13 research outputs found

    Chromosomal protein HMGN1 enhances the rate of DNA repair in chromatin

    No full text
    We report that HMGN1, a nucleosome binding protein that destabilizes the higher-order chromatin structure, modulates the repair rate of ultraviolet light (UV)-induced DNA lesions in chromatin. Hmgn1(–/–) mouse embryonic fibroblasts (MEFs) are hypersensitive to UV, and the removal rate of photoproducts from the chromatin of Hmgn1(–/–) MEFs is decreased as compared with the chromatin of Hmgn1(+/+) MEFs; yet, host cell reactivation assays and DNA array analysis indicate that the nucleotide excision repair (NER) pathway in the Hmgn1(–/–) MEFs remains intact. The UV hypersensitivity of Hmgn1(–/–) MEFs could be rescued by transfection with plasmids expressing wild-type HMGN1 protein, but not with plasmids expressing HMGN1 mutants that do not bind to nucleosomes or do not unfold chromatin. Transcriptionally active genes, the main target of the NER pathways in mice, contain HMGN1 protein, and loss of HMGN1 protein reduces the accessibility of transcribed genes to nucleases. By reducing the compaction of the higher-order chromatin structure, HMGN1 facilitates access to UV-damaged DNA sites and enhances the rate of DNA repair in chromatin

    Increased tumorigenicity and sensitivity to ionizing radiation upon loss of chromosomal protein HMGN1

    No full text
    We report that loss of HMGN1, a nucleosome-binding protein that alters the compaction of the chromatin fiber, increases the cellular sensitivity to ionizing radiation and the tumor burden of mice. The mortality and tumor burden of ionizing radiation–treated Hmgn1(−/−) mice is higher than that of their Hmgn1(+/+) littermates. Hmgn1(−/−) fibroblasts have an altered G(2)-M checkpoint activation and are hypersensitive to ionizing radiation. The ionizing radiation hypersensitivity and the aberrant G(2)-M checkpoint activation of Hmgn1(−/−) fibroblasts can be reverted by transfections with plasmids expressing wild-type HMGN1, but not with plasmids expressing mutant HMGN proteins that do not bind to chromatin. Transformed Hmgn1(−/−) fibroblasts grow in soft agar and produce tumors in nude mice with a significantly higher efficiency than Hmgn1(+/+) fibroblasts, suggesting that loss of HMGN1 protein disrupts cellular events controlling proliferation and growth. Hmgn1(−/−) mice have a higher incidence of multiple malignant tumors and metastases than their Hmgn1(+/+) littermates. We suggest that HMGN1 optimizes the cellular response to ionizing radiation and to other tumorigenic events; therefore, loss of this protein increases the tumor burden in mice
    corecore