200 results on '"Ohi R"'
Search Results
2. Systematic Identification of Microtubule Posttranslational Modification "Readers" by Quantitative Proteomics.
- Author
-
Hotta T and Ohi R
- Subjects
- Humans, Microtubule-Associated Proteins metabolism, Proteome metabolism, Animals, Mass Spectrometry methods, Tandem Mass Spectrometry methods, Protein Processing, Post-Translational, Microtubules metabolism, Proteomics methods, Tubulin metabolism
- Abstract
Microtubules, dynamic polymers assembled from α, β-tubulin dimers, contribute to myriad cellular processes. This is largely attributed to microtubule-associated proteins (MAPs). How MAPs selectively bind microtubules to carry out various functions is not known. The "Tubulin Code" theory proposes that posttranslational modifications (PTMs) of microtubules serve as signs that can be read by specific MAPs, thereby conferring specific functional properties to the microtubules. In support of this hypothesis, "reader" MAPs have been identified for various tubulin PTMs, but, until recently, no systematic screening had been performed to identify readers in an unbiased manner. We addressed this by developing a reader identification pipeline that uses quantitative mass spectrometry to interrogate the microtubule proteome of cells programmed to express specific PTMs. This pipeline can be used to identify readers for any tubulin PTM from various cell types as long as the writer enzymes are known. We also provide an alternative, complementary approach to obtain modified microtubules using a generic writer enzyme in vitro., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
3. Antiparallel microtubule bundling supports KIF15-driven mitotic spindle assembly.
- Author
-
Salazar BM and Ohi R
- Subjects
- Humans, Cell Cycle Proteins metabolism, Microtubule-Associated Proteins metabolism, Mitosis physiology, HeLa Cells, Chromosome Segregation, Kinesins metabolism, Spindle Apparatus metabolism, Microtubules metabolism
- Abstract
The spindle is a bipolar microtubule-based machine that is crucial for accurate chromosome segregation. Spindle bipolarity is generated by Eg5 (a kinesin-5), a conserved motor that drives spindle assembly by localizing to and sliding apart antiparallel microtubules. In the presence of Eg5 inhibitors (K5Is), KIF15 (a kinesin-12) can promote spindle assembly, resulting in K5I-resistant cells (KIRCs). However, KIF15 is a less potent motor than Eg5, suggesting that other factors may contribute to spindle formation in KIRCs. Protein Regulator of Cytokinesis 1 (PRC1) preferentially bundles antiparallel microtubules, and we previously showed that PRC1 promotes KIF15-microtubule binding, leading us to hypothesize that PRC1 may enhance KIF15 activity in KIRCs. Here, we demonstrate that: 1) loss of PRC1 in KIRCs decreases spindle bipolarity, 2) overexpression of PRC1 increases spindle formation efficiency in KIRCs, 3) overexpression of PRC1 protects K5I naïve cells against the K5I S-trityl-L-cysteine (STLC), and 4) PRC1 overexpression promotes the establishment of K5I resistance. These effects are not fully reproduced by a TPX2, a microtubule bundler with no known preference for microtubule orientation. These results suggest a model wherein PRC1-mediated bundling of microtubules creates a more favorable microtubule architecture for KIF15-driven mitotic spindle assembly in the context of Eg5 inhibition.
- Published
- 2024
- Full Text
- View/download PDF
4. Microtubule detyrosination by VASH1/SVBP is regulated by the conformational state of tubulin in the lattice.
- Author
-
Yue Y, Hotta T, Higaki T, Verhey KJ, and Ohi R
- Subjects
- Paclitaxel, Tyrosine metabolism, Guanosine Triphosphate metabolism, Tubulin metabolism, Microtubules metabolism
- Abstract
Tubulin, a heterodimer of α- and β-tubulin, is a GTPase that assembles into microtubule (MT) polymers whose dynamic properties are intimately coupled to nucleotide hydrolysis. In cells, the organization and dynamics of MTs are further tuned by post-translational modifications (PTMs), which control the ability of MT-associated proteins (MAPs) and molecular motors to engage MTs. Detyrosination is a PTM of α-tubulin, wherein its C-terminal tyrosine residue is enzymatically removed by either the vasohibin (VASH) or MT-associated tyrosine carboxypeptidase (MATCAP) peptidases. How these enzymes generate specific patterns of MT detyrosination in cells is not known. Here, we use a novel antibody-based probe to visualize the formation of detyrosinated MTs in real time and employ single-molecule imaging of VASH1 bound to its regulatory partner small-vasohibin binding protein (SVBP) to understand the process of MT detyrosination in vitro and in cells. We demonstrate that the activity, but not binding, of VASH1/SVBP is much greater on mimics of guanosine triphosphate (GTP)-MTs than on guanosine diphosphate (GDP)-MTs. Given emerging data showing that tubulin subunits in GTP-MTs are in expanded conformation relative to tubulin subunits in GDP-MTs, we reasoned that the lattice conformation of MTs is a key factor that gates the activity of VASH1/SVBP. We show that Taxol, a drug known to expand the MT lattice, promotes MT detyrosination and that CAMSAP2 and CAMSAP3 are two MAPs that spatially regulate detyrosination in cells. Collectively, our work shows that VASH1/SVBP detyrosination is regulated by the conformational state of tubulin in the MT lattice and that this is spatially determined in cells by the activity of MAPs., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. SETD2 safeguards the genome against isochromosome formation.
- Author
-
Mason FM, Kounlavong ES, Tebeje AT, Dahiya R, Guess T, Khan A, Vlach L, Norris SR, Lovejoy CA, Dere R, Strahl BD, Ohi R, Ly P, Walker CL, and Rathmell WK
- Subjects
- Humans, Centromere, Chromosome Aberrations, Cytogenetics, DNA Replication, Genomic Instability, Isochromosomes
- Abstract
Isochromosomes are mirror-imaged chromosomes with simultaneous duplication and deletion of genetic material which may contain two centromeres to create isodicentric chromosomes. Although isochromosomes commonly occur in cancer and developmental disorders and promote genome instability, mechanisms that prevent isochromosomes are not well understood. We show here that the tumor suppressor and methyltransferase SETD2 is essential to prevent these errors. Using cellular and cytogenetic approaches, we demonstrate that loss of SETD2 or its epigenetic mark, histone H3 lysine 36 trimethylation (H3K36me3), results in the formation of isochromosomes as well as isodicentric and acentric chromosomes. These defects arise during DNA replication and are likely due to faulty homologous recombination by RAD52. These data provide a mechanism for isochromosome generation and demonstrate that SETD2 and H3K36me3 are essential to prevent the formation of this common mutable chromatin structure known to initiate a cascade of genomic instability in cancer.
- Published
- 2023
- Full Text
- View/download PDF
6. β-Catenin-Driven Differentiation Is a Tissue-Specific Epigenetic Vulnerability in Adrenal Cancer.
- Author
-
Mohan DR, Borges KS, Finco I, LaPensee CR, Rege J, Solon AL, Little DW, Else T, Almeida MQ, Dang D, Haggerty-Skeans J, Apfelbaum AA, Vinco M, Wakamatsu A, Mariani BMP, Amorim LC, Latronico AC, Mendonca BB, Zerbini MCN, Lawlor ER, Ohi R, Auchus RJ, Rainey WE, Marie SKN, Giordano TJ, Venneti S, Fragoso MCBV, Breault DT, Lerario AM, and Hammer GD
- Subjects
- Humans, beta Catenin genetics, beta Catenin metabolism, Epigenesis, Genetic, Chromatin genetics, Adrenocortical Carcinoma genetics, Adrenocortical Carcinoma metabolism, Adrenocortical Carcinoma pathology, Adrenal Cortex Neoplasms genetics, Adrenal Cortex Neoplasms pathology
- Abstract
Adrenocortical carcinoma (ACC) is a rare cancer in which tissue-specific differentiation is paradoxically associated with dismal outcomes. The differentiated ACC subtype CIMP-high is prevalent, incurable, and routinely fatal. CIMP-high ACC possess abnormal DNA methylation and frequent β-catenin-activating mutations. Here, we demonstrated that ACC differentiation is maintained by a balance between nuclear, tissue-specific β-catenin-containing complexes, and the epigenome. On chromatin, β-catenin bound master adrenal transcription factor SF1 and hijacked the adrenocortical super-enhancer landscape to maintain differentiation in CIMP-high ACC; off chromatin, β-catenin bound histone methyltransferase EZH2. SF1/β-catenin and EZH2/β-catenin complexes present in normal adrenals persisted through all phases of ACC evolution. Pharmacologic EZH2 inhibition in CIMP-high ACC expelled SF1/β-catenin from chromatin and favored EZH2/β-catenin assembly, erasing differentiation and restraining cancer growth in vitro and in vivo. These studies illustrate how tissue-specific programs shape oncogene selection, surreptitiously encoding targetable therapeutic vulnerabilities., Significance: Oncogenic β-catenin can use tissue-specific partners to regulate cellular differentiation programs that can be reversed by epigenetic therapies, identifying epigenetic control of differentiation as a viable target for β-catenin-driven cancers., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
