Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Open Biol ; 14(3): 230376, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38503329

ABSTRACT

Fascin-1-mediated actin-bundling activity is central to the generation of plasma membrane protrusions required for cell migration. Dysregulated formation of cellular protrusions is observed in metastatic cancers, where they are required for increased invasiveness, and is often correlated with increased Fascin-1 abundance. Therefore, there is interest in generating therapeutic Fascin-1 inhibitors. We present the identification of Nb 3E11, a nanobody inhibitor of Fascin-1 actin-bundling activity and filopodia formation. The crystal structure of the Fascin-1/Nb 3E11 complex reveals the structural mechanism of inhibition. Nb 3E11 occludes an actin-binding site on the third ß-trefoil domain of Fascin-1 that is currently not targeted by chemical inhibitors. Binding of Nb 3E11 to Fascin-1 induces a conformational change in the adjacent domains to stabilize Fascin-1 in an inhibitory state similar to that adopted in the presence of small-molecule inhibitors. Nb 3E11 could be used as a tool inhibitor molecule to aid in the development of Fascin-1 targeted therapeutics.


Subject(s)
Actins , Carrier Proteins , Microfilament Proteins , Pseudopodia , Actins/metabolism , Pseudopodia/metabolism , Protein Binding , Cell Movement
2.
Redox Biol ; 28: 101318, 2020 01.
Article in English | MEDLINE | ID: mdl-31546169

ABSTRACT

Aurora A kinase is a master mitotic regulator whose functions are controlled by several regulatory interactions and post-translational modifications. It is frequently dysregulated in cancer, making Aurora A inhibition a very attractive antitumor target. However, recently uncovered links between Aurora A, cellular metabolism and redox regulation are not well understood. In this study, we report a novel mechanism of Aurora A regulation in the cellular response to oxidative stress through CoAlation. A combination of biochemical, biophysical, crystallographic and cell biology approaches revealed a new and, to our knowledge, unique mode of Aurora A inhibition by CoA, involving selective binding of the ADP moiety of CoA to the ATP binding pocket and covalent modification of Cys290 in the activation loop by the thiol group of the pantetheine tail. We provide evidence that covalent CoA modification (CoAlation) of Aurora A is specific, and that it can be induced by oxidative stress in human cells. Oxidising agents, such as diamide, hydrogen peroxide and menadione were found to induce Thr 288 phosphorylation and DTT-dependent dimerization of Aurora A. Moreover, microinjection of CoA into fertilized mouse embryos disrupts bipolar spindle formation and the alignment of chromosomes, consistent with Aurora A inhibition. Altogether, our data reveal CoA as a new, rather selective, inhibitor of Aurora A, which locks this kinase in an inactive state via a "dual anchor" mechanism of inhibition that might also operate in cellular response to oxidative stress. Finally and most importantly, we believe that these novel findings provide a new rationale for developing effective and irreversible inhibitors of Aurora A, and perhaps other protein kinases containing appropriately conserved Cys residues.


Subject(s)
Aurora Kinase A/chemistry , Aurora Kinase A/metabolism , Coenzyme A/administration & dosage , Animals , Coenzyme A/chemistry , Coenzyme A/pharmacology , Crystallography, X-Ray , HEK293 Cells , Hep G2 Cells , Humans , Mice , Models, Molecular , Oxidative Stress , Phosphorylation , Protein Conformation , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
3.
EMBO J ; 37(8)2018 04 13.
Article in English | MEDLINE | ID: mdl-29510984

ABSTRACT

Aurora-A regulates the recruitment of TACC3 to the mitotic spindle through a phospho-dependent interaction with clathrin heavy chain (CHC). Here, we describe the structural basis of these interactions, mediated by three motifs in a disordered region of TACC3. A hydrophobic docking motif binds to a previously uncharacterized pocket on Aurora-A that is blocked in most kinases. Abrogation of the docking motif causes a delay in late mitosis, consistent with the cellular distribution of Aurora-A complexes. Phosphorylation of Ser558 engages a conformational switch in a second motif from a disordered state, needed to bind the kinase active site, into a helical conformation. The helix extends into a third, adjacent motif that is recognized by a helical-repeat region of CHC, not a recognized phospho-reader domain. This potentially widespread mechanism of phospho-recognition provides greater flexibility to tune the molecular details of the interaction than canonical recognition motifs that are dominated by phosphate binding.


Subject(s)
Aurora Kinase A/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Cell Line , Humans , Microtubule-Associated Proteins/genetics , Protein Conformation, alpha-Helical
4.
Biomol NMR Assign ; 12(2): 221-224, 2018 10.
Article in English | MEDLINE | ID: mdl-29582386

ABSTRACT

The microtubule regulatory protein colonic and hepatic tumor overexpressed gene (chTOG), also known as cytoskeleleton associated protein 5 (CKAP5) plays an important role in organizing the cytoskeleton and in particular in the assembly of k-fibres in mitosis. Recently, we dissected the hitherto poorly understood C-terminus of this protein by discovering two new domains-a cryptic TOG domain (TOG6) and a smaller, helical domain at the very C-terminus. It was shown that the C-terminal domain is important for the interaction with the TACC domain in TACC3 during the assembly of k-fibres in a ternary complex that also includes clathrin. Here we now present the solution NMR assignment of the chTOG C-terminal domain which confirms our earlier prediction that it is mainly made of α-helices. However, the appearance of the 1H-15N HSQC spectrum is indicative of the presence of a considerable amount of unstructured and possibly flexible portions of protein in the domain.


Subject(s)
Microtubule-Associated Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Humans , Protein Domains , Solutions
5.
ACS Chem Biol ; 12(11): 2906-2914, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29045126

ABSTRACT

The mitotic kinase Aurora-A and its partner protein TPX2 (Targeting Protein for Xenopus kinesin-like protein 2) are overexpressed in cancers, and it has been proposed that they work together as an oncogenic holoenzyme. TPX2 is responsible for activating Aurora-A during mitosis, ensuring proper cell division. Disruption of the interface with TPX2 is therefore a potential target for novel anticancer drugs that exploit the increased sensitivity of cancer cells to mitotic stress. Here, we investigate the interface using coprecipitation assays and isothermal titration calorimetry to quantify the energetic contribution of individual residues of TPX2. Residues Tyr8, Tyr10, Phe16, and Trp34 of TPX2 are shown to be crucial for robust complex formation, suggesting that the interaction could be abrogated through blocking any of the three pockets on Aurora-A that complement these residues. Phosphorylation of Aurora-A on Thr288 is also necessary for high-affinity binding, and here we identify arginine residues that communicate the phosphorylation of Thr288 to the TPX2 binding site. With these findings in mind, we conducted a high-throughput X-ray crystallography-based screen of 1255 fragments against Aurora-A and identified 59 hits. Over three-quarters of these hits bound to the pockets described above, both validating our identification of hotspots and demonstrating the druggability of this protein-protein interaction. Our study exemplifies the potential of high-throughput crystallography facilities such as XChem to aid drug discovery. These results will accelerate the development of chemical inhibitors of the Aurora-A/TPX2 interaction.


Subject(s)
Aurora Kinase A/metabolism , Cell Cycle Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Protein Interaction Maps/drug effects , Aurora Kinase A/chemistry , Binding Sites/drug effects , Cell Cycle Proteins/chemistry , Crystallography, X-Ray , Drug Discovery , Humans , Ligands , Microtubule-Associated Proteins/chemistry , Molecular Docking Simulation , Nuclear Proteins/chemistry , Protein Binding/drug effects , Thiazolidines/chemistry , Thiazolidines/pharmacology
6.
Biochem Soc Trans ; 45(3): 709-717, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28620032

ABSTRACT

The Myc proteins comprise a family of ubiquitous regulators of gene expression implicated in over half of all human cancers. They interact with a large number of other proteins, such as transcription factors, chromatin-modifying enzymes and kinases. Remarkably, few of these interactions have been characterized structurally. This is at least in part due to the intrinsically disordered nature of Myc proteins, which adopt a defined conformation only in the presence of binding partners. Owing to this behaviour, crystallographic studies on Myc proteins have been limited to short fragments in complex with other proteins. Most recently, we determined the crystal structure of Aurora-A kinase domain bound to a 28-amino acid fragment of the N-Myc transactivation domain. The structure reveals an α-helical segment within N-Myc capped by two tryptophan residues that recognize the surface of Aurora-A. The kinase domain acts as a molecular scaffold, independently of its catalytic activity, upon which this region of N-Myc becomes ordered. The binding site for N-Myc on Aurora-A is disrupted by certain ATP-competitive inhibitors, such as MLN8237 (alisertib) and CD532, and explains how these kinase inhibitors are able to disrupt the protein-protein interaction to affect Myc destabilization. Structural studies on this and other Myc complexes will lead to the design of protein-protein interaction inhibitors as chemical tools to dissect the complex pathways of Myc regulation and function, which may be developed into Myc inhibitors for the treatment of cancer.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/chemistry , Azepines/pharmacology , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/chemistry , Pyrimidines/pharmacology , Aurora Kinase A/metabolism , Azepines/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phenylurea Compounds/therapeutic use , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/therapeutic use
7.
FEBS J ; 284(18): 2947-2954, 2017 09.
Article in English | MEDLINE | ID: mdl-28342286

ABSTRACT

Protein kinases are central players in the regulation of cell cycle and signalling pathways. Their catalytic activities are strictly regulated through post-translational modifications and protein-protein interactions that control switching between inactive and active states. These states have been studied extensively using protein crystallography, although the dynamic nature of protein kinases makes it difficult to capture all relevant states. Here, we describe two recent structures of Aurora-A kinase that trap its active and inactive states. In both cases, Aurora-A is trapped through interaction with a synthetic protein, either a single-domain antibody that inhibits the kinase or a hydrocarbon-stapled peptide that activates the kinase. These structures show how the distinct synthetic proteins target the same allosteric pocket with opposing effects on activity. These studies pave the way for the development of tools to probe these allosteric mechanisms in cells.


Subject(s)
Aurora Kinase A/chemistry , Cell Cycle Proteins/chemistry , Microtubule-Associated Proteins/chemistry , Nuclear Proteins/chemistry , Peptides/chemistry , Protein Processing, Post-Translational , Single-Domain Antibodies/chemistry , Allosteric Regulation , Allosteric Site , Amino Acid Motifs , Aurora Kinase A/metabolism , Binding Sites , Cell Cycle Proteins/metabolism , Crystallography, X-Ray , Humans , Kinetics , Microtubule-Associated Proteins/metabolism , Models, Molecular , Nuclear Proteins/metabolism , Peptides/metabolism , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Single-Domain Antibodies/metabolism , Substrate Specificity
8.
ChemistryOpen ; 5(6): 531-534, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28032021

ABSTRACT

The structure of protein kinases has been extensively studied by protein crystallography. Conformational movement of the kinase activation loop is thought to be crucial for regulation of activity; however, in many cases the position of the activation loop in solution is unknown. Protein kinases are an important class of therapeutic target and kinase inhibitors are classified by their effect on the activation loop. Here, we report the use of pulsed electron double resonance (PELDOR) and site-directed spin labeling to monitor conformational changes through the insertion of MTSL [S-(1-oxyl-2,2,5,5-tetramethyl-2,5-dihydro-1 H-pyrrol-3-yl)methyl methanesulfonothioate] on the dynamic activation loop and a stable site on the outer surface of the enzyme. The action of different ligands such as microtubule-associated protein (TPX2) and inhibitors could be discriminated as well as their ability to lock the activation loop in a fixed conformation. This study provides evidence for structural adaptations that could be used for drug design and a methodological approach that has potential to characterize inhibitors in development.

9.
Proc Natl Acad Sci U S A ; 113(48): 13726-13731, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27837025

ABSTRACT

Myc family proteins promote cancer by inducing widespread changes in gene expression. Their rapid turnover by the ubiquitin-proteasome pathway is regulated through phosphorylation of Myc Box I and ubiquitination by the E3 ubiquitin ligase SCFFbxW7 However, N-Myc protein (the product of the MYCN oncogene) is stabilized in neuroblastoma by the protein kinase Aurora-A in a manner that is sensitive to certain Aurora-A-selective inhibitors. Here we identify a direct interaction between the catalytic domain of Aurora-A and a site flanking Myc Box I that also binds SCFFbxW7 We determined the crystal structure of the complex between Aurora-A and this region of N-Myc to 1.72-Å resolution. The structure indicates that the conformation of Aurora-A induced by compounds such as alisertib and CD532 is not compatible with the binding of N-Myc, explaining the activity of these compounds in neuroblastoma cells and providing a rational basis for the design of cancer therapeutics optimized for destabilization of the complex. We also propose a model for the stabilization mechanism in which binding to Aurora-A alters how N-Myc interacts with SCFFbxW7 to disfavor the generation of Lys48-linked polyubiquitin chains.


Subject(s)
Aurora Kinase A/chemistry , N-Myc Proto-Oncogene Protein/chemistry , Neoplasms/drug therapy , SKP Cullin F-Box Protein Ligases/chemistry , Aurora Kinase A/genetics , Azepines/pharmacology , Binding Sites , Catalytic Domain/drug effects , Crystallography, X-Ray , Humans , N-Myc Proto-Oncogene Protein/genetics , Neoplasms/genetics , Neoplasms/pathology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Polyubiquitin/chemistry , Polyubiquitin/genetics , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , SKP Cullin F-Box Protein Ligases/genetics
10.
Open Biol ; 6(7)2016 07.
Article in English | MEDLINE | ID: mdl-27411893

ABSTRACT

The vast majority of clinically approved protein kinase inhibitors target the ATP-binding pocket directly. Consequently, many inhibitors have broad selectivity profiles and most have significant off-target effects. Allosteric inhibitors are generally more selective, but are difficult to identify because allosteric binding sites are often unknown or poorly characterized. Aurora-A is activated through binding of TPX2 to an allosteric site on the kinase catalytic domain, and this knowledge could be exploited to generate an inhibitor. Here, we generated an allosteric inhibitor of Aurora-A kinase based on a synthetic, vNAR single domain scaffold, vNAR-D01. Biochemical studies and a crystal structure of the Aurora-A/vNAR-D01 complex show that the vNAR domain overlaps with the TPX2 binding site. In contrast with the binding of TPX2, which stabilizes an active conformation of the kinase, binding of the vNAR domain stabilizes an inactive conformation, in which the αC-helix is distorted, the canonical Lys-Glu salt bridge is broken and the regulatory (R-) spine is disrupted by an additional hydrophobic side chain from the activation loop. These studies illustrate how single domain antibodies can be used to characterize the regulatory mechanisms of kinases and provide a rational basis for structure-guided design of allosteric Aurora-A kinase inhibitors.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/chemistry , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Allosteric Regulation , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary , Structure-Activity Relationship
11.
Theor Chem Acc ; 135: 97, 2016.
Article in English | MEDLINE | ID: mdl-27069413

ABSTRACT

A quantum mechanical (QM) method rooted on density functional theory (DFT) has been employed to determine conformations of the methane-thiosulfonate spin label (MTSL) attached to a fragment extracted from the activation loop of Aurora-A kinase. The features of the calculated energy surface revealed low energy barriers between isoenergetic minima, and the system could be described in a population of 76 rotamers that can be also considered for other systems since it was found that the [Formula: see text], [Formula: see text] and [Formula: see text] do not depend on the previous two dihedral angles. Conformational states obtained were seen to be comparable to those obtained in the α-helix systems studied previously, indicating that the protein backbone does not affect the torsional profiles significantly and suggesting the possibility to use determined conformations for other protein systems for further modelling studies.

12.
Nat Commun ; 6: 8771, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26522158

ABSTRACT

Mitotic spindle assembly requires the regulated activities of protein kinases such as Nek7 and Nek9. Nek7 is autoinhibited by the protrusion of Tyr97 into the active site and activated by the Nek9 non-catalytic C-terminal domain (CTD). CTD binding apparently releases autoinhibition because mutation of Tyr97 to phenylalanine increases Nek7 activity independently of Nek9. Here we find that self-association of the Nek9-CTD is needed for Nek7 activation. We map the minimal Nek7 binding region of Nek9 to residues 810-828. A crystal structure of Nek7(Y97F) bound to Nek9(810-828) reveals a binding site on the C-lobe of the Nek7 kinase domain. Nek7(Y97F) crystallizes as a back-to-back dimer between kinase domain N-lobes, in which the specific contacts within the interface are coupled to the conformation of residue 97. Hence, we propose that the Nek9-CTD activates Nek7 through promoting back-to-back dimerization that releases the autoinhibitory tyrosine residue, a mechanism conserved in unrelated kinase families.


Subject(s)
Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dimerization , HeLa Cells , Humans , NIMA-Related Kinases , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics
13.
PLoS Genet ; 11(7): e1005345, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26134678

ABSTRACT

The essential mammalian gene TACC3 is frequently mutated and amplified in cancers and its fusion products exhibit oncogenic activity in glioblastomas. TACC3 functions in mitotic spindle assembly and chromosome segregation. In particular, phosphorylation on S558 by the mitotic kinase, Aurora-A, promotes spindle recruitment of TACC3 and triggers the formation of a complex with ch-TOG-clathrin that crosslinks and stabilises kinetochore microtubules. Here we map the Aurora-A-binding interface in TACC3 and show that TACC3 potently activates Aurora-A through a domain centered on F525. Vertebrate cells carrying homozygous F525A mutation in the endogenous TACC3 loci exhibit defects in TACC3 function, namely perturbed localization, reduced phosphorylation and weakened interaction with clathrin. The most striking feature of the F525A cells however is a marked shortening of mitosis, at least in part due to rapid spindle assembly. F525A cells do not exhibit chromosome missegregation, indicating that they undergo fast yet apparently faithful mitosis. By contrast, mutating the phosphorylation site S558 to alanine in TACC3 causes aneuploidy without a significant change in mitotic duration. Our work has therefore defined a regulatory role for the Aurora-A-TACC3 interaction beyond the act of phosphorylation at S558. We propose that the regulatory relationship between Aurora-A and TACC3 enables the transition from the microtubule-polymerase activity of TACC3-ch-TOG to the microtubule-crosslinking activity of TACC3-ch-TOG-clathrin complexes as mitosis progresses. Aurora-A-dependent control of TACC3 could determine the balance between these activities, thereby influencing not only spindle length and stability but also the speed of spindle formation with vital consequences for chromosome alignment and segregation.


Subject(s)
Aurora Kinase A/metabolism , Carrier Proteins/genetics , Fetal Proteins/genetics , Microtubule-Associated Proteins/genetics , Spindle Apparatus/metabolism , Transcription Factors/genetics , Xenopus Proteins/genetics , Aneuploidy , Animals , Binding Sites/genetics , Cell Line, Tumor , Chickens , Chromosome Segregation/genetics , Clathrin/metabolism , HeLa Cells , Humans , Kinetochores , Mice , Microtubules/metabolism , Mitosis/genetics , Phosphorylation/genetics , Protein Binding/genetics , Protein Structure, Tertiary , Spindle Apparatus/genetics , Xenopus laevis
14.
Biomol NMR Assign ; 9(2): 411-3, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25971232

ABSTRACT

TOG domains contribute to the organisation of microtubules through their ability to bind tubulin. They are found in members of the XMAP215 family of proteins, which act as microtubule polymerases and fulfill important roles in the formation of the mitotic spindle and in the assembly of kinetochore fibres. We recently identified a cryptic TOG domain in the XMAP215 family proteins, chTOG and its Drosophila homologue, mini spindles. This domain is not part of the well-established array of TOG domains involved in tubulin polymerisation. Instead it forms part of a binding site for TACC3 family proteins. This interaction is required for the assembly of kinetochore bridges in a trimeric complex with clathrin. Here we present the first NMR assignment of a sixth TOG domain from mini spindles as a first step to elucidate its structure and function.


Subject(s)
Drosophila Proteins/chemistry , Microtubule-Associated Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Animals , Drosophila melanogaster/metabolism , Protein Structure, Tertiary , Proton Magnetic Resonance Spectroscopy , Solutions
15.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 3): 315-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25760707

ABSTRACT

Aurora A is a Ser/Thr protein kinase that functions in cell-cycle regulation and is implicated in cancer development. During mitosis, Aurora A is activated by autophosphorylation on its activation loop at Thr288. The Aurora A catalytic domain (amino acids 122-403) expressed in Escherichia coli autophosphorylates on two activation-loop threonine residues (Thr288 and Thr287), whereas a C290A,C393A double point mutant of the Aurora A catalytic domain autophosphorylates only on Thr288. The structure of the complex of this mutant with ADP and magnesium was determined to 2.1 Šresolution using molecular replacement. This is an improvement on the existing 2.75 Šresolution structure of the equivalent wild-type complex. The structure confirms that single phosphorylation of the activation loop on Thr288 is insufficient to stabilize a `fully active' conformation of the activation loop in the absence of binding to TPX2.


Subject(s)
Aurora Kinase A/chemistry , Aurora Kinase A/genetics , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Mutation, Missense , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Secondary
16.
Biol Open ; 4(2): 170-9, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25596274

ABSTRACT

The interaction between TACC3 (transforming acidic coiled coil protein 3) and the microtubule polymerase ch-TOG (colonic, hepatic tumor overexpressed gene) is evolutionarily conserved. Loading of TACC3-ch-TOG onto mitotic spindle microtubules requires the phosphorylation of TACC3 by Aurora-A kinase and the subsequent interaction of TACC3 with clathrin to form a microtubule-binding surface. Recent work indicates that TACC3 can track the plus-ends of microtubules and modulate microtubule dynamics in non-dividing cells via its interaction with ch-TOG. Whether there is a pool of TACC3-ch-TOG that is independent of clathrin in human cells, and what is the function of this pool, are open questions. Here, we describe the molecular interaction between TACC3 and ch-TOG that permits TACC3 recruitment to the plus-ends of microtubules. This TACC3-ch-TOG pool is independent of EB1, EB3, Aurora-A phosphorylation and binding to clathrin. We also describe the distinct combinatorial subcellular pools of TACC3, ch-TOG and clathrin. TACC3 is often described as a centrosomal protein, but we show that there is no significant population of TACC3 at centrosomes. The delineation of distinct protein pools reveals a simplified view of how these proteins are organized and controlled by post-translational modification.

