Skip to main content
Top
Published in: BMC Cancer 1/2024

Open Access 01-12-2024 | Endometrial Cancer | Research

DOCK1 regulates the malignant biological behavior of endometrial cancer through c-Raf/ERK pathway

Authors: Shangdan Xie, Yanshan Jin, Jiakun Wang, Jingwei Li, Mengjia Peng, Xueqiong Zhu

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

Background

The effect of DOCK1 gene on the biological behavior of endometrial carcinoma cells and its related pathway has not been reported.

Methods

The immunohistochemical method and western blot were utilized to analyze DOCK1 protein expression in endometrial tissues and cells, respectively. CCK-8, BrdU, transwell and flow cytometry were performed to analyze the effect of DOCK1 expression changes on the viability, proliferation, invasion, migration and apoptosis of endometrial cancer cells, respectively. The effects of DOCK1 gene on Bcl-2, MMP9, Ezrin, E-cadherin and c-RAF/ERK1/2 signaling pathway were evaluated by western blot. The xenograft models were constructed to analyze the effect of DOCK1 in vivo.

Results

DOCK1 expression was increased in endometrial cancer tissues and cells compared with those in normal adjacent tissues and cells. DOCK1 knockout could inhibit the malignant biological behavior of endometrial cancer cells, while DOCK1 overexpression played the opposite effect. The expression of E-cadherin was upregulated and those of MMP9, Ezrin, Bcl-2, p-c-RAF (S338) and p-ERK1/2 (T202/Y204) were downregulated after DOCK1 knockout, while DOCK1 overexpression played the opposite effect. Additionally, Raf inhibitor LY3009120 reversed the function of DOCK1 on malignant biological behavior. In vivo experiment results showed that the growth and weight of transplanted tumors in nude mice were inhibited after DOCK1 knockout. The changes of E-cadherin, MMP9, Ezrin and Bcl-2 expressions in the transplanted tumors were consistent with those in vitro.