7. Causes, costs and consequences of kinesin motors communicating through the microtubule lattice.
- Author
-
Verhey KJ and Ohi R
- Subjects
- Microtubules, Microtubule-Associated Proteins, Cytoskeleton, Kinesins, Tubulin
- Abstract
Microtubules are critical for a variety of important functions in eukaryotic cells. During intracellular trafficking, molecular motor proteins of the kinesin superfamily drive the transport of cellular cargoes by stepping processively along the microtubule surface. Traditionally, the microtubule has been viewed as simply a track for kinesin motility. New work is challenging this classic view by showing that kinesin-1 and kinesin-4 proteins can induce conformational changes in tubulin subunits while they are stepping. These conformational changes appear to propagate along the microtubule such that the kinesins can work allosterically through the lattice to influence other proteins on the same track. Thus, the microtubule is a plastic medium through which motors and other microtubule-associated proteins (MAPs) can communicate. Furthermore, stepping kinesin-1 can damage the microtubule lattice. Damage can be repaired by the incorporation of new tubulin subunits, but too much damage leads to microtubule breakage and disassembly. Thus, the addition and loss of tubulin subunits are not restricted to the ends of the microtubule filament but rather, the lattice itself undergoes continuous repair and remodeling. This work leads to a new understanding of how kinesin motors and their microtubule tracks engage in allosteric interactions that are critical for normal cell physiology., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
8. Mechanistic Analysis of CCP1 in Generating ΔC2 α-Tubulin in Mammalian Cells and Photoreceptor Neurons.
- Author
-
Hotta T, Plemmons A, Gebbie M, Ziehm TA, Blasius TL, Johnson C, Verhey KJ, Pearring JN, and Ohi R
- Subjects
- Humans, Mice, Animals, HeLa Cells, Peptide Hydrolases metabolism, Tyrosine metabolism, Microtubules metabolism, Protein Processing, Post-Translational, Mammals metabolism, Tubulin metabolism, Neurons metabolism
- Abstract
An important post-translational modification (PTM) of α-tubulin is the removal of amino acids from its C-terminus. Removal of the C-terminal tyrosine residue yields detyrosinated α-tubulin, and subsequent removal of the penultimate glutamate residue produces ΔC2-α-tubulin. These PTMs alter the ability of the α-tubulin C-terminal tail to interact with effector proteins and are thereby thought to change microtubule dynamics, stability, and organization. The peptidase(s) that produces ΔC2-α-tubulin in a physiological context remains unclear. Here, we take advantage of the observation that ΔC2-α-tubulin accumulates to high levels in cells lacking tubulin tyrosine ligase (TTL) to screen for cytosolic carboxypeptidases (CCPs) that generate ΔC2-α-tubulin. We identify CCP1 as the sole peptidase that produces ΔC2-α-tubulin in TTLΔ HeLa cells. Interestingly, we find that the levels of ΔC2-α-tubulin are only modestly reduced in photoreceptors of ccp1
-/- mice, indicating that other peptidases act synergistically with CCP1 to produce ΔC2-α-tubulin in post-mitotic cells. Moreover, the production of ΔC2-α-tubulin appears to be under tight spatial control in the photoreceptor cilium: ΔC2-α-tubulin persists in the connecting cilium of ccp1-/- but is depleted in the distal portion of the photoreceptor. This work establishes the groundwork to pinpoint the function of ΔC2-α-tubulin in proliferating and post-mitotic mammalian cells.- Published
- 2023
- Full Text
- View/download PDF
9. Synergy between inhibitors of two mitotic spindle assembly motors undermines an adaptive response.
- Author
-
Solon AL, Zaniewski TM, O'Brien P, Clasby M, Hancock WO, and Ohi R
- Subjects
- Microtubules metabolism, Mitosis, Cell Cycle, Kinesins, Spindle Apparatus metabolism
- Abstract
Mitosis is the cellular process that ensures accurate segregation of the cell's genetic material into two daughter cells. Mitosis is often deregulated in cancer; thus drugs that target mitosis-specific proteins represent attractive targets for anticancer therapy. Numerous inhibitors have been developed against kinesin-5 Eg5, a kinesin essential for bipolar spindle assembly. Unfortunately, Eg5 inhibitors (K5Is) have been largely ineffective in the clinic, possibly due to the activity of a second kinesin, KIF15, that can suppress the cytotoxic effect of K5Is by driving spindle assembly through an Eg5-independent pathway. We hypothesized that pairing of K5Is with small molecule inhibitors of KIF15 will be more cytotoxic than either inhibitor alone. Here we present the results of a high-throughput screen from which we identified two inhibitors that inhibit the motor activity of KIF15 both in vitro and in cells. These inhibitors selectively inhibit KIF15 over other molecular motors and differentially affect the ability of KIF15 to bind microtubules. Finally, we find that chemical inhibition of KIF15 reduces the ability of cells to acquire resistance to K5Is, highlighting the centrality of KIF15 to K5I resistance and the value of these inhibitors as tools with which to study KIF15 in a physiological context.
- Published
- 2022
- Full Text
- View/download PDF
10. EML2-S constitutes a new class of proteins that recognizes and regulates the dynamics of tyrosinated microtubules.
- Author
-
Hotta T, McAlear TS, Yue Y, Higaki T, Haynes SE, Nesvizhskii AI, Sept D, Verhey KJ, Bechstedt S, and Ohi R
- Subjects
- Humans, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Protein Processing, Post-Translational, Tyrosine metabolism, Microtubules metabolism, Tubulin metabolism
- Abstract
Tubulin post-translational modifications (PTMs) alter microtubule properties by affecting the binding of microtubule-associated proteins (MAPs). Microtubule detyrosination, which occurs by proteolytic removal of the C-terminal tyrosine from ɑ-tubulin, generates the oldest known tubulin PTM, but we lack comprehensive knowledge of MAPs that are regulated by this PTM. We developed a screening pipeline to identify proteins that discriminate between Y- and ΔY-microtubules and found that echinoderm microtubule-associated protein-like 2 (EML2) preferentially interacts with Y-microtubules. This activity depends on a Y-microtubule interaction motif built from WD40 repeats. We show that EML2 tracks the tips of shortening microtubules, a behavior not previously seen among human MAPs in vivo, and influences dynamics to increase microtubule stability. Our screening pipeline is readily adapted to identify proteins that specifically recognize a wide range of microtubule PTMs., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
11. Tubulin Carboxypeptidase Activity Promotes Focal Gelatin Degradation in Breast Tumor Cells and Induces Apoptosis in Breast Epithelial Cells That Is Overcome by Oncogenic Signaling.
- Author
-
Mathias TJ, Ju JA, Lee RM, Thompson KN, Mull ML, Annis DA, Chang KT, Ory EC, Stemberger MB, Hotta T, Ohi R, Vitolo MI, Moutin MJ, and Martin SS
- Abstract
Post-translational modifications (PTMs) of the microtubule network impart differential functions across normal cell types and their cancerous counterparts. The removal of the C-terminal tyrosine of α-tubulin (deTyr-Tub) as performed by the tubulin carboxypeptidase (TCP) is of particular interest in breast epithelial and breast cancer cells. The recent discovery of the genetic identity of the TCP to be a vasohibin ( VASH1/2 ) coupled with a small vasohibin-binding protein ( SVBP ) allows for the functional effect of this tubulin PTM to be directly tested for the first time. Our studies revealed the immortalized breast epithelial cell line MCF10A undergoes apoptosis following transfection with TCP constructs, but the addition of oncogenic KRas or Bcl-2/Bcl-xL overexpression prevents subsequent apoptotic induction in the MCF10A background. Functionally, an increase in deTyr-Tub via TCP transfection in MDA-MB-231 and Hs578t breast cancer cells leads to enhanced focal gelatin degradation. Given the elevated deTyr-Tub at invasive tumor fronts and the correlation with poor breast cancer survival, these new discoveries help clarify how the TCP synergizes with oncogene activation, increases focal gelatin degradation, and may correspond to increased tumor cell invasion. These connections could inform more specific microtubule-directed therapies to target deTyr-tubulin.
- Published
- 2022
- Full Text
- View/download PDF
12. Chemical Biology of Mitotic Spindle Assembly Motors.
- Author
-
Solon A and Ohi R
- Subjects
- Biology, Cell Division, Chromosome Segregation, Microtubules, Mitosis, Kinesins, Spindle Apparatus
- Abstract
Mitotic kinesins play essential roles during mitotic spindle assembly and in ensuring proper chromosome segregation. Chemical inhibitors of mitotic kinesins are therefore valuable tools to study kinesin function in vitro and in cells. Because cancer is a disease of unregulated cell division, inhibitors also represent potential chemotherapeutic agents. Here, we present assays that can be used to evaluate the potency and specificity of mitotic kinesin inhibitors identified from high-throughput screening. By evaluating their effects in a variety of in vitro, fixed-cell, and live cell assays, screening hits can be prioritized and optimized to produce effective, on-target inhibitors., (© 2022. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
13. K-fiber bundles in the mitotic spindle are mechanically reinforced by Kif15.
- Author
-
Begley MA, Solon AL, Davis EM, Sherrill MG, Ohi R, and Elting MW
- Subjects
- Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Humans, Kidney cytology, Kinesins antagonists & inhibitors, Kinesins genetics, Kinetochores, Time-Lapse Imaging, Kinesins metabolism, Microtubules metabolism, Spindle Apparatus physiology
- Abstract
The mitotic spindle, a self-constructed microtubule-based machine, segregates chromosomes during cell division. In mammalian cells, microtubule bundles called kinetochore fibers (k-fibers) connect chromosomes to the spindle poles. Chromosome segregation thus depends on the mechanical integrity of k-fibers. Here we investigate the physical and molecular basis of k-fiber bundle cohesion. We detach k-fibers from poles by laser ablation-based cutting, thus revealing the contribution of pole-localized forces to k-fiber cohesion. We then measure the physical response of the remaining kinetochore-bound segments of the k-fibers. We observe that microtubules within ablated k-fibers often splay apart from their minus-ends. Furthermore, we find that minus-end clustering forces induced by ablation seem at least partially responsible for k-fiber splaying. We also investigate the role of the k-fiber-binding kinesin-12 Kif15. We find that pharmacological inhibition of Kif15-microtubule binding reduces the mechanical integrity of k-fibers. In contrast, inhibition of its motor activity but not its microtubule binding ability, i.e., locking Kif15 into a rigor state, does not greatly affect splaying. Altogether, the data suggest that forces holding k-fibers together are of similar magnitude to other spindle forces, and that Kif15, acting as a microtubule cross-linker, helps fortify and repair k-fibers. This feature of Kif15 may help support robust k-fiber function and prevent chromosome segregation errors.
