Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

01-12-2021 | Prostate Cancer | Research

ASC-J9® suppresses prostate cancer cell proliferation and invasion via altering the ATF3-PTK2 signaling

Authors: Hao Tian, Fu-ju Chou, Jing Tian, Yong Zhang, Bosen You, Chi-Ping Huang, Shuyuan Yeh, Yuanjie Niu, Chawnshang Chang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Early studies indicated that ASC-J9®, an androgen receptor (AR) degradation enhancer, could suppress the prostate cancer (PCa) progression. Here we found ASC-J9® could also suppress the PCa progression via an AR-independent mechanism, which might involve modulating the tumor suppressor ATF3 expression.

Methods

The lentiviral system was used to modify gene expression in C4–2, CWR22Rv1 and PC-3 cells. Western blot and Immunohistochemistry were used to detect protein expression. MTT and Transwell assays were used to test the proliferation and invasion ability.

Results

ASC-J9® can suppress PCa cell proliferation and invasion in both PCa C4–2 and CWR22Rv1 cells via altering the ATF3 expression. Further mechanistic studies reveal that ASC-J9® can increase the ATF3 expression via decreasing Glutamate-cysteine ligase catalytic (GCLC) subunit expression, which can then lead to decrease the PTK2 expression. Human clinical studies further linked the ATF3 expression to the PCa progression. Preclinical studies using in vivo mouse model also proved ASC-J9® could suppress AR-independent PCa cell invasion, which could be reversed after suppressing ATF3.

