Elsevier

Gene

Volume 531, Issue 2, 1 December 2013, Pages 133-149
Gene

Review
Kinesin-5: Cross-bridging mechanism to targeted clinical therapy

https://doi.org/10.1016/j.gene.2013.08.004Get rights and content

Highlights

  • Kinesin-5 is primarily a mitotic microtubule motor that exhibits complex regulation.

  • Biophysical studies of the motor core alter the paradigm of kinesin catalysis.

  • Toolbox of small-molecule inhibitors opens basic and clinical research avenues.

  • Kinesin-5 inhibitors provide translational lessons about targeted patient therapies.

Abstract

Kinesin motor proteins comprise an ATPase superfamily that works hand in hand with microtubules in every eukaryote. The mitotic kinesins, by virtue of their potential therapeutic role in cancerous cells, have been a major focus of research for the past 28 years since the discovery of the canonical Kinesin-1 heavy chain. Perhaps the simplest player in mitotic spindle assembly, Kinesin-5 (also known as Kif11, Eg5, or kinesin spindle protein, KSP) is a plus-end-directed motor localized to interpolar spindle microtubules and to the spindle poles. Comprised of a homotetramer complex, its function primarily is to slide anti-parallel microtubules apart from one another. Based on multi-faceted analyses of this motor from numerous laboratories over the years, we have learned a great deal about the function of this motor at the atomic level for catalysis and as an integrated element of the cytoskeleton. These data have, in turn, informed the function of motile kinesins on the whole, as well as spearheaded integrative models of the mitotic apparatus in particular and regulation of the microtubule cytoskeleton in general. We review what is known about how this nanomotor works, its place inside the cytoskeleton of cells, and its small-molecule inhibitors that provide a toolbox for understanding motor function and for anticancer treatment in the clinic.

Introduction

Kinesins are the smallest and most abundant nanomotor in cells and the only canonical motor protein that is ubiquitous in all eukaryotes. Like dynein and myosin, these proteins hydrolyze ATP and convert chemical energy into mechanical energy. This permits transport and movement along the cytoskeletal track. Of the > 16 different kinesin isoforms (Wickstead and Gull, 2006), Kinesin-5 (BimC/Eg5/N-2/Kif11) family members were the first identified to be essential in mitosis. Genetic analysis provided the initial insight into the key mitotic role of this motor family. In the search for strains that were defective in cellular division at a restrictive temperature, screens of fungal libraries and fission yeast uncovered BimC (Enos and Morris, 1990) and Cut7 (Hagan and Yanagida, 1990), respectively, in the early 1990s. These mutants had malfunctions in spindle pole body separation and nuclear division and were unable to undergo mitosis. Cloning and sequencing of these mutants (Hagan and Yanagida, 1990, Kashina et al., 1997) revealed that the gene product encoded a 130 kDa protein with high similarity to the conventional kinesin (KHC/Kinesin-1) involved in motility in squid and mammalian brains. Similarly, simultaneous loss of function in Cin8p and Kip1p in Saccharomyces cerevisiae revealed a redundant function of these Kinesin-5 family members for spindle assembly (Hoyt et al., 1992, Roof et al., 1992).

The gene family has expanded from the early days of the molecular and bioinformatics era. Many groups (Bannigan et al., 2007, Bishop et al., 2005, Blangy et al., 1995, Chauviere et al., 2008, Heck et al., 1993, Hoyt et al., 1992, Sawin et al., 1992, Tihy et al., 1992) initially identified orthologs in Xenopus, S. cerevisiae, Drosophila melanogaster, Homo sapiens, Mus musculus, and Caenorhabditis elegans (Table 1). Currently, there are over 70 different Kinesin-5 proteins identified by sequence homology in 66 eukaryotes (Fig. 1). Subsequently classified as the Kinesin-5 family (Lawrence et al., 2004), this group of related kinesins localizes to spindle microtubules and structures present at spindle poles.