- Published
- 2021
- Full Text
- View/download PDF
14. Kinesin-binding protein remodels the kinesin motor to prevent microtubule binding.
- Author
-
Solon AL, Tan Z, Schutt KL, Jepsen L, Haynes SE, Nesvizhskii AI, Sept D, Stumpff J, Ohi R, and Cianfrocco MA
- Abstract
Kinesins are regulated in space and time to ensure activation only in the presence of cargo. Kinesin-binding protein (KIFBP), which is mutated in Goldberg-Shprintzen syndrome, binds to and inhibits the catalytic motor heads of 8 of 45 kinesin superfamily members, but the mechanism remains poorly defined. Here, we used cryo–electron microscopy and cross-linking mass spectrometry to determine high-resolution structures of KIFBP alone and in complex with two mitotic kinesins, revealing structural remodeling of kinesin by KIFBP. We find that KIFBP remodels kinesin motors and blocks microtubule binding (i) via allosteric changes to kinesin and (ii) by sterically blocking access to the microtubule. We identified two regions of KIFBP necessary for kinesin binding and cellular regulation during mitosis. Together, this work further elucidates the molecular mechanism of KIFBP-mediated kinesin inhibition and supports a model in which structural rearrangement of kinesin motor domains by KIFBP abrogates motor protein activity.
- Published
- 2021
- Full Text
- View/download PDF
15. A cytoskeletal function for PBRM1 reading methylated microtubules.
- Author
-
Karki M, Jangid RK, Anish R, Seervai RNH, Bertocchio JP, Hotta T, Msaouel P, Jung SY, Grimm SL, Coarfa C, Weissman BE, Ohi R, Verhey KJ, Hodges HC, Burggren W, Dere R, Park IY, Prasad BVV, Rathmell WK, Walker CL, and Tripathi DN
- Subjects
- Chromatin metabolism, Chromatin Assembly and Disassembly, Cytoskeleton metabolism, Microtubules metabolism, Reading
- Abstract
Epigenetic effectors "read" marks "written" on chromatin to regulate function and fidelity of the genome. Here, we show that this coordinated read-write activity of the epigenetic machinery extends to the cytoskeleton, with PBRM1 in the PBAF chromatin remodeling complex reading microtubule methyl marks written by the SETD2 histone methyltransferase. PBRM1 binds SETD2 methyl marks via BAH domains, recruiting PBAF components to the mitotic spindle. This read-write activity was required for normal mitosis: Loss of SETD2 methylation or pathogenic BAH domain mutations disrupt PBRM1 microtubule binding and PBAF recruitment and cause genomic instability. These data reveal PBRM1 functions beyond chromatin remodeling with domains that allow it to integrate chromatin and cytoskeletal activity via its acetyl-binding BD and methyl-binding BAH domains, respectively. Conserved coordinated activity of the epigenetic machinery on the cytoskeleton opens a previously unknown window into how chromatin remodeler defects can drive disease via both epigenetic and cytoskeletal dysfunction., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2021
- Full Text
- View/download PDF
16. Parthenolide Destabilizes Microtubules by Covalently Modifying Tubulin.
- Author
-
Hotta T, Haynes SE, Blasius TL, Gebbie M, Eberhardt EL, Sept D, Cianfrocco M, Verhey KJ, Nesvizhskii AI, and Ohi R
- Subjects
- Carboxypeptidases metabolism, Carrier Proteins, Cell Cycle Proteins metabolism, Cysteine, Microtubules metabolism, Sesquiterpenes pharmacology, Tubulin drug effects, Tubulin metabolism
- Abstract
Detyrosination of the α-tubulin C-terminal tail is a post-translational modification (PTM) of microtubules that is key for many biological processes.
1 Although detyrosination is the oldest known microtubule PTM,2-7 the carboxypeptidase responsible for this modification, VASH1/2-SVBP, was identified only 3 years ago,8 , 9 precluding genetic approaches to prevent detyrosination. Studies examining the cellular functions of detyrosination have therefore relied on a natural product, parthenolide, which is widely believed to block detyrosination of α-tubulin in cells, presumably by inhibiting the activity of the relevant carboxypeptidase(s).10 Parthenolide is a sesquiterpene lactone that forms covalent linkages predominantly with exposed thiol groups; e.g., on cysteine residues.11-13 Using mass spectrometry, we show that parthenolide forms adducts on both cysteine and histidine residues on tubulin itself, in vitro and in cells. Parthenolide causes tubulin protein aggregation and prevents the formation of microtubules. In contrast to epoY, an epoxide inhibitor of VASH1/2-SVBP,9 parthenolide does not block VASH1-SVBP activity in vitro. Lastly, we show that epoY is an efficacious inhibitor of microtubule detyrosination in cells, providing an alternative chemical means to block detyrosination. Collectively, our work supports the notion that parthenolide is a promiscuous inhibitor of many cellular processes and suggests that its ability to block detyrosination may be an indirect consequence of reducing the polymerization-competent pool of tubulin in cells., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
17. Impact of the 'tubulin economy' on the formation and function of the microtubule cytoskeleton.
- Author
-
Ohi R, Strothman C, and Zanic M
- Subjects
- Animals, Cytoskeleton metabolism, Humans, Kinesins metabolism, Microtubule-Associated Proteins metabolism, Tubulin metabolism, Microtubules metabolism
- Abstract
The microtubule cytoskeleton is assembled from a finite pool of α,β-tubulin, the size of which is controlled by an autoregulation mechanism. Cells also tightly regulate the architecture and dynamic behavior of microtubule arrays. Here, we discuss progress in our understanding of how tubulin autoregulation is achieved and highlight work showing that tubulin, in its unassembled state, is relevant for regulating the formation and organization of microtubules. Emerging evidence suggests that tubulin regulates microtubule-associated proteins and kinesin motors that are critical for microtubule nucleation, dynamics, and function. These relationships create feedback loops that connect the tubulin assembly cycle to the organization and dynamics of microtubule networks. We term this concept the 'tubulin economy', which emphasizes the idea that tubulin is a resource that can be deployed for the immediate purpose of creating polymers, or alternatively as a signaling molecule that has more far-reaching consequences for the organization of microtubule arrays., Competing Interests: Conflict of interest statement Nothing declared., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. Discovery of a selective inhibitor of doublecortin like kinase 1.
- Author
-
Ferguson FM, Nabet B, Raghavan S, Liu Y, Leggett AL, Kuljanin M, Kalekar RL, Yang A, He S, Wang J, Ng RWS, Sulahian R, Li L, Poulin EJ, Huang L, Koren J, Dieguez-Martinez N, Espinosa S, Zeng Z, Corona CR, Vasta JD, Ohi R, Sim T, Kim ND, Harshbarger W, Lizcano JM, Robers MB, Muthaswamy S, Lin CY, Look AT, Haigis KM, Mancias JD, Wolpin BM, Aguirre AJ, Hahn WC, Westover KD, and Gray NS
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, Doublecortin Protein, Doublecortin-Like Kinases, Drug Screening Assays, Antitumor, Gene Expression Regulation, Humans, Intracellular Signaling Peptides and Proteins metabolism, Male, Mice, Molecular Docking Simulation, Molecular Structure, Protein Kinase Inhibitors pharmacokinetics, Proteomics, Rats, Structure-Activity Relationship, Zebrafish, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal drug therapy, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, Pancreatic Neoplasms drug therapy, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer.
- Published
- 2020
- Full Text
- View/download PDF
19. Precise Tuning of Cortical Contractility Regulates Cell Shape during Cytokinesis.
- Author
-
Taneja N, Bersi MR, Baillargeon SM, Fenix AM, Cooper JA, Ohi R, Gama V, Merryman WD, and Burnette DT
- Subjects
- Actin Cytoskeleton metabolism, Actins metabolism, Actins physiology, Animals, COS Cells, Cell Division, Cell Movement, Chlorocebus aethiops, Cytoskeletal Proteins metabolism, HeLa Cells, Humans, Morphogenesis, Muscle Contraction, Myosin Type II physiology, Nonmuscle Myosin Type IIA metabolism, Nonmuscle Myosin Type IIB metabolism, Cell Shape physiology, Cytokinesis physiology, Myosin Type II metabolism
- Abstract
The mechanical properties of the actin cortex regulate shape changes during cell division, cell migration, and tissue morphogenesis. We show that modulation of myosin II (MII) filament composition allows tuning of surface tension at the cortex to maintain cell shape during cytokinesis. Our results reveal that MIIA generates cortex tension, while MIIB acts as a stabilizing motor and its inclusion in MII hetero-filaments reduces cortex tension. Tension generation by MIIA drives faster cleavage furrow ingression and bleb formation. We also show distinct roles for the motor and tail domains of MIIB in maintaining cytokinetic fidelity. Maintenance of cortical stability by the motor domain of MIIB safeguards against shape instability-induced chromosome missegregation, while its tail domain mediates cortical localization at the terminal stages of cytokinesis to mediate cell abscission. Because most non-muscle contractile systems are cortical, this tuning mechanism will likely be applicable to numerous processes driven by myosin-II contractility., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
20. Microtubule minus-end stability is dictated by the tubulin off-rate.
- Author
-
Strothman C, Farmer V, Arpağ G, Rodgers N, Podolski M, Norris S, Ohi R, and Zanic M
- Subjects
- Animals, Cattle, Kinesins metabolism, Microtubules metabolism, Tubulin metabolism, Kinesins chemistry, Microtubules chemistry, Tubulin chemistry
- Abstract
Dynamic organization of microtubule minus ends is vital for the formation and maintenance of acentrosomal microtubule arrays. In vitro, both microtubule ends switch between phases of assembly and disassembly, a behavior called dynamic instability. Although minus ends grow slower, their lifetimes are similar to those of plus ends. The mechanisms underlying these distinct dynamics remain unknown. Here, we use an in vitro reconstitution approach to investigate minus-end dynamics. We find that minus-end lifetimes are not defined by the mean size of the protective GTP-tubulin cap. Rather, we conclude that the distinct tubulin off-rate is the primary determinant of the difference between plus- and minus-end dynamics. Further, our results show that the minus-end-directed kinesin-14 HSET/KIFC1 suppresses tubulin off-rate to specifically suppress minus-end catastrophe. HSET maintains its protective minus-end activity even when challenged by a known microtubule depolymerase, kinesin-13 MCAK. Our results provide novel insight into the mechanisms of minus-end dynamics, essential for our understanding of microtubule minus-end regulation in cells., (© 2019 Strothman et al.)
- Published
- 2019
- Full Text
- View/download PDF
21. Polyglutamylation of tubulin's C-terminal tail controls pausing and motility of kinesin-3 family member KIF1A.
- Author
-
Lessard DV, Zinder OJ, Hotta T, Verhey KJ, Ohi R, and Berger CL
- Subjects
- Animals, Cattle, HeLa Cells, Humans, Kinesins genetics, Microtubules genetics, Peptides genetics, Protein Domains, Protein Structure, Secondary, Axonal Transport, Axons metabolism, Kinesins metabolism, Microtubules metabolism, Peptides metabolism, Protein Processing, Post-Translational
- Abstract
The kinesin-3 family member KIF1A plays a critical role in site-specific neuronal cargo delivery during axonal transport. KIF1A cargo is mislocalized in many neurodegenerative diseases, indicating that KIF1A's highly efficient, superprocessive motility along axonal microtubules needs to be tightly regulated. One potential regulatory mechanism may be through posttranslational modifications (PTMs) of axonal microtubules. These PTMs often occur on the C-terminal tails of the microtubule tracks, act as molecular "traffic signals" helping to direct kinesin motor cargo delivery, and include C-terminal tail polyglutamylation important for KIF1A cargo transport. KIF1A initially interacts with microtubule C-terminal tails through its K-loop, a positively charged surface loop of the KIF1A motor domain. However, the role of the K-loop in KIF1A motility and response to perturbations in C-terminal tail polyglutamylation is underexplored. Using single-molecule imaging, we present evidence that KIF1A pauses on different microtubule lattice structures, linking multiple processive segments together and contributing to KIF1A's characteristic superprocessive run length. Furthermore, modifications of the KIF1A K-loop or tubulin C-terminal tail polyglutamylation reduced KIF1A pausing and overall run length. These results suggest a new mechanism to regulate KIF1A motility via pauses mediated by K-loop/polyglutamylated C-terminal tail interactions, providing further insight into KIF1A's role in axonal transport., (© 2019 Lessard et al.)
- Published
- 2019
- Full Text
- View/download PDF
22. Mitotic chromosome alignment ensures mitotic fidelity by promoting interchromosomal compaction during anaphase.
- Author
-
Fonseca CL, Malaby HLH, Sepaniac LA, Martin W, Byers C, Czechanski A, Messinger D, Tang M, Ohi R, Reinholdt LG, and Stumpff J
- Subjects
- Animals, Cell Line, Cell Proliferation, Epithelial Cells physiology, Humans, Kinesins genetics, Kinesins metabolism, Mice, Knockout, Retinal Pigment Epithelium physiology, Spindle Apparatus genetics, Spindle Apparatus metabolism, Time Factors, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Anaphase, Chromosome Segregation, Chromosomes, Human, Spindle Apparatus physiology
- Abstract
Chromosome alignment at the equator of the mitotic spindle is a highly conserved step during cell division; however, its importance to genomic stability and cellular fitness is not understood. Normal mammalian somatic cells lacking KIF18A function complete cell division without aligning chromosomes. These alignment-deficient cells display normal chromosome copy numbers in vitro and in vivo, suggesting that chromosome alignment is largely dispensable for maintenance of euploidy. However, we find that loss of chromosome alignment leads to interchromosomal compaction defects during anaphase, abnormal organization of chromosomes into a single nucleus at mitotic exit, and the formation of micronuclei in vitro and in vivo. These defects slow cell proliferation and are associated with impaired postnatal growth and survival in mice. Our studies support a model in which the alignment of mitotic chromosomes promotes proper organization of chromosomes into a single nucleus and continued proliferation by ensuring that chromosomes segregate as a compact mass during anaphase., (© 2019 Fonseca et al.)
- Published
- 2019
- Full Text
- View/download PDF
23. Kinesin-binding protein ensures accurate chromosome segregation by buffering KIF18A and KIF15.
- Author
-
Malaby HLH, Dumas ME, Ohi R, and Stumpff J
- Subjects
- Cell Nucleus genetics, HeLa Cells, Humans, Kinesins genetics, Nerve Tissue Proteins genetics, Retinal Pigment Epithelium enzymology, Signal Transduction, Anaphase, Cell Nucleus enzymology, Chromosome Segregation, Chromosomes, Human, Kinesins metabolism, Nerve Tissue Proteins metabolism
- Abstract
Mitotic kinesins must be regulated to ensure a precise balance of spindle forces and accurate segregation of chromosomes into daughter cells. Here, we demonstrate that kinesin-binding protein (KBP) reduces the activity of KIF18A and KIF15 during metaphase. Overexpression of KBP disrupts the movement and alignment of mitotic chromosomes and decreases spindle length, a combination of phenotypes observed in cells deficient for KIF18A and KIF15, respectively. We show through gliding filament and microtubule co-pelleting assays that KBP directly inhibits KIF18A and KIF15 motor activity by preventing microtubule binding. Consistent with these effects, the mitotic localizations of KIF18A and KIF15 are altered by overexpression of KBP. Cells depleted of KBP exhibit lagging chromosomes in anaphase, an effect that is recapitulated by KIF15 and KIF18A overexpression. Based on these data, we propose a model in which KBP acts as a protein buffer in mitosis, protecting cells from excessive KIF18A and KIF15 activity to promote accurate chromosome segregation., (© 2019 Malaby et al.)
- Published
- 2019
- Full Text
- View/download PDF
24. Dual inhibition of Kif15 by oxindole and quinazolinedione chemical probes.
- Author
-
Dumas ME, Chen GY, Kendrick ND, Xu G, Larsen SD, Jana S, Waterson AG, Bauer JA, Hancock W, Sulikowski GA, and Ohi R
- Subjects
- Dose-Response Relationship, Drug, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Humans, Kinesins metabolism, Molecular Probes chemical synthesis, Molecular Probes chemistry, Molecular Structure, Oxindoles chemical synthesis, Oxindoles chemistry, Quinazolinones chemical synthesis, Quinazolinones chemistry, Structure-Activity Relationship, Enzyme Inhibitors pharmacology, Kinesins antagonists & inhibitors, Molecular Probes pharmacology, Oxindoles pharmacology, Quinazolinones pharmacology
- Abstract
The mitotic spindle is a microtubule-based machine that segregates a replicated set of chromosomes during cell division. Many cancer drugs alter or disrupt the microtubules that form the mitotic spindle. Microtubule-dependent molecular motors that function during mitosis are logical alternative mitotic targets for drug development. Eg5 (Kinesin-5) and Kif15 (Kinesin-12), in particular, are an attractive pair of motor proteins, as they work in concert to drive centrosome separation and promote spindle bipolarity. Furthermore, we hypothesize that the clinical failure of Eg5 inhibitors may be (in part) due to compensation by Kif15. In order to test this idea, we screened a small library of kinase inhibitors and identified GW108X, an oxindole that inhibits Kif15 in vitro. We show that GW108X has a distinct mechanism of action compared with a commercially available Kif15 inhibitor, Kif15-IN-1 and may serve as a lead with which to further develop Kif15 inhibitors as clinically relevant agents., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
