a growth suppressor mutation in eIF2ß that prevents eIF5 GDI and alters cellular responses to reduced eIF2B activity, including control of GCN4 translation. By monitoring the binding of fluorescent nucleotides and initiator tRNA to purified eIF2 we show that the eIF2ß mutation does not affect intrinsic eIF2 affinities for these ligands, neither does it interfere with eIF2 binding to 43S pre-initiation complex components. Instead we show that the eIF2ß mutation prevents eIF5 GDI stabilizing nucleotide binding to eIF2, thereby altering the off-rate of GDP from eIF2•GDP/eIF5 complexes. This enables cells to grow with reduced eIF2B GEF activity but impairs activation of GCN4 targets in response to amino acid starvation. These findings provide support for the importance of eIF5 GDI activity in vivo and demonstrate that eIF2ß acts in concert with eIF5 to prevent premature release of GDP from eIF2γ and thereby ensure tight control of protein synthesis initiation.


Eukaryotic Initiation Factor-2B/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Protein Biosynthesis , Repressor Proteins/metabolism , Amino Acid Sequence , Conserved Sequence , Eukaryotic Initiation Factor-2B/chemistry , Eukaryotic Initiation Factor-2B/genetics , Evolution, Molecular , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Mutation , Protein Binding , Protein Conformation , RNA, Transfer/genetics , RNA, Transfer/metabolism , Repressor Proteins/chemistry , Yeasts/drug effects , Yeasts/genetics , Yeasts/metabolism
13.
Angew Chem Int Ed Engl ; 55(28): 8129-33, 2016 07 04.
Article En | MEDLINE | ID: mdl-27213482

Weak transient protein-protein interactions (PPIs) play an essential role in cellular dynamics. However, it is challenging to obtain weak protein complexes owing to their short lifetime. Herein we present a general and facile method for trapping weak PPIs in an unbiased manner using proximity-induced ligations. To expand the chemical ligation spectrum, we developed novel N2N (N-terminus to N-terminus) and C2C (C-terminus to C-terminus) ligation approaches. By using N2C (N-terminus to C-terminus), N2N, and C2C ligations in one pot, the interacting proteins were linked. The weak Ypt1:GDI interaction drove C2C ligation with t1/2 of 4.8 min and near quantitative conversion. The Ypt1-GDI conjugate revealed that binding of Ypt1 G-domain causes opening of the lipid-binding site of GDI, which can accommodate one prenyl group, giving insights into Rab membrane recycling. Moreover, we used this strategy to trap the KRas homodimer, which plays an important role in Ras signaling.


Guanine Nucleotide Dissociation Inhibitors/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , rab GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , Molecular Dynamics Simulation , Protein Binding , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , rab GTP-Binding Proteins/chemistry
14.
Biosci Rep ; 35(6)2015 Oct 20.
Article En | MEDLINE | ID: mdl-26487707

Drosophila GoLoco motif-containing protein Pins is unusual in its highly efficient interaction with both GDP- and the GTP-loaded forms of the α-subunit of the heterotrimeric Go protein. We analysed the interactions of Gαo in its two nucleotide forms with GoLoco1-the first of the three GoLoco domains of Pins-and the possible structures of the resulting complexes, through combination of conventional fluorescence and FRET measurements as well as through molecular modelling. Our data suggest that the orientation of the GoLoco1 motif on Gαo significantly differs between the two nucleotide states of the latter. In other words, a rotation of the GoLoco1 peptide in respect with Gαo must accompany the nucleotide exchange in Gαo. The sterical hindrance requiring such a rotation probably contributes to the guanine nucleotide exchange inhibitor activity of GoLoco1 and Pins as a whole. Our data have important implications for the mechanisms of Pins regulation in the process of asymmetric cell divisions.


Drosophila Proteins/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanine Nucleotides/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Amino Acid Motifs/genetics , Animals , Asymmetric Cell Division/genetics , Cell Cycle Proteins , Drosophila/genetics , Drosophila Proteins/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Guanine Nucleotide Dissociation Inhibitors/chemistry , Heterotrimeric GTP-Binding Proteins/chemistry , Peptides/chemistry , Peptides/genetics
15.
Endocr J ; 62(2): 153-60, 2015.
Article En | MEDLINE | ID: mdl-25346144

Isolated adrenocorticotropin deficiency (IAD) is characterized by low or absent adrenocorticotropic hormone (ACTH) production. IAD is presumed to be caused in part by an autoimmune mechanism, and several lines of evidence have suggested the presence of anti-pituitary antibodies in IAD. However, the exact autoantigens remain unknown. The present study was designed to identify the autoantigen(s) in IAD using chromatography-tandem mass spectrometry (LC-MS/MS) analysis. Rat anterior pituitary lysate was subjected to SDS-PAGE, and immunoblotting was performed using the sera from two patients with IAD and from a healthy subject. The bands detected by the patient serum samples, but not by the healthy subject sample, were excised, in-gel digested using trypsin, and subjected to LC-MS/MS analysis. On immunoblots, a 51-kDa band in the insoluble pellet was detected by the sera from the IAD patients but not from the healthy subject. Mass spectrometric analysis revealed the 51-kDa band contained Rab guanine nucleotide dissociation inhibitor (GDI) alpha. Consistent with the mass spectrometric analysis, a recombinant full-length human Rab GDI alpha was recognized by the two IAD patient samples but not by the healthy subject sample using immunoblotting. In total, anti-Rab GDI alpha antibodies were detected in serum samples from three of five patients with IAD (60%) but were absent in 5 healthy subjects. In addition, Rab GDI alpha was expressed in the anterior pituitary. In conclusion, it appears that Rab GDI alpha is a candidate autoantigen involved in IAD, and that anti-Rab GDI alpha antibodies are present predominantly in patients with IAD.