The Kif11 gene product has four domains (Fig. 2A), three of which are assigned to roles resulting in differentiation of kinesin function. An N-terminal globular motor domain performs conserved functions of binding microtubules (MTs) and nucleotide hydrolysis. Variations in sequence in the tail, interrupted coiled-coil region, and neck-linker/cover neck (Hesse et al., 2013) are thought to dictate how these motor proteins bind specific cargo, have particular oligomerization states, and control directionality of net motion of the motor domain, respectively. Unlike the canonical dimeric kinesin motors, electron microscopy showed that native Kinesin-5 proteins are bipolar homotetramers (Blangy et al., 1995, Cole et al., 1994), with their motor domains positioned at the ends of the tetramer's long axis. The arrangement of two sets of antiparallel dimers is hypothesized to result in Kinesin-5 motors crosslinking and sliding antiparallel microtubules (Fig. 2B).

The human Eg5 gene product (HsEg5) is of particular interest amongst the Kinesin-5 proteins, because of its potential as a therapeutic target for cancer treatment. It is sensitive to a battery of small-molecule inhibitors (DeBonis et al., 2004, Hotha et al., 2003, Luo et al., 2007) that allosterically block Eg5 activity (DeBonis et al., 2003, Maliga et al., 2002). The specificity of these synthetic inhibitors is exceptional: although all eukaryotic organisms examined to date contain at least one Kinesin-5 protein, not all Kinesin-5 proteins are sensitive to these compounds (DeBonis et al., 2003, Maliga et al., 2002). Of the inhibitors most frequently employed, monastrol (Mayer et al., 1999) and S-trityl-l-cysteine [STC; (DeBonis et al., 2003)], were uncovered from independent chemical screens, inclusive for mitotic inhibition and exclusive for microtubule interactions. They induced mitotic arrest in human tissue culture cells by inhibiting Eg5-dependent MT motility (Kapoor et al., 2000, Mayer et al., 1999) and resulted in a spectacular reorganization of the mitotic spindle to aberrant monoastral form with no apparent effect on interphase MT arrays (Fig. 3). Failure either in chromosome segregation or in the timing of cell division events can result in aneuploidy that is strongly linked with developmental defects and cancer (Kops et al., 2005).

The discovery of these chemical inhibitors of HsEg5 is important on two fronts. First, they can be used as tools to dissect mechanotransduction in this mitotic kinesin and provide answers to still open questions of how catalysis is used and converted into force and motion. Second, numerous small-molecule agents that solely target this human mitotic Kinesin-5 protein with high specificity are leads for anti-cancer therapy; several are in trials as clinical anti-cancer agents [for example, see (Carol et al., 2009, Kathman et al., 2007, Purcell et al., 2010)].

Section snippets

Cellular functions of Kinesin-5

Kinesin-5 motors assemble into a bipolar homotetrameric structure that is capable of modulating the dynamics and organization of eukaryotic microtubule arrays (Kashina et al., 1996). Although an essential role for this enzyme in mitosis has been the focus of considerable research effort, recent data also implicate this motor in certain processes within non-dividing cells, such as neurons. Although classical genetic analysis of Kinesin-5 family members has pioneered the investigation of the

Mechanism of Kinesin-5

Knowledge of motor mechanism, or mechanotransduction, relies heavily on two types of in vitro investigations. In the first class, chemical-kinetic measurements of molecular motor ensembles in bulk solution largely define our current understanding of the various biochemical processes in kinesin proteins. Such experiments conclude that the catalytic cycle of kinesin includes multiple conformational states coupled to a complex biochemical network. In the second class, single-molecule experiments

Translational studies

Mitosis is a validated point of intervention for cancer therapy and a variety of antimitotic drugs against cytoskeleton proteins are successfully being used in the clinic (Rath and Kozielski, 2012). There is one report that little or no HsEg5 is detectable in normal non-proliferating cells, while expression is prominent in proliferating cells (Hegde et al., 2003). In addition, over-expression of HsEg5 has been noted in a variety of human solid tumors implicating the role of the enzyme in

Conflict of interest

None.