25. Promoting Diversity in the Cytoskeleton through STEM-Structural Transitions and Energetics of Microtubule Subunits.
- Author
-
Ohi R and Verhey KJ
- Subjects
- Cytoskeleton, Humans, Microtubules, Tubulin
- Abstract
All microtubules assemble from tubulin subunits, but the functional impact of tubulin isotypes has been unclear. Reporting in Developmental Cell, Chaaban et al. (2018) and Ti et al. (2018) show that tubulin isotypes differ in their conformational flexibility, which alters microtubule dynamics and architecture, yet maintain "lock-and-key" interactions with neighbors., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
26. Processive Kinesin-14 HSET Exhibits Directional Flexibility Depending on Motor Traffic.
- Author
-
Reinemann DN, Norris SR, Ohi R, and Lang MJ
- Subjects
- Dyneins metabolism, Spindle Apparatus metabolism, Tubulin metabolism, Centrosome metabolism, Kinesins metabolism, Microtubules metabolism
- Abstract
A common mitotic defect observed in cancer cells that possess supernumerary (more than two) centrosomes is multipolar spindle formation [1, 2]. Such structures are resolved into a bipolar geometry by minus-end-directed motor proteins, such as cytoplasmic dynein and the kinesin-14 HSET [3-8]. HSET is also thought to antagonize plus-end-directed kinesin-5 Eg5 to balance spindle forces [4, 5, 7, 9]. However, the biomechanics of this force opposition are unclear, as HSET has previously been defined as a non-processive motor [10-16]. Here, we use optical trapping to elucidate the mechanism of force generation by HSET. We show that a single HSET motor has a processive nature with the ability to complete multiple steps while trapped along a microtubule and when unloaded can move in both directions for microns. Compared to other kinesins, HSET has a relatively weak stall force of 1.1 pN [17, 18]. Moreover, HSET's tail domain and its interaction with the E-hook of tubulin are necessary for long-range motility. In vitro polarity-marked bundle assays revealed that HSET selectively generates force in anti-parallel bundles on the order of its stall force. When combined with varied ratios of Eg5, HSET adopts Eg5's directionality while acting as an antagonizing force brake, requiring at least a 10-fold higher Eg5 concentration to surpass HSET's sliding force. These results reveal HSET's ability to change roles within the spindle from acting as an adjustable microtubule slider and force regulator to a processive motor that aids in minus end focusing., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Microtubule minus-end aster organization is driven by processive HSET-tubulin clusters.
- Author
-
Norris SR, Jung S, Singh P, Strothman CE, Erwin AL, Ohi MD, Zanic M, and Ohi R
- Subjects
- Animals, Cell Tracking methods, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, HeLa Cells, Humans, Kinesins genetics, Microscopy, Fluorescence methods, Protein Binding, Time-Lapse Imaging methods, Kinesins metabolism, Microtubules metabolism, Molecular Motor Proteins metabolism, Tubulin metabolism
- Abstract
Higher-order structures of the microtubule (MT) cytoskeleton are comprised of two architectures: bundles and asters. Although both architectures are critical for cellular function, the molecular pathways that drive aster formation are poorly understood. Here, we study aster formation by human minus-end-directed kinesin-14 (HSET/KIFC1). We show that HSET is incapable of forming asters from preformed, nongrowing MTs, but rapidly forms MT asters in the presence of soluble (non-MT) tubulin. HSET binds soluble (non-MT) tubulin via its N-terminal tail domain to form heterogeneous HSET-tubulin clusters containing multiple motors. Cluster formation induces motor processivity and rescues the formation of asters from nongrowing MTs. We then show that excess soluble (non-MT) tubulin stimulates aster formation in HeLa cells overexpressing HSET during mitosis. We propose a model where HSET can toggle between MT bundle and aster formation in a manner governed by the availability of soluble (non-MT) tubulin.
- Published
- 2018
- Full Text
- View/download PDF
28. SETD2 Haploinsufficiency for Microtubule Methylation Is an Early Driver of Genomic Instability in Renal Cell Carcinoma.
- Author
-
Chiang YC, Park IY, Terzo EA, Tripathi DN, Mason FM, Fahey CC, Karki M, Shuster CB, Sohn BH, Chowdhury P, Powell RT, Ohi R, Tsai YS, de Cubas AA, Khan A, Davis IJ, Strahl BD, Parker JS, Dere R, Walker CL, and Rathmell WK
- Subjects
- Animals, Carcinogenesis genetics, Carcinoma, Renal Cell pathology, Cell Line, Tumor, Fibroblasts, Gene Knockdown Techniques, Genomic Instability, Haploinsufficiency, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Humans, Kidney Neoplasms pathology, Kidney Tubules, Proximal cytology, Kidney Tubules, Proximal pathology, Lysine metabolism, Methylation, Mice, Micronuclei, Chromosome-Defective, Carcinoma, Renal Cell genetics, Chromosomes, Human, Pair 3 genetics, Histone-Lysine N-Methyltransferase genetics, Kidney Neoplasms genetics, Microtubules metabolism
- Abstract
Loss of the short arm of chromosome 3 (3p) occurs early in >95% of clear cell renal cell carcinoma (ccRCC). Nearly ubiquitous 3p loss in ccRCC suggests haploinsufficiency for 3p tumor suppressors as early drivers of tumorigenesis. We previously reported methyltransferase SETD2 , which trimethylates H3 histones on lysine 36 (H3K36me3) and is located in the 3p deletion, to also trimethylate microtubules on lysine 40 (αTubK40me3) during mitosis, with αTubK40me3 required for genomic stability. We now show that monoallelic, Setd2 -deficient cells retaining H3K36me3, but not αTubK40me3, exhibit a dramatic increase in mitotic defects and micronuclei count, with increased viability compared with biallelic loss. In SETD2 -inactivated human kidney cells, rescue with a pathogenic SETD2 mutant deficient for microtubule (αTubK40me3), but not histone (H3K36me3) methylation, replicated this phenotype. Genomic instability (micronuclei) was also a hallmark of patient-derived cells from ccRCC. These data show that the SETD2 tumor suppressor displays a haploinsufficiency phenotype disproportionately impacting microtubule methylation and serves as an early driver of genomic instability. Significance: Loss of a single allele of a chromatin modifier plays a role in promoting oncogenesis, underscoring the growing relevance of tumor suppressor haploinsufficiency in tumorigenesis. Cancer Res; 78(12); 3135-46. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
29. Two mechanisms coordinate the recruitment of the chromosomal passenger complex to the plane of cell division.
- Author
-
Landino J, Norris SR, Li M, Ballister ER, Lampson MA, and Ohi R
- Subjects
- Anaphase physiology, Aurora Kinase B metabolism, Cell Division genetics, Centromere metabolism, Chromosomal Proteins, Non-Histone metabolism, Chromosomes metabolism, Cytokinesis physiology, DNA-Binding Proteins metabolism, HeLa Cells, Humans, Kinesins metabolism, Kinesins physiology, Metaphase physiology, Microfilament Proteins metabolism, Microtubules metabolism, Spindle Apparatus metabolism, Cell Division physiology, Chromosomal Proteins, Non-Histone physiology, Chromosome Segregation physiology
- Abstract
During cytokinesis, the chromosomal passenger complex (CPC) promotes midzone organization, specifies the cleavage plane, and regulates furrow contractility. The localizations of the CPC are coupled to its cytokinetic functions. At the metaphase-to-anaphase transition, the CPC dissociates from centromeres and localizes to midzone microtubules and the equatorial cortex. CPC relocalization to the cell middle is thought to depend on MKlp2-driven, plus end-directed transport. In support of this idea, MKlp2 depletion impairs cytokinesis; however, cytokinesis failure stems from furrow regression rather than failed initiation of furrowing. This suggests that an alternative mechanism(s) may concentrate the CPC at the division plane. We show here that direct actin binding, via the inner centromere protein (INCENP), enhances CPC enrichment at the equatorial cortex, thus acting in tandem with MKlp2. INCENP overexpression rescues furrowing in MKlp2-depleted cells in an INCENP-actin binding-dependent manner. Using live-cell imaging, we also find that MKlp2-dependent targeting of the CPC is biphasic. MKlp2 targets the CPC to the anti-parallel microtubule overlap of the midzone, after which the MKlp2-CPC complex moves in a nondirected manner. Collectively, our work suggests that both actin binding and MKlp2-dependent midzone targeting cooperate to precisely position the CPC during mitotic exit, and that these pathways converge to ensure successful cleavage furrow ingression., (© 2017 Landino et al. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).)
- Published
- 2017
- Full Text
- View/download PDF
30. Collective Force Regulation in Anti-parallel Microtubule Gliding by Dimeric Kif15 Kinesin Motors.
- Author
-
Reinemann DN, Sturgill EG, Das DK, Degen MS, Vörös Z, Hwang W, Ohi R, and Lang MJ
- Subjects
- Biomechanical Phenomena, Centrosome metabolism, Humans, Protein Binding, Kinesins metabolism, Microtubules metabolism
- Abstract
During cell division, the mitotic kinesin-5 Eg5 generates most of the force required to separate centrosomes during spindle assembly. However, Kif15, another mitotic kinesin, can replace Eg5 function, permitting mammalian cells to acquire resistance to Eg5 poisons. Unlike Eg5, the mechanism by which Kif15 generates centrosome separation forces is unknown. Here we investigated the mechanical properties and force generation capacity of Kif15 at the single-molecule level using optical tweezers. We found that the non-motor microtubule-binding tail domain interacts with the microtubule's E-hook tail with a rupture force higher than the stall force of the motor. This allows Kif15 dimers to productively and efficiently generate forces that could potentially slide microtubules apart. Using an in vitro optical trapping and fluorescence assay, we found that Kif15 slides anti-parallel microtubules apart with gradual force buildup while parallel microtubule bundles remain stationary with a small amount of antagonizing force generated. A stochastic simulation shows the essential role of Kif15's tail domain for load storage within the Kif15-microtubule system. These results suggest a mechanism for how Kif15 rescues bipolar spindle assembly., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
31. Cell Division: Centrosomes Have Separation Anxiety.
- Author
-
Norris SR and Ohi R
- Subjects
- Anxiety, Separation, Centrosome, Humans, Microtubules, Mitosis, Kinesins genetics, Spindle Apparatus
- Abstract
Prior to mitosis, duplicated centrosomes are tethered together, which is thought to prevent mitotic defects. A new study establishes the role of tetrameric Kif25, a microtubule minus-end-directed kinesin-14 motor, in preventing premature centrosome separation through a microtubule-dependent pathway., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. Eg5 Inhibitors Have Contrasting Effects on Microtubule Stability and Metaphase Spindle Integrity.