Adrenocorticotropic Hormone/deficiency , Autoantibodies/analysis , Autoantigens/metabolism , Autoimmune Diseases/metabolism , Autoimmunity , Endocrine System Diseases/metabolism , Genetic Diseases, Inborn/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Hypoglycemia/metabolism , Pituitary Gland, Anterior/metabolism , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/immunology , Adrenocorticotropic Hormone/metabolism , Adult , Aged , Animals , Antibody Specificity , Autoantigens/chemistry , Autoantigens/genetics , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Endocrine System Diseases/blood , Endocrine System Diseases/immunology , Female , Genetic Diseases, Inborn/blood , Genetic Diseases, Inborn/immunology , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Hypoglycemia/blood , Hypoglycemia/immunology , Japan , Male , Middle Aged , Molecular Weight , Peptide Mapping , Pituitary Gland, Anterior/immunology , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Specific Pathogen-Free Organisms
16.
PLoS One ; 9(7): e102425, 2014.
Article En | MEDLINE | ID: mdl-25014207

Small Rho GTPases are well known to regulate a variety of cellular processes by acting as molecular switches. The regulatory function of Rho GTPases is critically dependent on their posttranslational modification at the carboxyl terminus by isoprenylation and association with proper cellular membranes. Despite numerous studies, the mechanisms of recycling and functional integration of Rho GTPases at the biological membranes are largely unclear. In this study, prenylated human Rac1, a prominent member of the Rho family, was purified in large amount from baculovirus-infected Spodoptera frugiperda insect cells using a systematic detergent screening. In contrast to non-prenylated human Rac1 purified from Escherichia coli, prenylated Rac1 from insect cells was able to associate with synthetic liposomes and to bind Rho-specific guanine nucleotide dissociation inhibitor 1 (GDI1). Subsequent liposome reconstitution experiments revealed that GDI1 efficiently extracts Rac1 from liposomes preferentially in the inactive GDP-bound state. The extraction was prevented when Rac1 was activated to its GTP-bound state by Rac-specific guanine nucleotide exchange factors (GEFs), such as Vav2, Dbl, Tiam1, P-Rex1 and TrioN, and bound by the downstream effector Pak1. We found that dissociation of Rac1-GDP from its complex with GDI1 strongly correlated with two distinct activities of especially Dbl and Tiam1, including liposome association and the GDP/GTP exchange. Taken together, our results provided first detailed insights into the advantages of the in vitro liposome-based reconstitution system to study both the integration of the signal transducing protein complexes and the mechanisms of regulation and signaling of small GTPases at biological membranes.


Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Liposomes/chemistry , Protein Processing, Post-Translational , p21-Activated Kinases/chemistry , rac1 GTP-Binding Protein/chemistry , Animals , Baculoviridae/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanine Nucleotide Dissociation Inhibitors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Liposomes/metabolism , Models, Biological , Protein Prenylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
17.
PLoS One ; 8(12): e83698, 2013.
Article En | MEDLINE | ID: mdl-24376733

Mammalian neuroglobin (Ngb) protects neuronal cells under conditions of oxidative stress. We previously showed that human Ngb acts as a guanine nucleotide dissociation inhibitor (GDI) for the α-subunits of heterotrimeric Gi/o proteins and inhibits reductions in cAMP concentration, leading to protection against cell death. In the present study, we created human E60Q Ngb mutant and clarified that Glu60 of human Ngb is a crucial residue for its GDI and neuroprotective activities. Moreover, we investigated structural and functional properties of several human Ngb mutants and demonstrated that the neuroprotective effect of human Ngb is due to its GDI activity and not due to its scavenging activity against reactive oxygen species.


Globins/chemistry , Globins/metabolism , Glutamic Acid , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Cell Differentiation , Cell Line , Globins/genetics , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Mutation , Nerve Tissue Proteins/genetics , Neuroglobin , Neurons/cytology
18.
J Am Chem Soc ; 135(44): 16418-28, 2013 Nov 06.
Article En | MEDLINE | ID: mdl-24079270

