Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Proteins ; 89(10): 1324-1332, 2021 10.
Article En | MEDLINE | ID: mdl-34056776

Rab GTPases constitute the largest branch of the Ras protein superfamily that regulate intra-cellular membrane trafficking. Their signaling activity is mediated by the transition between an active GTP-bound state and an inactive GDP-bound state. In the inactive state the switch I and II segments adopt largely disordered flexible conformations, whereas in the active state these regions are in well-defined conformations. The switch I and II states are central for recognition of Rab GTPases by interacting partners. Phosphorylation of the Rab1b-GTPase at residue Ser111 (pS111) results in modulation of the signaling activity due to alterations of the protein interaction interface and also due to modulation of the conformational flexibility. We have studied the flexibility of native and pS111-Rab1b in complex with GTP or GDP using extensive Molecular Dynamics (MD) simulations and an advanced sampling method called DIhedral Angle-biasing potential Replica-Exchange Molecular dynamics (DIA-REMD). The DIA-REMD method promotes backbone and side chain dihedral transitions along a series of replica simulations in selected protein segments and through exchanges also improves sampling in an unbiased reference simulation. Application to the Rab1b system results in significantly enhanced sampling of different switch I/II conformational states in the GDP-bound Rab1b state. The pS111 modification is found to reduce the conformational flexibility even in the presence of GDP, which may influence signaling activities. The stabilizing effect can be attributed to the formation of additional surface salt bridges between Arg-residues and pS111 not present in the native structure. The DIA-REMD method could be a valuable approach for studying also other signaling proteins that contain flexible segments.


rab1 GTP-Binding Proteins/chemistry , Humans , Phosphorylation , Protein Conformation , Serine/chemistry
2.
EMBO J ; 40(12): e107608, 2021 06 15.
Article En | MEDLINE | ID: mdl-34018214

The TRAPP complexes are nucleotide exchange factors that play essential roles in membrane traffic and autophagy. TRAPPII activates Rab11, and TRAPPIII activates Rab1, with the two complexes sharing a core of small subunits that affect nucleotide exchange but being distinguished by specific large subunits that are essential for activity in vivo. Crystal structures of core subunits have revealed the mechanism of Rab activation, but how the core and the large subunits assemble to form the complexes is unknown. We report a cryo-EM structure of the entire Drosophila TRAPPIII complex. The TRAPPIII-specific subunits TRAPPC8 and TRAPPC11 hold the catalytic core like a pair of tongs, with TRAPPC12 and TRAPPC13 positioned at the joint between them. TRAPPC2 and TRAPPC2L link the core to the two large arms, with the interfaces containing residues affected by disease-causing mutations. The TRAPPC8 arm is positioned such that it would contact Rab1 that is bound to the core, indicating how the arm could determine the specificity of the complex. A lower resolution structure of TRAPPII shows a similar architecture and suggests that the TRAPP complexes evolved from a single ur-TRAPP.


Drosophila Proteins/chemistry , Vesicular Transport Proteins/chemistry , rab1 GTP-Binding Proteins/chemistry , Cryoelectron Microscopy , Drosophila Proteins/ultrastructure , Guanine Nucleotide Exchange Factors/chemistry , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Protein Conformation , Vesicular Transport Proteins/ultrastructure , rab1 GTP-Binding Proteins/ultrastructure
3.
Nat Commun ; 12(1): 1564, 2021 03 10.
Article En | MEDLINE | ID: mdl-33692360

