Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 137(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38323935

ABSTRACT

Robust linkage between adherens junctions and the actomyosin cytoskeleton allows cells to change shape and move during morphogenesis without tearing tissues apart. The Drosophila multidomain protein Canoe and its mammalian homolog afadin are crucial for this, as in their absence many events of morphogenesis fail. To define the mechanism of action for Canoe, we are taking it apart. Canoe has five folded protein domains and a long intrinsically disordered region. The largest is the Dilute domain, which is shared by Canoe and myosin V. To define the roles of this domain in Canoe, we combined biochemical, genetic and cell biological assays. AlphaFold was used to predict its structure, providing similarities and contrasts with Myosin V. Biochemical data suggested one potential shared function - the ability to dimerize. We generated Canoe mutants with the Dilute domain deleted (CnoΔDIL). Surprisingly, they were viable and fertile. CnoΔDIL localized to adherens junctions and was enriched at junctions under tension. However, when its dose was reduced, CnoΔDIL did not provide fully wild-type function. Furthermore, canoeΔDIL mutants had defects in the orchestrated cell rearrangements of eye development. This reveals the robustness of junction-cytoskeletal connections during morphogenesis and highlights the power of natural selection to maintain protein structure.


Subject(s)
Drosophila Proteins , Myosin Type V , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Myosin Type V/metabolism , Cytoskeleton/metabolism , Intercellular Junctions/metabolism , Adherens Junctions/metabolism , Morphogenesis , Cadherins/metabolism , Mammals/metabolism
2.
Plant Cell ; 36(4): 1056-1071, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38011314

ABSTRACT

The acentrosomal cortical microtubules (MTs) of higher plants dynamically assemble into specific array patterns that determine the axis of cell expansion. Recently, the Arabidopsis (Arabidopsis thaliana) SPIRAL2 (SPR2) protein was shown to regulate cortical MT length and light-induced array reorientation by stabilizing MT minus ends. SPR2 autonomously localizes to both the MT lattice and MT minus ends, where it decreases the minus end depolymerization rate. However, the structural determinants that contribute to the ability of SPR2 to target and stabilize MT minus ends remain unknown. Here, we present the crystal structure of the SPR2 N-terminal domain, which reveals a unique tumor overexpressed gene (TOG) domain architecture with 7 HEAT repeats. We demonstrate that a coiled-coil domain mediates the multimerization of SPR2, which provides avidity for MT binding, and is essential to bind soluble tubulin. In addition, we found that an SPR2 construct spanning the TOG domain, basic region, and coiled-coil domain targets and stabilizes MT minus ends similar to full-length SPR2 in plants. These results reveal how a TOG domain, which is typically found in microtubule plus-end regulators, has been appropriated in plants to regulate MT minus ends.


Subject(s)
Arabidopsis , Neoplasms , Microtubule-Associated Proteins/metabolism , Protein Binding , Microtubules/metabolism , Tubulin/genetics , Tubulin/metabolism , Arabidopsis/metabolism , Neoplasms/metabolism
3.
PLoS One ; 18(12): e0290024, 2023.
Article in English | MEDLINE | ID: mdl-38157339

ABSTRACT

Epidermal cells of dark-grown plant seedlings reorient their cortical microtubule arrays in response to blue light from a net lateral orientation to a net longitudinal orientation with respect to the long axis of cells. The molecular mechanism underlying this microtubule array reorientation involves katanin, a microtubule severing enzyme, and a plant-specific microtubule associated protein called SPIRAL2. Katanin preferentially severs longitudinal microtubules, generating seeds that amplify the longitudinal array. Upon severing, SPIRAL2 binds nascent microtubule minus ends and limits their dynamics, thereby stabilizing the longitudinal array while the lateral array undergoes net depolymerization. To date, no experimental structural information is available for SPIRAL2 to help inform its mechanism. To gain insight into SPIRAL2 structure and function, we determined a 1.8 Å resolution crystal structure of the Arabidopsis thaliana SPIRAL2 C-terminal domain. The domain is composed of seven core α-helices, arranged in an α-solenoid. Amino-acid sequence conservation maps primarily to one face of the domain involving helices α1, α3, α5, and an extended loop, the α6-α7 loop. The domain fold is similar to, yet structurally distinct from the C-terminal domain of Ge-1 (an mRNA decapping complex factor involved in P-body localization) and, surprisingly, the C-terminal domain of the katanin p80 regulatory subunit. The katanin p80 C-terminal domain heterodimerizes with the MIT domain of the katanin p60 catalytic subunit, and in metazoans, binds the microtubule minus-end factors CAMSAP3 and ASPM. Structural analysis predicts that SPIRAL2 does not engage katanin p60 in a mode homologous to katanin p80. The SPIRAL2 structure highlights an interesting evolutionary convergence of domain architecture and microtubule minus-end localization between SPIRAL2 and katanin complexes, and establishes a foundation upon which structure-function analysis can be conducted to elucidate the role of this domain in the regulation of plant microtubule arrays.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Katanin/chemistry , Microtubules/metabolism , Plants/metabolism
4.
bioRxiv ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37905001

ABSTRACT

Robust linkage between cell-cell adherens junctions and the actomyosin cytoskeleton allows cells to change shape and move during morphogenesis without tearing tissues apart. The multidomain protein Drosophila Canoe and its mammalian homolog Afadin are critical for this linkage, and in their absence many events of morphogenesis fail. To define underlying mechanisms, we are taking Canoe apart, using Drosophila as our model. Canoe and Afadin share five folded protein domains, followed by a large intrinsically disordered region. The largest of these folded domains is the Dilute domain, which is found in Canoe/Afadin, their paralogs, and members of the MyosinV family. To define the roles of Canoe's Dilute domain we have combined biochemical, genetic and cell biological assays. Use of the AlphaFold tools revealed the predicted structure of the Canoe/Afadin Dilute domain, providing similarities and contrasts with that of MyosinV. Our biochemical data suggest one potential shared function: the ability to dimerize. We next generated Drosophila mutants with the Dilute domain cleanly deleted. Surprisingly, these mutants are viable and fertile, and CanoeΔDIL protein localizes to adherens junctions and is enriched at junctions under tension. However, when we reduce the dose of CanoeΔDIL protein in a sensitized assay, it becomes clear it does not provide full wildtype function. Further, canoeΔDIL mutants have defects in pupal eye development, another process that requires orchestrated cell rearrangements. Together, these data reveal the robustness in AJ-cytoskeletal connections during multiple embryonic and postembryonic events, and the power of natural selection to maintain protein structure even in robust systems.

5.
Mol Biol Cell ; 34(8): ar80, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37163316

ABSTRACT

Polo-like kinase 4 (Plk4) is the master-regulator of centriole assembly, and cell cycle-dependent regulation of its activity maintains proper centrosome number. During most of the cell cycle, Plk4 levels are nearly undetectable due to its ability to autophosphorylate and trigger its own ubiquitin-mediated degradation. However, during mitotic exit, Plk4 forms a single aggregate on the centriole surface to stimulate centriole duplication. Whereas most Polo-like kinase family members are monomeric, Plk4 is unique because it forms homodimers. Notably, Plk4 trans-autophosphorylates a degron near its kinase domain, a critical step in autodestruction. While it is thought that the purpose of homodimerization is to promote trans-autophosphorylation, this has not been tested. Here, we generated separation-of-function Plk4 mutants that fail to dimerize and show that homodimerization creates a binding site for the Plk4 activator, Asterless. Surprisingly, however, Plk4 dimer mutants are catalytically active in cells, promote centriole assembly, and can trans-autophosphorylate through concentration-dependent condensate formation. Moreover, we mapped and then deleted the weak-interacting regions within Plk4 that mediate condensation and conclude that dimerization and condensation are not required for centriole assembly. Our findings suggest that Plk4 dimerization and condensation function simply to down-regulate Plk4 and suppress centriole overduplication.


Subject(s)
Cell Cycle Proteins , Centrioles , Centrioles/metabolism , Dimerization , Cell Line , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Phosphorylation
6.
J Cell Biol ; 220(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34762121

ABSTRACT

Embryogenesis requires cells to change shape and move without disrupting epithelial integrity. This requires robust, responsive linkage between adherens junctions and the actomyosin cytoskeleton. Using Drosophila morphogenesis, we define molecular mechanisms mediating junction-cytoskeletal linkage and explore the role of mechanosensing. We focus on the junction-cytoskeletal linker Canoe, a multidomain protein. We engineered the canoe locus to define how its domains mediate its mechanism of action. To our surprise, the PDZ and FAB domains, which we thought connected junctions and F-actin, are not required for viability or mechanosensitive recruitment to junctions under tension. The FAB domain stabilizes junctions experiencing elevated force, but in its absence, most cells recover, suggesting redundant interactions. In contrast, the Rap1-binding RA domains are critical for all Cno functions and enrichment at junctions under tension. This supports a model in which junctional robustness derives from a large protein network assembled via multivalent interactions, with proteins at network nodes and some node connections more critical than others.


Subject(s)
Adherens Junctions/metabolism , Cytoskeleton/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Morphogenesis , Alleles , Animals , Cell Survival , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Epithelium/metabolism , Loss of Function Mutation/genetics , Protein Domains
7.
J Med Genet ; 58(8): 526-533, 2021 08.
Article in English | MEDLINE | ID: mdl-32747439

ABSTRACT

BACKGROUND: Dysfunction in non-motile cilia is associated with a broad spectrum of developmental disorders characterised by clinical heterogeneity. While over 100 genes have been associated with primary ciliopathies, with wide phenotypic overlap, some patients still lack a molecular diagnosis. OBJECTIVE: To investigate and functionally characterise the molecular cause of a malformation disorder observed in two sibling fetuses characterised by microphthalmia, cleft lip and palate, and brain anomalies. METHODS: A trio-based whole exome sequencing (WES) strategy was used to identify candidate variants in the TOGARAM1 gene. In silico, in vitro and in vivo (Caenorhabditis elegans) studies were carried out to explore the impact of mutations on protein structure and function, and relevant biological processes. RESULTS: TOGARAM1 encodes a member of the Crescerin1 family of proteins regulating microtubule dynamics. Its orthologue in C. elegans, che-12, is expressed in a subset of sensory neurons and localises in the dendritic cilium where it is required for chemosensation. Nematode lines harbouring the corresponding missense variant in TOGARAM1 were generated by CRISPR/Cas9 technology. Although chemotaxis ability on a NaCl gradient was not affected, che-12 point mutants displayed impaired lipophilic dye uptake, with shorter and altered cilia in sensory neurons. Finally, in vitro analysis of microtubule polymerisation in the presence of wild-type or mutant TOG2 domain revealed a faster polymerisation associated with the mutant protein, suggesting aberrant tubulin binding. CONCLUSIONS: Our data are in favour of a causative role of TOGARAM1 variants in the pathogenesis of this novel disorder, connecting this gene with primary ciliopathy.


Subject(s)
Cilia/pathology , Ciliopathies/genetics , Mutation/genetics , Nervous System Malformations/genetics , Animals , Caenorhabditis elegans/genetics , Cleft Lip/pathology , Cleft Palate/pathology , Female , Humans , Male , Nervous System Malformations/pathology
8.
Curr Biol ; 30(14): 2874, 2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32693064
9.
Curr Biol ; 30(11): R646-R649, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32516614

ABSTRACT

While the dynamics of microtubule ends are well characterized, the mechanism that repairs breaks in the lattice interior is poorly understood. A new in vitro study finds that the microtubule-associated protein CLASP repairs lattice damage by regulating GTP-tubulin incorporation into the break site.


Subject(s)
Orthopedics , Tubulin , Microtubule-Associated Proteins , Microtubules
10.
PLoS One ; 14(7): e0219823, 2019.
Article in English | MEDLINE | ID: mdl-31323070

ABSTRACT

Tubulin-binding TOG domains are found arrayed in a number of proteins that regulate microtubule dynamics. While much is known about the structure and function of TOG domains from the XMAP215 microtubule polymerase family, less in known about the TOG domain array found in animal CLASP family members. The animal CLASP TOG array promotes microtubule pause, potentiates rescue, and limits catastrophe. How structurally distinct the TOG domains of animal CLASP are from one another, from XMAP215 family TOG domains, and whether a specific order of structurally distinct TOG domains in the TOG array is conserved across animal CLASP family members is poorly understood. We present the x-ray crystal structures of Homo sapiens (H.s.) CLASP1 TOG1 and TOG2. The structures of H.s. CLASP1 TOG1 and TOG2 are distinct from each other and from the previously determined structure of Mus musculus (M.m.) CLASP2 TOG3. Comparative analyses of CLASP family TOG domain structures determined to date across species and paralogs supports a conserved CLASP TOG array paradigm in which structurally distinct TOG domains are arrayed in a specific order. H.s. CLASP1 TOG1 bears structural similarity to the free-tubulin binding TOG domains of the XMAP215 family but lacks many of the key tubulin-binding determinants found in XMAP215 family TOG domains. This aligns with studies that report that animal CLASP family TOG1 domains cannot bind free tubulin or microtubules. In contrast, animal CLASP family TOG2 and TOG3 domains have reported microtubule-binding activity but are structurally distinct from the free-tubulin binding TOG domains of the XMAP215 family. H.s. CLASP1 TOG2 has a convex architecture, predicted to engage a hyper-curved tubulin state that may underlie its ability to limit microtubule catastrophe and promote rescue. M.m. CLASP2 TOG3 has unique structural elements in the C-terminal half of its α-solenoid domain that our modeling studies implicate in binding to laterally-associated tubulin subunits in the microtubule lattice in a mode similar to, yet distinct from those predicted for the XMAP215 family TOG4 domain. The potential ability of the animal CLASP family TOG3 domain to engage lateral tubulin subunits may underlie the microtubule rescue activity ascribed to the domain. These findings highlight the structural diversity of TOG domains within the CLASP family TOG array and provide a molecular foundation for understanding CLASP-dependent effects on microtubule dynamics.


Subject(s)
Microtubule-Associated Proteins/chemistry , Models, Molecular , Protein Conformation , Amino Acid Sequence , Animals , Conserved Sequence , Humans , Microtubule-Associated Proteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins , Structure-Activity Relationship
12.
J Biol Chem ; 294(3): 918-931, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30455356

ABSTRACT

Cytoplasmic linker protein 170 (CLIP-170) is a microtubule plus-end factor that links vesicles to microtubules and recruits the dynein-dynactin complex to microtubule plus ends. CLIP-170 plus-end localization is end binding 1 (EB1)-dependent. CLIP-170 contains two N-terminal cytoskeleton-associated protein glycine-rich (CAP-Gly) domains flanked by serine-rich regions. The CAP-Gly domains are known EB1-binding domains, and the serine-rich regions have also been implicated in CLIP-170's microtubule plus-end localization mechanism. However, the determinants in these serine-rich regions have not been identified. Here we elucidated multiple EB1-binding modules in the CLIP-170 N-terminal region. Using isothermal titration calorimetry and size-exclusion chromatography, we mapped and biophysically characterized these EB1-binding modules, including the two CAP-Gly domains, a bridging SXIP motif, and a unique array of divergent SXIP-like motifs located N-terminally to the first CAP-Gly domain. We found that, unlike the EB1-binding mode of the CAP-Gly domain in the dynactin-associated protein p150Glued, which dually engages the EB1 C-terminal EEY motif as well as the EB homology domain and sterically occludes SXIP motif binding, the CLIP-170 CAP-Gly domains engage only the EEY motif, enabling the flanking SXIP and SXIP-like motifs to bind the EB homology domain. These multivalent EB1-binding modules provided avidity to the CLIP-170-EB1 interaction, likely clarifying why CLIP-170 preferentially binds EB1 rather than the α-tubulin C-terminal EEY motif. Our finding that CLIP-170 has multiple non-CAP-Gly EB1-binding modules may explain why autoinhibition of CLIP-170 GAP-Gly domains does not fully abrogate its microtubule plus-end localization. This work expands our understanding of EB1-binding motifs and their multivalent networks.


Subject(s)
Microtubule-Associated Proteins/chemistry , Multiprotein Complexes/chemistry , Neoplasm Proteins/chemistry , Tubulin/chemistry , Amino Acid Motifs , Humans , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Protein Domains , Tubulin/metabolism
13.
Mol Biol Cell ; 29(23): 2874-2886, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30256714

ABSTRACT

Centriole assembly initiates when Polo-like kinase 4 (Plk4) interacts with a centriole "targeting-factor." In Drosophila, Asterless/Asl (Cep152 in humans) fulfills the targeting role. Interestingly, Asl also regulates Plk4 levels. The N-terminus of Asl (Asl-A; amino acids 1-374) binds Plk4 and promotes Plk4 self-destruction, although it is unclear how this is achieved. Moreover, Plk4 phosphorylates the Cep152 N-terminus, but the functional consequence is unknown. Here, we show that Plk4 phosphorylates Asl and mapped 13 phospho-residues in Asl-A. Nonphosphorylatable alanine (13A) and phosphomimetic (13PM) mutants did not alter Asl function, presumably because of the dominant role of the Asl C-terminus in Plk4 stabilization and centriolar targeting. To address how Asl-A phosphorylation specifically affects Plk4 regulation, we generated Asl-A fragment phospho-mutants and expressed them in cultured Drosophila cells. Asl-A-13A stimulated kinase activity by relieving Plk4 autoinhibition. In contrast, Asl-A-13PM inhibited Plk4 activity by a novel mechanism involving autophosphorylation of Plk4's kinase domain. Thus, Asl-A's phosphorylation state determines which of Asl-A's two opposing effects are exerted on Plk4. Initially, nonphosphorylated Asl binds Plk4 and stimulates its kinase activity, but after Asl is phosphorylated, a negative-feedback mechanism suppresses Plk4 activity. This dual regulatory effect by Asl-A may limit Plk4 to bursts of activity that modulate centriole duplication.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Line , Centrioles/metabolism , Drosophila , Phosphorylation , Protein Binding
14.
Dev Cell ; 46(1): 5-7, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29974863

ABSTRACT

Like permutating motifs in music, similar protein folds are employed across biology for distinct functions. In this issue of Developmental Cell, Aher et al. (2018) provide insight into how variable TOG domains within an array in the microtubule regulator CLASP are used to prevent microtubule catastrophe and potentiate rescue.


Subject(s)
Microtubule-Associated Proteins , Tubulin , Amino Acid Sequence , Microtubules , Models, Molecular
15.
J Cell Biol ; 217(4): 1217-1231, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29496738

ABSTRACT

Polo-like kinase 4 (Plk4) initiates an early step in centriole assembly by phosphorylating Ana2/STIL, a structural component of the procentriole. Here, we show that Plk4 binding to the central coiled-coil (CC) of Ana2 is a conserved event involving Polo-box 3 and a previously unidentified putative CC located adjacent to the kinase domain. Ana2 is then phosphorylated along its length. Previous studies showed that Plk4 phosphorylates the C-terminal STil/ANa2 (STAN) domain of Ana2/STIL, triggering binding and recruitment of the cartwheel protein Sas6 to the procentriole assembly site. However, the physiological relevance of N-terminal phosphorylation was unknown. We found that Plk4 first phosphorylates the extreme N terminus of Ana2, which is critical for subsequent STAN domain modification. Phosphorylation of the central region then breaks the Plk4-Ana2 interaction. This phosphorylation pattern is important for centriole assembly and integrity because replacement of endogenous Ana2 with phospho-Ana2 mutants disrupts distinct steps in Ana2 function and inhibits centriole duplication.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , Centrioles/enzymology , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line , Centrioles/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein Transport , Signal Transduction
16.
Mol Biol Cell ; 29(3): 285-294, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29187574

ABSTRACT

XMAP215/Dis1 family proteins are potent microtubule polymerases, critical for mitotic spindle structure and dynamics. While microtubule polymerase activity is driven by an N-terminal tumor overexpressed gene (TOG) domain array, proper cellular localization is a requisite for full activity and is mediated by a C-terminal domain. Structural insight into the C-terminal domain's architecture and localization mechanism remain outstanding. We present the crystal structure of the Saccharomyces cerevisiae Stu2 C-terminal domain, revealing a 15-nm parallel homodimeric coiled coil. The parallel architecture of the coiled coil has mechanistic implications for the arrangement of the homodimer's N-terminal TOG domains during microtubule polymerization. The coiled coil has two spatially distinct conserved regions: CRI and CRII. Mutations in CRI and CRII perturb the distribution and localization of Stu2 along the mitotic spindle and yield defects in spindle morphology including increased frequencies of mispositioned and fragmented spindles. Collectively, these data highlight roles for the Stu2 dimerization domain as a scaffold for factor binding that optimally positions Stu2 on the mitotic spindle to promote proper spindle structure and dynamics.