17.
J Cell Biol ; 202(3): 463-78, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23918938

ABSTRACT

A complex of transforming acidic coiled-coil protein 3 (TACC3), colonic and hepatic tumor overexpressed gene (ch-TOG), and clathrin has been implicated in mitotic spindle assembly and in the stabilization of kinetochore fibers by cross-linking microtubules. It is unclear how this complex binds microtubules and how the proteins in the complex interact with one another. TACC3 and clathrin have each been proposed to be the spindle recruitment factor. We have mapped the interactions within the complex and show that TACC3 and clathrin were interdependent for spindle recruitment, having to interact in order for either to be recruited to the spindle. The N-terminal domain of clathrin and the TACC domain of TACC3 in tandem made a microtubule interaction surface, coordinated by TACC3-clathrin binding. A dileucine motif and Aurora A-phosphorylated serine 558 on TACC3 bound to the "ankle" of clathrin. The other interaction within the complex involved a stutter in the TACC3 coiled-coil and a proposed novel sixth TOG domain in ch-TOG, which was required for microtubule localization of ch-TOG but not TACC3-clathrin.


Subject(s)
Carrier Proteins/metabolism , Clathrin/metabolism , Fetal Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Binding Sites , HEK293 Cells , Humans , Mice
18.
Biochemistry ; 47(18): 5168-81, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18407658

ABSTRACT

We have used multiple solution state techniques and crystallographic analysis to investigate the importance of a putative transient interaction formed between Arg-alpha237 in electron transferring flavoprotein (ETF) and Tyr-442 in trimethylamine dehydrogenase (TMADH) in complex assembly, electron transfer, and structural imprinting of ETF by TMADH. We have isolated four mutant forms of ETF altered in the identity of the residue at position 237 (alphaR237A, alphaR237K, alphaR237C, and alphaR237E) and with each form studied electron transfer from TMADH to ETF, investigated the reduction potentials of the bound ETF cofactor, and analyzed complex formation. We show that mutation of Arg-alpha237 substantially destabilizes the semiquinone couple of the bound FAD and impedes electron transfer from TMADH to ETF. Crystallographic structures of the mutant ETF proteins indicate that mutation does not perturb the overall structure of ETF, but leads to disruption of an electrostatic network at an ETF domain boundary that likely affects the dynamic properties of ETF in the crystal and in solution. We show that Arg-alpha237 is required for TMADH to structurally imprint the as-purified semiquinone form of wild-type ETF and that the ability of TMADH to facilitate this structural reorganization is lost following (i) redox cycling of ETF, or simple conversion to the oxidized form, and (ii) mutagenesis of Arg-alpha237. We discuss this result in light of recent apparent conflict in the literature relating to the structural imprinting of wild-type ETF. Our studies support a mechanism of electron transfer by conformational sampling as advanced from our previous analysis of the crystal structure of the TMADH-2ETF complex [Leys, D. , Basran, J. , Sutcliffe, M. J., and Scrutton, N. S. (2003) Nature Struct. Biol. 10, 219-225] and point to a key role for the Tyr-442 (TMADH) and Arg-alpha237 (ETF) residue pair in transiently stabilizing productive electron transfer configurations. Our work also points to the importance of Arg-alpha237 in controlling the thermodynamics of electron transfer, the dynamics of ETF, and the protection of reducing equivalents following disassembly of the TMADH-2ETF complex.


Subject(s)
Electron-Transferring Flavoproteins/chemistry , Electron-Transferring Flavoproteins/metabolism , Oxidoreductases, N-Demethylating/chemistry , Oxidoreductases, N-Demethylating/metabolism , Arginine/genetics , Arginine/metabolism , Catalysis , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Electron-Transferring Flavoproteins/genetics , Methylophilus methylotrophus/chemistry , Methylophilus methylotrophus/genetics , Methylophilus methylotrophus/metabolism , Models, Molecular , Mutation/genetics , Oxidation-Reduction , Oxidoreductases, N-Demethylating/genetics , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Titrimetry , Tyrosine/genetics , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...