- Author
-
Chen GY, Kang YJ, Gayek AS, Youyen W, Tüzel E, Ohi R, and Hancock WO
- Subjects
- Animals, Cell Line, Humans, Microtubules metabolism, Spindle Apparatus metabolism, Kinesins antagonists & inhibitors, Metaphase drug effects, Microtubules drug effects, Spindle Apparatus drug effects
- Abstract
To uncover their contrasting mechanisms, antimitotic drugs that inhibit Eg5 (kinesin-5) were analyzed in mixed-motor gliding assays of kinesin-1 and Eg5 motors in which Eg5 "braking" dominates motility. Loop-5 inhibitors (monastrol, STLC, ispinesib, and filanesib) increased gliding speeds, consistent with inducing a weak-binding state in Eg5, whereas BRD9876 slowed gliding, consistent with locking Eg5 in a rigor state. Biochemical and single-molecule assays demonstrated that BRD9876 acts as an ATP- and ADP-competitive inhibitor with 4 nM K
I . Consistent with its microtubule polymerase activity, Eg5 was shown to stabilize microtubules against depolymerization. This stabilization activity was eliminated in monastrol but was enhanced by BRD9876. Finally, in metaphase-arrested RPE-1 cells, STLC promoted spindle collapse, whereas BRD9876 did not. Thus, different Eg5 inhibitors impact spindle assembly and architecture through contrasting mechanisms, and rigor inhibitors may paradoxically have the capacity to stabilize microtubule arrays in cells.- Published
- 2017
- Full Text
- View/download PDF
33. Resistance is not futile: Surviving Eg5 inhibition.
- Author
-
Dumas ME, Sturgill EG, and Ohi R
- Subjects
- Microtubules, Mitosis, Spindle Apparatus, Kinesins metabolism
- Published
- 2016
- Full Text
- View/download PDF
34. Loss of CENP-F results in distinct microtubule-related defects without chromosomal abnormalities.
- Author
-
Pfaltzgraff ER, Roth GM, Miller PM, Gintzig AG, Ohi R, and Bader DM
- Subjects
- Animals, Cell Cycle genetics, Cell Cycle physiology, Centromere metabolism, Chromosome Aberrations, Chromosome Segregation, Fibroblasts, Interphase genetics, Kinetochores metabolism, Mice, Mice, Knockout, Microtubules physiology, Mitosis genetics, Protein Binding, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Microfilament Proteins genetics, Microfilament Proteins metabolism
- Abstract
Microtubule (MT)-binding centromere protein F (CENP-F) was previously shown to play a role exclusively in chromosome segregation during cellular division. Many cell models of CENP-F depletion show a lag in the cell cycle and aneuploidy. Here, using our novel genetic deletion model, we show that CENP-F also regulates a broader range of cellular functions outside of cell division. We characterized CENP-F(+/+) and CENP-F(-/-) mouse embryonic fibroblasts (MEFs) and found drastic differences in multiple cellular functions during interphase, including cell migration, focal adhesion dynamics, and primary cilia formation. We discovered that CENP-F(-/-) MEFs have severely diminished MT dynamics, which underlies the phenotypes we describe. These data, combined with recent biochemical research demonstrating the strong binding of CENP-F to the MT network, support the conclusion that CENP-F is a powerful regulator of MT dynamics during interphase and affects heterogeneous cell functions., (© 2016 Pfaltzgraff et al. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).)
- Published
- 2016
- Full Text
- View/download PDF
35. CDK-1 Inhibition in G2 Stabilizes Kinetochore-Microtubules in the following Mitosis.
- Author
-
Gayek AS and Ohi R
- Subjects
- CDC2 Protein Kinase, Cell Line, Transformed, Chromosomes, Human genetics, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Humans, Kinesins genetics, Kinesins metabolism, Microtubules genetics, Anaphase, Chromosomes, Human metabolism, Cyclin-Dependent Kinases antagonists & inhibitors, G2 Phase, Kinetochores metabolism, Microtubules metabolism
- Abstract
Cell proliferation is driven by cyclical activation of cyclin-dependent kinases (CDKs), which produce distinct biochemical cell cycle phases. Mitosis (M phase) is orchestrated by CDK-1, complexed with mitotic cyclins. During M phase, chromosomes are segregated by a bipolar array of microtubules called the mitotic spindle. The essential bipolarity of the mitotic spindle is established by the kinesin-5 Eg5, but factors influencing the maintenance of spindle bipolarity are not fully understood. Here, we describe an unexpected link between inhibiting CDK-1 before mitosis and bipolar spindle maintenance. Spindles in human RPE-1 cells normally collapse to monopolar structures when Eg5 is inhibited at metaphase. However, we found that inhibition of CDK-1 in the G2 phase of the cell cycle improved the ability of RPE-1 cells to maintain spindle bipolarity without Eg5 activity in the mitosis immediately after release from CDK-1 inhibition. This improved bipolarity maintenance correlated with an increase in the stability of kinetochore-microtubules, the subset of microtubules that link chromosomes to the spindle. The improvement in bipolarity maintenance after CDK-1 inhibition in G2 required both the kinesin-12 Kif15 and increased stability of kinetochore-microtubules. Consistent with increased kinetochore-microtubule stability, we find that inhibition of CDK-1 in G2 impairs mitotic fidelity by increasing the incidence of lagging chromosomes in anaphase. These results suggest that inhibition of CDK-1 in G2 causes unpredicted effects in mitosis, even after CDK-1 inhibition is relieved.
- Published
- 2016
- Full Text
- View/download PDF
36. Kinesin-5 inhibitor resistance is driven by kinesin-12.
- Author
-
Sturgill EG, Norris SR, Guo Y, and Ohi R
- Subjects
- Cysteine analogs & derivatives, Cysteine pharmacology, HeLa Cells, Humans, Kinesins antagonists & inhibitors, Kinesins genetics, Kinesins metabolism, Spindle Apparatus ultrastructure, Kinesins physiology, Spindle Apparatus metabolism
- Abstract
The microtubule (MT) cytoskeleton bipolarizes at the onset of mitosis to form the spindle. In animal cells, the kinesin-5 Eg5 primarily drives this reorganization by actively sliding MTs apart. Its primacy during spindle assembly renders Eg5 essential for mitotic progression, demonstrated by the lethal effects of kinesin-5/Eg5 inhibitors (K5Is) administered in cell culture. However, cultured cells can acquire resistance to K5Is, indicative of alternative spindle assembly mechanisms and/or pharmacological failure. Through characterization of novel K5I-resistant cell lines, we unveil an Eg5 motility-independent spindle assembly pathway that involves both an Eg5 rigor mutant and the kinesin-12 Kif15. This pathway centers on spindle MT bundling instead of Kif15 overexpression, distinguishing it from those previously described. We further show that large populations (∼10(7) cells) of HeLa cells require Kif15 to survive K5I treatment. Overall, this study provides insight into the functional plasticity of mitotic kinesins during spindle assembly and has important implications for the development of antimitotic regimens that target this process., (© 2016 Sturgill et al.)
- Published
- 2016
- Full Text
- View/download PDF
37. Ahead of the Curve: New Insights into Microtubule Dynamics.
- Author
-
Ohi R and Zanic M
- Abstract
Microtubule dynamics are fundamental for many aspects of cell physiology, but their mechanistic underpinnings remain unclear despite 40 years of intense research. In recent years, the continued union of reconstitution biochemistry, structural biology, and modeling has yielded important discoveries that deepen our understanding of microtubule dynamics. These studies, which we review here, underscore the importance of GTP hydrolysis-induced changes in tubulin structure as microtubules assemble, and highlight the fact that each aspect of microtubule behavior is the output of complex, multi-step processes. Although this body of work moves us closer to appreciating the key features of microtubule biochemistry that drive dynamic instability, the divide between our understanding of microtubules in isolation versus within the cellular milieu remains vast. Bridging this gap will serve as fertile grounds of cytoskeleton-focused research for many years to come.
- Published
- 2016
- Full Text
- View/download PDF
38. Expansion and concatenation of non-muscle myosin IIA filaments drive cellular contractile system formation during interphase and mitosis.