Conclusions

ASC-J9® can function via altering ATF3/PTK2 signaling to suppress the PCa progression in an AR-independent manner.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Huggins C, Hodges CV. Studies on prostatic cancer - I the effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res. 1941;1(4):293–7. Huggins C, Hodges CV. Studies on prostatic cancer - I the effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res. 1941;1(4):293–7.
3.
go back to reference Logothetis CJ, Lin SH. Osteoblasts in prostate cancer metastasis to bone. Nat Rev Cancer. 2005;5(1):21–8.PubMedCrossRef Logothetis CJ, Lin SH. Osteoblasts in prostate cancer metastasis to bone. Nat Rev Cancer. 2005;5(1):21–8.PubMedCrossRef
5.
go back to reference Lin TH, et al. Differential androgen deprivation therapies with anti-androgens casodex/bicalutamide or MDV3100/Enzalutamide versus anti-androgen receptor ASC-J9(R) Lead to promotion versus suppression of prostate cancer metastasis. J Biol Chem. 2013;288(27):19359–69.PubMedPubMedCentralCrossRef Lin TH, et al. Differential androgen deprivation therapies with anti-androgens casodex/bicalutamide or MDV3100/Enzalutamide versus anti-androgen receptor ASC-J9(R) Lead to promotion versus suppression of prostate cancer metastasis. J Biol Chem. 2013;288(27):19359–69.PubMedPubMedCentralCrossRef
6.
go back to reference Lin TH, et al. Anti-androgen receptor ASC-J9 versus anti-androgens MDV3100 (Enzalutamide) or Casodex (Bicalutamide) leads to opposite effects on prostate cancer metastasis via differential modulation of macrophage infiltration and STAT3-CCL2 signaling. Cell Death Dis. 2013;4:e764.PubMedPubMedCentralCrossRef Lin TH, et al. Anti-androgen receptor ASC-J9 versus anti-androgens MDV3100 (Enzalutamide) or Casodex (Bicalutamide) leads to opposite effects on prostate cancer metastasis via differential modulation of macrophage infiltration and STAT3-CCL2 signaling. Cell Death Dis. 2013;4:e764.PubMedPubMedCentralCrossRef
7.
go back to reference Kregel S, et al. Sox2 is an androgen receptor-repressed gene that promotes castration-resistant prostate cancer. PLoS One. 2013;8:1.CrossRef Kregel S, et al. Sox2 is an androgen receptor-repressed gene that promotes castration-resistant prostate cancer. PLoS One. 2013;8:1.CrossRef
8.
go back to reference Takayama K, et al. Castration-induced acceleration of bone metastasis prevented by rank inhibitor Osteoprotegerin in murine castration-resistant prostate cancer model. J Urol. 2015;193(4):E549–50. Takayama K, et al. Castration-induced acceleration of bone metastasis prevented by rank inhibitor Osteoprotegerin in murine castration-resistant prostate cancer model. J Urol. 2015;193(4):E549–50.
9.
go back to reference Chen WY, et al. Loss of SPDEF and gain of TGFBI activity after androgen deprivation therapy promote EMT and bone metastasis of prostate cancer. Sci Signal. 2017;10:492. Chen WY, et al. Loss of SPDEF and gain of TGFBI activity after androgen deprivation therapy promote EMT and bone metastasis of prostate cancer. Sci Signal. 2017;10:492.
10.
go back to reference Chen WY, et al. Inhibition of the androgen receptor induces a novel tumor promoter, ZBTB46, for prostate cancer metastasis. Oncogene. 2017;36(45):6213–24.PubMedCrossRef Chen WY, et al. Inhibition of the androgen receptor induces a novel tumor promoter, ZBTB46, for prostate cancer metastasis. Oncogene. 2017;36(45):6213–24.PubMedCrossRef
11.
go back to reference Wu W, et al. Phenylbutyl isoselenocyanate induces reactive oxygen species to inhibit androgen receptor and to initiate p53-mediated apoptosis in LNCaP prostate cancer cells. Mol Carcinog. 2018;57(8):1055.PubMedCrossRef Wu W, et al. Phenylbutyl isoselenocyanate induces reactive oxygen species to inhibit androgen receptor and to initiate p53-mediated apoptosis in LNCaP prostate cancer cells. Mol Carcinog. 2018;57(8):1055.PubMedCrossRef
12.
go back to reference Qin X, et al. Theranostic Pt(IV) conjugate with target selectivity for androgen receptor. Inorg Chem. 2018;57(9):5019.PubMedCrossRef Qin X, et al. Theranostic Pt(IV) conjugate with target selectivity for androgen receptor. Inorg Chem. 2018;57(9):5019.PubMedCrossRef
13.
14.
go back to reference Tatsumi K, et al. Effect of propofol on androgen receptor activity in prostate cancer cells. Eur J Pharmacol. 2017;809:242–52.PubMedCrossRef Tatsumi K, et al. Effect of propofol on androgen receptor activity in prostate cancer cells. Eur J Pharmacol. 2017;809:242–52.PubMedCrossRef
16.
go back to reference Wang R, et al. ASC-J9((R)) suppresses castration resistant prostate cancer progression via degrading the enzalutamide-induced androgen receptor mutant AR-F876L. Cancer Lett. 2016;379(1):154–60.PubMedCrossRef Wang R, et al. ASC-J9((R)) suppresses castration resistant prostate cancer progression via degrading the enzalutamide-induced androgen receptor mutant AR-F876L. Cancer Lett. 2016;379(1):154–60.PubMedCrossRef
