Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33602807

ABSTRACT

Site-selective chemical bioconjugation reactions are enabling tools for the chemical biologist. Guided by a careful study of the selenomethionine (SeM) benzylation, we have refined the reaction to meet the requirements of practical protein bioconjugation. SeM is readily introduced through auxotrophic expression and exhibits unique nucleophilic properties that allow it to be selectively modified even in the presence of cysteine. The resulting benzylselenonium adduct is stable at physiological pH, is selectively labile to glutathione, and embodies a broadly tunable cleavage profile. Specifically, a 4-bromomethylphenylacetyl (BrMePAA) linker has been applied for efficient conjugation of complex organic molecules to SeM-containing proteins. This expansion of the bioconjugation toolkit has broad potential in the development of chemically enhanced proteins.


Subject(s)
Glutathione/metabolism , Selenomethionine/chemistry , Selenomethionine/metabolism , Selenoproteins/metabolism , Catalysis , Selenoproteins/chemistry
2.
Proc Natl Acad Sci U S A ; 115(42): E9792-E9801, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30185561

ABSTRACT

Autophagy is an enigmatic cellular process in which double-membrane compartments, called "autophagosomes, form de novo adjacent to the endoplasmic reticulum (ER) and package cytoplasmic contents for delivery to lysosomes. Expansion of the precursor membrane phagophore requires autophagy-related 2 (ATG2), which localizes to the PI3P-enriched ER-phagophore junction. We combined single-particle electron microscopy, chemical cross-linking coupled with mass spectrometry, and biochemical analyses to characterize human ATG2A in complex with the PI3P effector WIPI4. ATG2A is a rod-shaped protein that can bridge neighboring vesicles through interactions at each of its tips. WIPI4 binds to one of the tips, enabling the ATG2A-WIPI4 complex to tether a PI3P-containing vesicle to another PI3P-free vesicle. These data suggest that the ATG2A-WIPI4 complex mediates ER-phagophore association and/or tethers vesicles to the ER-phagophore junction, establishing the required organization for phagophore expansion via the transfer of lipid membranes from the ER and/or the vesicles to the phagophore.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Phosphatidylinositol Phosphates/metabolism , Amino Acid Sequence , Autophagy-Related Proteins/chemistry , Humans , Membrane Proteins/chemistry , Multiprotein Complexes/chemistry , Phosphatidylinositol Phosphates/chemistry , Protein Conformation , Sequence Homology
3.
Angew Chem Int Ed Engl ; 60(50): 26105-26114, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34590387

ABSTRACT

The autophagic ubiquitin-like protein LC3 functions through interactions with LC3-interaction regions (LIRs) of other autophagy proteins, including autophagy receptors, which stands out as a promising protein-protein interaction (PPI) target for the intervention of autophagy. Post-translational modifications like acetylation of Lys49 on the LIR-interacting surface could disrupt the interaction, offering an opportunity to design covalent small molecules interfering with the interface. Through screening covalent compounds, we discovered a small molecule modulator of LC3A/B that covalently modifies LC3A/B protein at Lys49. Activity-based protein profiling (ABPP) based evaluations reveal that a derivative molecule DC-LC3in-D5 exhibits a potent covalent reactivity and selectivity to LC3A/B in HeLa cells. DC-LC3in-D5 compromises LC3B lipidation in vitro and in HeLa cells, leading to deficiency in the formation of autophagic structures and autophagic substrate degradation. DC-LC3in-D5 could serve as a powerful tool for autophagy research as well as for therapeutic interventions.


Subject(s)
Autophagy/drug effects , Microtubule-Associated Proteins/metabolism , Small Molecule Libraries/pharmacology , HeLa Cells , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry
4.
Proc Natl Acad Sci U S A ; 110(47): 18844-9, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24191030

