Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2282, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35477723

ABSTRACT

Treatment with androgen receptor pathway inhibitors (ARPIs) in prostate cancer leads to the emergence of resistant tumors characterized by lineage plasticity and differentiation toward neuroendocrine lineage. Here, we find that ARPIs induce a rapid epigenetic alteration mediated by large-scale chromatin remodeling to support activation of stem/neuronal transcriptional programs. We identify the proneuronal transcription factor ASCL1 motif to be enriched in hyper-accessible regions. ASCL1 acts as a driver of the lineage plastic, neuronal transcriptional program to support treatment resistance and neuroendocrine phenotype. Targeting ASCL1 switches the neuroendocrine lineage back to the luminal epithelial state. This effect is modulated by disruption of the polycomb repressive complex-2 through UHRF1/AMPK axis and change the chromatin architecture in favor of luminal phenotype. Our study provides insights into the epigenetic alterations induced by ARPIs, governed by ASCL1, provides a proof of principle of targeting ASCL1 to reverse neuroendocrine phenotype, support luminal conversion and re-addiction to ARPIs.


Subject(s)
Chromatin , Prostatic Neoplasms , Androgen Receptor Antagonists , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Chromatin/genetics , Chromatin/metabolism , Humans , Male , Neurons/metabolism , Prostatic Neoplasms/pathology , Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
J Clin Invest ; 130(2): 699-714, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31845908

ABSTRACT

HSP27 is highly expressed in, and supports oncogene addiction of, many cancers. HSP27 phosphorylation is a limiting step for activation of this protein and a target for inhibition, but its highly disordered structure challenges rational structure-guided drug discovery. We performed multistep biochemical, structural, and computational experiments to define a spherical 24-monomer complex composed of 12 HSP27 dimers with a phosphorylation pocket flanked by serine residues between their N-terminal domains. Ivermectin directly binds this pocket to inhibit MAPKAP2-mediated HSP27 phosphorylation and depolymerization, thereby blocking HSP27-regulated survival signaling and client-oncoprotein interactions. Ivermectin potentiated activity of anti-androgen receptor and anti-EGFR drugs in prostate and EGFR/HER2-driven tumor models, respectively, identifying a repurposing approach for cotargeting stress-adaptive responses to overcome resistance to inhibitors of oncogenic pathway signaling.


Subject(s)
Heat-Shock Proteins , Ivermectin , Molecular Chaperones , Neoplasms, Experimental , Receptor, ErbB-2 , A549 Cells , Animals , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ivermectin/chemistry , Ivermectin/pharmacology , Mice , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Domains , Protein Multimerization , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
3.
PLoS One ; 12(10): e0186869, 2017.
Article in English | MEDLINE | ID: mdl-29088253

ABSTRACT

Cathepsin K (CatK) is a cysteine protease that plays an important role in mammalian intra- and extracellular protein turnover and is known for its unique and potent collagenase activity. Through studies on the mechanism of its collagenase activity, selective ectosteric sites were identified that are remote from the active site. Inhibitors targeting these ectosteric sites are collagenase selective and do not interfere with other proteolytic activities of the enzyme. Potential ectosteric inhibitors were identified using a computational approach to screen the druggable subset of and the entire 281,987 compounds comprising Chemical Repository library of the National Cancer Institute-Developmental Therapeutics Program (NCI-DTP). Compounds were scored based on their affinity for the ectosteric site. Here we compared the scores of three individual molecular docking methods with that of a composite score of all three methods together. The composite docking method was up to five-fold more effective at identifying potent collagenase inhibitors (IC50 < 20 µM) than the individual methods. Of 160 top compounds tested in enzymatic assays, 28 compounds revealed blocking of the collagenase activity of CatK at 100 µM. Two compounds exhibited IC50 values below 5 µM corresponding to a molar protease:inhibitor concentration of <1:12. Both compounds were subsequently tested in osteoclast bone resorption assays where the most potent inhibitor, 10-[2-[bis(2-hydroxyethyl)amino]ethyl]-7,8-diethylbenzo[g]pteridine-2,4-dione, (NSC-374902), displayed an inhibition of bone resorption with an IC50-value of approximately 300 nM and no cell toxicity effects.


