Chromatin structure is highly dynamic and can be modulated through a number of different mechanisms, including nucleosome remodeling, histone post-translational modifications and, relevant to this proposal, the nucleosomal incorporation of histone variants. This proposal focuses on the macroH2A histone variants, which are primarily associated with condensed chromatin and inactive genes, and act as tumor suppressors across a spectrum of cancers (Vardabasso et al., 2014). We showed that macroH2A variants inhibit tumor growth and metastatic potential of melanoma cells (Kapoor et al., 2010). Here we will use a unique macroH2A double knockout (dKO) mouse model (Pehrson et al., 2014) to study cancer initiation and progression in the absence of macroH2A variants in vivo. We previously used these mice to demonstrate that macroH2A isoforms present a barrier to the reprogramming process of somatic cells towards a pluripotent state (Gaspar-Maia et al., 2013). However, macroH2A-deficient animals have yet to be crossed with any genetically engineered mouse models of cancer. Owing to the importance of macroH2A in suppressing melanoma progression and our unpublished studies showing a defect in mammary gland differentiation coupled to increased stem cell activity, we hereby propose to model melanoma and breast cancer in the context of a living organism.
In Aim 1, we will investigate the role of macroH2A in suppressing melanoma pathogenesis by crossing macroH2A dKO mice to an established melanoma mouse model (Tyr-Cre/BRAFV600E/Pten-flox) (Dankort et al., 2009). We will investigate whether loss of macroH2A isoforms promotes tumor growth and/or metastasis in the induced tumors. These analyses will be coupled to xenograft models, as well as genetically defined melanocytes and melanoma cell lines in vitro for probing the underlying molecular mechanisms of macroH2A tumor suppression via genomic approaches.
In Aim 2, we will challenge the macroH2A-deficient mice by crossing them to an established mammary tumor model (MMTV- NeuY1227D). Given the mammary gland phenotype we identified in the dKO mice, as well as the loss of macroH2A isoforms in human breast cancer specimens, we hypothesize that in the presence of mammary driver mutations, macroH2A dKO mice will develop more proliferative and invasive cancers. These studies will also be coupled to genomics approaches. In both aims, we will determine which macroH2A isoforms, domains and modified residues allow inhibition of tumor development by reconstituting macroH2A in mouse and human cells devoid of this histone variant. This unique mouse colony provides us with exciting opportunities for studying macroH2A histone variant regulation in development and cancer, allowing for a deeper understanding of human disease.

Public Health Relevance

Emerging evidence points towards a role for histone variants in contributing to tumor progression. We propose to dissect the role of macroH2A variants in melanoma progression and mammary carcinoma development in vivo using novel mouse models. Our studies will elucidate the epigenetic and transcriptional programs regulated by macroH2A that support its tumor suppressive functions.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA154683-09
Application #
9924501
Study Section
Cancer Genetics Study Section (CG)
Program Officer
Okano, Paul
Project Start
2017-06-16
Project End
2022-05-31
Budget Start
2020-06-01
Budget End
2021-05-31
Support Year
9
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Icahn School of Medicine at Mount Sinai
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
078861598
City
New York
State
NY
Country
United States
Zip Code
10029
Sun, Zhen; Filipescu, Dan; Andrade, Joshua et al. (2018) Transcription-associated histone pruning demarcates macroH2A chromatin domains. Nat Struct Mol Biol 25:958-970
Scaglione, Antonella; Patzig, Julia; Liang, Jialiang et al. (2018) PRMT5-mediated regulation of developmental myelination. Nat Commun 9:2840
Fontanals-Cirera, Barbara; Hasson, Dan; Vardabasso, Chiara et al. (2017) Harnessing BET Inhibitor Sensitivity Reveals AMIGO2 as a Melanoma Survival Gene. Mol Cell 68:731-744.e9
Pünzeler, Sebastian; Link, Stephanie; Wagner, Gabriele et al. (2017) Multivalent binding of PWWP2A to H2A.Z regulates mitosis and neural crest differentiation. EMBO J 36:2263-2279
Badal, Brateil; Solovyov, Alexander; Di Cecilia, Serena et al. (2017) Transcriptional dissection of melanoma identifies a high-risk subtype underlying TP53 family genes and epigenome deregulation. JCI Insight 2:
Valle-García, David; Qadeer, Zulekha A; McHugh, Domhnall S et al. (2016) ATRX binds to atypical chromatin domains at the 3' exons of zinc finger genes to preserve H3K9me3 enrichment. Epigenetics 11:398-414
Vardabasso, Chiara; Gaspar-Maia, Alexandre; Hasson, Dan et al. (2015) Histone Variant H2A.Z.2 Mediates Proliferation and Drug Sensitivity of Malignant Melanoma. Mol Cell 59:75-88
Vardabasso, Chiara; Hasson, Dan; Ratnakumar, Kajan et al. (2014) Histone variants: emerging players in cancer biology. Cell Mol Life Sci 71:379-404
Qadeer, Zulekha A; Harcharik, Sara; Valle-Garcia, David et al. (2014) Decreased expression of the chromatin remodeler ATRX associates with melanoma progression. J Invest Dermatol 134:1768-1772
Duarte, Luis F; Young, Andrew R J; Wang, Zichen et al. (2014) Histone H3.3 and its proteolytically processed form drive a cellular senescence programme. Nat Commun 5:5210

Showing the most recent 10 out of 13 publications