دورية أكاديمية

Autochthonous tumors driven by Rb1 loss have an ongoing requirement for the RBP2 histone demethylase.

التفاصيل البيبلوغرافية
العنوان: Autochthonous tumors driven by Rb1 loss have an ongoing requirement for the RBP2 histone demethylase.
المؤلفون: McBrayer, Samuel K., Olenchock, Benjamin A., DiNatale, Gabriel J., Shi, Diana D., Khanal, Januka, Jennings, Rebecca B., Novak, Jesse S., Oser, Matthew G., Robbins, Alissa K., Modiste, Rebecca, Bonal, Dennis, Moslehi, Javid, Bronson, Roderick T., Neuberg, Donna, Quang-De Nguyen, Signoretti, Sabina, Losman, Julie-Aurore, Kaelin Jr., William G.
المصدر: Proceedings of the National Academy of Sciences of the United States of America; 4/17/2018, Vol. 115 Issue 16, pE3741-E3748, 8p
مصطلحات موضوعية: HISTONE demethylases, RETINOBLASTOMA gene, CELLULAR aging, TUMOR growth, CELL culture
مستخلص: Inactivation of the retinoblastoma gene (RBI) product, pRB, is common in many human cancers. Targeting downstream effectors of pRB that are central to tumorigenesis is a promising strategy to block the growth of tumors harboring loss-of-function RBI mutations. One such effector is retinoblastoma-binding protein 2 (RBP2, also called JARID1A or KDM5A), which encodes an H3K4 demethylase. Binding of pRB to RBP2 has been linked to the ability of pRB to promote senescence and differentiation. Importantly, genetic ablation of RBP2 is sufficient to phenocopy pRB's ability to induce these cellular changes in cell culture experiments. Moreover, germline Rbp2 deletion significantly impedes tumorigenesis in Rb1+/- mice. The value of RBP2 as a therapeutic target in cancer, however, hinges on whether loss of RBP2 could block the growth of established tumors as opposed to simply delaying their onset. Here we show that conditional, systemic ablation of RBP2 in tumor-bearing Rb1+/- mice is sufficient to slow tumor growth and significantly extend survival without causing obvious toxicity to the host. These findings show that established Rb1-null tumors require RBP2 for growth and further credential RBP2 as a therapeutic target in human cancers driven by RBI inactivation. [ABSTRACT FROM AUTHOR]
Copyright of Proceedings of the National Academy of Sciences of the United States of America is the property of National Academy of Sciences and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
قاعدة البيانات: Complementary Index
الوصف
تدمد:00278424
DOI:10.1073/pnas.1716029115