Cholesterol depletion sensitizes gallbladder cancer to cisplatin by impairing DNA damage response

Cell Cycle. 2019 Dec;18(23):3337-3350. doi: 10.1080/15384101.2019.1676581. Epub 2019 Oct 10.

Abstract

Gallbladder cancer (GBC) is the common malignancy of the bile tract system with extremely poor clinical outcomes, owing to its metastatic property and intrinsic resistance to the first-line drugs. Although it is well-established that cholesterol abnormity contributes to gallstone formation, a leading risk factor for GBC, the link of cholesterol homeostasis with GBC has not been investigated. The present study systematically examined the genes implicated in cholesterol homeostasis, and revealed altered gene expressions of de novo cholesterol biosynthesis and sterol sulfonation (SULT2B1), reduced bile acid synthesis (CYP7B1 and CYP39A1) and impaired sterol efflux (ABCA1, ABCG5, LCAT, and CETP) in GBC tissues. Suppression of cholesterol biosynthesis by lovastatin inhibited GBC cell proliferation possibly through attenuating the DNA repair process. Further investigation revealed lovastatin sensitized GBC cells to cisplatin-induced apoptosis and suppressed the activation of CHK1, CHK2, and H2AX during DNA damage response. By using chemically distinct statins, HMGCR depletion or supplementing mevalonate, the product of HMGCR, we showed the inhibitory effects on DNA repair process of lovastatin were due to the blockage of the mevalonate pathway. Subcutaneous xenograft mice model suggested lovastatin promoted the therapeutic efficacy of cisplatin, and significantly prolonged the survival times of tumor-bearing mice. Moreover, HMGCR ablation repressed tumor growth in vivo, which can be rescued partially by restored expression of HMGCR, suggesting the on-target effects of lovastatin. Therefore, our study provides the clinical relevance of cholesterol homeostasis with GBC progression, and highlights a novel intervention of combined use of lovastatin and cisplatin for GBC.

Keywords: Cholesterol; DNA Damage response; gallbladder cancer; statins; the mevalonate pathway.

MeSH terms

  • ATP Binding Cassette Transporter 1 / genetics
  • ATP Binding Cassette Transporter, Subfamily G, Member 5 / genetics
  • Animals
  • Apoptosis / drug effects
  • Cholesterol / biosynthesis
  • Cholesterol / genetics*
  • Cholesterol Ester Transfer Proteins / genetics
  • Cisplatin / adverse effects*
  • Cisplatin / pharmacology
  • Cytochrome P450 Family 7 / genetics
  • DNA Damage / drug effects
  • DNA Repair / drug effects
  • Female
  • Gallbladder Neoplasms / drug therapy*
  • Gallbladder Neoplasms / genetics
  • Gallbladder Neoplasms / pathology
  • Gallstones / drug therapy*
  • Gallstones / genetics
  • Gallstones / pathology
  • Heterografts
  • Humans
  • Male
  • Mice
  • Phosphatidylcholine-Sterol O-Acyltransferase / genetics
  • Risk Factors
  • Steroid Hydroxylases / genetics
  • Sulfotransferases / genetics

Substances

  • ABCA1 protein, human
  • ATP Binding Cassette Transporter 1
  • ATP Binding Cassette Transporter, Subfamily G, Member 5
  • CETP protein, human
  • Cholesterol Ester Transfer Proteins
  • Cholesterol
  • Steroid Hydroxylases
  • Cytochrome P450 Family 7
  • CYP39A1 protein, human
  • CYP7B1 protein, human
  • LCAT protein, human
  • Phosphatidylcholine-Sterol O-Acyltransferase
  • Sulfotransferases
  • SULT2B1 protein, human
  • Cisplatin

Grants and funding

This work was supported by the National Natural Science Foundation of China [81472240]; National Natural Science Foundation of China [81773184]; National Natural Science Foundation of China [81702864]; Research Fund of Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine [TRYJ201702]; Shanghai Sailing Program for Young Talents [17YF1411200]; Innovative Research Team of High-level Local Universities in Shanghai [NA]; Medical-Engineering Joint Fund of Shanghai Jiaotong University [YG2016QN51]; Shanghai Outstanding Academic Leaders Plan.