The goal of this proposal is to define the nature of the kinetochore-DNA interface and determine how centromere-specific chromatin directs the assembly of a functional kinetochore structure in human cells. During each cell division, the entire complement of genetic material must be accurately partitioned to the daughter cells. Even a single chromosome mis-segregation event can be catastrophic, resulting in the loss or gain of hundreds of genes, with severe consequences for development and disease, including tumorigenesis. The central player in directing chromosome segregation is the kinetochore, a large macromolecular structure that mediates attachments between spindle microtubules and a region of each chromosome termed the centromere. Determining the molecular basis for kinetochore function is crucial to understand the defective processes that can give rise to tumor cells, and to evaluate the best targets for the diagnosis and treatment of disease. In vertebrates, centromeres are specified by sequence-independent epigenetic mechanisms that involve the targeted deposition of nucleosomes containing the histone H3-variant, CENP-A. A fundamental unanswered question is how the remainder of the kinetochore is assembled downstream of CENP-A. Our previous work demonstrated that CENP-A is not sufficient to direct assembly of a complete, functional kinetochore structure in human cells. We have identified a heterotetrameric complex comprised of the histone fold proteins CENP-T, -W, -S, and -X as a critical additional component of the kinetochore-DNA interface. The CENP-T-W-S-X complex displays structural similarity to a nucleosome and possesses sequence-independent DNA binding activity. In addition, CENP-T interacts directly with outer kinetochore microtubule-binding proteins to direct kinetochore assembly. Thus, the CENP-T-W-S-X complex plays a pivotal role in connecting the DNA and microtubule interfaces at kinetochores. However, it remains unknown how the CENP-T-W-S-X complex is targeted exclusively to centromeres to generate a single, functional microtubule attachment site on each chromosome. This proposed work will define the mechanisms that direct the centromere localization of the CENP-T-W-S-X complex by assessing: 1) The intrinsic sequence features of the CENP-T-W-S-X complex that are required for its localization, 2) The extrinsic factors that associate with the CENP-T-W-S-X complex to deposit or maintain it at centromeres, and 3) The regulatory modifications and chromatin features that control CENP-T-W-S-X complex localization.

Public Health Relevance

The failure to form proper physical connections with chromosomes during mitosis can result in errors in chromosome numbers or cause chromosome rearrangements. In both cases, these defects are thought to contribute to tumor progression. Understanding the means by which these units of DNA, and the genetic information that they contain, are properly distributed to new cells is critical for the diagnosis nd treatment of cancer. This proposed work will analyze the mechanisms that specify the proper association of cell division structures (termed kinetochores) with chromosomal DNA to direct and control chromosome segregation in human cells.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM108718-03
Application #
9315900
Study Section
Nuclear and Cytoplasmic Structure/Function and Dynamics Study Section (NCSD)
Program Officer
Deatherage, James F
Project Start
2015-08-01
Project End
2018-07-31
Budget Start
2017-08-01
Budget End
2018-07-31
Support Year
3
Fiscal Year
2017
Total Cost
Indirect Cost
Name
Whitehead Institute for Biomedical Research
Department
Type
DUNS #
120989983
City
Cambridge
State
MA
Country
United States
Zip Code
02142
Rodriguez-Rodriguez, Jose-Antonio; Lewis, Clare; McKinley, Kara L et al. (2018) Distinct Roles of RZZ and Bub1-KNL1 in Mitotic Checkpoint Signaling and Kinetochore Expansion. Curr Biol 28:3422-3429.e5
Omer Javed, Attya; Li, Yun; Muffat, Julien et al. (2018) Microcephaly Modeling of Kinetochore Mutation Reveals a Brain-Specific Phenotype. Cell Rep 25:368-382.e5
Monda, Julie K; Cheeseman, Iain M (2018) Nde1 promotes diverse dynein functions through differential interactions and exhibits an isoform-specific proteasome association. Mol Biol Cell 29:2336-2345
Monda, Julie K; Cheeseman, Iain M (2018) The kinetochore-microtubule interface at a glance. J Cell Sci 131:
Su, Kuan-Chung; Tsang, Mary-Jane; Emans, Neil et al. (2018) CRISPR/Cas9-based gene targeting using synthetic guide RNAs enables robust cell biological analyses. Mol Biol Cell 29:2370-2377
McKinley, Kara L; Cheeseman, Iain M (2017) Large-Scale Analysis of CRISPR/Cas9 Cell-Cycle Knockouts Reveals the Diversity of p53-Dependent Responses to Cell-Cycle Defects. Dev Cell 40:405-420.e2
Guo, Lucie Y; Allu, Praveen Kumar; Zandarashvili, Levani et al. (2017) Centromeres are maintained by fastening CENP-A to DNA and directing an arginine anchor-dependent nucleosome transition. Nat Commun 8:15775
McKinley, Kara L; Sekulic, Nikolina; Guo, Lucie Y et al. (2015) The CENP-L-N Complex Forms a Critical Node in an Integrated Meshwork of Interactions at the Centromere-Kinetochore Interface. Mol Cell 60:886-98