Acknowledgments

We thank members of the Huckaba, Kim, Wojcik, and Worthylake laboratories for their intellectual input and criticism. This work was funded by grants from the National Institutes of Health (R01GM097350 to S.K.; R01GM066328 to E.W.; and P20GM103424 and 5G12RR026260 to T.H.) and the School of Graduate Studies at LSU Health Sciences Center — New Orleans (R.B. and J.R.).

References (230)

  • D.G. Cole et al.

    A slow homotetrameric kinesin-related motor protein purified from Drosophila embryos

    J. Biol. Chem.

    (1994)
  • C.D. Cox

    Kinesin spindle protein (KSP) inhibitors. Part 1: the discovery of 3,5-diaryl-4,5-dihydropyrazoles as potent and selective inhibitors of the mitotic kinesin KSP

    Bioorg. Med. Chem. Lett.

    (2005)
  • I.M.T.C. Crevel et al.

    Kinetic evidence for low chemical processivity in ncd and Eg5

    J. Mol. Biol.

    (1997)
  • I.M.-T.C. Crevel et al.

    Monastrol stabilises an attached low-friction mode of Eg5

    Curr. Biol.

    (2004)
  • A. Duselder et al.

    Neck-linker length dependence of processive Kinesin-5 motility

    J. Mol. Biol.

    (2012)
  • F. Eckerdt et al.

    Spindle pole regulation by a discrete Eg5-interacting domain in TPX2

    Curr. Biol.

    (2008)
  • H.B. El-Nassan

    Advances in the discovery of kinesin spindle protein (Eg5) inhibitors as antitumor agents

    Eur. J. Med. Chem.

    (2013)
  • A. Enos et al.

    Mutation of a gene that encodes a kinesin-like protein blocks nuclear division in A. nidulans

    Cell

    (1990)
  • N.P. Ferenz et al.

    Mitotic functions of kinesin-5

    Semin. Cell Dev. Biol.

    (2010)
  • S. Florian et al.

    The functional antagonism between Eg5 and dynein in spindle bipolarization is not compatible with a simple push–pull model

    Cell Rep.

    (2012)
  • M.E. Fraley

    Kinesin spindle protein (KSP) inhibitors. Part 2: the design, synthesis, and characterization of 2,4-diaryl-2,5-dihydropyrrole inhibitors of the mitotic kinesin KSP

    Bioorg. Med. Chem. Lett.

    (2006)
  • R.M. Garbaccio

    Kinesin spindle protein (KSP) inhibitors. Part 3: synthesis and evaluation of phenolic 2,4-diaryl-2,5-dihydropyrroles with reduced hERG binding and employment of a phosphate prodrug strategy for aqueous solubility

    Bioorg. Med. Chem. Lett.

    (2006)
  • K. Garcia et al.

    Tyrosines in the kinesin-5 head domain are necessary for phosphorylation by Wee1 and for mitotic spindle integrity

    Curr. Biol.

    (2009)
  • M.K. Gardner

    Chromosome congression by Kinesin-5 motor-mediated disassembly of longer kinetochore microtubules

    Cell

    (2008)
  • R. Giet et al.

    The Xenopus laevis aurora-related protein kinase pEg2 associates with and phosphorylates the kinesin-related protein XlEg5

    J. Biol. Chem.

    (1999)
  • G. Goshima et al.

    Length control of the metaphase spindle

    Curr. Biol.

    (2005)
  • A. Goulet et al.

    The structural basis of force generation by the mitotic motor kinesin-5

    J. Biol. Chem.

    (2012)
  • W.R. Hesse et al.

    Modular aspects of kinesin force generation machinery

    Biophys. J.

    (2013)
  • E.R. Hildebrandt et al.

    Homotetrameric form of Cin8p, a Saccharomyces cerevisiae kinesin-5 motor, is essential for its in vivo function

    J. Biol. Chem.

    (2006)
  • K. Hirose et al.

    Large conformational changes in a kinesin motor catalyzed by interaction with microtubules

    Mol. Cell

    (2006)
  • W. Hwang et al.