The lipid phosphatidylinositol-3-phosphate (PI3P) is a regulator of two fundamental but distinct cellular processes, endocytosis and autophagy, so its generation needs to be under precise temporal and spatial control. PI3P is generated by two complexes that both contain the lipid kinase VPS34: complex II on endosomes (VPS34/VPS15/Beclin 1/UVRAG), and complex I on autophagosomes (VPS34/VPS15/Beclin 1/ATG14L). The endosomal GTPase Rab5 binds complex II, but the mechanism of VPS34 activation by Rab5 has remained elusive, and no GTPase is known to bind complex I. Here we show that Rab5a-GTP recruits endocytic complex II to membranes and activates it by binding between the VPS34 C2 and VPS15 WD40 domains. Electron cryotomography of complex II on Rab5a-decorated vesicles shows that the VPS34 kinase domain is released from inhibition by VPS15 and hovers over the lipid bilayer, poised for catalysis. We also show that the GTPase Rab1a, which is known to be involved in autophagy, recruits and activates the autophagy-specific complex I, but not complex II. Both Rabs bind to the same VPS34 interface but in a manner unique for each. These findings reveal how VPS34 complexes are activated on membranes by specific Rab GTPases and how they are recruited to unique cellular locations.


Cell Membrane/metabolism , Class III Phosphatidylinositol 3-Kinases/chemistry , Class III Phosphatidylinositol 3-Kinases/metabolism , rab1 GTP-Binding Proteins/chemistry , rab1 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/chemistry , rab5 GTP-Binding Proteins/metabolism , Beclin-1/chemistry , Beclin-1/genetics , Beclin-1/metabolism , Class III Phosphatidylinositol 3-Kinases/genetics , Endosomes/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Structure, Secondary , Tomography , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vacuolar Sorting Protein VPS15/chemistry , Vacuolar Sorting Protein VPS15/genetics , Vacuolar Sorting Protein VPS15/metabolism , rab1 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics
5.
Proc Natl Acad Sci U S A ; 117(11): 5772-5781, 2020 03 17.
Article En | MEDLINE | ID: mdl-32123090

Posttranslational modifications (PTMs) are important physiological means to regulate the activities and structures of central regulatory proteins in health and disease. Small GTPases have been recognized as important molecules that are targeted by PTMs during infections of mammalian cells by bacterial pathogens. The enzymes DrrA/SidM and AnkX from Legionella pneumophila AMPylate and phosphocholinate Rab1b during infection, respectively. Cdc42 is AMPylated by IbpA from Histophilus somni at tyrosine 32 or by VopS from Vibrio parahaemolyticus at threonine 35. These modifications take place in the important regulatory switch I or switch II regions of the GTPases. Since Rab1b and Cdc42 are central regulators of intracellular vesicular trafficking and of the actin cytoskeleton, their modifications by bacterial pathogens have a profound impact on the course of infection. Here, we addressed the biochemical and structural consequences of GTPase AMPylation and phosphocholination. By combining biochemical experiments and NMR analysis, we demonstrate that AMPylation can overrule the activity state of Rab1b that is commonly dictated by binding to guanosine diphosphate or guanosine triphosphate. Thus, PTMs may exert conformational control over small GTPases and may add another previously unrecognized layer of activity control to this important regulatory protein family.


Adenosine Monophosphate/metabolism , Bacterial Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , cdc42 GTP-Binding Protein/chemistry , rab1 GTP-Binding Proteins/chemistry , Adenosine Monophosphate/chemistry , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Processing, Post-Translational , cdc42 GTP-Binding Protein/metabolism , rab1 GTP-Binding Proteins/metabolism
6.
Chembiochem ; 20(18): 2336-2340, 2019 09 16.
Article En | MEDLINE | ID: mdl-31054261

Site-specific protein functionalization has become an indispensable tool in modern life sciences. Here, tag-based enzymatic protein functionalization techniques are among the most versatilely applicable approaches. However, many chemo-enzymatic functionalization strategies suffer from low substrate scopes of the enzymes utilized for functional labeling probes. We report on the wide substrate scope of the bacterial enzyme AnkX towards derivatized CDP-choline analogues and demonstrate that AnkX-catalyzed phosphocholination can be used for site-specific one- and two-step protein labeling with a broad array of different functionalities, displaying fast second-order transfer rates of 5×102 to 1.8×104 m-1 s-1 . Furthermore, we also present a strategy for the site-specific dual labeling of proteins of interest, based on the exploitation of AnkX and the delabeling function of the enzyme Lem3. Our results contribute to the wide field of protein functionalization, offering an attractive chemo-enzymatic tag-based modification strategy for in vitro labeling.


