Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
NAR Cancer ; 6(2): zcae016, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38596431

ABSTRACT

With its ligand estrogen, the estrogen receptor (ER) initiates a global transcriptional program, promoting cell growth. This process involves topoisomerase 2 (TOP2), a key protein in resolving topological issues during transcription by cleaving a DNA duplex, passing another duplex through the break, and repairing the break. Recent studies revealed the involvement of various DNA repair proteins in the repair of TOP2-induced breaks, suggesting potential alternative repair pathways in cases where TOP2 is halted after cleavage. However, the contribution of these proteins in ER-induced transcriptional regulation remains unclear. We investigated the role of tyrosyl-DNA phosphodiesterase 2 (TDP2), an enzyme for the removal of halted TOP2 from the DNA ends, in the estrogen-induced transcriptome using both targeted and global transcription analyses. MYC activation by estrogen, a TOP2-dependent and transient event, became prolonged in the absence of TDP2 in both TDP2-deficient cells and mice. Bulk and single-cell RNA-seq analyses defined MYC and CCND1 as oncogenes whose estrogen response is tightly regulated by TDP2. These results suggest that TDP2 may inherently participate in the repair of estrogen-induced breaks at specific genomic loci, exerting precise control over oncogenic gene expression.

2.
Cell Rep ; 42(1): 111909, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640339

ABSTRACT

ATM gene mutation carriers are predisposed to estrogen-receptor-positive breast cancer (BC). ATM prevents BC oncogenesis by activating p53 in every cell; however, much remains unknown about tissue-specific oncogenesis after ATM loss. Here, we report that ATM controls the early transcriptional response to estrogens. This response depends on topoisomerase II (TOP2), which generates TOP2-DNA double-strand break (DSB) complexes and rejoins the breaks. When TOP2-mediated ligation fails, ATM facilitates DSB repair. After estrogen exposure, TOP2-dependent DSBs arise at the c-MYC enhancer in human BC cells, and their defective repair changes the activation profile of enhancers and induces the overexpression of many genes, including the c-MYC oncogene. CRISPR/Cas9 cleavage at the enhancer also causes c-MYC overexpression, indicating that this DSB causes c-MYC overexpression. Estrogen treatment induced c-Myc protein overexpression in mammary epithelial cells of ATM-deficient mice. In conclusion, ATM suppresses the c-Myc-driven proliferative effects of estrogens, possibly explaining such tissue-specific oncogenesis.


Subject(s)
DNA Breaks, Double-Stranded , Genes, myc , Humans , Mice , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Repair , Estrogens/pharmacology , Epithelium/metabolism , Carcinogenesis/genetics , Cell Cycle Proteins/metabolism
3.
Genes Cells ; 25(7): 450-465, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32277721

ABSTRACT

Androgens stimulate the proliferation of epithelial cells in the prostate by activating topoisomerase 2 (TOP2) and regulating the transcription of target genes. TOP2 resolves the entanglement of genomic DNA by transiently generating double-strand breaks (DSBs), where TOP2 homodimers covalently bind to 5' DSB ends, called TOP2-DNA cleavage complexes (TOP2ccs). When TOP2 fails to rejoin TOP2ccs generating stalled TOP2ccs, tyrosyl DNA phosphodiesterase-2 (TDP2) removes 5' TOP2 adducts from stalled TOP2ccs prior to the ligation of the DSBs by nonhomologous end joining (NHEJ), the dominant DSB repair pathway in G0 /G1 phases. We previously showed that estrogens frequently generate stalled TOP2ccs in G0 /G1 phases. Here, we show that physiological concentrations of androgens induce several DSBs in individual human prostate cancer cells during G1 phase, and loss of TDP2 causes a five times higher number of androgen-induced chromosome breaks in mitotic chromosome spreads. Intraperitoneally injected androgens induce several DSBs in individual epithelial cells of the prostate in TDP2-deficient mice, even at 20 hr postinjection. In conclusion, physiological concentrations of androgens have very strong genotoxicity, most likely by generating stalled TOP2ccs.


Subject(s)
Androgens/toxicity , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Genomic Instability/genetics , Phosphoric Diester Hydrolases/metabolism , Prostate/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromosome Breakage , DNA End-Joining Repair/drug effects , DNA End-Joining Repair/genetics , DNA-Binding Proteins/genetics , Epithelial Cells/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/genetics , Genomic Instability/drug effects , Histones/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoric Diester Hydrolases/genetics , Prostate/drug effects , Prostatic Neoplasms/genetics , RNA, Small Interfering , Receptors, Androgen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL