RRC ID 74992
著者 Parvin S, Akter J, Takenobu H, Katai Y, Satoh S, Okada R, Haruta M, Mukae K, Wada T, Ohira M, Ando K, Kamijo T.
タイトル ATM depletion induces proteasomal degradation of FANCD2 and sensitizes neuroblastoma cells to PARP inhibitors.
ジャーナル BMC Cancer
Abstract BACKGROUND:Genomic alterations, including loss of function in chromosome band 11q22-23, are frequently observed in neuroblastoma, which is the most common extracranial childhood tumour. In neuroblastoma, ATM, a DNA damage response-associated gene located on 11q22-23, has been linked to tumorigenicity. Genetic changes in ATM are heterozygous in most tumours. However, it is unclear how ATM is associated with tumorigenesis and cancer aggressiveness.
METHODS:To elucidate its molecular mechanism of action, we established ATM-inactivated NGP and CHP-134 neuroblastoma cell lines using CRISPR/Cas9 genome editing. The knock out cells were rigorously characterized by analyzing proliferation, colony forming abilities and responses to PARP inhibitor (Olaparib). Western blot analyses were performed to detect different protein expression related to DNA repair pathway. ShRNA lentiviral vectors were used to knockdown ATM expression in SK-N-AS and SK-N-SH neuroblastoma cell lines. ATM knock out cells were stably transfected with FANCD2 expression plasmid to over-expressed the FANCD2. Moreover, knock out cells were treated with proteasome inhibitor MG132 to determine the protein stability of FANCD2. FANCD2, RAD51 and γH2AX protein expressions were determined by Immunofluorescence microscopy.
RESULTS:Haploinsufficient ATM resulted in increased proliferation (p < 0.01) and cell survival following PARP inhibitor (olaparib) treatment. However, complete ATM knockout decreased proliferation (p < 0.01) and promoted cell susceptibility to olaparib (p < 0.01). Complete loss of ATM suppressed the expression of DNA repair-associated molecules FANCD2 and RAD51 and induced DNA damage in neuroblastoma cells. A marked downregulation of FANCD2 expression was also observed in shRNA-mediated ATM-knockdown neuroblastoma cells. Inhibitor experiments demonstrated that the degradation of FANCD2 was regulated at the protein level through the ubiquitin-proteasome pathway. Reintroduction of FANCD2 expression is sufficient to reverse decreased proliferation mediated by ATM depletion.
CONCLUSIONS:Our study revealed the molecular mechanism underlying ATM heterozygosity in neuroblastomas and elucidated that ATM inactivation enhances the susceptibility of neuroblastoma cells to olaparib treatment. These findings might be useful in the treatment of high-risk NB patients showing ATM zygosity and aggressive cancer progression in future.
巻・号 23(1)
ページ 313
公開日 2023-4-5
DOI 10.1186/s12885-023-10772-y
PII 10.1186/s12885-023-10772-y
PMID 37020276
PMC PMC10077671
MeSH Antineoplastic Agents* / therapeutic use Ataxia Telangiectasia Mutated Proteins / genetics Cell Line, Tumor Child Fanconi Anemia* Fanconi Anemia Complementation Group D2 Protein Humans Neuroblastoma* / pathology Poly(ADP-ribose) Polymerase Inhibitors / therapeutic use RNA, Small Interfering / therapeutic use
IF 3.15
リソース情報
ヒト・動物細胞 SK-N-SH(RCB0426)