Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
1.
J Mol Biol ; 435(22): 168293, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37775038

ABSTRACT

Arl8b, a specific Arf-like family GTPase present on lysosome, and plays critical roles in many lysosome-related cellular processes such as autophagy. The active Arl8b can be specifically recognized by the RUN domains of two Arl8b-effectors PLEKHM1 and SKIP, thereby regulating the autophagosome/lysosome membrane fusion and the intracellular lysosome positioning, respectively. However, the mechanistic bases underlying the interactions of Arl8b with the RUN domains of PLEKHM1 and SKIP remain elusive. Here, we report the two high-resolution crystal structures of the active Arl8b in complex with the RUN domains of PLEKHM1 and SKIP. In addition to elucidating the detailed molecular mechanism governing the specific interactions of the active Arl8b with the RUN domains of PLEKHM1 and SKIP, the determined complex structures also reveal a general binding mode shared by the PLEKHM1 and SKIP RUN domains for interacting with the active Arl8b. Furthermore, we uncovered a competitive relationship between the RUN domains of PLEKHM1 and SKIP in binding to the active Arl8b as well as a unique small GTPase-binding mode adopted by the PLEKHM1 and SKIP RUN domains, thereby enriching the repertoire of the RUN domain/small GTPase interaction modes. In all, our findings provide new mechanistic insights into the interactions of the active Arl8b with PLEKHM1 and SKIP, and are valuable for further understanding the working modes of these proteins in relevant cellular processes.


Subject(s)
ADP-Ribosylation Factors , Adaptor Proteins, Signal Transducing , Autophagy-Related Proteins , Nuclear Receptor Coactivators , Protein Interaction Domains and Motifs , Adaptor Proteins, Signal Transducing/chemistry , Lysosomes/metabolism , Membrane Fusion , ADP-Ribosylation Factors/chemistry , Autophagy-Related Proteins/chemistry , Nuclear Receptor Coactivators/chemistry , Crystallography, X-Ray , Humans
2.
J Biomol Struct Dyn ; 40(10): 4626-4641, 2022 07.
Article in English | MEDLINE | ID: mdl-33356902

ABSTRACT

The ARL15 gene (ADP ribosylation factor like protein 15) encodes for an uncharacterized small GTP-binding protein. Its exact role in human physiology remains unknown, but a number of genetic association studies have recognised different variants in this gene to be statistically associated with numerous traits and complex diseases. We have previously reported a novel association of ARL15 with rheumatoid arthritis (RA) based on a genome-wide association study in a north Indian cohort. Subsequent investigations have provided leads for its involvement in RA pathophysiology, especially its potential as a novel therapeutic target. However, the absence of an experimentally determined tertiary structure for ARL15 significantly hinders the understanding of its biochemical and physiological functions, as well as development of potential lead molecules. We, therefore, aimed to derive a high quality, refined model of the three dimensional structure of human ARL15 protein using two different computational protein structure prediction methods - template-based threading and ab initio modelling. The best model each from among the five each derived from both the approaches was selected based on stringent quality assessment and refinement. Molecular dynamics simulations over long timescales revealed the ab initio model to be relatively more stable, and it marginally outperformed the template-based model in the quality assessment as well. A putative GTP-binding site was also predicted using homology for the ARL15 protein, where potential competitive inhibitors can be targeted. This high quality predicted model may provide insights to the biological role(s) of ARL15 and inform and guide further experimental, structural and biochemical characterization efforts.Communicated by Ramaswamy H. Sarma.


Subject(s)
ADP-Ribosylation Factors , Arthritis, Rheumatoid , Genome-Wide Association Study , ADP-Ribosylation Factors/chemistry , Arthritis, Rheumatoid/genetics , Humans , Membrane Proteins , Molecular Dynamics Simulation
3.
Molecules ; 26(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34208932

ABSTRACT

We report the results of calculations of the Gibbs energy profiles of the guanosine triphosphate (GTP) hydrolysis by the Arl3-RP2 protein complex using molecular dynamics (MD) simulations with ab initio type QM/MM potentials. The chemical reaction of GTP hydrolysis to guanosine diphosphate (GDP) and inorganic phosphate (Pi) is catalyzed by GTPases, the enzymes, which are responsible for signal transduction in live cells. A small GTPase Arl3, catalyzing the GTP → GDP reaction in complex with the activating protein RP2, constitute an essential part of the human vision cycle. To simulate the reaction mechanism, a model system is constructed by motifs of the crystal structure of the Arl3-RP2 complexed with a substrate analog. After selection of reaction coordinates, energy profiles for elementary steps along the reaction pathway GTP + H2O → GDP + Pi are computed using the umbrella sampling and umbrella integration procedures. QM/MM MD calculations are carried out, interfacing the molecular dynamics program NAMD and the quantum chemistry program TeraChem. Ab initio type QM(DFT)/MM potentials are computed with atom-centered basis sets 6-31G** and two hybrid functionals (PBE0-D3 and ωB97x-D3) of the density functional theory, describing a large QM subsystem. Results of these simulations of the reaction mechanism are compared to those obtained with QM/MM calculations on the potential energy surface using a similar description of the QM part. We find that both approaches, QM/MM and QM/MM MD, support the mechanism of GTP hydrolysis by GTPases, according to which the catalytic glutamine side chain (Gln71, in this system) actively participates in the reaction. Both approaches distinguish two parts of the reaction: the cleavage of the phosphorus-oxygen bond in GTP coupled with the formation of Pi, and the enzyme regeneration. Newly performed QM/MM MD simulations confirmed the profile predicted in the QM/MM minimum energy calculations, called here the pathway-I, and corrected its relief at the first elementary step from the enzyme-substrate complex. The QM/MM MD simulations also revealed another mechanism at the part of enzyme regeneration leading to pathway-II. Pathway-II is more consistent with the experimental kinetic data of the wild-type complex Arl3-RP2, whereas pathway-I explains the role of the mutation Glu138Gly in RP2 slowing down the hydrolysis rate.


Subject(s)
ADP-Ribosylation Factors/chemistry , GTP-Binding Proteins/chemistry , Guanosine Triphosphate/chemistry , Membrane Proteins/chemistry , Models, Chemical , Molecular Dynamics Simulation , Catalysis , Hydrolysis
4.
J Biol Chem ; 295(49): 16643-16654, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32972971

ABSTRACT

The subcellular localization of Arf family proteins is generally thought to be determined by their corresponding guanine nucleotide exchange factors. By promoting GTP binding, guanine nucleotide exchange factors induce conformational changes of Arf proteins exposing their N-terminal amphipathic helices, which then insert into the membranes to stabilize the membrane association process. Here, we found that the N-terminal amphipathic motifs of the Golgi-localized Arf family protein, Arfrp1, and the endosome- and plasma membrane-localized Arf family protein, Arl14, play critical roles in spatial determination. Exchanging the amphipathic helix motifs between these two Arf proteins causes the switch of their localizations. Moreover, the amphipathic helices of Arfrp1 and Arl14 are sufficient for cytosolic proteins to be localized into a specific cellular compartment. The spatial determination mediated by the Arfrp1 helix requires its binding partner Sys1. In addition, the residues that are required for the acetylation of the Arfrp1 helix and the myristoylation of the Arl14 helix are important for the specific subcellular localization. Interestingly, Arfrp1 and Arl14 are recruited to their specific cellular compartments independent of GTP binding. Our results demonstrate that the amphipathic motifs of Arfrp1 and Arl14 are sufficient for determining specific subcellular localizations in a GTP-independent manner, suggesting that the membrane association and activation of some Arf proteins are uncoupled.


Subject(s)
ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/genetics , Amino Acid Sequence , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Protein Conformation, alpha-Helical , Sequence Alignment , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
5.
Genes Genet Syst ; 95(3): 119-131, 2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32611934

ABSTRACT

A homolog of the bacterial ribosome rescue factor ArfB was identified in Arabidopsis thaliana. The factor, named AtArfB for Arabidopsis thaliana ArfB, showed ribosome rescue activity in both in vivo and in vitro assays based on the bacterial translation system. As has been shown for ArfB, the ribosome rescue activity of AtArfB was dependent on the GGQ motif, the crucial motif for the function of class I release factors and ArfB. The C-terminal region of AtArfB was also important for its function. The N-terminal region of AtArfB, which is absent in bacterial ArfB, functioned as a transit peptide for chloroplast targeting in tobacco cells. These results strongly suggest that AtArfB is a ribosome rescue factor that functions in chloroplasts.


Subject(s)
ADP-Ribosylation Factors/metabolism , Arabidopsis Proteins/metabolism , Ribosomes/metabolism , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/genetics , Arabidopsis , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Chloroplasts/metabolism , Protein Domains , Protein Sorting Signals , Protein Transport
6.
Tissue Barriers ; 8(1): 1710429, 2020.
Article in English | MEDLINE | ID: mdl-31928299

ABSTRACT

Cholix (Chx) is expressed by the intestinal pathogen Vibrio cholerae as a single chain of 634 amino acids (~70.7 kDa protein) that folds into three distinct domains, with elements of the second and third domains being involved in accessing the cytoplasm of nonpolarized cells and inciting cell death via ADP-ribosylation of elongation factor 2, respectively. In order to reach nonpolarized cells within the intestinal lamina propria, however, Chx must cross the polarized epithelial barrier in an intact form. Here, we provide invitro and invivo demonstrations that a nontoxic Chx transports across intestinal epithelium via a vesicular trafficking pathway that rapidly achieves vesicular apical to basal (A→B) transcytosis and avoids routing to lysosomes. Specifically, Chx traffics in apical endocytic Rab7+ vesicles and in basal exocytic Rab11+ vesicles with a transition between these domains occurring in the ER-Golgi intermediate compartment (ERGIC) through interactions with the lectin mannose-binding protein 1 (LMAN1) protein that undergoes an intracellular re-distribution that coincides with the re-organization of COPI+ and COPII+ vesicular structures. Truncation studies demonstrated that domain I of Chx alone was sufficient to efficiently complete A→B transcytosis and capable of ferrying genetically conjoined human growth hormone (hGH). These studies provide evidence for a pathophysiological strategy where native Chx exotoxin secreted in the intestinal lumen by nonpandemic V. cholerae can reach nonpolarized cells within the lamina propria in an intact form by using a nondestructive pathway to cross in the intestinal epithelial that appears useful for oral delivery of biopharmaceuticals.One-Sentence Summary: Elements within the first domain of the Cholix exotoxin protein are essential and sufficient for the apical to basal transcytosis of this Vibrio cholerae-derived virulence factor across polarized intestinal epithelial cells.


Subject(s)
ADP-Ribosylation Factors/chemistry , Bacterial Toxins/chemistry , Protein Domains/physiology , Transcytosis/physiology , Humans
7.
Elife ; 92020 01 17.
Article in English | MEDLINE | ID: mdl-31951201

ABSTRACT

The BBSome is a heterooctameric protein complex that plays a central role in primary cilia homeostasis. Its malfunction causes the severe ciliopathy Bardet-Biedl syndrome (BBS). The complex acts as a cargo adapter that recognizes signaling proteins such as GPCRs and links them to the intraflagellar transport machinery. The underlying mechanism is poorly understood. Here we present a high-resolution cryo-EM structure of a human heterohexameric core subcomplex of the BBSome. The structure reveals the architecture of the complex in atomic detail. It explains how the subunits interact with each other and how disease-causing mutations hamper this interaction. The complex adopts a conformation that is open for binding to membrane-associated GTPase Arl6 and a large positively charged patch likely strengthens the interaction with the membrane. A prominent negatively charged cleft at the center of the complex is likely involved in binding of positively charged signaling sequences of cargo proteins.


Subject(s)
ADP-Ribosylation Factors/metabolism , Bardet-Biedl Syndrome/metabolism , ADP-Ribosylation Factors/chemistry , Cilia/metabolism , Cryoelectron Microscopy , Humans , Models, Molecular , Protein Binding , Protein Conformation , Signal Transduction
8.
J Biol Chem ; 294(46): 17354-17370, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31591270

ABSTRACT

Arf GAP with Src homology 3 domain, ankyrin repeat, and pleckstrin homology (PH) domain 1 (ASAP1) is a multidomain GTPase-activating protein (GAP) for ADP-ribosylation factor (ARF)-type GTPases. ASAP1 affects integrin adhesions, the actin cytoskeleton, and invasion and metastasis of cancer cells. ASAP1's cellular function depends on its highly-regulated and robust ARF GAP activity, requiring both the PH and the ARF GAP domains of ASAP1, and is modulated by phosphatidylinositol 4,5-bisphosphate (PIP2). The mechanistic basis of PIP2-stimulated GAP activity is incompletely understood. Here, we investigated whether PIP2 controls binding of the N-terminal extension of ARF1 to ASAP1's PH domain and thereby regulates its GAP activity. Using [Δ17]ARF1, lacking the N terminus, we found that PIP2 has little effect on ASAP1's activity. A soluble PIP2 analog, dioctanoyl-PIP2 (diC8PIP2), stimulated GAP activity on an N terminus-containing variant, [L8K]ARF1, but only marginally affected activity on [Δ17]ARF1. A peptide comprising residues 2-17 of ARF1 ([2-17]ARF1) inhibited GAP activity, and PIP2-dependently bound to a protein containing the PH domain and a 17-amino acid-long interdomain linker immediately N-terminal to the first ß-strand of the PH domain. Point mutations in either the linker or the C-terminal α-helix of the PH domain decreased [2-17]ARF1 binding and GAP activity. Mutations that reduced ARF1 N-terminal binding to the PH domain also reduced the effect of ASAP1 on cellular actin remodeling. Mutations in the ARF N terminus that reduced binding also reduced GAP activity. We conclude that PIP2 regulates binding of ASAP1's PH domain to the ARF1 N terminus, which may partially regulate GAP activity.


Subject(s)
ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Phosphatidylinositol 4,5-Diphosphate/genetics , ADP-Ribosylation Factor 1/chemistry , ADP-Ribosylation Factors/chemistry , Actins/chemistry , Actins/genetics , Adaptor Proteins, Signal Transducing/chemistry , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Humans , Neoplasms/genetics , Phosphatidylinositol 4,5-Diphosphate/chemistry , Pleckstrin Homology Domains/genetics , Point Mutation/genetics , Protein Binding/genetics
9.
Toxicon ; 170: 1-9, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31494206

ABSTRACT

Cholera is a major global threat, affecting millions each year. The ADP ribosyltransferase activity of the active cholera toxin catalyses the massive loss of water and electrolytes during cholera infections. The active toxin heterodimer comprises the A1 subunit from Vibrio cholerae and ARF (ADP Ribosylation Factor) from the human host. Although the active toxin is a potential target for drug discovery against cholera, it has been scarcely targeted to date. The A1-ARF interface contains a potential druggable site for small molecule inhibitors. By combining a sequential docking and scoring strategy with molecular dynamics (MD) simulations, this study identified hits against the protein-protein interface (PPI) of the active cholera toxin from an in-house library of 9,175 ADMET-screened alkaloids. The docking algorithms and scoring functions of Glide SP, Glide XP, and AutoDock were employed for initial library screening. Three alkaloids were initially selected by docking-based virtual screening. The stability of the hit-toxin complexes was validated by MD simulations. Two of the three hits, namely, A6225 (7-formyldehydrothalicsimidine) and A16503 (1,2,7,8-tetrahydroxy dibenz[cd,f]indol-4(5H)-one), formed stable complexes with the toxin. Analyses of the hydrogen bond occupancies revealed that the hits formed stable hydrogen bonds with the toxin PPI. The hits identified herein can serve as reference compounds for drug discovery against cholera in the future.


Subject(s)
ADP-Ribosylation Factors/chemistry , Alkaloids/pharmacology , Cholera Toxin/chemistry , Drug Discovery/methods , Molecular Dynamics Simulation , Alkaloids/chemistry , Binding Sites , Formaldehyde/chemistry , Formaldehyde/pharmacology , Humans , Hydrogen Bonding , Indoles/chemistry , Indoles/pharmacology , Protein Binding , Vibrio cholerae
10.
Proc Natl Acad Sci U S A ; 116(36): 18031-18040, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31427506

ABSTRACT

Upon invading target cells, multifunctional autoprocessing repeats-in-toxin (MARTX) toxins secreted by bacterial pathogens release their disease-related modularly structured effector domains. However, it is unclear how a diverse repertoire of effector domains within these toxins are processed and activated. Here, we report that Makes caterpillars floppy-like effector (MCF)-containing MARTX toxins require ubiquitous ADP-ribosylation factor (ARF) proteins for processing and activation of intermediate effector modules, which localize in different subcellular compartments following limited processing of holo effector modules by the internal cysteine protease. Effector domains structured tandemly with MCF in intermediate modules become disengaged and fully activated by MCF, which aggressively interacts with ARF proteins present at the same location as intermediate modules and is converted allosterically into a catalytically competent protease. MCF-mediated effector processing leads ultimately to severe virulence in mice via an MCF-mediated ARF switching mechanism across subcellular compartments. This work provides insight into how bacteria take advantage of host systems to induce systemic pathogenicity.


Subject(s)
ADP-Ribosylation Factors , ADP-Ribosylation , Bacterial Toxins , Vibrio vulnificus , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/metabolism , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Protein Domains , Vibrio vulnificus/genetics , Vibrio vulnificus/metabolism , Vibrio vulnificus/pathogenicity
11.
Cell Microbiol ; 21(8): e13033, 2019 08.
Article in English | MEDLINE | ID: mdl-31009148

ABSTRACT

Vibrio cholerae produced-Cholix toxin (Cholix) is a cytotoxin that ADP-ribosylates eukaryotic elongation factor 2, inhibiting protein synthesis, and inducing apoptosis. Here, we identified prohibitin (PHB) 1 and 2 as novel Cholix-interacting membrane proteins in immortalised human hepatocytes and HepG2 cells by Cholix immunoprecipitation assays. The expression level of PHB1 was decreased by Cholix after a 12hr incubation. Cholix-induced poly (ADP-ribose) polymerase (PARP) cleavage was significantly enhanced in PHB (PHB1 or PHB2) knockdown cells. In contrast, transiently overexpressed PHB in hepatocytes attenuated Cholix-induced Bax/Bak conformational changes and PARP cleavage. In addition, Cholix-induced reactive oxygen species production and accumulation of fragmented mitochondria were enhanced in PHB-knockdown cells. Furthermore, Cholix induced activation of Rho-associated coiled coil-containing protein kinase 1 (ROCK1), which was enhanced in PHB-knockdown cells, followed by actin filament depolymerisation and accumulation of tubulin in the blebbing cells. Inhibition of ROCK1 by siRNA or its inhibitor suppressed Cholix-induced PARP cleavage and reactive oxygen species generation. Our findings identify PHB as a new protein that interacts with Cholix and is involved in Cholix-induced mitochondrial dysfunction and cytoskeletal rearrangement by ROCK1 activation during apoptosis.


Subject(s)
ADP-Ribosylation Factors/chemistry , Bacterial Toxins/chemistry , Eukaryotic Initiation Factor-2/metabolism , Host-Pathogen Interactions/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Vibrio cholerae/genetics , ADP-Ribosylation , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/microbiology , Actin Cytoskeleton/ultrastructure , Amino Acid Sequence , Apoptosis/genetics , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Line, Transformed , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/microbiology , Hepatocytes/pathology , Humans , Mitochondria/metabolism , Mitochondria/microbiology , Mitochondria/ultrastructure , Prohibitins , Protein Binding , Protein Isoforms/deficiency , Protein Isoforms/genetics , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/deficiency , Signal Transduction , Tubulin/genetics , Tubulin/metabolism , Vibrio cholerae/metabolism , Vibrio cholerae/pathogenicity , Virulence , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
12.
Structure ; 27(5): 729-740.e3, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30905672

ABSTRACT

Rab35, a master regulator of membrane trafficking, regulates diverse cellular processes and is associated with various human diseases. Although a number of effectors have been identified, the molecular basis of Rab35-effector interactions remains unclear. Here, we provide the high-resolution crystal structures of Rab35 in complex with its two specific effectors ACAP2 and RUSC2, respectively. In the Rab35/ACAP2 complex structure, Rab35 binds to the terminal ankyrin repeat and a C-terminal extended α helix of ACAP2, revealing a previously uncharacterized binding mode both for Rabs and ankyrin repeats. In the Rab35/RUSC2 complex structure, Arg1015 of RUSC2 functions as a "pseudo-arginine finger" that stabilizes the GTP-bound Rab35, thus facilitating the assembly of Rab35/RUSC2 complex. The structural analysis allows us to design specific Rab35 mutants capable of eliminating Rab35/ACAP2 and Rab35/RUSC2 interactions, but not interfering with other effector bindings. The atomic structures also offer possible explanations to disease-associated mutants identified at the Rab35-effector interfaces.


Subject(s)
Carrier Proteins/chemistry , Membrane Proteins/chemistry , Mutation , rab GTP-Binding Proteins/chemistry , ADP-Ribosylation Factors/chemistry , Arginine/chemistry , HEK293 Cells , Humans , Protein Binding , Protein Domains , Protein Structure, Secondary
13.
Int J Mol Sci ; 20(2)2019 01 21.
Article in English | MEDLINE | ID: mdl-30669557

ABSTRACT

Arf GTPase-activating proteins (Arf GAPs) control the activity of ADP-ribosylation factors (Arfs) by inducing GTP hydrolysis and participate in a diverse array of cellular functions both through mechanisms that are dependent on and independent of their Arf GAP activity. A number of these functions hinge on the remodeling of actin filaments. Accordingly, some of the effects exerted by Arf GAPs involve proteins known to engage in regulation of the actin dynamics and architecture, such as Rho family proteins and nonmuscle myosin 2. Circular dorsal ruffles (CDRs), podosomes, invadopodia, lamellipodia, stress fibers and focal adhesions are among the actin-based structures regulated by Arf GAPs. Arf GAPs are thus important actors in broad functions like adhesion and motility, as well as the specialized functions of bone resorption, neurite outgrowth, and pathogen internalization by immune cells. Arf GAPs, with their multiple protein-protein interactions, membrane-binding domains and sites for post-translational modification, are good candidates for linking the changes in actin to the membrane. The findings discussed depict a family of proteins with a critical role in regulating actin dynamics to enable proper cell function.


