RRC ID 63709
著者 Yuan G, Flores NM, Hausmann S, Lofgren SM, Kharchenko V, Angulo-Ibanez M, Sengupta D, Lu X, Czaban I, Azhibek D, Vicent S, Fischle W, Jaremko M, Fang B, Wistuba II, Chua KF, Roth JA, Minna JD, Shao NY, Jaremko Ł, Mazur PK, Gozani O.
タイトル Elevated NSD3 histone methylation activity drives squamous cell lung cancer.
ジャーナル Nature
Abstract Amplification of chromosomal region 8p11-12 is a common genetic alteration that has been implicated in the aetiology of lung squamous cell carcinoma (LUSC)1-3. The FGFR1 gene is the main candidate driver of tumorigenesis within this region4. However, clinical trials evaluating FGFR1 inhibition as a targeted therapy have been unsuccessful5. Here we identify the histone H3 lysine 36 (H3K36) methyltransferase NSD3, the gene for which is located in the 8p11-12 amplicon, as a key regulator of LUSC tumorigenesis. In contrast to other 8p11-12 candidate LUSC drivers, increased expression of NSD3 correlated strongly with its gene amplification. Ablation of NSD3, but not of FGFR1, attenuated tumour growth and extended survival in a mouse model of LUSC. We identify an LUSC-associated variant NSD3(T1232A) that shows increased catalytic activity for dimethylation of H3K36 (H3K36me2) in vitro and in vivo. Structural dynamic analyses revealed that the T1232A substitution elicited localized mobility changes throughout the catalytic domain of NSD3 to relieve auto-inhibition and to increase accessibility of the H3 substrate. Expression of NSD3(T1232A) in vivo accelerated tumorigenesis and decreased overall survival in mouse models of LUSC. Pathological generation of H3K36me2 by NSD3(T1232A) reprograms the chromatin landscape to promote oncogenic gene expression signatures. Furthermore, NSD3, in a manner dependent on its catalytic activity, promoted transformation in human tracheobronchial cells and growth of xenografted human LUSC cell lines with amplification of 8p11-12. Depletion of NSD3 in patient-derived xenografts from primary LUSCs containing NSD3 amplification or the NSD3(T1232A)-encoding variant attenuated neoplastic growth in mice. Finally, NSD3-regulated LUSC-derived xenografts were hypersensitive to bromodomain inhibition. Thus, our work identifies NSD3 as a principal 8p11-12 amplicon-associated oncogenic driver in LUSC, and suggests that NSD3-dependency renders LUSC therapeutically vulnerable to bromodomain inhibition.
巻・号 590(7846)
ページ 504-508
公開日 2021-2-1
DOI 10.1038/s41586-020-03170-y
PII 10.1038/s41586-020-03170-y
PMID 33536620
PMC PMC7895461
MeSH Animals Biocatalysis Carcinogenesis / genetics Carcinoma, Squamous Cell / genetics Carcinoma, Squamous Cell / metabolism* Carcinoma, Squamous Cell / pathology* Female Histone-Lysine N-Methyltransferase / deficiency Histone-Lysine N-Methyltransferase / genetics Histone-Lysine N-Methyltransferase / metabolism* Histones / chemistry* Histones / metabolism* Humans Lung Neoplasms / genetics Lung Neoplasms / metabolism* Lung Neoplasms / pathology* Male Methylation Mice Models, Molecular Mutation Nuclear Proteins / deficiency Nuclear Proteins / genetics Nuclear Proteins / metabolism* Receptor, Fibroblast Growth Factor, Type 1 / deficiency Receptor, Fibroblast Growth Factor, Type 1 / genetics Receptor, Fibroblast Growth Factor, Type 1 / metabolism Xenograft Model Antitumor Assays
IF 42.779
リソース情報
ヒト・動物細胞 EBC-1(RCB1965) LK-2(RCB1970)