ABSTRACT

The autophagic ubiquitin-like protein (ublp) autophagy-related (ATG)12 is a component of the ATG12∼ATG5-ATG16L1 E3 complex that promotes lipid conjugation of members of the LC3 ublp family. A role of ATG12 in the E3 complex is to recruit the E2 enzyme ATG3. Here we report the identification of the ATG12 binding sequence in the flexible region of human ATG3 and the crystal structure of the minimal E3 complexed with the identified binding fragment of ATG3. The structure shows that 13 residues of the ATG3 fragment form a short ß-strand followed by an α-helix on a surface area that is exclusive to ATG12. Mutational analyses of ATG3 confirm that four residues whose side chains make contacts with ATG12 are important for E3 interaction as well as LC3 lipidation. Conservation of these four critical residues is high in metazoan organisms and plants but lower in fungi. A structural comparison reveals that the ATG3 binding surface on ATG12 contains a hydrophobic pocket corresponding to the binding pocket of LC3 that accommodates the leucine of the LC3-interacting region motif. These findings establish the mechanism of ATG3 recruitment by ATG12 in higher eukaryotes and place ATG12 among the members of signaling ublps that bind liner sequences.


Subject(s)
Autophagy/physiology , Models, Molecular , Protein Conformation , Small Ubiquitin-Related Modifier Proteins/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Autophagy-Related Protein 12 , Autophagy-Related Proteins , Blotting, Western , Crystallization , Humans , Immunoprecipitation , Magnetic Resonance Spectroscopy , Mutation/genetics , Protein Binding , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , X-Ray Diffraction
5.
Biochem Biophys Res Commun ; 463(3): 447-52, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26043688

ABSTRACT

Transfer of GABARAP thioester from the E1 ATG7 to the E2 ATG3 requires the interaction between the N-terminal domain of ATG7 and the flexible region (FR) of ATG3. This interaction has been visualized in the yeast Atg7-Atg3 complex crystal structure, but remains to be defined in higher eukaryotes. Here, our NMR data precisely define the region of the FR of human ATG3 that interacts with ATG7 (RIA7) and demonstrate RIA7 partially overlaps with the E3-interacting region, explaining how the E1-E2 and E2-E3 interactions are mutually exclusive. Mutational analyses identify critical residues of the RIA7 for the E1 interaction and GABARAP transfer, advancing our understanding of a molecular mechanism of the autophagic conjugation cascade in higher eukaryotes.


Subject(s)
Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Amino Acid Sequence , Animals , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Conserved Sequence , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Interaction Domains and Motifs , Protein Interaction Maps , Sequence Alignment , Ubiquitin-Activating Enzymes/chemistry
6.
Nat Struct Mol Biol ; 30(6): 778-784, 2023 06.
Article in English | MEDLINE | ID: mdl-37127820

ABSTRACT

Argonaute (AGO) proteins use small RNAs to recognize transcripts targeted for silencing in plants and animals. Many AGOs cleave target RNAs using an endoribonuclease activity termed 'slicing'. Slicing by DNA-guided prokaryotic AGOs has been studied in detail, but structural insights into RNA-guided slicing by eukaryotic AGOs are lacking. Here we present cryogenic electron microscopy structures of the Arabidopsis thaliana Argonaute10 (AtAgo10)-guide RNA complex with and without a target RNA representing a slicing substrate. The AtAgo10-guide-target complex adopts slicing-competent and slicing-incompetent conformations that are unlike known prokaryotic AGO structures. AtAgo10 slicing activity is licensed by docking target (t) nucleotides t9-t13 into a surface channel containing the AGO endoribonuclease active site. A ß-hairpin in the L1 domain secures the t9-t13 segment and coordinates t9-t13 docking with extended guide-target pairing. Results show that prokaryotic and eukaryotic AGOs use distinct mechanisms for achieving target slicing and provide insights into small interfering RNA potency.


Subject(s)
Arabidopsis , Argonaute Proteins , Animals , Argonaute Proteins/metabolism , RNA, Small Interfering/genetics , RNA Interference , Arabidopsis/genetics , Arabidopsis/metabolism , Catalytic Domain , RNA, Plant/genetics , RNA, Plant/metabolism
7.
Elife ; 112022 11 14.
Article in English | MEDLINE | ID: mdl-36373674

ABSTRACT

The human SMC5/6 complex is a conserved guardian of genome stability and an emerging component of antiviral responses. These disparate functions likely require distinct mechanisms of SMC5/6 regulation. In yeast, Smc5/6 is regulated by its Nse5/6 subunits, but such regulatory subunits for human SMC5/6 are poorly defined. Here, we identify a novel SMC5/6 subunit called SIMC1 that contains SUMO interacting motifs (SIMs) and an Nse5-like domain. We isolated SIMC1 from the proteomic environment of SMC5/6 within polyomavirus large T antigen (LT)-induced subnuclear compartments. SIMC1 uses its SIMs and Nse5-like domain to localize SMC5/6 to polyomavirus replication centers (PyVRCs) at SUMO-rich PML nuclear bodies. SIMC1's Nse5-like domain binds to the putative Nse6 orthologue SLF2 to form an anti-parallel helical dimer resembling the yeast Nse5/6 structure. SIMC1-SLF2 structure-based mutagenesis defines a conserved surface region containing the N-terminus of SIMC1's helical domain that regulates SMC5/6 localization to PyVRCs. Furthermore, SLF1, which recruits SMC5/6 to DNA lesions via its BRCT and ARD motifs, binds SLF2 analogously to SIMC1 and forms a separate Nse5/6-like complex. Thus, two Nse5/6-like complexes with distinct recruitment domains control human SMC5/6 localization.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Humans , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Proteomics , Viral Replication Compartments
8.
Nature ; 433(7025): 488-94, 2005 Feb 03.
Article in English | MEDLINE | ID: mdl-15635372

ABSTRACT

The conserved formin homology 2 (FH2) domain nucleates actin filaments and remains bound to the barbed end of the growing filament. Here we report the crystal structure of the yeast Bni1p FH2 domain in complex with tetramethylrhodamine-actin. Each of the two structural units in the FH2 dimer binds two actins in an orientation similar to that in an actin filament, suggesting that this structure could function as a filament nucleus. Biochemical properties of heterodimeric FH2 mutants suggest that the wild-type protein equilibrates between two bound states at the barbed end: one permitting monomer binding and the other permitting monomer dissociation. Interconversion between these states allows processive barbed-end polymerization and depolymerization in the presence of bound FH2 domain. Kinetic and/or thermodynamic differences in the conformational and binding equilibria can explain the variable activity of different FH2 domains as well as the effects of the actin-binding protein profilin on FH2 function.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Dimerization , Kinetics , Microfilament Proteins/genetics , Models, Biological , Models, Molecular , Mutation/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Rhodamines/chemistry , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship , Thermodynamics
9.
J Cell Biol ; 170(6): 889-93, 2005 Sep 12.
Article in English | MEDLINE | ID: mdl-16157699

ABSTRACT

The FH2 domains of formin family proteins act as processive cappers of actin filaments. Previously suggested stair-stepping mechanisms of processive capping imply that a formin cap rotates persistently in one direction with respect to the filament. This challenges the formin-mediated mechanism of intracellular cable formation. We suggest a novel scenario of processive capping that is driven by developing and relaxing torsion elastic stresses. Based on the recently discovered crystal structure of an FH2-actin complex, we propose a second mode of processive capping-the screw mode. Within the screw mode, the formin dimer rotates with respect to the actin filament in the direction opposite to that generated by the stair-stepping mode so that a combination of the two modes prevents persistent torsion strain accumulation. We determine an optimal regime of processive capping, whose essence is a periodic switch between the stair-stepping and screw modes. In this regime, elastic energy does not exceed feasible values, and supercoiling of actin filaments is prevented.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Microfilament Proteins , Rotation , Actin Cytoskeleton/genetics , Actins/metabolism , Dimerization , Elasticity , Fetal Proteins , Formins , Models, Biological , Nuclear Proteins , Protein Structure, Tertiary
10.
Nat Struct Mol Biol ; 27(12): 1194-1201, 2020 12.
Article in English | MEDLINE | ID: mdl-33106659

ABSTRACT

De novo formation of the double-membrane compartment autophagosome is seeded by small vesicles carrying membrane protein autophagy-related 9 (ATG9), the function of which remains unknown. Here we find that ATG9A scrambles phospholipids of membranes in vitro. Cryo-EM structures of human ATG9A reveal a trimer with a solvated central pore, which is connected laterally to the cytosol through the cavity within each protomer. Similarities to ABC exporters suggest that ATG9A could be a transporter that uses the central pore to function. Moreover, molecular dynamics simulation suggests that the central pore opens laterally to accommodate lipid headgroups, thereby enabling lipids to flip. Mutations in the pore reduce scrambling activity and yield markedly smaller autophagosomes, indicating that lipid scrambling by ATG9A is essential for membrane expansion. We propose ATG9A acts as a membrane-embedded funnel to facilitate lipid flipping and to redistribute lipids added to the outer leaflet of ATG9 vesicles, thereby enabling growth into autophagosomes.