Subject(s)
Cathepsin K/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Molecular Docking Simulation/methods , Allosteric Regulation , Allosteric Site , Binding Sites , Catalytic Domain , Cathepsin K/chemistry , Cathepsin K/metabolism , Cells, Cultured , Collagenases/chemistry , Collagenases/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/metabolism , Humans , Molecular Structure , Osteoclasts/drug effects , Osteoclasts/metabolism , Protein Binding , Protein Domains
4.
Biochem J ; 474(5): 851-864, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28049758

ABSTRACT

Cathepsin K (CatK) is the predominant mammalian bone-degrading protease and thus an ideal target for antiosteoporotic drug development. Rodent models of osteoporosis are preferred due to their close reflection of the human disease and their ease of handling, genetic manipulation and economic affordability. However, large differences in the potency of CatK inhibitors for the mouse/rat vs. the human protease orthologs have made it impossible to use rodent models. This is even more of a problem considering that the most advanced CatK inhibitors, including odanacatib (ODN) and balicatib, failed in human clinical trials due to side effects and rodent models are not available to investigate the mechanism of these failures. Here, we elucidated the structural elements of the potency differences between mouse and human CatK (hCatK) using ODN. We determined and compared the structures of inhibitor-free mouse CatK (mCatK), hCatK and ODN bound to hCatK. Two structural differences were identified and investigated by mutational analysis. Humanizing subsite 2 in mCatK led to a 5-fold improvement of ODN binding, whereas the replacement of Tyr61 in mCatK with Asp resulted in an hCatK with comparable ODN potency. Combining both sites further improved the inhibition of the mCatK variant. Similar results were obtained for balicatib. These findings will allow the generation of transgenic CatK mice that will facilitate the evaluation of CatK inhibitor adverse effects and to explore routes to avoid them.


Subject(s)
Benzamides/chemistry , Biphenyl Compounds/chemistry , Bone Density Conservation Agents/chemistry , Cathepsin K/antagonists & inhibitors , Piperazines/chemistry , Protease Inhibitors/chemistry , Amino Acid Sequence , Animals , Benzamides/metabolism , Binding Sites , Biphenyl Compounds/metabolism , Bone Density Conservation Agents/metabolism , Cathepsin K/chemistry , Cathepsin K/genetics , Cathepsin K/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Ligands , Mice , Mutagenesis, Site-Directed , Piperazines/metabolism , Protease Inhibitors/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Structural Homology, Protein
5.
J Nat Prod ; 79(8): 1962-70, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27498895

ABSTRACT

Natural products are an important source of novel drug scaffolds. The highly variable and unpredictable timelines associated with isolating novel compounds and elucidating their structures have led to the demise of exploring natural product extract libraries in drug discovery programs. Here we introduce affinity crystallography as a new methodology that significantly shortens the time of the hit to active structure cycle in bioactive natural product discovery research. This affinity crystallography approach is illustrated by using semipure fractions of an actinomycetes culture extract to isolate and identify a cathepsin K inhibitor and to compare the outcome with the traditional assay-guided purification/structural analysis approach. The traditional approach resulted in the identification of the known inhibitor antipain (1) and its new but lower potency dehydration product 2, while the affinity crystallography approach led to the identification of a new high-affinity inhibitor named lichostatinal (3). The structure and potency of lichostatinal (3) was verified by total synthesis and kinetic characterization. To the best of our knowledge, this is the first example of isolating and characterizing a potent enzyme inhibitor from a partially purified crude natural product extract using a protein crystallographic approach.


Subject(s)
Biological Products/pharmacology , Cathepsin K/antagonists & inhibitors , Lichens/chemistry , Peptides/isolation & purification , Peptides/pharmacology , Antipain/chemistry , Antipain/pharmacology , Biological Products/chemical synthesis , Biological Products/chemistry , British Columbia , Crystallography, X-Ray , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry
6.
ACS Cent Sci ; 2(3): 154-161, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27066537

ABSTRACT

Selective inhibitors of human pancreatic α-amylase (HPA) are an effective means of controlling blood sugar levels in the management of diabetes. A high-throughput screen of marine natural product extracts led to the identification of a potent (Ki = 10 pM) peptidic HPA inhibitor, helianthamide, from the Caribbean sea anemone Stichodactyla helianthus. Active helianthamide was produced in Escherichia coli via secretion as a barnase fusion protein. X-ray crystallographic analysis of the complex of helianthamide with porcine pancreatic α-amylase revealed that helianthamide adopts a ß-defensin fold and binds into and across the amylase active site, utilizing a contiguous YIYH inhibitory motif. Helianthamide represents the first of a novel class of glycosidase inhibitors and provides an unusual example of functional malleability of the ß-defensin fold, which is rarely seen outside of its traditional role in antimicrobial peptides.

