Precise cell division is essential for life. Mistakes during this process lead to many diseases, including cancer. The mitotic spindle is the engine that moves chromosomes, and it therefore plays an essential role in ensuring that newly-born cells inherit a complete genetic blueprint. A key feature of the spindle is its bipolarity, a geometry that ensures bi-directional chromosome movements. Non-bipolar geometries, such as monopolarity or multipolarity, cause errors in chromosome segregation that are incompatible with life. Therefore, non-bipolar structures must be reorganized to be bipolar prior to the onset of the anaphase stage of mitosis. Spindle formation is a complex process that is mediated by many microtubule-associated proteins (MAPs) and molecular motors (i.e., kinesins and dynein). The mechanisms of spindle assembly can vary among organisms, and can show remarkable plasticity even within a single organism. The malleability of spindle assembly is derived from how MAPs and motor proteins engage each other, either directly or indirectly through a network of dynamic MTs. The sophisticated nature of these systems-level relationships has made it difficult to fully understand the mechanisms that drive spindle formation, despite decades of research. Our unique approach has been to isolate and characterize human cell lines that survive in the absence of the major spindle assembly pathway driven by the kinesin Eg5 in eukaryotes. Our work has taught us that human cells can assemble a proper mitotic spindle using an auxiliary pathway. Furthermore, we identified a kinesin (Kif15) that is essential for this alternate mechanism. Ongoing work in our lab has unveiled systems-level changes in cells that require Kif15 for cell division, motivating efforts to obtain a better understanding of how spindle motors function at a systems level within the spindle. In this renewal application, we will address three key questions: 1) How does Kif15 drive spindle assembly?; 2) How is Kif15 activity regulated during cell division?; and 3) What roles do motors and chromosomes play in centrosome clustering? This work will advance our understanding of spindle mechanics and have immediate relevance to the development of anti-mitotic chemotherapeutic strategies.

Public Health Relevance

Regulation of microtubule dynamics and organization during cell division Ryoma Ohi, Ph.D. PROJECT NARRATIVE Bipolarity is an essential feature of the mitotic spindle, a microtubule-based machine that segregates a replicated set of chromosomes equally among two daughter cells during mitosis. Using chemical genetics, we discovered a new spindle assembly pathway, the protein factor (Kif15) that drives it, tools to study how the new spindle assembly pathway works, and relevance of Kif15 to other aspects of mitosis. In this renewal application, we will address three key questions: 1) How does Kif15 drive spindle assembly?; 2) How is Kif15 activity regulated during cell division?; and 3) What roles do motors and chromosomes play in centrosome clustering?

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
2R01GM086610-11A1
Application #
10118298
Study Section
Nuclear and Cytoplasmic Structure/Function and Dynamics Study Section (NCSD)
Program Officer
Gindhart, Joseph G
Project Start
2010-05-01
Project End
2024-08-31
Budget Start
2020-09-14
Budget End
2021-08-31
Support Year
11
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of Michigan Ann Arbor
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Chiang, Yun-Chen; Park, In-Young; Terzo, Esteban A et al. (2018) SETD2 Haploinsufficiency for Microtubule Methylation Is an Early Driver of Genomic Instability in Renal Cell Carcinoma. Cancer Res 78:3135-3146
Landino, Jennifer; Norris, Stephen R; Li, Muyi et al. (2017) Two mechanisms coordinate the recruitment of the chromosomal passenger complex to the plane of cell division. Mol Biol Cell 28:3634-3646
Reinemann, Dana N; Sturgill, Emma G; Das, Dibyendu Kumar et al. (2017) Collective Force Regulation in Anti-parallel Microtubule Gliding by Dimeric Kif15 Kinesin Motors. Curr Biol 27:2810-2820.e6
Chen, Geng-Yuan; Kang, You Jung; Gayek, A Sophia et al. (2017) Eg5 Inhibitors Have Contrasting Effects on Microtubule Stability and Metaphase Spindle Integrity. ACS Chem Biol 12:1038-1046
Pfaltzgraff, Elise R; Roth, Gretchen M; Miller, Paul M et al. (2016) Loss of CENP-F results in distinct microtubule-related defects without chromosomal abnormalities. Mol Biol Cell 27:1990-9
Sturgill, Emma G; Norris, Stephen R; Guo, Yan et al. (2016) Kinesin-5 inhibitor resistance is driven by kinesin-12. J Cell Biol 213:213-27
Landino, Jennifer; Ohi, Ryoma (2016) The Timing of Midzone Stabilization during Cytokinesis Depends on Myosin II Activity and an Interaction between INCENP and Actin. Curr Biol 26:698-706
Shin, Yongdae; Du, Yaqing; Collier, Scott E et al. (2015) Biased Brownian motion as a mechanism to facilitate nanometer-scale exploration of the microtubule plus end by a kinesin-8. Proc Natl Acad Sci U S A 112:E3826-35
Sturgill, Emma G; Das, Dibyendu Kumar; Takizawa, Yoshimasa et al. (2014) Kinesin-12 Kif15 targets kinetochore fibers through an intrinsic two-step mechanism. Curr Biol 24:2307-13
Gayek, A Sophia; Ohi, Ryoma (2014) Kinetochore-microtubule stability governs the metaphase requirement for Eg5. Mol Biol Cell 25:2051-60

Showing the most recent 10 out of 18 publications