4.5 Article

ATM-JAK-PD-L1 signaling pathway inhibition decreases EMT and metastasis of androgen-independent prostate cancer

Journal

MOLECULAR MEDICINE REPORTS
Volume 17, Issue 5, Pages 7045-7054

Publisher

SPANDIDOS PUBL LTD
DOI: 10.3892/mmr.2018.8781

Keywords

castration-resistant prostate cancer; metastasis; programmed cell death 1 ligand 1; ataxia telangiectasia mutated kinase; Janus kinase

Funding

  1. National Natural Science Foundation of China [81472776]
  2. Second Affiliated Hospital of Soochow University [XKQ2015008]

Ask authors/readers for more resources

Castration-resistant prostate cancer (CRPC), also known as androgen-independent prostate cancer, frequently develops local and distant metastases, the underlying mechanisms of which remain undetermined. In the present study, surgical specimens obtained from patients with clinical prostate cancer were investigated, and it was revealed that the expression levels of ataxia telangiectasia mutated kinase (ATM) were significantly enhanced in prostate cancer tissues isolated from patients with CRPC compared with from patients with hormone-dependent prostate cancer. CRPC C4-2 and CWR22Rv1 cells lines were subsequently selected to establish prostate cancer models, and ATM knockout cells were established via lentivirus infection. The results of the present study demonstrated that the migration and epithelial-mesenchymal transition (EMT) of ATM knockout cells were significantly decreased, which suggested that ATM is closely associated with CRPC cell migration and EMT. To further investigate the mechanisms underlying this process, programmed cell death 1 ligand 1 (PD-L1) expression was investigated in ATM knockout cells. In addition, inhibitors of Janus kinase (JAK) and signal transducer and activator of transcription 3 (STAT3; Stattic) were added to C4-2-Sc and CWR22Rv1-Sc cells, and the results demonstrated that PD-L1 expression was significantly decreased following the addition of JAK inhibitor 1; however, no significant change was observed following the addition of Stattic. Furthermore, a PD-L1 antibody and JAK inhibitor 1 were added to C4-2-Sc and CWR22Rv1-Sc cells, and it was revealed that cell migration ability was significantly decreased and the expression of EMT-associated markers was effectively reversed. The results of the present study suggested that via inhibition of the ATM-JAK-PD-L1 signaling pathway, EMT, metastasis and progression of CRPC may be effectively suppressed, which may represent a novel therapeutic approach for targeted therapy for patients with CRPC.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

Article Oncology

Screening key microRNAs for castration-resistant prostate cancer based on miRNA/mRNA functional synergistic network

Jin Zhu, Sugui Wang, Wenyu Zhang, Junyi Qiu, Yuxi Shan, Dongrong Yang, Bairong Shen

ONCOTARGET (2015)

Article Biotechnology & Applied Microbiology

miR-137-3p Modulates the Progression of Prostate Cancer by Regulating the JNK3/EZH2 Axis

Yachen Zang, Jin Zhu, Qin Li, Jian Tu, Xiaoqing Li, Rongkuan Hu, Dongrong Yang

ONCOTARGETS AND THERAPY (2020)

Article Andrology

Development and validation of an integrative methylation signature and nomogram for predicting survival in clear cell renal cell carcinoma

Qiliang Peng, Yibin Zhou, Lu Jin, Cheng Cao, Cheng Gao, Jianfang Zhou, Dongrong Yang, Jin Zhu

TRANSLATIONAL ANDROLOGY AND UROLOGY (2020)

Article Oncology

Oncolytic Adenovirus with SPAG9 shRNA Driven by DD3 Promoter Improved the Efficacy of Docetaxil for Prostate Cancer

Meng Lu, Fu-kun Wei, Chuang Wu, Zi-yang Xu, Li-jun Mao, Dong-rong Yang

Summary: In this study, a new oncolytic adenovirus, DD3-ZD55-SPAG9, was constructed to enhance the efficacy and safety of adenovirus using the DD3 promoter. The combined use of DD3-ZD55-SPAG9 and docetaxel significantly improved the anti-tumor efficacy of docetaxel in prostate cancer both in vitro and in vivo, possibly due to the induction of tumor cell apoptosis and the inhibition of tumor cell invasion.

JOURNAL OF ONCOLOGY (2022)

No Data Available