    Force generation in kinesin hinges on cover-neck bundle formation

    Structure

    (2008)
  • B. Jun et al.

    Real-time structural transitions are coupled to chemical steps in ATP hydrolysis by Eg5 kinesin

    J. Biol. Chem.

    (2010)
  • A.S. Kashina et al.

    The bimC family of kinesins: essential bipolar mitotic motors driving centrosome separation

    Biochim. Biophys. Acta

    (1997)
  • A.B. Asenjo et al.

    Configuration of the two kinesin motor domains during ATP hydrolysis

    Nat. Struct. Biol.

    (2003)
  • S. Atherton-Fessler et al.

    Mechanisms of p34cdc2 regulation

    Mol. Cell. Biol.

    (1993)
  • R. Avunie-Masala

    Phospho-regulation of kinesin-5 during anaphase spindle elongation

    J. Cell Sci.

    (2011)
  • A. Bannigan

    A conserved role for kinesin-5 in plant mitosis

    J. Cell Sci.

    (2007)
  • J.D. Bishop et al.

    The Caenorhabditis elegans Aurora B kinase AIR-2 phosphorylates and is required for the localization of a BimC kinesin to meiotic and mitotic spindles

    Mol. Biol. Cell

    (2005)
  • S.P. Blagden

    A phase I trial of ispinesib, a kinesin spindle protein inhibitor, with docetaxel in patients with advanced solid tumours

    Br. J. Cancer

    (2008)
  • S.M. Block et al.

    Bead movement by single kinesin molecules studied with optical tweezers

    Nature

    (1990)
  • S.M. Block et al.

    Probing the kinesin reaction cycle with a 2D optical force clamp

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • B. Bodenmiller

    An integrated chemical, mass spectrometric and computational strategy for (quantitative) phosphoproteomics: application to Drosophila melanogaster Kc167 cells

    Mol. Biosyst.

    (2007)
  • I. Brust-Mascher et al.

    Kinesin-5-dependent poleward flux and spindle length control in Drosophila embryo mitosis

    Mol. Biol. Cell

    (2009)
  • H.A. Burris

    A phase I study of ispinesib, a kinesin spindle protein inhibitor, administered weekly for three consecutive weeks of a 28-day cycle in patients with solid tumors

    Invest. New Drugs

    (2011)
  • J. Cahu

    Phosphorylation by Cdk1 increases the binding of Eg5 to microtubules in vitro and in Xenopus egg extract spindles

    PLoS One

    (2008)
  • H. Carol

    Initial testing (stage 1) of the kinesin spindle protein inhibitor ispinesib by the pediatric preclinical testing program

    Pediatr. Blood Cancer

    (2009)
  • A. Castillo et al.

    Overexpression of Eg5 causes genomic instability and tumor formation in mice

    Cancer Res.

    (2007)
  • K.S. Chan et al.

    Mitosis-targeted anti-cancer therapies: where they stand

    Cell Death Dis.

    (2012)
  • M.K. Chee et al.

    B-cyclin/CDKs regulate mitotic spindle assembly by phosphorylating kinesins-5 in budding yeast

    PLoS Genet.

    (2010)
  • D.K. Cheerambathur et al.

    Dynamic partitioning of mitotic kinesin-5 cross-linkers between microtubule-bound and freely diffusing states

    J. Cell Biol.

    (2008)
  • Cited by (54)

    • The classification and therapeutic applications of molecular motors

      2021, European Journal of Medicinal Chemistry Reports
    • Various effects of two types of kinesin-5 inhibitors on mitosis and cell proliferation

      2021, Biochemical Pharmacology
      Citation Excerpt :

      Kinesin-5 (encoded by the Kif11 gene, also called Eg5 or kinesin spindle protein) is a microtubule-associated and ATP-dependent motor protein [1,2].

    • Protein function | Allostery in proteins: Canonical models and new insights

      2021, Encyclopedia of Biological Chemistry: Third Edition
    View all citing articles on Scopus
    View full text