- Author
-
Fenix AM, Taneja N, Buttler CA, Lewis J, Van Engelenburg SB, Ohi R, and Burnette DT
- Abstract
Cell movement and cytokinesis are facilitated by contractile forces generated by the molecular motor, non-muscle myosin II (NMII). NMII molecules form a filament (NMII-F) through interactions of their C-terminal rod domains, positioning groups of N-terminal motor domains on opposite sides. The NMII motors then bind and pull actin filaments toward the NMII-F, thus driving contraction. Inside of crawling cells, NMIIA-Fs form large macromolecular ensembles (i.e., NMIIA-F stacks) but how this occurs is unknown. Here we show NMIIA-F stacks are formed through two non-mutually exclusive mechanisms: expansion and concatenation. During expansion, NMIIA molecules within the NMIIA-F spread out concurrent with addition of new NMIIA molecules. Concatenation occurs when multiple NMIIA-F/NMIIA-F stacks move together and align. We found NMIIA-F stack formation was regulated by both motor-activity and the availability of surrounding actin filaments. Furthermore, our data showed expansion and concatenation also formed the contractile ring in dividing cells. Thus, interphase and mitotic cells share similar mechanisms for creating large contractile units, and these are likely to underlie how other myosin II-based contractile systems are assembled., (© 2016 by The American Society for Cell Biology.)
- Published
- 2016
- Full Text
- View/download PDF
39. The Timing of Midzone Stabilization during Cytokinesis Depends on Myosin II Activity and an Interaction between INCENP and Actin.
- Author
-
Landino J and Ohi R
- Subjects
- Actins metabolism, Anaphase, Cell Line, HeLa Cells, Humans, Myosin Type II metabolism, Actin Cytoskeleton physiology, Cell Division, Cytokinesis, Microtubules physiology
- Abstract
The final steps of cell division are tightly coordinated in space and time, but whether mechanisms exist to couple the actin and microtubule (MT) cytoskeletons during anaphase and cytokinesis (C phase) is largely unknown. During anaphase, MTs are incorporated into an anti-parallel array termed the spindle midzone (midzone MTs), whereas F-actin and non-muscle myosin II, together with other factors, organize into the cleavage furrow [1]. Previous studies in somatic cells have shown that midzone MTs become highly stable after furrows have begun ingression [2], indicating that furrow-to-MT communication may occur. Midzone formation is also inhibited in fly spermatocytes that fail to form a cleavage furrow [3] and during monopolar cytokinesis when myosin contractility is blocked by blebbistatin [4]. We show here that midzone MT stabilization is dependent on actomyosin contraction, suggesting that there is active coordination between furrow ingression and microtubule dynamics. Midzone microtubule stabilization also depends on the kinase activity of Aurora B, the catalytic subunit of the chromosomal passenger complex (CPC), uncovering a feedback mechanism that couples furrowing with microtubule dynamics. We further show that the CPC scaffolding protein INCENP (inner centromere protein) binds actin, an interaction that is important for cytokinesis and for midzone MT stabilization following furrow ingression. Stabilization of midzone MTs with low amounts of Taxol rescues cytokinesis in INCENP actin-binding mutant-expressing cells. Collectively, our work demonstrates that the actin and microtubule cytoskeletons are coordinated during cytokinesis and suggests that the CPC is integral for coupling furrow ingression with midzone microtubule stabilization., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Biased Brownian motion as a mechanism to facilitate nanometer-scale exploration of the microtubule plus end by a kinesin-8.
- Author
-
Shin Y, Du Y, Collier SE, Ohi MD, Lang MJ, and Ohi R
- Subjects
- Cell Tracking, Diffusion, HeLa Cells, Humans, Kinesins chemistry, Kinetics, Mutant Proteins chemistry, Mutant Proteins metabolism, Protein Binding, Protein Multimerization, Protein Structure, Tertiary, Video Recording, Biophysical Phenomena, Kinesins metabolism, Microtubules metabolism, Motion
- Abstract
Kinesin-8s are plus-end-directed motors that negatively regulate microtubule (MT) length. Well-characterized members of this subfamily (Kip3, Kif18A) exhibit two important properties: (i) They are "ultraprocessive," a feature enabled by a second MT-binding site that tethers the motors to a MT track, and (ii) they dissociate infrequently from the plus end. Together, these characteristics combined with their plus-end motility cause Kip3 and Kif18A to enrich preferentially at the plus ends of long MTs, promoting MT catastrophes or pausing. Kif18B, an understudied human kinesin-8, also limits MT growth during mitosis. In contrast to Kif18A and Kip3, localization of Kif18B to plus ends relies on binding to the plus-end tracking protein EB1, making the relationship between its potential plus-end-directed motility and plus-end accumulation unclear. Using single-molecule assays, we show that Kif18B is only modestly processive and that the motor switches frequently between directed and diffusive modes of motility. Diffusion is promoted by the tail domain, which also contains a second MT-binding site that decreases the off rate of the motor from the MT lattice. In cells, Kif18B concentrates at the extreme tip of a subset of MTs, superseding EB1. Our data demonstrate that kinesin-8 motors use diverse design principles to target MT plus ends, which likely target them to the plus ends of distinct MT subpopulations in the mitotic spindle.
- Published
- 2015
- Full Text
- View/download PDF
41. Fanning the flames of CIN.
- Author
-
Gayek AS and Ohi R
- Subjects
- Humans, Microtubules metabolism, Spindle Apparatus metabolism
- Published
- 2015
- Full Text
- View/download PDF
42. Structural and functional insights into the N-terminus of Schizosaccharomyces pombe Cdc5.
- Author
-
Collier SE, Voehler M, Peng D, Ohi R, Gould KL, Reiter NJ, and Ohi MD
- Subjects
- Binding Sites, Catalytic Domain, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Gene Deletion, Mutant Proteins chemistry, Mutant Proteins metabolism, Nuclear Magnetic Resonance, Biomolecular, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Protein Conformation, Protein Folding, Protein Interaction Domains and Motifs, Protein Stability, RNA, Fungal chemistry, RNA, Fungal metabolism, RNA, Small Nuclear chemistry, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Schizosaccharomyces pombe Proteins genetics, Schizosaccharomyces pombe Proteins metabolism, Spliceosomes genetics, Spliceosomes metabolism, Titrimetry, Cell Cycle Proteins chemistry, Models, Molecular, RNA Splicing, RNA, Double-Stranded metabolism, RNA, Small Nuclear metabolism, RNA-Binding Proteins chemistry, Schizosaccharomyces pombe Proteins chemistry, Spliceosomes chemistry
- Abstract
The spliceosome is a dynamic macromolecular machine composed of five small nuclear ribonucleoparticles (snRNPs), the NineTeen Complex (NTC), and other proteins that catalyze the removal of introns mature to form the mature message. The NTC, named after its founding member Saccharomyces cerevisiae Prp19, is a conserved spliceosome subcomplex composed of at least nine proteins. During spliceosome assembly, the transition to an active spliceosome correlates with stable binding of the NTC, although the mechanism of NTC function is not understood. Schizosaccharomyces pombe Cdc5, a core subunit of the NTC, is an essential protein required for pre-mRNA splicing. The highly conserved Cdc5 N-terminus contains two canonical Myb (myeloblastosis) repeats (R1 and R2) and a third domain (D3) that was previously classified as a Myb-like repeat. Although the N-terminus of Cdc5 is required for its function, how R1, R2, and D3 each contribute to functionality is unclear. Using a combination of yeast genetics, structural approaches, and RNA binding assays, we show that R1, R2, and D3 are all required for the function of Cdc5 in cells. We also show that the N-terminus of Cdc5 binds RNA in vitro. Structural and functional analyses of Cdc5-D3 show that, while this domain does not adopt a Myb fold, Cdc5-D3 preferentially binds double-stranded RNA. Our data suggest that the Cdc5 N-terminus interacts with RNA structures proposed to be near the catalytic core of the spliceosome.
- Published
- 2014
- Full Text
- View/download PDF
43. Kinesin-12 Kif15 targets kinetochore fibers through an intrinsic two-step mechanism.
- Author
-
Sturgill EG, Das DK, Takizawa Y, Shin Y, Collier SE, Ohi MD, Hwang W, Lang MJ, and Ohi R
- Subjects
- HeLa Cells, Humans, Immunoblotting, Kinesins metabolism, Kinetochores ultrastructure, Microscopy, Fluorescence, Microtubules ultrastructure, Mitosis, Spindle Apparatus ultrastructure, Cell Cycle, Kinesins genetics, Kinetochores metabolism, Microtubules metabolism, Spindle Apparatus metabolism
- Abstract
Proteins that recognize and act on specific subsets of microtubules (MTs) enable the varied functions of the MT cytoskeleton. We recently discovered that Kif15 localizes exclusively to kinetochore fibers (K-fibers) or bundles of kinetochore-MTs within the mitotic spindle. It is currently speculated that the MT-associated protein TPX2 loads Kif15 onto spindle MTs, but this model has not been rigorously tested. Here, we show that Kif15 accumulates on MT bundles as a consequence of two inherent biochemical properties. First, Kif15 is self-repressed by its C terminus. Second, Kif15 harbors a nonmotor MT-binding site, enabling dimeric Kif15 to crosslink and slide MTs. Two-MT binding activates Kif15, resulting in its accumulation on and motility within MT bundles but not on individual MTs. We propose that Kif15 targets K-fibers via an intrinsic two-step mechanism involving molecular unfolding and two-MT binding. This work challenges the current model of Kif15 regulation and provides the first account of a kinesin that specifically recognizes a higher-order MT array., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Kinetochore-microtubule stability governs the metaphase requirement for Eg5.