7.
Proc Natl Acad Sci U S A ; 111(49): 17474-9, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25422423

ABSTRACT

Cathepsin K is the major collagenolytic protease in bone that facilitates physiological as well as pathological bone degradation. Despite its key role in bone remodeling and for being a highly sought-after drug target for the treatment of osteoporosis, the mechanism of collagen fiber degradation by cathepsin K remained elusive. Here, we report the structure of a collagenolytically active cathepsin K protein dimer. Cathepsin K is organized into elongated C-shaped protease dimers that reveal a putative collagen-binding interface aided by glycosaminoglycans. Molecular modeling of collagen binding to the dimer indicates the participation of nonactive site amino acid residues, Q21 and Q92, in collagen unfolding. Mutations at these sites as well as perturbation of the dimer protein-protein interface completely inhibit cathepsin-K-mediated fiber degradation without affecting the hydrolysis of gelatin or synthetic peptide. Using scanning electron microscopy, we demonstrate the specific binding of cathepsin K at the edge of the fibrillar gap region of collagen fibers, which suggest initial cleavage events at the N- and C-terminal ends of tropocollagen molecules. Edman degradation analysis of collagen fiber degradation products revealed those initial cleavage sites. We propose that one cathepsin K molecule binds to collagen-bound glycosaminoglycans at the gap region and recruits a second protease molecule that provides an unfolding and cleavage mechanism for triple helical collagen. Removal of collagen-associated glycosaminoglycans prevents cathepsin K binding and subsequently fiber hydrolysis. Cathepsin K dimer and glycosaminoglycan binding sites represent novel targeting sites for the development of nonactive site-directed second-generation inhibitors of this important drug target.


Subject(s)
Cathepsin K/chemistry , Collagen/chemistry , Amino Acids/chemistry , Binding Sites , Bone Remodeling , Bone and Bones/metabolism , Crystallography, X-Ray , Glycosaminoglycans/chemistry , Humans , Hydrolysis , Microscopy, Electron , Models, Molecular , Mutagenesis , Osteoporosis , Peptide Hydrolases/chemistry , Pichia , Protein Denaturation , Protein Folding , Protein Multimerization , Protein Structure, Tertiary , Substrate Specificity
8.
PLoS One ; 9(7): e103598, 2014.
Article in English | MEDLINE | ID: mdl-25072253

ABSTRACT

Due to the emergence of resistance toward current antibiotics, there is a pressing need to develop the next generation of antibiotics as therapeutics against infectious and opportunistic diseases of microbial origins. The shikimate pathway is exclusive to microbes, plants and fungi, and hence is an attractive and logical target for development of antimicrobial therapeutics. The Gram-positive commensal microbe, Enterococcus faecalis, is a major human pathogen associated with nosocomial infections and resistance to vancomycin, the "drug of last resort". Here, we report the identification of several polyketide-based inhibitors against the E. faecalis shikimate pathway enzyme, 3-dehydroquinate dehydratase (DHQase). In particular, marein, a flavonoid polyketide, both inhibited DHQase and retarded the growth of Enterococcus faecalis. The purification, crystallization and structural resolution of recombinant DHQase from E. faecalis (at 2.2 Å resolution) are also reported. This study provides a route in the development of polyketide-based antimicrobial inhibitors targeting the shikimate pathway of the human pathogen E. faecalis.


Subject(s)
Enterococcus faecalis/enzymology , Enzyme Inhibitors/chemistry , Hydro-Lyases/antagonists & inhibitors , Shikimic Acid/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Enterococcus faecalis/metabolism , Enzyme Inhibitors/metabolism , Flavonoids/chemistry , Flavonoids/metabolism , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Kinetics , Polyketides/chemistry , Polyketides/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Shikimic Acid/chemistry
9.
Glycobiology ; 23(9): 1075-83, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23735230

ABSTRACT

Trehalose synthase (TreS) catalyzes the reversible conversion of maltose into trehalose in mycobacteria as one of three biosynthetic pathways to this nonreducing disaccharide. Given the importance of trehalose to survival of mycobacteria, there has been considerable interest in understanding the enzymes involved in its production; indeed the structures of the key enzymes in the other two pathways have already been determined. Herein, we present the first structure of TreS from Mycobacterium smegmatis, thereby providing insights into the catalytic machinery involved in this intriguing intramolecular reaction. This structure, which is of interest both mechanistically and as a potential pharmaceutical target, reveals a narrow and enclosed active site pocket within which intramolecular substrate rearrangements can occur. We also present the structure of a complex of TreS with acarbose, revealing a hitherto unsuspected oligosaccharide-binding site within the C-terminal domain. This may well provide an anchor point for the association of TreS with glycogen, thereby enhancing its role in glycogen biosynthesis and degradation.