Subject(s)
Kinetochores/physiology , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Kinetochores/metabolism , Microtubules/metabolism , Protein Binding , Protein Domains/physiology , Protein Structural Elements/physiology , Saccharomyces cerevisiae/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/physiology , Tubulin/metabolism
17.
J Cell Biol ; 217(2): 779-793, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29259096

ABSTRACT

We developed a novel optogenetic tool, SxIP-improved light-inducible dimer (iLID), to facilitate the reversible recruitment of factors to microtubule (MT) plus ends in an end-binding protein-dependent manner using blue light. We show that SxIP-iLID can track MT plus ends and recruit tgRFP-SspB upon blue light activation. We used this system to investigate the effects of cross-linking MT plus ends and F-actin in Drosophila melanogaster S2 cells to gain insight into spectraplakin function and mechanism. We show that SxIP-iLID can be used to temporally recruit an F-actin binding domain to MT plus ends and cross-link the MT and F-actin networks. Cross-linking decreases MT growth velocities and generates a peripheral MT exclusion zone. SxIP-iLID facilitates the general recruitment of specific factors to MT plus ends with temporal control enabling researchers to systematically regulate MT plus end dynamics and probe MT plus end function in many biological processes.


Subject(s)
Actins/metabolism , Cross-Linking Reagents/metabolism , Microtubules/metabolism , Optogenetics , Animals , Cells, Cultured , Drosophila melanogaster
18.
Structure ; 25(7): 1130-1138.e6, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28602822

ABSTRACT

Spectraplakins are large molecules that cross-link F-actin and microtubules (MTs). Mutations in spectraplakins yield defective cell polarization, aberrant focal adhesion dynamics, and dystonia. We present the 2.8 Å crystal structure of the hACF7 EF1-EF2-GAR MT-binding module and delineate the GAR residues critical for MT binding. The EF1-EF2 and GAR domains are autonomous domains connected by a flexible linker. The EF1-EF2 domain is an EFß-scaffold with two bound Ca2+ ions that straddle an N-terminal α helix. The GAR domain has a unique α/ß sandwich fold that coordinates Zn2+. While the EF1-EF2 domain is not sufficient for MT binding, the GAR domain is and likely enhances EF1-EF2-MT engagement. Residues in a conserved basic patch, distal to the GAR domain's Zn2+-binding site, mediate MT binding.


Subject(s)
Microfilament Proteins/chemistry , Microtubules/metabolism , Binding Sites , EF Hand Motifs , HEK293 Cells , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Protein Binding , Zinc/metabolism
19.
J Cell Biol ; 216(6): 1641-1657, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28512144

ABSTRACT

XMAP215, CLASP, and Crescerin use arrayed tubulin-binding tumor overexpressed gene (TOG) domains to modulate microtubule dynamics. We hypothesized that TOGs have distinct architectures and tubulin-binding properties that underlie each family's ability to promote microtubule polymerization or pause. As a model, we investigated the pentameric TOG array of a Drosophila melanogaster XMAP215 member, Msps. We found that Msps TOGs have distinct architectures that bind either free or polymerized tubulin, and that a polarized array drives microtubule polymerization. An engineered TOG1-2-5 array fully supported Msps-dependent microtubule polymerase activity. Requisite for this activity was a TOG5-specific N-terminal HEAT repeat that engaged microtubule lattice-incorporated tubulin. TOG5-microtubule binding maintained mitotic spindle formation as deleting or mutating TOG5 compromised spindle architecture and increased the mitotic index. Mad2 knockdown released the spindle assembly checkpoint triggered when TOG5-microtubule binding was compromised, indicating that TOG5 is essential for spindle function. Our results reveal a TOG5-specific role in mitotic fidelity and support our hypothesis that architecturally distinct TOGs arranged in a sequence-specific order underlie TOG array microtubule regulator activity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis , Spindle Apparatus/metabolism , Tubulin/metabolism , Animals , Animals, Genetically Modified , Cell Line , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Mitotic Index , Models, Molecular , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Time Factors , Transfection
20.
J Cell Biol ; 215(3): 305-307, 2016 Nov 07.
Article in English | MEDLINE | ID: mdl-27821492

ABSTRACT

In vitro reconstitution is the fundamental test for identification of the core components of a biological process. In this issue, Moriwaki and Goshima (2016. J. Cell Biol. https://doi.org/10.1083/jcb.201604118) reconstitute all phases of microtubule dynamics through the inclusion of five key regulators and demonstrate that Polo kinase activity shifts the system from an interphase mode into an enhanced mitotic mode.


Subject(s)
Microtubules/metabolism , Animals , Humans , Microtubule-Associated Proteins , Models, Biological , Spindle Apparatus/metabolism , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...