- Author
-
Gayek AS and Ohi R
- Subjects
- Cell Line, Tumor, Humans, Kinesins metabolism, Protein Stability, Spindle Apparatus metabolism, Kinesins physiology, Kinetochores metabolism, Metaphase, Microtubules metabolism
- Abstract
The mitotic spindle is a bipolar, microtubule (MT)-based cellular machine that segregates the duplicated genome into two daughter cells. The kinesin-5 Eg5 establishes the bipolar geometry of the mitotic spindle, but previous work in mammalian cells suggested that this motor is unimportant for the maintenance of spindle bipolarity. Although it is known that Kif15, a second mitotic kinesin, enforces spindle bipolarity in the absence of Eg5, how Kif15 functions in this capacity and/or whether other biochemical or physical properties of the spindle promote its bipolarity have been poorly studied. Here we report that not all human cell lines can efficiently maintain bipolarity without Eg5, despite their expressing Kif15. We show that the stability of chromosome-attached kinetochore-MTs (K-MTs) is important for bipolar spindle maintenance without Eg5. Cells that efficiently maintain bipolar spindles without Eg5 have more stable K-MTs than those that collapse without Eg5. Consistent with this observation, artificial destabilization of K-MTs promotes spindle collapse without Eg5, whereas stabilizing K-MTs improves bipolar spindle maintenance without Eg5. Our findings suggest that either rapid K-MT turnover pulls poles inward or slow K-MT turnover allows for greater resistance to inward-directed forces., (© 2014 Gayek and Ohi. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).)
- Published
- 2014
- Full Text
- View/download PDF
45. Microtubule-regulating kinesins.
- Author
-
Sturgill EG and Ohi R
- Subjects
- Eukaryota cytology, Eukaryota metabolism, Humans, Tubulin metabolism, Yeasts cytology, Yeasts metabolism, Kinesins metabolism, Microtubules metabolism
- Published
- 2013
- Full Text
- View/download PDF
46. Kinesin-12 differentially affects spindle assembly depending on its microtubule substrate.
- Author
-
Sturgill EG and Ohi R
- Subjects
- HeLa Cells, Humans, Immunoblotting, Kinesins metabolism, Kinetochores ultrastructure, Microscopy, Fluorescence, Microtubules ultrastructure, Mitosis, Spindle Apparatus ultrastructure, Cell Cycle, Kinesins genetics, Kinetochores metabolism, Microtubules metabolism, Spindle Apparatus metabolism
- Abstract
Background: During mitosis, the microtubule (MT) cytoskeleton rearranges into a bipolar spindle that drives chromosome segregation. Two kinesin subtypes, kinesin-5 and kinesin-12, help build this bipolar array by separating the spindle poles. However, unlike kinesin-5, the kinesin-12 mechanism is not well understood., Results: At physiologically normal protein levels, we demonstrate that the human kinesin-12 Kif15 acts predominantly on kinetochore fibers to regulate their lengths. This activity limits the extent to which spindle poles separate, leading to transient spindle length instabilities when the motor is absent. Using a novel cell line wherein Kif15 usurps kinesin-5 function, we further show that Kif15 can assume a commanding role in spindle pole separation as a consequence of its mislocalization to nonkinetochore MTs. This Kif15-dependent mechanism is inefficient, however, as spindles assemble through a perilous monopolar intermediate., Conclusions: By examining Kif15 activity in two cellular contexts, we found that Kif15 bound to kinetochore fibers antagonizes centrosome separation while Kif15 bound to nonkinetochore MTs mediates centrosome separation. Our work demonstrates that Kif15 acts on parallel MT arrays and clarifies its role under both normal and pathological conditions., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
47. Microtubule-depolymerizing kinesins.
- Author
-
Walczak CE, Gayek S, and Ohi R
- Subjects
- Animals, Humans, Kinesins chemistry, Molecular Motor Proteins metabolism, Phylogeny, Spindle Apparatus metabolism, Kinesins metabolism, Microtubules metabolism
- Abstract
The microtubule (MT) cytoskeleton supports a broad range of cellular functions, from providing tracks for intracellular transport, to supporting movement of cilia and flagella, to segregating chromosomes in mitosis. These functions are facilitated by the organizational and dynamic plasticity of MT networks. An important class of enzymes that alters MT dynamics is the depolymerizing kinesin-like proteins, which use their catalytic activities to regulate MT end dynamics. In this review, we discuss four topics surrounding these MT-depolymerizing kinesins. We provide a historical overview of studies focused on these motors and discuss their phylogeny. In the second half, we discuss their enzymology and biophysics and give an overview of their known cellular functions. This discussion highlights the fact that MT-depolymerizing kinesins exhibit a diverse range of design principles, which in turn increases their functional versatility in cells.
- Published
- 2013
- Full Text
- View/download PDF
48. Move in for the kill: motile microtubule regulators.
- Author
-
Su X, Ohi R, and Pellman D
- Subjects
- Animals, Biological Transport, Humans, Kinesins chemistry, Protein Binding, Kinesins metabolism, Microtubules metabolism
- Abstract
The stereotypical function of kinesin superfamily motors is to transport cargo along microtubules. However, some kinesins also shape the microtubule track by regulating microtubule assembly and disassembly. Recent work has shown that the kinesin-8 family of motors emerge as key regulators of cellular microtubule length. The studied kinesin-8s are highly processive motors that walk towards the microtubule plus-end. Once at plus-ends, they have complex effects on polymer dynamics; kinesin-8s either destabilize or stabilize microtubules, depending on the context. This review focuses on the mechanisms underlying kinesin-8-microtubule interactions and microtubule length control. We compare and contrast kinesin-8s with the other major microtubule-regulating kinesins (kinesin-4 and kinesin-13), to survey the current understanding of the diverse ways that kinesins control microtubule dynamics., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
49. Cdk1 phosphorylation of the kinetochore protein Nsk1 prevents error-prone chromosome segregation.
- Author
-
Chen JS, Lu LX, Ohi MD, Creamer KM, English C, Partridge JF, Ohi R, and Gould KL
- Subjects
- Dyneins metabolism, Microtubules metabolism, Mitosis, Phosphorylation, Saccharomyces cerevisiae cytology, Spindle Apparatus metabolism, CDC2 Protein Kinase metabolism, Cell Cycle Proteins metabolism, Chromosome Segregation, Chromosomes, Fungal metabolism, Kinetochores metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Cdk1 controls many aspects of mitotic chromosome behavior and spindle microtubule (MT) dynamics to ensure accurate chromosome segregation. In this paper, we characterize a new kinetochore substrate of fission yeast Cdk1, Nsk1, which promotes proper kinetochore-MT (k-MT) interactions and chromosome movements in a phosphoregulated manner. Cdk1 phosphorylation of Nsk1 antagonizes Nsk1 kinetochore and spindle localization during early mitosis. A nonphosphorylatable Nsk1 mutant binds prematurely to kinetochores and spindle, cementing improper k-MT attachments and leading to high rates of lagging chromosomes that missegregate. Accordingly, cells lacking nsk1 exhibit synthetic growth defects with mutations that disturb MT dynamics and/or kinetochore structure, and lack of proper phosphoregulation leads to even more severe defects. Intriguingly, Nsk1 is stabilized by binding directly to the dynein light chain Dlc1 independently of the dynein motor, and Nsk1-Dlc1 forms chainlike structures in vitro. Our findings establish new roles for Cdk1 and the Nsk1-Dlc1 complex in regulating the k-MT interface and chromosome segregation., (© 2011 Chen et al.)
- Published
- 2011
- Full Text
- View/download PDF
50. The Chlamydia effector chlamydial outer protein N (CopN) sequesters tubulin and prevents microtubule assembly.
- Author
-
Archuleta TL, Du Y, English CA, Lory S, Lesser C, Ohi MD, Ohi R, and Spiller BW
- Subjects
- Animals, Bacterial Outer Membrane Proteins chemistry, Bacterial Outer Membrane Proteins genetics, Cattle, Chlamydia chemistry, Chlamydia genetics, Chlamydia Infections genetics, Chlamydia Infections metabolism, Metaphase, Microtubules chemistry, Microtubules genetics, Spindle Apparatus genetics, Spindle Apparatus metabolism, Tubulin chemistry, Tubulin genetics, Virulence Factors chemistry, Virulence Factors genetics, Bacterial Outer Membrane Proteins metabolism, Chlamydia metabolism, Chlamydia pathogenicity, Microtubules metabolism, Tubulin metabolism, Virulence Factors metabolism
- Abstract
Chlamydia species are obligate intracellular pathogens that utilize a type three secretion system to manipulate host cell processes. Genetic manipulations are currently not possible in Chlamydia, necessitating study of effector proteins in heterologous expression systems and severely complicating efforts to relate molecular strategies used by Chlamydia to the biochemical activities of effector proteins. CopN is a chlamydial type three secretion effector that is essential for virulence. Heterologous expression of CopN in cells results in loss of microtubule spindles and metaphase plate formation and causes mitotic arrest. CopN is a multidomain protein with similarity to type three secretion system "plug" proteins from other organisms but has functionally diverged such that it also functions as an effector protein. We show that CopN binds directly to αβ-tubulin but not to microtubules (MTs). Furthermore, CopN inhibits tubulin polymerization by sequestering free αβ-tubulin, similar to one of the mechanisms utilized by stathmin. Although CopN and stathmin share no detectable sequence identity, both influence MT formation by sequestration of αβ-tubulin. CopN displaces stathmin from preformed stathmin-tubulin complexes, indicating that the proteins bind overlapping sites on tubulin. CopN is the first bacterial effector shown to disrupt MT formation directly. This recognition affords a mechanistic understanding of a strategy Chlamydia species use to manipulate the host cell cycle.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.