Subject(s)
Autophagosomes/chemistry , Autophagy-Related Proteins/chemistry , Membrane Proteins/chemistry , Phospholipids/chemistry , Proteolipids/chemistry , Vesicular Transport Proteins/chemistry , Animals , Autophagosomes/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Binding Sites , Biological Transport , Cell Line , Cryoelectron Microscopy , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Expression , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Dynamics Simulation , Phospholipids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Proteolipids/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Red Fluorescent Protein
11.
Autophagy ; 15(11): 2031-2032, 2019 11.
Article in English | MEDLINE | ID: mdl-31441376

ABSTRACT

ATG2 is a rod-shaped membrane-tethering protein suggested to mediate the endoplasmic reticulum (ER)-phagophore association and the expansion of the phagophore. We recently demonstrated that human ATG2A transfers lipids between membranes in vitro, which led us to propose a model that the phagophore expands upon the transfer of lipids from the ER by ATG2. Here, we summarize our findings and arising issues that need to be addressed to establish the mechanism of phagophore expansion.


Subject(s)
Autophagy , Lipids , Autophagy-Related Proteins , Endoplasmic Reticulum , Humans , Membrane Proteins
12.
Elife ; 82019 07 04.
Article in English | MEDLINE | ID: mdl-31271352

ABSTRACT

An enigmatic step in de novo formation of the autophagosome membrane compartment is the expansion of the precursor membrane phagophore, which requires the acquisition of lipids to serve as building blocks. Autophagy-related 2 (ATG2), the rod-shaped protein that tethers phosphatidylinositol 3-phosphate (PI3P)-enriched phagophores to the endoplasmic reticulum (ER), is suggested to be essential for phagophore expansion, but the underlying mechanism remains unclear. Here, we demonstrate that human ATG2A is a lipid transfer protein. ATG2A can extract lipids from membrane vesicles and unload them to other vesicles. Lipid transfer by ATG2A is more efficient between tethered vesicles than between untethered vesicles. The PI3P effectors WIPI4 and WIPI1 associate ATG2A stably to PI3P-containing vesicles, thereby facilitating ATG2A-mediated tethering and lipid transfer between PI3P-containing vesicles and PI3P-free vesicles. Based on these results, we propose that ATG2-mediated transfer of lipids from the ER to the phagophore enables phagophore expansion.


Subject(s)
Autophagy-Related Proteins/metabolism , Lipid Metabolism , Membranes/metabolism , Humans , Membrane Proteins/metabolism , Protein Binding
13.
Methods Mol Biol ; 1880: 57-75, 2019.
Article in English | MEDLINE | ID: mdl-30610689

ABSTRACT

Members of the autophagy-related protein 8 (Atg8) family of ubiquitin-like proteins (ublps), including mammalian LC3 and GABARAP proteins, play crucial roles in autophagosome biogenesis, as well as selective autophagy. Upon induction of autophagy, the autophagic ublps are covalently attached to a phosphatidylethanolamine (PE) molecule of the autophagosomal membrane. This unique lipid conjugation of the autophagic ublps, which is essential for their functions, occurs in a ubiquitination-like reaction cascade consisting of the E1 enzyme ATG7, the E2 ATG3, and the E3 ATG12~ATG5-ATG16L1 complex (~denotes a covalent linkage). These enzymes are structurally unique among those of the canonical ubiquitination cascades, necessitating structural and biochemical studies of these molecules for understanding the molecular mechanisms underlying the lipidation cascade. Here, we will describe methods that were employed in our previous studies (Otomo et al., Nat Struct Mol Biol 20:59-66, 2013; Metlagel et al., Proc Natl Acad Sci U S A 110:18844-18849, 2013; Ohashi and Otomo, Biochem Biophys Res Commun 463:447-452, 2015), including the production of recombinant enzymes, in vitro enzymatic reactions, the crystallization of the E3 complexes, and the NMR-based investigations of E1-E2 and E2-E3 interactions.


