AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.4 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

RECQL4 regulates DNA damage response and redox homeostasis in esophageal cancer

Guosheng Lyu1Peng Su2Xiaohe Hao1Shiming Chen2Shuai Ren1Zixiao Zhao1Yaoqin Gong1Qiao Liu1 ( )Changshun Shao3 ( )
Key Laboratory of Experimental Teratology, Ministry of Education, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
Department of Pathology, Qilu Hospital of Shandong University, Jinan 250012, China
State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, China
Show Author Information

Abstract

Objective

RECQL4 (a member of the RECQ helicase family) upregulation has been reported to be associated with tumor progression in several malignancies. However, whether RECQL4 sustains esophageal squamous cell carcinoma (ESCC) has not been elucidated. In this study, we determined the functional role for RECQL4 in ESCC progression.

Methods

RECQL4 expression in clinical samples of ESCC was examined by immunohistochemistry. Cell proliferation, cellular senescence, the epithelial-mesenchymal transition (EMT), DNA damage, and reactive oxygen species in ESCC cell lines with RECQL4 depletion or overexpression were analyzed. The levels of proteins involved in the DNA damage response (DDR), cell cycle progression, survival, and the EMT were determined by Western blot analyses.

Results

RECQL4 was highly expressed in tumor tissues when compared to adjacent non-tumor tissues in ESCC (P < 0.001) and positively correlated with poor differentiation (P = 0.011), enhanced invasion (P = 0.033), and metastasis (P = 0.048). RECQL4 was positively associated with proliferation and migration in ESCC cells. Depletion of RECQL4 also inhibited growth of tumor xenografts in vivo. RECQL4 depletion induced G0/G1 phase arrest and cellular senescence. Importantly, the levels of DNA damage and reactive oxygen species were increased when RECQL4 was depleted. DDR, as measured by the activation of ATM, ATR, CHK1, and CHK2, was impaired. RECQL4 was also shown to promote the activation of AKT, ERK, and NF-kB in ESCC cells.

Conclusions

The results indicated that RECQL4 was highly expressed in ESCC and played critical roles in the regulation of DDR, redox homeostasis, and cell survival.

Electronic Supplementary Material

Download File(s)
cbm-18-1-120_ESM.pdf (276.3 KB)

References

1

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020; 70: 7-30.

2

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016; 66: 115-32.

3

Zeng H, Zheng R, Zhang S, Zuo T, Xia C, Zou X, et al. Esophageal cancer statistics in China, 2011: estimates based on 177 cancer registries. Thorac Cancer. 2016; 7: 232-7.

4

Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics? Cancer Commun (Lond). 2019; 39: 22.

5

Croteau DL, Singh DK, Ferrarelli LH, Lu HM, Bohr VA. RECQL4 in genomic instability and aging. Trends Genet. 2012; 28: 624-31.

6

Croteau DL, Popuri V, Opresko PL, Bohr VA. Human RecQ helicases in DNA repair, recombination, and replication. Annu Rev Biochem. 2014; 83: 519-52.

7

Mo D, Zhao Y, Balajee AS. Human RecQL4 helicase plays multifaceted roles in the genomic stability of normal and cancer cells. Cancer Lett. 2018; 413: 1-10.

8

Schurman SH, Hedayati M, Wang ZM, Singh DK, Speina E, Zhang YQ, et al. Direct and indirect roles of RECQL4 in modulating base excision repair capacity. Hum Mol Genet. 2009; 18: 3470-83.

9

Singh DK, Karmakar P, Aamann M, Schurman SH, May A, Croteau DL, et al. The involvement of human RECQL4 in DNA doublestrand break repair. Aging Cell. 2010; 9: 358-71.

10

Shamanna RA, Singh DK, Lu HM, Mirey G, Keijzers G, Salles B, et al. RECQ helicase RECQL4 participates in non-homologous end joining and interacts with the Ku complex. Carcinogenesis. 2014; 35: 2415-24.

11

Gupta S, De S, Srivastava V, Hussain M, Kumari J, Muniyappa K, et al. RECQL4 and p53 potentiate the activity of polymerase and maintain the integrity of the human mitochondrial genome. Carcinogenesis. 2014; 35: 34-45.

12

Lu HM, Shamanna RA, Keijzers G, Anand R, Rasmussen LJ, Cejka P, et al. RECQL4 promotes DNA end resection in repair of DNA double-strand breaks. Cell Rep. 2016; 16: 161-73.

13

Masai H. RecQL4: a helicase linking formation and maintenance of a replication fork. J Biochem. 2011; 149: 629-31.

14

Ghosh AK, Rossi ML, Singh DK, Dunn C, Ramamoorthy M, Croteau DL, et al. RECQL4, the protein mutated in Rothmund-Thomson syndrome, functions in telomere maintenance. J Biol Chem. 2012; 287: 196-209.

15

Sangrithi MN, Bernal JA, Madine M, Philpott A, Lee J, Dunphy WG, et al. Initiation of DNA replication requires the RECQL4 protein mutated in Rothmund-Thomson syndrome. Cell. 2005; 121: 887-98.

16

Kitao S, Shimamoto A, Goto M, Miller RW, Smithson WA, Lindor NM, et al. Mutations in RECQL4 cause a subset of cases of Rothmund-Thomson syndrome. Nat Genet. 1999; 22: 82-4.

17

Smeets MF, DeLuca E, Wall M, Quach JM, Chalk AM, Deans AJ, et al. The Rothmund-Thomson syndrome helicase RECQL4 is essential for hematopoiesis. J Clin Invest. 2014; 124: 3551-65.

18

Castillo-Tandazo W, Smeets MF, Murphy V, Liu R, Hodson C, Heierhorst J, et al. ATP-dependent helicase activity is dispensable for the physiological functions of Recql4. PLoS Genet. 2019; 15: e1008266.

19

Lao VV, Welcsh P, Luo Y, Carter KT, Dzieciatkowski S, Dintzis S, et al. Altered RECQ helicase expression in sporadic primary colorectal cancers. Transl Oncol. 2013; 6: 458-69.

20

Su Y, Meador JA, Calaf GM, Proietti De-Santis L, Zhao Y, Bohr VA, et al. Human RecQL4 helicase plays critical roles in prostate carcinogenesis. Cancer Res. 2010; 70: 9207-17.

21

Chen H, Yuan K, Wang X, Wang H, Wu Q, Wu X, et al. Overexpression of RECQL4 is associated with poor prognosis in patients with gastric cancer. Oncol Lett. 2018; 16: 5419-25.

22

Arora A, Agarwal D, Abdel-Fatah TMA, Lu HM, Croteau DL, Moseley P, et al. RECQL4 helicase has oncogenic potential in sporadic breast cancers. J Pathol. 2016; 238: 495-501.

23

Li J, Jin JF, Liao MJ, Dang W, Chen XH, Wu YF, et al. Upregulation of RECQL4 expression predicts poor prognosis in hepatocellular carcinoma. Oncol Lett. 2018; 15: 4248-54.

24

Fang HB, Nie LH, Chi ZF, Liu J, Guo D, Lu XM, et al. RecQL4 helicase amplification is involved in human breast tumorigenesis. PLoS One. 2013; 8: e69600.

25

Chen HL, Wu XB, Wang XY, Lin YJ, Wang HS, Peng JS. Downregulation of RECQL4 inhibits gastric cancer cell proliferation and induces cell cycle arrest at G0/G1 phase. Int J Clin Exp Med. 2018; 11: 10504-15.

26

Mo D, Fang H, Niu K, Liu J, Wu M, Li S, et al. Human helicase RECQL4 drives cisplatin resistance in gastric cancer by activating an AKT-YB1-MDR1 signaling pathway. Cancer Res. 2016; 76: 3057-66.

27

Ng AJ, Walia MK, Smeets MF, Mutsaers AJ, Sims NA, Purton LE, et al. The DNA helicase recql4 is required for normal osteoblast expansion and osteosarcoma formation. PLoS Genet. 2015; 11: e1005160.

28

Xu X, Liu Q, Zhang C, Ren S, Xu L, Zhao Z, et al. Inhibition of DYRK1A-EGFR axis by p53-MDM2 cascade mediates the induction of cellular senescence. Cell Death Dis. 2019; 10: 282.

29

Lu H, Fang EF, Sykora P, Kulikowicz T, Zhang Y, Becker KG, et al. Senescence induced by RECQL4 dysfunction contributes to Rothmund-Thomson syndrome features in mice. Cell Death Dis. 2014; 5: e1226.

30

Thiery JP, Sleeman JP. Complex networks orchestrate epithelialmesenchymal transitions. Nat Rev Mol Cell Biol. 2006; 7: 131-42.

31

Christofori G. New signals from the invasive front. Nature. 2006; 441: 444-50.

32

Miyamoto S. Nuclear initiated NF-kappaB signaling: NEMO and ATM take center stage. Cell Res. 2011; 21: 116-30.

33

Chua HL, Bhat-Nakshatri P, Clare SE, Morimiya A, Badve S, Nakshatri H. NF-kappaB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: potential involvement of ZEB-1 and ZEB-2. Oncogene. 2007; 26: 711-24.

34

Peter B, Falkenberg M. TWINKLE and other human mitochondrial DNA helicases: structure, function and disease. Genes (Basel). 2020; 11: 408.

35

Mishra A, Saxena S, Kaushal A, Nagaraju G. RAD51C/XRCC3 facilitates mitochondrial DNA replication and maintains integrity of the mitochondrial genome. Mol Cell Biol. 2018; 38: e00489-17.

36

Billard P, Poncet DA. Replication stress at telomeric and mitochondrial DNA: common origins and consequences on ageing. Int J Mol Sci. 2019; 20: 4959.

37

Chu WK, Hickson ID. RecQ helicases: multifunctional genome caretakers. Nat Rev Cancer. 2009; 9: 644-54.

38

Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, Linn S. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem. 2004; 73: 39-85.

39

Zhou BB, Elledge SJ. The DNA damage response: putting checkpoints in perspective. Nature. 2000; 408: 433-9.

40

Rouse J, Jackson SP. Interfaces between the detection, signaling, and repair of DNA damage. Science. 2002; 297: 547-51.

41

Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 2009; 8: 627-44.

42

Taniguchi K, Karin M. NF-kappaB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018; 18: 309-24.

43

Samatar AA, Poulikakos PI. Targeting RAS-ERK signalling in cancer: promises and challenges. Nat Rev Drug Discov. 2014; 13: 928-42.

44

Park SY, Kim H, Im JS, Lee JK. ATM activation is impaired in human cells defective in RecQL4 helicase activity. Biochem Biophys Res Commun. 2019; 509: 379-83.

Cancer Biology & Medicine
Pages 120-138
Cite this article:
Lyu G, Su P, Hao X, et al. RECQL4 regulates DNA damage response and redox homeostasis in esophageal cancer. Cancer Biology & Medicine, 2021, 18(1): 120-138. https://doi.org/10.20892/j.issn.2095-3941.2020.0105

78

Views

0

Downloads

5

Crossref

6

Web of Science

6

Scopus

Altmetrics

Received: 11 March 2020
Accepted: 10 July 2020
Published: 01 February 2021
©2021 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return