17.
go back to reference Yamashita S, et al. ASC-J9 suppresses castration-resistant prostate cancer growth through degradation of full-length and splice variant androgen receptors. Neoplasia. 2012;14(1):74–83.PubMedPubMedCentralCrossRef Yamashita S, et al. ASC-J9 suppresses castration-resistant prostate cancer growth through degradation of full-length and splice variant androgen receptors. Neoplasia. 2012;14(1):74–83.PubMedPubMedCentralCrossRef
18.
go back to reference Lai KP, et al. New therapeutic approach to suppress castration-resistant prostate cancer using ASC-J9 via targeting androgen receptor in selective prostate cells. Am J Pathol. 2013;182(2):460–73.PubMedPubMedCentralCrossRef Lai KP, et al. New therapeutic approach to suppress castration-resistant prostate cancer using ASC-J9 via targeting androgen receptor in selective prostate cells. Am J Pathol. 2013;182(2):460–73.PubMedPubMedCentralCrossRef
19.
go back to reference Izumi K, Chang C. Targeting inflammatory cytokines-androgen receptor (AR) signaling with ASC-J9((R)) to better battle prostate cancer progression. Oncoimmunology. 2013;2(12):e26853.PubMedPubMedCentralCrossRef Izumi K, Chang C. Targeting inflammatory cytokines-androgen receptor (AR) signaling with ASC-J9((R)) to better battle prostate cancer progression. Oncoimmunology. 2013;2(12):e26853.PubMedPubMedCentralCrossRef
20.
go back to reference Jiang Q, et al. Targeting androgen receptor leads to suppression of prostate cancer via induction of autophagy. J Urol. 2012;188(4):1361–8.PubMedCrossRef Jiang Q, et al. Targeting androgen receptor leads to suppression of prostate cancer via induction of autophagy. J Urol. 2012;188(4):1361–8.PubMedCrossRef
21.
go back to reference Wang R, et al. Preclinical study using Malat1 small interfering RNA or androgen receptor splicing variant 7 degradation enhancer ASC-J9((R)) to suppress Enzalutamide-resistant prostate cancer progression. Eur Urol. 2017;72(5):835–44.PubMedPubMedCentralCrossRef Wang R, et al. Preclinical study using Malat1 small interfering RNA or androgen receptor splicing variant 7 degradation enhancer ASC-J9((R)) to suppress Enzalutamide-resistant prostate cancer progression. Eur Urol. 2017;72(5):835–44.PubMedPubMedCentralCrossRef
22.
go back to reference Chen J, et al. Androgen-deprivation therapy with enzalutamide enhances prostate cancer metastasis via decreasing the EPHB6 suppressor expression. Cancer Lett. 2017;408:155–63.PubMedCrossRef Chen J, et al. Androgen-deprivation therapy with enzalutamide enhances prostate cancer metastasis via decreasing the EPHB6 suppressor expression. Cancer Lett. 2017;408:155–63.PubMedCrossRef
23.
go back to reference Sha K, et al. TNF signaling mediates an enzalutamide-induced metastatic phenotype of prostate cancer and microenvironment cell co-cultures. Oncotarget. 2015;6(28):25726–40.PubMedPubMedCentralCrossRef Sha K, et al. TNF signaling mediates an enzalutamide-induced metastatic phenotype of prostate cancer and microenvironment cell co-cultures. Oncotarget. 2015;6(28):25726–40.PubMedPubMedCentralCrossRef
24.
go back to reference Xu M, et al. Up-regulation of LAT1 during Antiandrogen therapy contributes to progression in prostate cancer cells. J Urol. 2016;195(5):1588–97.PubMedCrossRef Xu M, et al. Up-regulation of LAT1 during Antiandrogen therapy contributes to progression in prostate cancer cells. J Urol. 2016;195(5):1588–97.PubMedCrossRef
25.
26.
go back to reference Thompson MR, Xu D, Williams BR. ATF3 transcription factor and its emerging roles in immunity and cancer. J Mol Med (Berl). 2009;87(11):1053–60.CrossRef Thompson MR, Xu D, Williams BR. ATF3 transcription factor and its emerging roles in immunity and cancer. J Mol Med (Berl). 2009;87(11):1053–60.CrossRef
27.
go back to reference Lim R, et al. ATF3 is a negative regulator of inflammation in human fetal membranes. Placenta. 2016;47:63–72.PubMedCrossRef Lim R, et al. ATF3 is a negative regulator of inflammation in human fetal membranes. Placenta. 2016;47:63–72.PubMedCrossRef
28.
go back to reference Yan F, et al. Overexpression of the transcription factor ATF3 with a regulatory molecular signature associates with the pathogenic development of colorectal cancer. Oncotarget. 2017;8(29):47020–36.PubMedPubMedCentralCrossRef Yan F, et al. Overexpression of the transcription factor ATF3 with a regulatory molecular signature associates with the pathogenic development of colorectal cancer. Oncotarget. 2017;8(29):47020–36.PubMedPubMedCentralCrossRef
29.
go back to reference Yuan X, et al. ATF3 suppresses metastasis of bladder cancer by regulating gelsolin-mediated remodeling of the actin cytoskeleton. Cancer Res. 2013;73(12):3625–37.PubMedCrossRef Yuan X, et al. ATF3 suppresses metastasis of bladder cancer by regulating gelsolin-mediated remodeling of the actin cytoskeleton. Cancer Res. 2013;73(12):3625–37.PubMedCrossRef
30.
go back to reference Xie JJ, et al. ATF3 functions as a novel tumor suppressor with prognostic significance in esophageal squamous cell carcinoma. Oncotarget. 2014;5(18):8569–82.PubMedPubMedCentralCrossRef Xie JJ, et al. ATF3 functions as a novel tumor suppressor with prognostic significance in esophageal squamous cell carcinoma. Oncotarget. 2014;5(18):8569–82.PubMedPubMedCentralCrossRef
31.
go back to reference Hung JY, et al. Subamolide a induces mitotic catastrophe accompanied by apoptosis in human lung cancer cells. Evid Based Complement Alternat Med. 2013;2013:828143.PubMedPubMedCentral Hung JY, et al. Subamolide a induces mitotic catastrophe accompanied by apoptosis in human lung cancer cells. Evid Based Complement Alternat Med. 2013;2013:828143.PubMedPubMedCentral
32.
go back to reference Edagawa M, et al. Role of activating transcription factor 3 (ATF3) in endoplasmic reticulum (ER) stress-induced sensitization of p53-deficient human colon cancer cells to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis through up-regulation of death receptor 5 (DR5) by zerumbone and celecoxib. J Biol Chem. 2014;289(31):21544–61.PubMedPubMedCentralCrossRef Edagawa M, et al. Role of activating transcription factor 3 (ATF3) in endoplasmic reticulum (ER) stress-induced sensitization of p53-deficient human colon cancer cells to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis through up-regulation of death receptor 5 (DR5) by zerumbone and celecoxib. J Biol Chem. 2014;289(31):21544–61.PubMedPubMedCentralCrossRef
33.
go back to reference Park GH, et al. Cytotoxic activity of the twigs of Cinnamomum cassia through the suppression of cell proliferation and the induction of apoptosis in human colorectal cancer cells. BMC Complement Altern Med. 2018;18(1):28.PubMedPubMedCentralCrossRef Park GH, et al. Cytotoxic activity of the twigs of Cinnamomum cassia through the suppression of cell proliferation and the induction of apoptosis in human colorectal cancer cells. BMC Complement Altern Med. 2018;18(1):28.PubMedPubMedCentralCrossRef
34.
35.
go back to reference Pelzer AE, et al. The expression of transcription factor activating transcription factor 3 in the human prostate and its regulation by androgen in prostate cancer. J Urol. 2006;175(4):1517–22.PubMedCrossRef Pelzer AE, et al. The expression of transcription factor activating transcription factor 3 in the human prostate and its regulation by androgen in prostate cancer. J Urol. 2006;175(4):1517–22.PubMedCrossRef
36.
go back to reference Liu Y, et al. Induction of DNA damage and ATF3 by retigeric acid B, a novel topoisomerase II inhibitor, promotes apoptosis in prostate cancer cells. Cancer Lett. 2013;337(1):66–76.PubMedCrossRef Liu Y, et al. Induction of DNA damage and ATF3 by retigeric acid B, a novel topoisomerase II inhibitor, promotes apoptosis in prostate cancer cells. Cancer Lett. 2013;337(1):66–76.PubMedCrossRef
37.
go back to reference Jiang X, et al. Potential dual role of activating transcription factor 3 in colorectal cancer. Anticancer Res. 2016;36(2):509–16.PubMed Jiang X, et al. Potential dual role of activating transcription factor 3 in colorectal cancer. Anticancer Res. 2016;36(2):509–16.PubMed
39.
go back to reference Wen S, et al. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. Am J Pathol. 2015;185(2):293–301.PubMedPubMedCentralCrossRef Wen S, et al. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. Am J Pathol. 2015;185(2):293–301.PubMedPubMedCentralCrossRef
40.
go back to reference Soh SF, et al. Determination of androgen receptor degradation enhancer ASC-J9((R)) in mouse sera and organs with liquid chromatography tandem mass spectrometry. J Pharm Biomed Anal. 2014;88:117–22.PubMedCrossRef Soh SF, et al. Determination of androgen receptor degradation enhancer ASC-J9((R)) in mouse sera and organs with liquid chromatography tandem mass spectrometry. J Pharm Biomed Anal. 2014;88:117–22.PubMedCrossRef
42.
go back to reference Chandran UR, et al. Gene expression profiles of prostate cancer reveal involvement of multiple molecular pathways in the metastatic process. BMC Cancer. 2007;7:64.PubMedPubMedCentralCrossRef Chandran UR, et al. Gene expression profiles of prostate cancer reveal involvement of multiple molecular pathways in the metastatic process. BMC Cancer. 2007;7:64.PubMedPubMedCentralCrossRef
43.
go back to reference Varambally S, et al. Integrative genomic and proteomic analysis of prostate cancer reveals signatures of metastatic progression. Cancer Cell. 2005;8(5):393–406.PubMedCrossRef Varambally S, et al. Integrative genomic and proteomic analysis of prostate cancer reveals signatures of metastatic progression. Cancer Cell. 2005;8(5):393–406.PubMedCrossRef
44.
go back to reference Tomlins SA, et al. Integrative molecular concept modeling of prostate cancer progression. Nat Genet. 2007;39(1):41–51.PubMedCrossRef Tomlins SA, et al. Integrative molecular concept modeling of prostate cancer progression. Nat Genet. 2007;39(1):41–51.PubMedCrossRef
45.
go back to reference Zhao Q, et al. Echinacoside protects against MPP(+)-induced neuronal apoptosis via ROS/ATF3/CHOP pathway regulation. Neurosci Bull. 2016;32(4):349–62.PubMedPubMedCentralCrossRef Zhao Q, et al. Echinacoside protects against MPP(+)-induced neuronal apoptosis via ROS/ATF3/CHOP pathway regulation. Neurosci Bull. 2016;32(4):349–62.PubMedPubMedCentralCrossRef
46.
go back to reference Xu J, et al. Sorafenib with ASC-J9((R)) synergistically suppresses the HCC progression via altering the pSTAT3-CCL2/Bcl2 signals. Int J Cancer. 2017;140(3):705–17.PubMedCrossRef Xu J, et al. Sorafenib with ASC-J9((R)) synergistically suppresses the HCC progression via altering the pSTAT3-CCL2/Bcl2 signals. Int J Cancer. 2017;140(3):705–17.PubMedCrossRef
47.
go back to reference Cheng MA, et al. Androgen receptor (AR) degradation enhancer ASC-J9((R)) in an FDA-approved formulated solution suppresses castration resistant prostate cancer cell growth. Cancer Lett. 2018;417:182–91.PubMedCrossRef Cheng MA, et al. Androgen receptor (AR) degradation enhancer ASC-J9((R)) in an FDA-approved formulated solution suppresses castration resistant prostate cancer cell growth. Cancer Lett. 2018;417:182–91.PubMedCrossRef
48.
go back to reference Verderio P, et al. Antiproliferative effect of ASC-J9 delivered by PLGA nanoparticles against estrogen-dependent breast cancer cells. Mol Pharm. 2014;11(8):2864–75.PubMedCrossRef Verderio P, et al. Antiproliferative effect of ASC-J9 delivered by PLGA nanoparticles against estrogen-dependent breast cancer cells. Mol Pharm. 2014;11(8):2864–75.PubMedCrossRef
49.
go back to reference Hilchie AL, et al. Curcumin-induced apoptosis in PC3 prostate carcinoma cells is caspase-independent and involves cellular ceramide accumulation and damage to mitochondria. Nutr Cancer. 2010;62(3):379–89.PubMedCrossRef Hilchie AL, et al. Curcumin-induced apoptosis in PC3 prostate carcinoma cells is caspase-independent and involves cellular ceramide accumulation and damage to mitochondria. Nutr Cancer. 2010;62(3):379–89.PubMedCrossRef
51.
go back to reference Jacobson JR, et al. Simvastatin attenuates vascular leak and inflammation in murine inflammatory lung injury. Am J Phys Lung Cell Mol Phys. 2005;288(6):L1026–32. Jacobson JR, et al. Simvastatin attenuates vascular leak and inflammation in murine inflammatory lung injury. Am J Phys Lung Cell Mol Phys. 2005;288(6):L1026–32.
52.
go back to reference Khan A, et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 2018;46(D1):D260–6.PubMedCrossRef Khan A, et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 2018;46(D1):D260–6.PubMedCrossRef
53.
go back to reference Stafford LJ, Vaidya KS, Welch DR. Metastasis suppressors genes in cancer. Int J Biochem Cell Biol. 2008;40(5):874–91.PubMedCrossRef Stafford LJ, Vaidya KS, Welch DR. Metastasis suppressors genes in cancer. Int J Biochem Cell Biol. 2008;40(5):874–91.PubMedCrossRef
55.
go back to reference Chan JYW, et al. RNA-Seq revealed ATF3-regulated inflammation induced by silica. Toxicology. 2018;393:34–41.PubMedCrossRef Chan JYW, et al. RNA-Seq revealed ATF3-regulated inflammation induced by silica. Toxicology. 2018;393:34–41.PubMedCrossRef
57.
go back to reference Lin W, et al. ASC-J9((R)) suppresses prostate cancer cell invasion via altering the sumoylation-phosphorylation of STAT3. Cancer Lett. 2018;425:21–30.PubMedCrossRef Lin W, et al. ASC-J9((R)) suppresses prostate cancer cell invasion via altering the sumoylation-phosphorylation of STAT3. Cancer Lett. 2018;425:21–30.PubMedCrossRef
58.
go back to reference Chou FJ, et al. Preclinical study using androgen receptor (AR) degradation enhancer to increase radiotherapy efficacy via targeting radiation-increased AR to better suppress prostate cancer progression. EBioMedicine. 2019;40:504.PubMedPubMedCentralCrossRef Chou FJ, et al. Preclinical study using androgen receptor (AR) degradation enhancer to increase radiotherapy efficacy via targeting radiation-increased AR to better suppress prostate cancer progression. EBioMedicine. 2019;40:504.PubMedPubMedCentralCrossRef
Metadata
Title
ASC-J9® suppresses prostate cancer cell proliferation and invasion via altering the ATF3-PTK2 signaling
Authors
Hao Tian
Fu-ju Chou
Jing Tian
Yong Zhang
Bosen You
Chi-Ping Huang
Shuyuan Yeh
Yuanjie Niu
Chawnshang Chang
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01760-2

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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