FtsZ is the key protein of bacterial cell-division and target for new antibiotics. Selective inhibition of FtsZ polymerization without impairing the assembly of the eukaryotic homologue tubulin was demonstrated with C8-substituted guanine nucleotides. By combining NMR techniques with biochemical and molecular modeling procedures, we have investigated the molecular recognition of C8-substituted-nucleotides by FtsZ from Methanococcus jannaschii (Mj-FtsZ) and Bacillus subtilis (Bs-FtsZ). STD epitope mapping and trNOESY bioactive conformation analysis of each nucleotide were employed to deduce differences in their recognition mode by each FtsZ species. GMP binds in the same anti conformation as GTP, whereas 8-pyrrolidino-GMP binds in the syn conformation. However, the anti conformation of 8-morpholino-GMP is selected by Bs-FtsZ, while Mj-FtsZ binds both anti- and syn-geometries. The inhibitory potencies of the C8-modified-nucleotides on the assembly of Bs-FtsZ, but not of Mj-FtsZ, correlate with their binding affinities. Thus, MorphGTP behaves as a nonhydrolyzable analog whose binding induces formation of Mj-FtsZ curved filaments, resembling polymers formed by the inactive forms of this protein. NMR data, combined with molecular modeling protocols, permit explanation of the mechanism of FtsZ assembly impairment by C8-substituted GTP analogs. The presence of the C8-substituent induces electrostatic remodeling and small structural displacements at the association interface between FtsZ monomers to form filaments, leading to complete assembly inhibition or to formation of abnormal FtsZ polymers. The inhibition of bacterial Bs-FtsZ assembly may be simply explained by steric clashes of the C8-GTP-analogs with the incoming FtsZ monomer. This information may facilitate the design of antibacterial FtsZ inhibitors replacing GTP.


Bacillus subtilis/chemistry , Bacterial Proteins/chemistry , Cytoskeletal Proteins/chemistry , Guanine Nucleotide Dissociation Inhibitors/chemistry , Methanocaldococcus/chemistry , Nuclear Magnetic Resonance, Biomolecular , Models, Molecular , Nucleic Acid Conformation
19.
Mol Biosyst ; 9(10): 2454-62, 2013 Oct.
Article En | MEDLINE | ID: mdl-23872884

A network of the Rho family GTPases, which cycle between inactive GDP-bound and active GTP-bound states, controls key cellular processes, including proliferation and migration. Activating and deactivating GTPase transitions are controlled by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs) and GDP dissociation inhibitors (GDIs) that sequester GTPases from the membrane to the cytoplasm. Here we show that a cascade of two Rho family GTPases, RhoA and Rac1, regulated by RhoGDI1, exhibits distinct modes of the dynamic behavior, including abrupt, bistable switches, excitable overshoot transitions and oscillations. The RhoGDI1 abundance and signal-induced changes in the RhoGDI1 affinity for GTPases control these different dynamics, enabling transitions from a single stable steady state to bistability, to excitable pulses and to sustained oscillations of GTPase activities. These RhoGDI1-controlled dynamic modes of RhoA and Rac1 activities form the basis of cell migration behaviors, including protrusion-retraction cycles at the leading edge of migrating cells.


Guanine Nucleotide Dissociation Inhibitors/chemistry , Monomeric GTP-Binding Proteins/chemistry , Enzyme Activation , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Guanine Nucleotide Dissociation Inhibitors/pharmacology , Kinetics , Models, Biological , Monomeric GTP-Binding Proteins/metabolism , Protein Binding/drug effects , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism
20.
J Biol Chem ; 288(33): 24091-103, 2013 Aug 16.
Article En | MEDLINE | ID: mdl-23770668

Group II activators of G-protein signaling play diverse functional roles through their interaction with Gαi, Gαt, and Gαo via a G-protein regulatory (GPR) motif that serves as a docking site for Gα-GDP. We recently reported the regulation of the AGS3-Gαi signaling module by a cell surface, seven-transmembrane receptor. Upon receptor activation, AGS3 reversibly dissociates from the cell cortex, suggesting that it may function as a signal transducer with downstream signaling implications, and this question is addressed in the current report. In HEK-293 and COS-7 cells expressing the α2A/D-AR and Gαi3, receptor activation resulted in the translocation of endogenous AGS3 and AGS3-GFP from the cell cortex to a juxtanuclear region, where it co-localized with markers of the Golgi apparatus (GA). The agonist-induced translocation of AGS3 was reversed by the α2-AR antagonist rauwolscine. The TPR domain of AGS3 was required for agonist-induced translocation of AGS3 from the cell cortex to the GA, and the translocation was blocked by pertussis toxin pretreatment or by the phospholipase Cß inhibitor U73122. Agonist-induced translocation of AGS3 to the GA altered the functional organization and protein sorting at the trans-Golgi network. The regulated movement of AGS3 between the cell cortex and the GA offers unexpected mechanisms for modulating protein secretion and/or endosome recycling events at the trans-Golgi network.


Guanine Nucleotide Dissociation Inhibitors/metabolism , Receptors, Cell Surface/metabolism , trans-Golgi Network/metabolism , Animals , Biomarkers/metabolism , Brimonidine Tartrate , COS Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Chlorocebus aethiops , Endosomes/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Green Fluorescent Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , HEK293 Cells , Humans , Lysosomes/metabolism , Protein Structure, Tertiary , Protein Transport/drug effects , Quinoxalines/pharmacology , Subcellular Fractions/metabolism , Time Factors , trans-Golgi Network/drug effects
...