Subject(s)
Autophagy-Related Proteins/chemistry , Cloning, Molecular/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Phosphatidylethanolamines/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Autophagy , Autophagy-Related Protein 12/chemistry , Autophagy-Related Protein 12/genetics , Autophagy-Related Protein 5/chemistry , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 8 Family/chemistry , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Proteins/genetics , Baculoviridae/genetics , Cell Line , Crystallization/methods , Escherichia coli/genetics , Humans , Insecta , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Protein Conformation , Transfection/methods , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics
14.
Article in English | MEDLINE | ID: mdl-30766969

ABSTRACT

The autophagosome precursor membrane, termed the "isolation membrane" or "phagophore," emerges adjacent to a PI3P-enriched transient subdomain of the ER called the "omegasome," thereafter expanding to engulf cytoplasmic content. Uncovering the molecular events that occur in the vicinity of the omegasome during phagophore biogenesis is imperative for understanding the mechanisms involved in this critical step of the autophagy pathway. We recently characterized the ATG2A-WIPI4 complex, one of the factors that localize to the omegasome and play a critical role in mediating phagophore expansion. Our structural and biochemical studies revealed that ATG2A is a rod-shaped protein with membrane-interacting properties at each end, endowing ATG2A with membrane-tethering capability. Association of the PI3P-binding protein WIPI4 at one of the ATG2A tips enables the ATG2A-WIPI4 complex to specifically tether PI3P-containing membranes to non-PI3P-containing membranes. We proposed models for the ATG2A-WIPI4 complex-mediated membrane associations between the omegasome and surrounding membranes, including the phagophore edge, the ER, ATG9 vesicles, and COPII vesicles.

15.
Autophagy ; 10(3): 522-3, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24413923

ABSTRACT

The members of the LC3/Atg8 family of proteins are covalently attached to phagophore and autophagosomal membranes. At the last step of the LC3 lipidation cascade, LC3 is transferred from the E2 enzyme ATG3 to phosphatidylethanolamine (PE). This transfer is stimulated by the ATG12-ATG5-ATG16L1 E3 complex, but the mechanism is not fully understood. We recently found that ATG12 of the E3 binds to a short sequence in the flexible region (FR) of ATG3 with high affinity, and that this interaction is critical for E2-E3 complex formation. These findings, together with detailed structural analyses of this interaction, define the properties of ATG12 and provide new insights of how LC3 transfer begins with ATG3 recruitment by ATG12.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy/physiology , Lipid Metabolism , Microfilament Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Humans , Protein Binding/physiology
16.
Nat Struct Mol Biol ; 20(1): 59-66, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23202584

ABSTRACT

The autophagy factor ATG12~ATG5 conjugate exhibits E3 ligase-like activity which facilitates the lipidation of members of the LC3 family. The crystal structure of the human ATG12~ATG5 conjugate bound to the N-terminal region of ATG16L1, the factor that recruits the conjugate to autophagosomal membranes, reveals an integrated architecture in which ATG12 docks onto ATG5 through conserved residues. ATG12 and ATG5 are oriented such that other conserved residues on each molecule, including the conjugation junction, form a continuous surface patch. Mutagenesis data support the importance of both the interface between ATG12 and ATG5 and the continuous patch for E3 activity. The ATG12~ATG5 conjugate interacts with the E2 enzyme ATG3 with high affinity through another surface location that is exclusive to ATG12, suggesting a different role of the continuous patch in E3 activity. These findings provide a foundation for understanding the mechanism of LC3 lipidation.


Subject(s)
Autophagy , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Binding Sites , Carrier Proteins/metabolism , Cell Line , Crystallography, X-Ray , HEK293 Cells , Humans , Mice , Mutation , Ubiquitin-Conjugating Enzymes/metabolism
18.
PLoS One ; 5(9)2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20927343

ABSTRACT

BACKGROUND: Formin proteins utilize a conserved formin homology 2 (FH2) domain to nucleate new actin filaments. In mammalian diaphanous-related formins (DRFs) the FH2 domain is inhibited through an unknown mechanism by intramolecular binding of the diaphanous autoinhibitory domain (DAD) and the diaphanous inhibitory domain (DID). METHODOLOGY/PRINCIPAL FINDINGS: Here we report the crystal structure of a complex between DID and FH2-DAD fragments of the mammalian DRF, mDia1 (mammalian diaphanous 1 also called Drf1 or p140mDia). The structure shows a tetrameric configuration (4 FH2 + 4 DID) in which the actin-binding sites on the FH2 domain are sterically occluded. However biochemical data suggest the full-length mDia1 is a dimer in solution (2 FH2 + 2 DID). Based on the crystal structure, we have generated possible dimer models and found that architectures of all of these models are incompatible with binding to actin filament but not to actin monomer. Furthermore, we show that the minimal functional monomeric unit in the FH2 domain, termed the bridge element, can be inhibited by isolated monomeric DID. NMR data on the bridge-DID system revealed that at least one of the two actin-binding sites on the bridge element is accessible to actin monomer in the inhibited state. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that autoinhibition in the native DRF dimer involves steric hindrance with the actin filament. Although the structure of a full-length DRF would be required for clarification of the presented models, our work here provides the first structural insights into the mechanism of the DRF autoinhibition.


Subject(s)
Carrier Proteins/chemistry , Homeostasis , Actins/chemistry , Actins/metabolism , Amino Acid Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallography, X-Ray , Dimerization , Formins , Molecular Conformation , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary
19.
Mol Cell ; 18(3): 273-81, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15866170

ABSTRACT

Diaphanous-related formins (DRFs) regulate dynamics of unbranched actin filaments during cell contraction and cytokinesis. DRFs are autoinhibited through intramolecular binding of a Diaphanous autoinhibitory domain (DAD) to a conserved N-terminal regulatory element. Autoinhibition is relieved through binding of the GTPase RhoA to the N-terminal element. We report the crystal structure of the dimeric regulatory domain of the DRF, mDia1. Dimerization is mediated by an intertwined six-helix bundle, from which extend two Diaphanous inhibitory domains (DIDs) composed of five armadillo repeats. NMR and biochemical mapping indicate the RhoA and DAD binding sites on the DID partially overlap, explaining activation of mDia1 by the GTPase. RhoA binding also requires an additional structurally independent segment adjacent to the DID. This regulatory construction, involving a GTPase binding site spanning a flexibly tethered arm and the inhibitory module, is observed in many autoinhibited effectors of Ras superfamily GTPases, suggesting evolutionary pressure for this design.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Protein Structure, Tertiary , rho GTP-Binding Proteins/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Crystallography, X-Ray , Formins , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Point Mutation
20.
Biochemistry ; 42(14): 3997-4008, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12680752

ABSTRACT

The temporal and spatial control of Rho GTPase signaling pathways is a central issue in understanding the molecular mechanisms that generate complex cellular movements. The Rho protein Cdc42 induces a significant conformational change in its downstream effector, the Wiskott-Aldrich syndrome protein (WASP). On the basis of this conformational change, we have created a series of single-molecule sensors for both active Cdc42 and Cdc42 guanine nucleotide exchange factors (GEFs) that utilize fluorescence resonance energy transfer (FRET) between cyan and yellow fluorescent proteins. In vitro, the Cdc42 sensors produce up to 3.2-fold FRET emission ratio changes upon binding active Cdc42. The GEF sensors yield up to 1.7-fold changes in FRET upon exchange of GDP for GTP. The GEF-catalyzed rate of nucleotide exchange for the GEF sensor is indistinguishable from that of wild-type Cdc42, but GAP-catalyzed nucleotide hydrolysis is slowed approximately 16-fold. In vivo, both sensors faithfully report on Cdc42 and/or Cdc42-GEF activity. These results establish the successful creation of rationally designed and genetically encoded tools that can be used to image the activity of biologically and medically important molecules in living systems.


Subject(s)
Energy Transfer , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , Fluorescence , Plasmids
SELECTION OF CITATIONS
SEARCH DETAIL