Subject(s)
Acarbose/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/metabolism , Mycobacterium smegmatis/enzymology , Acarbose/chemistry , Acarbose/pharmacology , Amino Acid Sequence , Biocatalysis/drug effects , Catalytic Domain/drug effects , Glucosyltransferases/antagonists & inhibitors , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Alignment , Structure-Activity Relationship
10.
Nat Struct Mol Biol ; 17(4): 497-503, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20357771

ABSTRACT

Capping protein (CP) regulates actin dynamics by binding the barbed ends of actin filaments. Removal of CP may be one means to harness actin polymerization for processes such as cell movement and endocytosis. Here we structurally and biochemically investigated a CP interaction (CPI) motif present in the otherwise unrelated proteins CARMIL and CD2AP. The CPI motif wraps around the stalk of the mushroom-shaped CP at a site distant from the actin-binding interface, which lies on the top of the mushroom cap. We propose that the CPI motif may act as an allosteric modulator, restricting CP to a low-affinity, filament-binding conformation. Structure-based sequence alignments extend the CPI motif-containing family to include CIN85, CKIP-1, CapZIP and a relatively uncharacterized protein, WASHCAP (FAM21). Peptides comprising these CPI motifs are able to inhibit CP and to uncap CP-bound actin filaments.


Subject(s)
Actin Capping Proteins/chemistry , Actin Capping Proteins/metabolism , Actins/metabolism , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
11.
J Struct Biol ; 162(3): 491-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18467126

ABSTRACT

This research describes four X-ray structures of Vibrio harveyi chitinase A and its catalytically inactive mutant (E315M) in the presence and absence of substrates. The overall structure of chitinase A is that of a typical family-18 glycosyl hydrolase comprising three distinct domains: (i) the amino-terminal chitin-binding domain; (ii) the main catalytic (alpha/beta)(8) TIM-barrel domain; and (iii) the small (alpha+beta) insertion domain. The catalytic cleft of chitinase A has a long, deep groove, which contains six chitooligosaccharide ring-binding subsites (-4)(-3)(-2)(-1)(+1)(+2). The binding cleft of the ligand-free E315M is partially blocked by the C-terminal (His)(6)-tag. Structures of E315M-chitooligosaccharide complexes display a linear conformation of pentaNAG, but a bent conformation of hexaNAG. Analysis of the final 2F(o)-F(c) omit map of E315M-NAG6 reveals the existence of the linear conformation of the hexaNAG at a lower occupancy with respect to the bent conformation. These crystallographic data provide evidence that the interacting sugars undergo conformational changes prior to hydrolysis by the wild-type enzyme.


Subject(s)
Chitinases/chemistry , Oligosaccharides/chemistry , Binding Sites , Catalysis , Catalytic Domain , Chitin/chemistry , Cloning, Molecular , Crystallography, X-Ray/methods , Hydrolysis , Molecular Conformation , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Vibrio/enzymology
12.
Ann N Y Acad Sci ; 1112: 56-66, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17468228

ABSTRACT

In recent years two structures have been reported that demonstrate how the two halves of a beta-thymosin repeat bind to actin monomers. Here we assess the validity of these structures and construct minimally biased models of the beta-thymosin:actin complexes. The models reveal that the beta-thymosins interact with actin throughout their length and that all the conserved residues are functional in this interface. These models are judged to be in excellent agreement with published biochemical and functional data. In particular, the models are consistent with the actin monomer sequestering and actin filament binding properties of beta-thymosins. The models also correctly predict competition between thymosin-beta4 with DNase I or profilin in binding actin while allowing ternary complexes at higher concentrations.


Subject(s)
Actins/chemistry , Actins/metabolism , Thymosin/analogs & derivatives , Thymosin/chemistry , Amino Acid Sequence , Animals , Consensus Sequence , Models, Molecular , Protein Binding , Protein Conformation , Thymosin/metabolism
13.
Article in English | MEDLINE | ID: mdl-17401197

ABSTRACT

Human filamin A is a 280 kDa protein involved in actin-filament cross-linking. It is structurally divided into an actin-binding headpiece (ABD) and a rod domain containing 24 immunoglobulin-like (Ig) repeats. A fragment of human filamin A (Ig repeats 14-16) was cloned and expressed in Escherichia coli and the purified protein was crystallized in 1.6 M ammonium sulfate, 2% PEG 1000 and 100 mM HEPES pH 7.5. The crystals diffracted to 1.95 A and belong to space group P2(1)2(1)2(1), with unit-cell parameters a = 50.63, b = 52.10, c = 98.46 A, alpha = beta = gamma = 90 degrees.


Subject(s)
Contractile Proteins/chemistry , Microfilament Proteins/chemistry , Base Sequence , Cloning, Molecular , Contractile Proteins/genetics , Crystallization , Crystallography, X-Ray , DNA Primers , DNA, Complementary , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Filamins , Humans , Microfilament Proteins/genetics , Protein Conformation
14.
Structure ; 14(3): 469-76, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16531231

ABSTRACT

Participation of actin in cellular processes relies on the dynamics of filament assembly. Filament elongation is fed by monomeric actin in complex with either profilin or a Wiscott-Aldrich syndrome protein (WASP) homology domain 2 (WH2)/beta-thymosin (betaT) domain. WH2/betaT motif repetition (typified by ciboulot) or combination with nonrelated domains (as found in N-WASP) results in proteins that yield their actin to filament elongation. Here, we report the crystal structures of actin bound hybrid proteins, constructed between gelsolin and WH2/betaT domains from ciboulot or N-WASP. We observe the C-terminal half of ciboulot domain 2 bound to actin. In solution, we show that cibolout domains 2 and 3 bind to both G- and F-actin, and that whole ciboulot forms a complex with two actin monomers. In contrast, the analogous portion of N-WASP WH2 domain 2 is detached from actin, indicating that the C-terminal halves of the betaT and WH2 motifs are not functionally analogous.


Subject(s)
Actins/metabolism , Drosophila Proteins/chemistry , Gelsolin/chemistry , Microfilament Proteins/chemistry , Nerve Tissue Proteins/chemistry , Thymosin/chemistry , Wiskott-Aldrich Syndrome Protein/chemistry , Actins/chemistry , Amino Acid Sequence , Animals , Drosophila , Drosophila Proteins/metabolism , Microfilament Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Structure-Activity Relationship , Thymosin/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism
15.
EMBO Rep ; 6(3): 220-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15741975

ABSTRACT

Movement is a defining characteristic of life. Macroscopic motion is driven by the dynamic interactions of myosin with actin filaments in muscle. Directed polymerization of actin behind the advancing membrane of a eukaryotic cell generates microscopic movement. Despite the fundamental importance of actin in these processes, the structure of the actin filament remains unknown. The Holmes model of the actin filament was published 15 years ago, and although it has been widely accepted, no high-resolution structural data have yet confirmed its veracity. Here, we review the implications of recently determined structures of F-actin-binding proteins for the structure of the actin filament and suggest a series of in silico tests for actin-filament models. We also review the significance of these structures for the arp2/3-mediated branched filament.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/metabolism , Actin-Related Protein 2 , Actin-Related Protein 3 , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Protein Binding , Protein Conformation
16.
EMBO J ; 23(18): 3599-608, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15329672

ABSTRACT

The WH2 (Wiscott-Aldridge syndrome protein homology domain 2) repeat is an actin interacting motif found in monomer sequestering and filament assembly proteins. We have stabilized the prototypical WH2 family member, thymosin-beta4 (Tbeta4), with respect to actin, by creating a hybrid between gelsolin domain 1 and the C-terminal half of Tbeta4 (G1-Tbeta4). This hybrid protein sequesters actin monomers, severs actin filaments and acts as a leaky barbed end cap. Here, we present the structure of the G1-Tbeta4:actin complex at 2 A resolution. The structure reveals that Tbeta4 sequesters by capping both ends of the actin monomer, and that exchange of actin between Tbeta4 and profilin is mediated by a minor overlap in binding sites. The structure implies that multiple WH2 motif-containing proteins will associate longitudinally with actin filaments. Finally, we discuss the role of the WH2 motif in arp2/3 activation.


Subject(s)
Actins/chemistry , Thymosin/chemistry , Actin-Related Protein 2 , Actin-Related Protein 3 , Actins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Crystallography, X-Ray , Cytoskeletal Proteins/chemistry , Cytoskeleton/metabolism , Gelsolin/chemistry , Gelsolin/genetics , Gelsolin/metabolism , Humans , Molecular Sequence Data , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Thymosin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...