透過您的圖書館登入
IP:18.118.25.174
  • 學位論文

人類組蛋白去甲基酵素KDM4B之天然抑制劑開發

Structure-based discovery of KDM4B inhibitors from natural compounds

指導教授 : 王雯靜
若您是本文的作者,可授權文章由華藝線上圖書館中協助推廣。

摘要


關遺傳學是在不改變DNA序列下,進一步調控基因的表現。包含DNA的甲基化和組蛋白後轉錄修飾。其中一種組蛋白修飾為組蛋白甲基和去甲基化,被甲基轉移酵素和組蛋白去甲基酵素調控。在許多種癌症中發現,甲基轉移酵素和組蛋白去甲基酵素的作用已經失調。主要的組蛋白去甲基酵素都包含一段Jumonji-C domain,同時利用2-oxoglutarate和二價鐵離子進行去甲基的功能。KDM4B是其中一種的去甲基酵素,作用於H3K9me3/me2以及H3K36me3/me2,另一方面KDM4B在許多種癌症中的表現量都偏高,例如:胃癌,腎臟癌,直腸癌,乳癌和前列腺癌。其中在前列腺癌上,KDM4B和AR receptor共同作用促進細胞增長因此我們假設KDM4B可以作為一個治療癌症的目標蛋白。在這篇論文中,我們希望可以探討可以抑制KDM4B活性的天然抑制劑。在蛋白質和分子結構的研究下,我們發現一個天然物M2可以抑制KDM4B(IC50:12.2 μM,Ki:2.72μM),同時會和反應物有一個競爭的關係。另一方面,M2也可以抑制其他KDM4的去甲基酵素。在細胞的實驗中,M2會在CWR22Rv1,LNCaP,LNCaP C4-2B和PC-3的細胞上,產生毒性,同時也發現KDM4B的主要反應物H3K9me3在細胞的表現量也提升。最後總結我們的實驗,認為M2可以被視為可以發展為抗癌藥物的方向,未來我們將開發M2的衍生物,進一步提升抑制性和靈敏度。

並列摘要


Epigenetics is the study of transcriptional and physiological trait variations that do not involve changes in the DNA sequence. DNA methylation and post-translational modification in the histone tail are major mechanisms that produce such changes. One of histone modifications, methylation/demethylation, is controlled by histone methyltransferase and histone demethylase has been recently studied. Dysregulation of histone methylation/demethylation is found to be associated with various types of cancer. The major type of lysine demethylase (KDM) consists of Jumonji-C (JmjC) domain (KDM2‒KDM8) and requires 2-oxoglutarate and Fe(II) as cofactors to remove the methyl moiety from the histone tail. Of those, KDM4B, a member of the KDM4 family can specifically remove the methyl group from H3K9me3/me2 and H3K36me3/me2. KDM4B are overexpressed in many types of cancer including gastric cancer, renal cancer, colon cancer, breast cancer and prostate cancer. In prostate cancer, KDM4B is a co-activator of androgen receptor (AR) to stimulate cell growth. Given the important role of AR in prostate carcinogenesis, we propose KDM4B as a useful target protein. In this study, we aim to discover natural compounds that inhibit KDM4B based on a structure-guided approach. We have successfully identified a compound M2 with IC50 = 12.2 ± 1.6 μM, Ki= 2.72 μM. M2 exhibits a competitive inhibition mode toward KDM4B. M2 also shows inhibition against other members of the KDM4 family but not against PHF8. In cell-based experiment, M2 had cytoxicity in AR-dependent (LNCaP) as well as AR-independent prostate cancer cells (CWR22Rv1, LNCaP C4-2B and PC-3). Addition of M2 in LNCaP and CWR22Rv1 cells led to the increase of H3K9m3 signal. Together, M2 is a specific KDM4 inhibitor and shows anti-prostate cancer effect. Future work is to derive M2-based compounds with higher specificity and potency.

參考文獻


Bannister, A. J. and T. Kouzarides. "Regulation of Chromatin by Histone Modifications." Cell Res 21, no. 3 (2011): 381-95.
Bassett, S. A. and M. P. Barnett. "The Role of Dietary Histone Deacetylases (Hdacs) Inhibitors in Health and Disease." Nutrients 6, no. 10 (2014): 4273-301.
Berger, S. L., T. Kouzarides, R. Shiekhattar and A. Shilatifard. "An Operational Definition of Epigenetics." Genes Dev 23, no. 7 (2009): 781-3.
Beyer, S., M. M. Kristensen, K. S. Jensen, J. V. Johansen and P. Staller. "The Histone Demethylases Jmjd1a and Jmjd2b Are Transcriptional Targets of Hypoxia-Inducible Factor Hif." J Biol Chem 283, no. 52 (2008): 36542-52.
Bhasin, M., E. L. Reinherz and P. A. Reche. "Recognition and Classification of Histones Using Support Vector Machine." J Comput Biol 13, no. 1 (2006): 102-12.

延伸閱讀