Conclusion

DOCK1 could enhance the malignant biological behavior of endometrial cancer cells, which might be through c-RAF/ERK1/2 signaling pathways in vitro and in vivo.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed
3.
go back to reference Winterhoff B, Thomaier L, Mullany S, Powell MA. Molecular characterization of endometrial cancer and therapeutic implications. Curr Opin Obstet Gynecol. 2020;32(1):76–83.CrossRefPubMed Winterhoff B, Thomaier L, Mullany S, Powell MA. Molecular characterization of endometrial cancer and therapeutic implications. Curr Opin Obstet Gynecol. 2020;32(1):76–83.CrossRefPubMed
5.
go back to reference Crosbie EJ, Kitson SJ, McAlpine JN, Mukhopadhyay A, Powell ME, Singh N. Endometrial cancer. Lancet. 2022;399(10333):1412–28.CrossRefPubMed Crosbie EJ, Kitson SJ, McAlpine JN, Mukhopadhyay A, Powell ME, Singh N. Endometrial cancer. Lancet. 2022;399(10333):1412–28.CrossRefPubMed
7.
8.
go back to reference Mohan JJ, Narayan P, Padmanabhan RA, Joseph S, Kumar PG, Laloraya M. Silencing of dedicator of cytokinesis (DOCK180) obliterates pregnancy by interfering with decidualization due to blockage of nuclear entry of autoimmune regulator (AIRE). Am J Reprod Immunol. 2018;80(1):e12844.CrossRefPubMed Mohan JJ, Narayan P, Padmanabhan RA, Joseph S, Kumar PG, Laloraya M. Silencing of dedicator of cytokinesis (DOCK180) obliterates pregnancy by interfering with decidualization due to blockage of nuclear entry of autoimmune regulator (AIRE). Am J Reprod Immunol. 2018;80(1):e12844.CrossRefPubMed
9.
go back to reference Grimsley CM, Kinchen JM, Tosello-Trampont AC, Brugnera E, Haney LB, Lu M, et al. Dock180 and ELMO1 proteins cooperate to promote evolutionarily conserved rac-dependent cell migration. J Biol Chem. 2004;279(7):6087–97.CrossRefPubMed Grimsley CM, Kinchen JM, Tosello-Trampont AC, Brugnera E, Haney LB, Lu M, et al. Dock180 and ELMO1 proteins cooperate to promote evolutionarily conserved rac-dependent cell migration. J Biol Chem. 2004;279(7):6087–97.CrossRefPubMed
10.
go back to reference Sanders MA, Ampasala D, Basson MD. DOCK5 and DOCK1 regulate Caco-2 intestinal epithelial cell spreading and migration on collagen IV. J Biol Chem. 2009;284(1):27–35.CrossRefPubMedPubMedCentral Sanders MA, Ampasala D, Basson MD. DOCK5 and DOCK1 regulate Caco-2 intestinal epithelial cell spreading and migration on collagen IV. J Biol Chem. 2009;284(1):27–35.CrossRefPubMedPubMedCentral
11.
go back to reference Bagci H, Laurin M, Huber J, Muller WJ, Cote JF. Impaired cell death and mammary gland involution in the absence of Dock1 and Rac1 signaling. Cell Death Dis. 2014;5:e1375.CrossRefPubMedPubMedCentral Bagci H, Laurin M, Huber J, Muller WJ, Cote JF. Impaired cell death and mammary gland involution in the absence of Dock1 and Rac1 signaling. Cell Death Dis. 2014;5:e1375.CrossRefPubMedPubMedCentral
12.
go back to reference Chen DJ, Chen W, Jiang H, Yang H, Wang YC, Chen JH. Downregulation of DOCK1 sensitizes bladder cancer cells to cisplatin through preventing epithelial-mesenchymal transition. Drug Des Devel Ther. 2016;10:2845–53.CrossRefPubMedPubMedCentral Chen DJ, Chen W, Jiang H, Yang H, Wang YC, Chen JH. Downregulation of DOCK1 sensitizes bladder cancer cells to cisplatin through preventing epithelial-mesenchymal transition. Drug Des Devel Ther. 2016;10:2845–53.CrossRefPubMedPubMedCentral
13.
go back to reference Misek SA, Chen J, Schroeder L, Rattanasinchai C, Sample A, Sarkaria JN, et al. EGFR signals through a DOCK180-MLK3 Axis to drive Glioblastoma Cell Invasion. Mol Cancer Res. 2017;15(8):1085–95.CrossRefPubMed Misek SA, Chen J, Schroeder L, Rattanasinchai C, Sample A, Sarkaria JN, et al. EGFR signals through a DOCK180-MLK3 Axis to drive Glioblastoma Cell Invasion. Mol Cancer Res. 2017;15(8):1085–95.CrossRefPubMed
14.
go back to reference Wang J, Dai JM, Che YL, Gao YM, Peng HJ, Liu B, et al. Elmo1 helps dock180 to regulate Rac1 activity and cell migration of ovarian cancer. Int J Gynecol Cancer. 2014;24(5):844–50.CrossRefPubMed Wang J, Dai JM, Che YL, Gao YM, Peng HJ, Liu B, et al. Elmo1 helps dock180 to regulate Rac1 activity and cell migration of ovarian cancer. Int J Gynecol Cancer. 2014;24(5):844–50.CrossRefPubMed
15.
go back to reference Xu Y, Chen X, Pan S, Wang ZW, Zhu X. TM7SF2 regulates cell proliferation and apoptosis by activation of C-Raf/ERK pathway in cervical cancer. Cell Death Discov. 2021;7(1):299.CrossRefPubMedPubMedCentral Xu Y, Chen X, Pan S, Wang ZW, Zhu X. TM7SF2 regulates cell proliferation and apoptosis by activation of C-Raf/ERK pathway in cervical cancer. Cell Death Discov. 2021;7(1):299.CrossRefPubMedPubMedCentral
16.
go back to reference Xu W, Xie S, Chen X, Pan S, Qian H, Zhu X. Effects of Quercetin on the efficacy of various chemotherapeutic drugs in Cervical Cancer cells. Drug Des Devel Ther. 2021;15:577–88.CrossRefPubMedPubMedCentral Xu W, Xie S, Chen X, Pan S, Qian H, Zhu X. Effects of Quercetin on the efficacy of various chemotherapeutic drugs in Cervical Cancer cells. Drug Des Devel Ther. 2021;15:577–88.CrossRefPubMedPubMedCentral
17.
go back to reference Zhao F, Siu MK, Jiang L, Tam KF, Ngan HY, Le XF, et al. Overexpression of dedicator of cytokinesis I (Dock180) in ovarian cancer correlated with aggressive phenotype and poor patient survival. Histopathology. 2011;59(6):1163–72.CrossRefPubMed Zhao F, Siu MK, Jiang L, Tam KF, Ngan HY, Le XF, et al. Overexpression of dedicator of cytokinesis I (Dock180) in ovarian cancer correlated with aggressive phenotype and poor patient survival. Histopathology. 2011;59(6):1163–72.CrossRefPubMed
18.
go back to reference Zhang B, Li H, Yin C, Sun X, Zheng S, Zhang C, et al. Dock1 promotes the mesenchymal transition of glioma and is modulated by MiR-31. Neuropathol Appl Neurobiol. 2017;43(5):419–32.CrossRefPubMed Zhang B, Li H, Yin C, Sun X, Zheng S, Zhang C, et al. Dock1 promotes the mesenchymal transition of glioma and is modulated by MiR-31. Neuropathol Appl Neurobiol. 2017;43(5):419–32.CrossRefPubMed
19.
go back to reference Chiang SK, Chang WC, Chen SE, Chang LC. DOCK1 regulates growth and motility through the RRP1B-Claudin-1 pathway in claudin-low breast Cancer cells. Cancers (Basel). 2019;11(11):1762.CrossRefPubMed Chiang SK, Chang WC, Chen SE, Chang LC. DOCK1 regulates growth and motility through the RRP1B-Claudin-1 pathway in claudin-low breast Cancer cells. Cancers (Basel). 2019;11(11):1762.CrossRefPubMed
20.
go back to reference Aznavoorian S, Murphy AN, Stetler-Stevenson WG, Liotta LA. Molecular aspects of tumor cell invasion and metastasis. Cancer. 1993;71(4):1368–83.CrossRefPubMed Aznavoorian S, Murphy AN, Stetler-Stevenson WG, Liotta LA. Molecular aspects of tumor cell invasion and metastasis. Cancer. 1993;71(4):1368–83.CrossRefPubMed
21.
go back to reference Barrett A, Pellet-Many C, Zachary IC, Evans IM, Frankel P. p130Cas: a key signalling node in health and disease. Cell Signal. 2013;25(4):766–77.CrossRefPubMed Barrett A, Pellet-Many C, Zachary IC, Evans IM, Frankel P. p130Cas: a key signalling node in health and disease. Cell Signal. 2013;25(4):766–77.CrossRefPubMed
22.
go back to reference Feng H, Li Y, Yin Y, Zhang W, Hou Y, Zhang L, et al. Protein kinase A-dependent phosphorylation of Dock180 at serine residue 1250 is important for glioma growth and invasion stimulated by platelet derived-growth factor receptor alpha. Neuro Oncol. 2015;17(6):832–42.CrossRefPubMed Feng H, Li Y, Yin Y, Zhang W, Hou Y, Zhang L, et al. Protein kinase A-dependent phosphorylation of Dock180 at serine residue 1250 is important for glioma growth and invasion stimulated by platelet derived-growth factor receptor alpha. Neuro Oncol. 2015;17(6):832–42.CrossRefPubMed
23.
go back to reference Mondal S, Adhikari N, Banerjee S, Amin SA, Jha T. Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: a minireview. Eur J Med Chem. 2020;194:112260.CrossRefPubMed Mondal S, Adhikari N, Banerjee S, Amin SA, Jha T. Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: a minireview. Eur J Med Chem. 2020;194:112260.CrossRefPubMed
24.
go back to reference Song Y, Ma X, Zhang M, Wang M, Wang G, Ye Y, et al. Ezrin mediates Invasion and Metastasis in Tumorigenesis: a review. Front Cell Dev Biol. 2020;8:588801.CrossRefPubMedPubMedCentral Song Y, Ma X, Zhang M, Wang M, Wang G, Ye Y, et al. Ezrin mediates Invasion and Metastasis in Tumorigenesis: a review. Front Cell Dev Biol. 2020;8:588801.CrossRefPubMedPubMedCentral
25.
go back to reference Kocher HM, Sandle J, Mirza TA, Li NF, Hart IR. Ezrin interacts with cortactin to form podosomal rosettes in pancreatic cancer cells. Gut. 2009;58(2):271–84.CrossRefPubMed Kocher HM, Sandle J, Mirza TA, Li NF, Hart IR. Ezrin interacts with cortactin to form podosomal rosettes in pancreatic cancer cells. Gut. 2009;58(2):271–84.CrossRefPubMed
26.
go back to reference Fang C, Kang Y, E-Cadherin. Context-dependent functions of a quintessential epithelial marker in Metastasis. Cancer Res. 2021;81(23):5800–2.CrossRefPubMed Fang C, Kang Y, E-Cadherin. Context-dependent functions of a quintessential epithelial marker in Metastasis. Cancer Res. 2021;81(23):5800–2.CrossRefPubMed
27.
go back to reference Riegel K, Schloder J, Sobczak M, Jonuleit H, Thiede B, Schild H, et al. RAF kinases are stabilized and required for dendritic cell differentiation and function. Cell Death Differ. 2020;27(4):1300–15.CrossRefPubMed Riegel K, Schloder J, Sobczak M, Jonuleit H, Thiede B, Schild H, et al. RAF kinases are stabilized and required for dendritic cell differentiation and function. Cell Death Differ. 2020;27(4):1300–15.CrossRefPubMed
28.
29.
go back to reference Yeh LT, Lin CW, Lu KH, Hsieh YH, Yeh CB, Yang SF, et al. Niclosamide suppresses Migration and Invasion of Human Osteosarcoma cells by repressing TGFBI expression via the ERK Signaling Pathway. Int J Mol Sci. 2022;23(1):484.CrossRefPubMedPubMedCentral Yeh LT, Lin CW, Lu KH, Hsieh YH, Yeh CB, Yang SF, et al. Niclosamide suppresses Migration and Invasion of Human Osteosarcoma cells by repressing TGFBI expression via the ERK Signaling Pathway. Int J Mol Sci. 2022;23(1):484.CrossRefPubMedPubMedCentral
30.
go back to reference Li B, Xiao Q, Shan L, Song Y. NCAPH promotes cell proliferation and inhibits cell apoptosis of bladder cancer cells through MEK/ERK signaling pathway. Cell Cycle. 2022;21(4):427–38.CrossRefPubMedPubMedCentral Li B, Xiao Q, Shan L, Song Y. NCAPH promotes cell proliferation and inhibits cell apoptosis of bladder cancer cells through MEK/ERK signaling pathway. Cell Cycle. 2022;21(4):427–38.CrossRefPubMedPubMedCentral
31.
go back to reference Feng HZ, Hu B, Liu KW, Li YX, Lu XH, Cheng T, et al. Activation of Rac1 by src-dependent phosphorylation of Dock180(Y1811) mediates PDGFRα-stimulated glioma tumorigenesis in mice and humans. J Clin Invest. 2011;121(12):4670–84.CrossRefPubMedPubMedCentral Feng HZ, Hu B, Liu KW, Li YX, Lu XH, Cheng T, et al. Activation of Rac1 by src-dependent phosphorylation of Dock180(Y1811) mediates PDGFRα-stimulated glioma tumorigenesis in mice and humans. J Clin Invest. 2011;121(12):4670–84.CrossRefPubMedPubMedCentral
32.
go back to reference Ebi H, Costa C, Faber AC, Nishtala M, Kotani H, Juric D, et al. PI3K regulates MEK/ERK signaling in breast cancer via the Rac-GEF, P-Rex1. Proc Natl Acad Sci. 2013;110(52):21124–29.CrossRefPubMedPubMedCentralADS Ebi H, Costa C, Faber AC, Nishtala M, Kotani H, Juric D, et al. PI3K regulates MEK/ERK signaling in breast cancer via the Rac-GEF, P-Rex1. Proc Natl Acad Sci. 2013;110(52):21124–29.CrossRefPubMedPubMedCentralADS
Metadata
Title
DOCK1 regulates the malignant biological behavior of endometrial cancer through c-Raf/ERK pathway
Authors
Shangdan Xie
Yanshan Jin
Jiakun Wang
Jingwei Li
Mengjia Peng
Xueqiong Zhu
Publication date
01-12-2024
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2024
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-12030-1

Other articles of this Issue 1/2024

BMC Cancer 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine