Cargando…

Histone variant H3F3A promotes lung cancer cell migration through intronic regulation

Although several somatic single nucleotide variations in histone H3.3 have been investigated as cancer drivers, other types of aberration have not been well studied. Here, we demonstrate that overexpression of H3F3A, encoding H3.3, is associated with lung cancer progression and promotes lung cancer...

Descripción completa

Detalles Bibliográficos
Autores principales: Park, Seong-Min, Choi, Eun-Young, Bae, Mingyun, Kim, Sunshin, Park, Jong Bae, Yoo, Heon, Choi, Jung Kyoon, Kim, Youn-Jae, Lee, Seung-Hoon, Kim, In-Hoo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5477500/
https://www.ncbi.nlm.nih.gov/pubmed/27694942
http://dx.doi.org/10.1038/ncomms12914
Descripción
Sumario:Although several somatic single nucleotide variations in histone H3.3 have been investigated as cancer drivers, other types of aberration have not been well studied. Here, we demonstrate that overexpression of H3F3A, encoding H3.3, is associated with lung cancer progression and promotes lung cancer cell migration by activating metastasis-related genes. H3.3 globally activates gene expression through the occupation of intronic regions in lung cancer cells. Moreover, H3.3 binding regions show characteristics of regulatory DNA elements. We show that H3.3 is deposited at a specific intronic region of GPR87, where it modifies the chromatin status and directly activates GPR87 transcription. The expression levels of H3F3A and GPR87, either alone or in combination, are robust prognostic markers for early-stage lung cancer, and may indicate potential for the development of treatments involving GPR87 antagonists. In summary, our results demonstrate that intronic regulation by H3F3A may be a target for the development of novel therapeutic strategies.