Subject(s)
ADP-Ribosylation Factors/metabolism , Actin Cytoskeleton/metabolism , GTPase-Activating Proteins/metabolism , ADP-Ribosylation Factors/chemistry , Actin Cytoskeleton/chemistry , Actins/chemistry , Actins/metabolism , Animals , Apoptosis , Cell Movement , Focal Adhesions , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Host-Pathogen Interactions , Humans , Multigene Family , Neuronal Outgrowth , Neurons/metabolism , Podosomes/metabolism , Protein Binding , Pseudopodia/metabolism , Structure-Activity Relationship , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
14.
J Cell Sci ; 131(4)2018 02 20.
Article in English | MEDLINE | ID: mdl-29361551

ABSTRACT

Retinitis pigmentosa 2 (RP2) is the causative gene for a form of X-linked retinal degeneration. RP2 was previously shown to have GTPase-activating protein (GAP) activity towards the small GTPase ARL3 via its N-terminus, but the function of the C-terminus remains elusive. Here, we report a novel interaction between RP2 and osteoclast-stimulating factor 1 (OSTF1), an intracellular protein that indirectly enhances osteoclast formation and activity and is a negative regulator of cell motility. Moreover, this interaction is abolished by a human pathogenic mutation in RP2. We utilized a structure-based approach to pinpoint the binding interface to a strictly conserved cluster of residues on the surface of RP2 that spans both the C- and N-terminal domains of the protein, and which is structurally distinct from the ARL3-binding site. In addition, we show that RP2 is a positive regulator of cell motility in vitro, recruiting OSTF1 to the cell membrane and preventing its interaction with the migration regulator Myo1E.


Subject(s)
ADP-Ribosylation Factors/genetics , Actins/genetics , Eye Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Proteins/genetics , Retinitis Pigmentosa/genetics , ADP-Ribosylation Factors/chemistry , Actins/chemistry , Binding Sites/genetics , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Movement/genetics , Cilia/genetics , Cilia/metabolism , Eye Proteins/chemistry , GTP-Binding Proteins , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Membrane Proteins/chemistry , Molecular Docking Simulation , Myosin Type I/chemistry , Myosin Type I/genetics , Protein Binding/genetics , Protein Conformation , Protein Domains/genetics , Protein Structure, Tertiary , Proteins/chemistry , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
15.
Structure ; 26(1): 106-117.e6, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29276036

ABSTRACT

Membrane dynamic processes including vesicle biogenesis depend on Arf guanosine triphosphatase (GTPase) activation by guanine nucleotide exchange factors (GEFs) containing a catalytic Sec7 domain and a membrane-targeting module such as a pleckstrin homology (PH) domain. The catalytic output of cytohesin family Arf GEFs is controlled by autoinhibitory interactions that impede accessibility of the exchange site in the Sec7 domain. These restraints can be relieved through activator Arf-GTP binding to an allosteric site comprising the PH domain and proximal autoinhibitory elements (Sec7-PH linker and C-terminal helix). Small-angle X-ray scattering and negative-stain electron microscopy were used to investigate the structural organization and conformational dynamics of cytohesin-3 (Grp1) in autoinhibited and active states. The results support a model in which hinge dynamics in the autoinhibited state expose the activator site for Arf-GTP binding, while subsequent C-terminal helix unlatching and repositioning unleash conformational entropy in the Sec7-PH linker to drive exposure of the exchange site.


Subject(s)
ADP-Ribosylation Factors/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Guanosine Triphosphate/chemistry , Pleckstrin Homology Domains , Receptors, Cytoplasmic and Nuclear/chemistry , Recombinant Fusion Proteins/chemistry , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Animals , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Triphosphate/metabolism , Humans , Kinetics , Mice , Molecular Dynamics Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
16.
J Biol Chem ; 292(26): 11091-11108, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28487361

ABSTRACT

Primary cilia play central roles in signaling during metazoan development. Several key regulators of ciliogenesis and ciliary signaling are mutated in humans, resulting in a number of ciliopathies, including Joubert syndrome (JS). ARL13B is a ciliary GTPase with at least three missense mutations identified in JS patients. ARL13B is a member of the ADP ribosylation factor family of regulatory GTPases, but is atypical in having a non-homologous, C-terminal domain of ∼20 kDa and at least one key residue difference in the consensus GTP-binding motifs. For these reasons, and to establish a solid biochemical basis on which to begin to model its actions in cells and animals, we developed preparations of purified, recombinant, murine Arl13b protein. We report results from assays for solution-based nucleotide binding, intrinsic and GTPase-activating protein-stimulated GTPase, and ARL3 guanine nucleotide exchange factor activities. Biochemical analyses of three human missense mutations found in JS and of two consensus GTPase motifs reinforce the atypical properties of this regulatory GTPase. We also discovered that murine Arl13b is a substrate for casein kinase 2, a contaminant in our preparation from human embryonic kidney cells. This activity, and the ability of casein kinase 2 to use GTP as a phosphate donor, may be a source of differences between our data and previously published results. These results provide a solid framework for further research into ARL13B on which to develop models for the actions of this clinically important cell regulator.


Subject(s)
ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/isolation & purification , ADP-Ribosylation Factors/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Amino Acid Motifs , Amino Acid Substitution , Animals , Casein Kinase II/metabolism , Cerebellum/abnormalities , Cerebellum/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Mice , Mutation, Missense , Retina/abnormalities , Retina/metabolism
17.
J Cell Sci ; 130(3): 563-576, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27927754

ABSTRACT

ARL13B (a small GTPase) and INPP5E (a phosphoinositide 5-phosphatase) are ciliary proteins encoded by causative genes of Joubert syndrome. We here showed, by taking advantage of a visible immunoprecipitation assay, that ARL13B interacts with the IFT46 -: IFT56 (IFT56 is also known as TTC26) dimer of the intraflagellar transport (IFT)-B complex, which mediates anterograde ciliary protein trafficking. However, the ciliary localization of ARL13B was found to be independent of its interaction with IFT-B, but dependent on the ciliary-targeting sequence RVEP in its C-terminal region. ARL13B-knockout cells had shorter cilia than control cells and exhibited aberrant localization of ciliary proteins, including INPP5E. In particular, in ARL13B-knockout cells, the IFT-A and IFT-B complexes accumulated at ciliary tips, and GPR161 (a negative regulator of Hedgehog signaling) could not exit cilia in response to stimulation with Smoothened agonist. This abnormal phenotype was rescued by the exogenous expression of wild-type ARL13B, as well as by its mutant defective in the interaction with IFT-B, but not by its mutants defective in INPP5E binding or in ciliary localization. Thus, ARL13B regulates IFT-A-mediated retrograde protein trafficking within cilia through its interaction with INPP5E.


Subject(s)
ADP-Ribosylation Factors/metabolism , Abnormalities, Multiple/metabolism , Cerebellum/abnormalities , Cilia/metabolism , Eye Abnormalities/metabolism , Kidney Diseases, Cystic/metabolism , Phosphoric Monoester Hydrolases/metabolism , Retina/abnormalities , ADP-Ribosylation Factors/chemistry , Amino Acid Sequence , Biological Transport , Cerebellum/metabolism , Flagella/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Mutation/genetics , Protein Binding , Protein Multimerization , Protein Sorting Signals , Protein Transport , Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Retina/metabolism
18.
J Biol Chem ; 291(39): 20766-78, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27481943

ABSTRACT

Primary cilia are highly specialized small antenna-like cellular protrusions that extend from the cell surface of many eukaryotic cell types. The protein content inside cilia and cytoplasm is very different, but details of the sorting process are not understood for most ciliary proteins. Recently, we have shown that prenylated proteins are sorted according to their affinity to the carrier protein PDE6δ and the ability of Arl3 but not Arl2 to release high affinity cargo inside the cilia (Fansa, E. K., Kösling, S. K., Zent, E., Wittinghofer, A., and Ismail, S. (2016) Nat. Commun. 7, 11366). Here we address the question whether a similar principle governs the transport of myristoylated cargo by the carrier proteins Unc119a and Unc119b. We thus analyzed the binding strength of N-terminal myristoylated cargo peptides (GNAT1, NPHP3, Cystin1, RP2, and Src) to Unc119a and Unc119b proteins. The affinity between myristoylated cargo and carrier protein, Unc119, varies between subnanomolar and micromolar. Peptides derived from ciliary localizing proteins (GNAT1, NPHP3, and Cystin1) bind with high affinity to Unc119 proteins, whereas a peptide derived from a non-ciliary localizing protein (Src) has low affinity. The peptide with intermediate affinity (RP2) is localized at the ciliary transition zone as a gate keeper. We show that the low affinity peptides are released by both Arl2·GppNHp and Arl3·GppNHp, whereas the high affinity peptides are exclusively released by only Arl3·GppNHp. Determination of the x-ray structure of myristoylated NPHP3 peptide in complex with Unc119a reveals the molecular details of high affinity binding and suggests the importance of the residues at the +2 and +3 positions relative to the myristoylated glycine for high and low affinities. The mutational analysis of swapping the residues at the +2 and +3 positions between high and low affinity peptides results in reversing their affinities for Unc119a and leads to a partial mislocalization of a low affinity mutant of NPHP3.


Subject(s)
ADP-Ribosylation Factors/chemistry , Adaptor Proteins, Signal Transducing/chemistry , GTP-Binding Proteins/chemistry , Kinesins/chemistry , Peptides/chemistry , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Kinesins/genetics , Kinesins/metabolism , Peptides/genetics , Peptides/metabolism , Protein Structure, Quaternary
19.
Small GTPases ; 7(4): 283-296, 2016 10.
Article in English | MEDLINE | ID: mdl-27449855

ABSTRACT

Arf GTPases assemble protein complexes on membranes to carry out major functions in cellular traffic. An essential step is their activation by guanine nucleotide exchange factors (GEFs), whose Sec7 domain stimulates GDP/GTP exchange. ArfGEFs form 2 major families: ArfGEFs with DCB, HUS and HDS domains (GBF1 and BIG1/BIG2 in humans), which act at the Golgi; and ArfGEFs with a C-terminal PH domain (cytohesin, EFA6 and BRAG), which function at the plasma membrane and endosomes. In addition, pathogenic bacteria encode an ArfGEF with a unique membrane-binding domain. Here we review the allosteric regulation of Arf GTPases and their GEFs at the membrane interface. Membranes contribute several regulatory layers: at the GTPase level, where activation by GTP is coupled to membrane recruitment by a built-in structural device; at the Sec7 domain, which manipulates this device to ensure that Arf-GTP is attached to membranes; and at the level of non-catalytic ArfGEF domains, which form direct or GTPase-mediated interactions with membranes that enable a spectacular diversity of regulatory regimes. Notably, we show here that membranes increase the efficiency of a large ArfGEF (human BIG1) by 32-fold by interacting directly with its N-terminal DCB and HUS domains. The diversity of allosteric regulatory regimes suggests that ArfGEFs can function in cascades and circuits to modulate the shape, amplitude and duration of Arf signals in cells. Because Arf-like GTPases feature autoinhibitory elements similar to those of Arf GTPases, we propose that their activation also requires allosteric interactions of these elements with membranes or other proteins.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Membrane/metabolism , Guanine Nucleotide Exchange Factors/metabolism , ADP-Ribosylation Factors/chemistry , Allosteric Regulation , Animals , Binding Sites , Endosomes/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Humans , Models, Molecular , Protein Binding
20.
J Biol Chem ; 291(32): 16709-19, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27311713

ABSTRACT

ARAP3 (Arf-GAP with Rho-GAP domain, ANK repeat, and PH domain-containing protein 3) is unique for its dual specificity GAPs (GTPase-activating protein) activity for Arf6 (ADP-ribosylation factor 6) and RhoA (Ras homolog gene family member A) regulated by phosphatidylinositol 3,4,5-trisphosphate and a small GTPase Rap1-GTP and is involved in regulation of cell shape and adhesion. However, the molecular interface between the ARAP3-RhoGAP domain and RhoA is unknown, as is the substrates specificity of the RhoGAP domain. In this study, we solved the crystal structure of RhoA in complex with the RhoGAP domain of ARAP3. The structure of the complex presented a clear interface between the RhoGAP domain and RhoA. By analyzing the crystal structure and in combination with in vitro GTPase activity assays and isothermal titration calorimetry experiments, we identified the crucial residues affecting RhoGAP activity and substrates specificity among RhoA, Rac1 (Ras-related C3 botulinum toxin substrate 1), and Cdc42 (cell division control protein 42 homolog).


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , GTPase-Activating Proteins/chemistry , rhoA GTP-Binding Protein/chemistry , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Crystallography, X-Ray , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Protein Domains , Shelterin Complex , Structure-Activity Relationship , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...