Microrchidia 3 (MORC3) has been linked to cancer-associated inflammations. Recent studies from our group demonstrate that MORC3 is deregulated in patients with Down syndrome (DS), however the precise role of MORC3 in disease or in normal cellular processes remains poorly understood. Our preliminary data show that MORC3 has an intrinsic ATPase activity and that the catalytic ATPase domain binds to DNA, whereas the adjacent CW domain recognizes histone H3 and mediates the ATPase activity of MORC3. The molecular mechanisms underlying these novel functions of MORC3 are unclear and will be elucidated in the proposed studies. We hypothesize that the DNA-stimulated ATPase activity of MORC3 is mediated through histone-binding activity of the CW domain, which in the MORC3 inactive state, binds to and autoinhibits the ATPase domain; and that PTMs of histone H3 alter binding of CW and fine-tune the ATPase activity of MORC3.
We aim to define the molecular basis and functional significance of the multivalent engagement of MORC3 with chromatin and to understand the mechanism of its autoregulation. By establishing the biological role and obtaining the mechanistic details, we will learn how this fundamental chromatin remodeling component can be controlled. In this research we will employ a powerful combination of in vitro and in vivo approaches to examine a newly identified epigenetic regulatory mechanism. We integrate X-ray crystallographic, advanced Frster Resonance Energy Transfer, NMR spectroscopic and high- throughput technological experiments with molecular and cell biology tools to gain insight into the role of the ATPase and CW domains in biological activities of MORC3. In-depth structural, biochemical and functional characterization of MORC3 will have significant impact on chromatin biology and basic molecular biology of chromatin-remodeling enzymes. It will also aid to our understanding of the epigenetic-driven signaling events that are deregulated in DS and other human diseases.

Public Health Relevance

Human MORC3 is found deregulated in Down syndrome, inflammation and cancer. The proposed studies will lead to a better understanding of how the MORC3 signaling pathways can be therapeutically manipulated and may help to develop new strategies to prevent or treat these diseases.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
1R01GM125195-01
Application #
9397841
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Barski, Oleg
Project Start
2017-09-01
Project End
2021-06-30
Budget Start
2017-09-01
Budget End
2018-06-30
Support Year
1
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Colorado Denver
Department
Pharmacology
Type
Schools of Medicine
DUNS #
041096314
City
Aurora
State
CO
Country
United States
Zip Code
80045
Zhang, Yi; Xue, Yongming; Shi, Jiejun et al. (2018) The ZZ domain of p300 mediates specificity of the adjacent HAT domain for histone H3. Nat Struct Mol Biol 25:841-849
Mi, Wenyi; Zhang, Yi; Lyu, Jie et al. (2018) The ZZ-type zinc finger of ZZZ3 modulates the ATAC complex-mediated histone acetylation and gene activation. Nat Commun 9:3759
Vann, Kendra R; Kutateladze, Tatiana G (2018) Architecture of PRC2 Holo Complexes. Trends Biochem Sci 43:487-489
Klein, Brianna J; Vann, Kendra R; Andrews, Forest H et al. (2018) Structural insights into the ?-?-? stacking mechanism and DNA-binding activity of the YEATS domain. Nat Commun 9:4574
Klein, Brianna J; Ahmad, Salar; Vann, Kendra R et al. (2018) Yaf9 subunit of the NuA4 and SWR1 complexes targets histone H3K27ac through its YEATS domain. Nucleic Acids Res 46:421-430
Tencer, Adam H; Cox, Khan L; Di, Luo et al. (2017) Covalent Modifications of Histone H3K9 Promote Binding of CHD3. Cell Rep 21:455-466
Gatchalian, Jovylyn; Wang, Xiaodong; Ikebe, Jinzen et al. (2017) Accessibility of the histone H3 tail in the nucleosome for binding of paired readers. Nat Commun 8:1489