Bacterial Proteins/metabolism , Diacylglycerol Cholinephosphotransferase/metabolism , rab1 GTP-Binding Proteins/chemistry , Cytidine Diphosphate Choline/analogs & derivatives , Cytidine Diphosphate Choline/metabolism , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Protein Engineering , Substrate Specificity
7.
Biochem Biophys Res Commun ; 495(2): 1708-1715, 2018 01 08.
Article En | MEDLINE | ID: mdl-29223392

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the major genetic cause of autosomal-dominantly inherited Parkinson's disease. LRRK2 is implicated in the regulation of intracellular trafficking, neurite outgrowth and PD risk in connection with Rab7L1, a putative interactor of LRRK2. Recently, a subset of Rab GTPases have been reported as substrates of LRRK2. Here we examine the kinase activity of LRRK2 on Rab7L1 in situ in cells. Phos-tag analyses and metabolic labeling assays revealed that LRRK2 readily phosphorylates Golgi-localized wild-type Rab7L1 but not mutant forms that are distributed in the cytoplasm. In vitro assays demonstrated direct phosphorylation of Rab7L1 by LRRK2. Subsequent screening using Rab7L1 mutants harboring alanine-substitution for every single Ser/Thr residue revealed that Ser72 is a major phosphorylation site, which was confirmed by using a phospho-Ser72-specific antibody. Moreover, LRRK2 pathogenic Parkinson mutants altogether markedly enhanced the phosphorylation at Ser72. The modulation of Ser72 phosphorylation in Rab7L1 resulted in an alteration of the morphology and distribution of the trans-Golgi network. These data collectively support the involvement of Rab7L1 phosphorylation in the LRRK2-mediated cellular and pathogenetic mechanisms.


Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , rab1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites/genetics , HEK293 Cells , HeLa Cells , Humans , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Parkinson Disease/pathology , Phosphorylation , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Serine/genetics , Substrate Specificity , rab GTP-Binding Proteins , rab1 GTP-Binding Proteins/chemistry , rab1 GTP-Binding Proteins/genetics , trans-Golgi Network/metabolism , trans-Golgi Network/pathology
8.
FEBS J ; 284(24): 4358-4375, 2017 12.
Article En | MEDLINE | ID: mdl-29095572

The classical GTP hydrolysis mechanism, as seen in Ras, employs a catalytic glutamine provided in cis by the GTPase and an arginine supplied in trans by a GTPase activating protein (GAP). The key idea emergent from a large body of research on small GTPases is that GTPases employ a variety of different hydrolysis mechanisms; evidently, these variations permit diverse rates of GTPase inactivation, crucial for temporal regulation of different biological processes. Recently, we unified these variations and argued that a steric clash between active site residues (corresponding to positions 12 and 61 of Ras) governs whether a GTPase utilizes the cis-Gln or the trans-Gln (from the GAP) for catalysis. As the cis-Gln encounters a steric clash, the Rab GTPases employ the so-called dual finger mechanism where the interacting GAP supplies a trans-Gln for catalysis. Using experimental and computational methods, we demonstrate how the cis-Gln of Rab33 overcomes the steric clash when it is stabilized by a residue in the vicinity. In effect, this demonstrates how both cis-Gln- and trans-Gln-mediated mechanisms could operate in the same GTPase in different contexts, i.e. depending on the GAP that regulates its action. Interestingly, in the case of Rab5, which possesses a higher intrinsic GTP hydrolysis rate, a similar stabilization of the cis-Gln appears to overcome the steric clash. Taken together with the mechanisms seen for Rab1, it is evident that the observed variations in Rab and their GAP partners allow structural plasticity, or in other words, the choice of different catalytic mechanisms.


GTPase-Activating Proteins/metabolism , Guanosine Triphosphate/metabolism , Molecular Dynamics Simulation , Protozoan Proteins/chemistry , rab GTP-Binding Proteins/chemistry , rab5 GTP-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Arginine/metabolism , Catalysis , Catalytic Domain , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Glutamine/metabolism , Humans , Kinetics , Mice , Models, Chemical , Models, Molecular , Mutagenesis, Site-Directed , Plasmodium falciparum/enzymology , Protein Conformation , Protein Stability , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Structure-Activity Relationship , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab1 GTP-Binding Proteins/chemistry , rab1 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism
9.
Proteins ; 85(5): 859-871, 2017 05.
Article En | MEDLINE | ID: mdl-28120477

Targeting non-native-ligand binding sites for potential investigative and therapeutic applications is an attractive strategy in proteins that share common native ligands, as in Rab1 protein. Rab1 is a subfamily member of Rab proteins, which are members of Ras GTPase superfamily. All Ras GTPase superfamily members bind to native ligands GTP and GDP, that switch on and off the proteins, respectively. Rab1 is physiologically essential for autophagy and transport between endoplasmic reticulum and Golgi apparatus. Pathologically, Rab1 is implicated in human cancers, a neurodegenerative disease, cardiomyopathy, and bacteria-caused infectious diseases. We have performed structural analyses on Rab1 protein using a unique ensemble of clustering methods, including multi-step principal component analysis, non-negative matrix factorization, and independent component analysis, to better identify representative Rab1 proteins than the application of a single clustering method alone does. We then used the identified representative Rab1 structures, resolved in multiple ligand states, to map their known and novel binding sites. We report here at least a novel binding site on Rab1, involving Rab1-specific residues that could be further explored for the rational design and development of investigative probes and/or therapeutic small molecules against the Rab1 protein. Proteins 2017; 85:859-871. © 2016 Wiley Periodicals, Inc.


Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , rab1 GTP-Binding Proteins/chemistry , Animals , Binding Sites , CREB-Binding Protein/chemistry , Carrier Proteins/chemistry , Cluster Analysis , Cockroaches/chemistry , Factor Analysis, Statistical , Humans , Insect Proteins/chemistry , Ligands , Molecular Docking Simulation , Phosphoric Diester Hydrolases/chemistry , Principal Component Analysis , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Pyrophosphatases/chemistry , Structural Homology, Protein , Thermodynamics
10.
Proc Natl Acad Sci U S A ; 113(50): 14348-14353, 2016 12 13.
Article En | MEDLINE | ID: mdl-27911813

Ras-like small GTPases function as molecular switches and regulate diverse cellular events. To examine the dynamics of signaling requires spatiotemporal visualization of their activity in the cell. Current small GTPase sensors rely on specific effector domains that are available for only a small number of GTPases and compete for endogenous regulator/effector binding. Here, we describe versatile conformational sensors for GTPase activity (COSGAs) based on the conserved GTPase fold. Conformational changes upon GDP/GTP exchange were directly observed in solution, on beads, and in live cells by Förster resonance energy transfer (FRET). The COSGAs allow for monitoring of Rab1 and K-Ras activity in live cells using fluorescence lifetime imaging microscopy. We found that Rab1 is largely active in the cytoplasm and inactive at the Golgi, suggesting that the Golgi serves as the terminal of the Rab1 functional cycle. K-Ras displays polarized activity at the plasma membrane, with less activity at the edge of the cell and membrane ruffles.


Monomeric GTP-Binding Proteins/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Dogs , Fluorescence Polarization , Fluorescence Resonance Energy Transfer , Fluorescent Dyes , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence , Models, Molecular , Monomeric GTP-Binding Proteins/chemistry , Protein Conformation , Signal Transduction , rab1 GTP-Binding Proteins/chemistry , rab1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism
11.
Oncogene ; 35(44): 5699-5704, 2016 11 03.
Article En | MEDLINE | ID: mdl-27041585

The endoplasmic reticulum (ER) and Golgi membrane system have major roles in cell signaling and regulation of the biosynthesis/transport of proteins and lipids in response to environmental cues such as amino acid and cholesterol levels. Rab1 is the founding member of the Rab small GTPase family, which is known to mediate dynamic membrane trafficking between ER and Golgi. Growing evidence indicate that Rab1 proteins have important functions beyond their classical vesicular transport functions, including nutrient sensing and signaling, cell migration and presentation of cell-surface receptors. Moreover, deregulation of RAB1 expression has been linked to a myriad of human diseases such as cancer, cardiomyopathy and Parkinson's disease. Further investigating these new physiological and pathological functions of Rab1 should provide new opportunities for better understanding of the disease processes and may lead to more effective therapeutic interventions.


Disease Susceptibility , Neoplasms/metabolism , Signal Transduction , rab1 GTP-Binding Proteins/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Carrier Proteins , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Protein Binding , Protein Interaction Domains and Motifs , rab1 GTP-Binding Proteins/chemistry , rab1 GTP-Binding Proteins/genetics
12.
J Mol Biol ; 428(8): 1544-57, 2016 Apr 24.
Article En | MEDLINE | ID: mdl-26953259

Alternative splicing often affects structured and highly conserved regions of proteins, generating so called non-trivial splicing variants of unknown structure and cellular function. The human small G-protein Rab1A is involved in the regulation of the vesicle transfer from the ER to Golgi. A conserved non-trivial splice variant lacks nearly 40% of the sequence of the native Rab1A, including most of the regulatory interaction sites. We show that this variant of Rab1A represents a stable and folded protein, which is still able to bind nucleotides and co-localizes with membranes. Nevertheless, it should be mentioned that compared to other wild-typeRabGTPases, the measured nucleotide binding affinities are dramatically reduced in the variant studied. Furthermore, the Rab1A variant forms hetero-dimers with wild-type Rab1A and its presence in the cell enhances the efficiency of alkaline phosphatase secretion. However, this variant shows no specificity for GXP nucleotides, a constantly enhanced GTP hydrolysis activity and is no longer controlled by GEF or GAP proteins, indicating a new regulatory mechanism for the Rab1A cycle via alternative non-trivial splicing.


rab1 GTP-Binding Proteins/chemistry , Alternative Splicing , Cell Membrane/metabolism , Evolution, Molecular , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Humans , Hydrolysis , Nucleotides/chemistry , Protein Binding , Protein Folding , Protein Isoforms/chemistry , Protein Multimerization , Protein Structure, Tertiary , Proteome , rab GTP-Binding Proteins/chemistry
13.
Sci Rep ; 6: 19896, 2016 Jan 28.
Article En | MEDLINE | ID: mdl-26818796

The pathogenic pathway of Legionella pneumophila exploits the intercellular vesicle transport system via the posttranslational attachment of adenosine monophosphate (AMP) to the Tyr77 sidechain of human Ras like GTPase Rab1b. The modification, termed adenylylation, is performed by the bacterial enzyme DrrA/SidM, however the effect on conformational properties of the molecular switch mechanism of Rab1b remained unresolved. In this study we find that the adenylylation of Tyr77 stabilizes the active Rab1b state by locking the switch in the active signaling conformation independent of bound GTP or GDP and that electrostatic interactions due to the additional negative charge in the switch region make significant contributions. The stacking interaction between adenine and Phe45 however, seems to have only minor influence on this stabilisation. The results may also have implications for the mechanistic understanding of conformational switching in other signaling proteins.


Molecular Dynamics Simulation , Protein Conformation , Tyrosine/chemistry , rab1 GTP-Binding Proteins/chemistry , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Protein Binding , Protein Stability , Static Electricity , Tyrosine/metabolism , rab1 GTP-Binding Proteins/metabolism
14.
J Biol Chem ; 290(50): 29993-30005, 2015 Dec 11.
Article En | MEDLINE | ID: mdl-26499792

Leishmania secretes a large number of its effectors to the extracellular milieu. However, regulation of the secretory pathway in Leishmania is not well characterized. Here, we report the cloning, expression, and characterization of the Rab1 homologue from Leishmania. We have found that LdRab1 localizes in Golgi in Leishmania. To understand the role of LdRab1 in the secretory pathway of Leishmania, we have generated transgenic parasites overexpressing GFP-LdRab1:WT, GFP-LdRab1:Q67L (a GTPase-deficient dominant positive mutant of Rab1), and GFP-LdRab1:S22N (a GDP-locked dominant negative mutant of Rab1). Surprisingly, our results have shown that overexpression of GFP-LdRab1:Q67L or GFP-LdRab1:S22N does not disrupt the trafficking and localization of hemoglobin receptor in Leishmania. To determine whether the Rab1-dependent secretory pathway is conserved in parasites, we have analyzed the role of LdRab1 in the secretion of secretory acid phosphatase and Ldgp63 in Leishmania. Our results have shown that overexpression of GFP-LdRab1:Q67L or GFP-LdRab1:S22N significantly inhibits the secretion of secretory acid phosphatase by Leishmania. We have also found that overexpression of GFP-LdRab1:Q67L or GFP-LdRab1:S22N retains RFP-Ldgp63 in Golgi and blocks the secretion of Ldgp63, whereas the trafficking of RFP-Ldgp63 in GFP-LdRab1:WT-expressing cells is unaltered in comparison with control cells. Taken together, our results have shown that the Rab1-regulated secretory pathway is well conserved, and hemoglobin receptor trafficking follows an Rab1-independent secretory pathway in Leishmania.


Leishmania/enzymology , rab1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Molecular Sequence Data , Protein Transport , Sequence Homology, Amino Acid , rab1 GTP-Binding Proteins/chemistry
15.
PLoS Pathog ; 9(5): e1003382, 2013.
Article En | MEDLINE | ID: mdl-23696742

The covalent attachment of adenosine monophosphate (AMP) to proteins, a process called AMPylation (adenylylation), has recently emerged as a novel theme in microbial pathogenesis. Although several AMPylating enzymes have been characterized, the only known virulence protein with de-AMPylation activity is SidD from the human pathogen Legionella pneumophila. SidD de-AMPylates mammalian Rab1, a small GTPase involved in secretory vesicle transport, thereby targeting the host protein for inactivation. The molecular mechanisms underlying Rab1 recognition and de-AMPylation by SidD are unclear. Here, we report the crystal structure of the catalytic region of SidD at 1.6 Å resolution. The structure reveals a phosphatase-like fold with additional structural elements not present in generic PP2C-type phosphatases. The catalytic pocket contains a binuclear metal-binding site characteristic of hydrolytic metalloenzymes, with strong dependency on magnesium ions. Subsequent docking and molecular dynamics simulations between SidD and Rab1 revealed the interface contacts and the energetic contribution of key residues to the interaction. In conjunction with an extensive structure-based mutational analysis, we provide in vivo and in vitro evidence for a remarkable adaptation of SidD to its host cell target Rab1 which explains how this effector confers specificity to the reaction it catalyses.


Adenosine Monophosphate/chemistry , Bacterial Proteins/chemistry , Legionella pneumophila/enzymology , Molecular Docking Simulation , rab1 GTP-Binding Proteins/chemistry , Adenosine Monophosphate/genetics , Adenosine Monophosphate/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Humans , Legionella pneumophila/genetics , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2C , Protein Structure, Quaternary , Structure-Activity Relationship , rab1 GTP-Binding Proteins/genetics , rab1 GTP-Binding Proteins/metabolism
16.
Proteomics ; 13(6): 955-63, 2013 Mar.
Article En | MEDLINE | ID: mdl-23335384

Although the addition of a 5'-adenosine phosphodiester group to proteins, called adenylylation, has been known for decades, the possibility that adenylylation could be a molecular switch in cellular signaling pathways has emerged recently. The distinct mass shift upon adenylation of threonine or tyrosine residues renders it a good target for MS detection and identification; however, the fragmentation of adenylylated peptides derived from proteolytic digestion of adenylylated proteins has not yet been systematically investigated. Here, we demonstrate that adenylylated peptides show loss of parts of the adenosine monophosphate (AMP) upon different fragmentation techniques. As expected, causing the least fragmentation of the AMP group, electron transfer dissociation yields less complicated spectra. In contrast, CID and higher energy collision (HCD) fragmentation caused AMP to fragment, generating characteristic ions that could be utilized in the specific identification of adenylylated peptides. The characteristic ions and losses upon CID and higher energy collision fragmentation from the AMP group turned out to be highly dependent on which amino acid was adenylylated, with different reporter ions for adenylylated threonine and tyrosine. We also investigated how adenylylation is best incorporated into search engines, exemplified by Mascot and showed that it is possible to identify adenylylation by search engines.


Adenosine Monophosphate/chemistry , Peptide Fragments/chemistry , Protein Processing, Post-Translational , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Bacterial Proteins/chemistry , Humans , Molecular Weight , Nucleotidyltransferases/chemistry , Proteomics/methods , cdc42 GTP-Binding Protein/chemistry , rab1 GTP-Binding Proteins/chemistry
17.
EMBO Rep ; 14(2): 199-205, 2013 Feb.
Article En | MEDLINE | ID: mdl-23288104

Legionella pneumophila is an intracellularly surviving pathogen that releases about 270 different proteins into the host cell during infection. A set of secreted proteins takes control of the vesicular trafficking regulator Rab1. Legionella LepB inactivates Rab1 by acting as a GTPase-activating protein (GAP). We present the crystal structure of the Rab1b:LepB complex together with a thorough biochemical analysis and show that the GAP domain of LepB consists of an unusual fold. LepB acts by an atypical RabGAP mechanism that is reminiscent of classical GAPs and therefore sets the protein apart from mammalian TBC-like GAPs. Surprisingly, LepB can function as a GAP for Rab3, Rab8, Rab13 and Rab35, too, suggesting that it has a broader cellular role than previously thought.


Bacterial Proteins/chemistry , Legionella pneumophila/enzymology , rab1 GTP-Binding Proteins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Bacterial Proteins/genetics , Conserved Sequence , Crystallography, X-Ray , Guanosine Triphosphate/chemistry , Host-Pathogen Interactions , Humans , Hydrolysis , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , rab GTP-Binding Proteins/chemistry
18.
Proc Natl Acad Sci U S A ; 109(52): 21348-53, 2012 Dec 26.
Article En | MEDLINE | ID: mdl-23236136

Rab GTPases, key regulators of vesicular transport, hydrolyze GTP very slowly unless assisted by Rab GTPase-activating proteins (RabGAPs). Dysfunction of RabGAPs is involved in many diseases. By combining X-ray structure analysis and time-resolved FTIR spectroscopy we reveal here the detailed molecular reaction mechanism of a complex between human Rab and RabGAP at the highest possible spatiotemporal resolution and in atomic detail. A glutamine residue of Rab proteins (cis-glutamine) that is essential for intrinsic activity is less important in the GAP-activated reaction. During generation of the RabGAP·Rab:GTP complex, there is a rapid conformational change in which the cis-glutamine is replaced by a glutamine from RabGAP (trans-glutamine); this differs from the RasGAP mechanism, where the cis-glutamine is also important for GAP catalysis. However, as in the case of Ras, a trans-arginine is also recruited to complete the active center during this conformational change. In contrast to the RasGAP mechanism, an accumulation of a state in which phosphate is bound is not observed, and bond breakage is the rate-limiting step. The movement of trans-glutamine and trans-arginine into the catalytic site and bond breakage during hydrolysis are monitored in real time. The combination of X-ray structure analysis and time-resolved FTIR spectroscopy provides detailed insight in the catalysis of human Rab GTPases.


Biocatalysis , GTPase-Activating Proteins/metabolism , Mammals/metabolism , Models, Molecular , rab1 GTP-Binding Proteins/metabolism , Animals , Catalytic Domain , DNA Mutational Analysis , GTPase-Activating Proteins/chemistry , Glutamine/metabolism , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Kinetics , Spectroscopy, Fourier Transform Infrared , rab1 GTP-Binding Proteins/chemistry
19.
J Biol Chem ; 287(42): 35036-35046, 2012 Oct 12.
Article En | MEDLINE | ID: mdl-22872634

After the pathogenic bacterium Legionella pneumophila is phagocytosed, it injects more than 250 different proteins into the cytoplasm of host cells to evade lysosomal digestion and to replicate inside the host cell. Among these secreted proteins is the protein DrrA/SidM, which has been shown to modify Rab1b, a main regulator of vesicular trafficking in eukaryotic cells, by transfer of adenosine monophosphate (AMP) to Tyr(77). In addition, Legionella provides the protein SidD that hydrolytically reverses the covalent modification, suggesting a tight spatial and temporal control of Rab1 function by Legionella during infection. Small angle x-ray scattering experiments of DrrA allowed us to validate a tentative complex model built by combining available crystallographic data. We have established the effects of adenylylation on Rab1 interactions and properties in a quantitative way. In addition, we have characterized the kinetics of DrrA-catalyzed adenylylation as well as SidD-catalyzed deadenylylation toward Rab1 and have determined the nucleotide specificities of both enzymes. This study enhances our knowledge of proteins subverting Rab1 function at the Legionella-containing vacuole.


Bacterial Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Legionella pneumophila/enzymology , Legionnaires' Disease/enzymology , Protein Processing, Post-Translational , rab1 GTP-Binding Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Legionella pneumophila/genetics , Legionnaires' Disease/genetics , rab1 GTP-Binding Proteins/chemistry , rab1 GTP-Binding Proteins/genetics
20.
PLoS Pathog ; 8(3): e1002528, 2012.
Article En | MEDLINE | ID: mdl-22416225

The intracellular pathogen Legionella pneumophila hijacks the endoplasmic reticulum (ER)-derived vesicles to create an organelle designated Legionella-containing vacuole (LCV) required for bacterial replication. Maturation of the LCV involved acquisition of Rab1, which is mediated by the bacterial effector protein SidM/DrrA. SidM/DrrA is a bifunctional enzyme having the activity of both Rab1-specific GDP dissociation inhibitor (GDI) displacement factor (GDF) and guanine nucleotide exchange factor (GEF). LidA, another Rab1-interacting bacterial effector protein, was reported to promote SidM/DrrA-mediated recruitment of Rab1 to the LCV as well. Here we report the crystal structures of LidA complexes with GDP- and GTP-bound Rab1 respectively. Structural comparison revealed that GDP-Rab1 bound by LidA exhibits an active and nearly identical conformation with that of GTP-Rab1, suggesting that LidA can disrupt the switch function of Rab1 and render it persistently active. As with GTP, LidA maintains GDP-Rab1 in the active conformation through interaction with its two conserved switch regions. Consistent with the structural observations, biochemical assays showed that LidA binds to GDP- and GTP-Rab1 equally well with an affinity approximately 7.5 nM. We propose that the tight interaction with Rab1 allows LidA to facilitate SidM/DrrA-catalyzed release of Rab1 from GDIs. Taken together, our results support a unique mechanism by which a bacterial effector protein regulates Rab1 recycling.


Guanine Nucleotide Dissociation Inhibitors/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Host-Pathogen Interactions , Legionella pneumophila/pathogenicity , rab1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Crystallization , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Humans , Legionella pneumophila/metabolism , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Sequence Analysis, Protein , rab1 GTP-Binding Proteins/chemistry , rho-Specific Guanine Nucleotide Dissociation Inhibitors
...