1
|
Zhang J, Wu Q, Xie Y, Li F, Wei H, Jiang Y, Qiao Y, Li Y, Sun Y, Huang H, Ge M, Zhao D, Dong Z, Liu K. Ribonucleotide reductase small subunit M2 promotes the proliferation of esophageal squamous cell carcinoma cells via HuR-mediated mRNA stabilization. Acta Pharm Sin B 2024; 14:4329-4344. [PMID: 39525580 PMCID: PMC11544187 DOI: 10.1016/j.apsb.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 11/16/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC), a malignancy of the digestive system, is highly prevalent and the primary cause of cancer-related deaths worldwide due to the lack of early diagnostic biomarkers and effective therapeutic targets. Dysregulated ribonucleotide reductase (RNR) expression has been confirmed to be causally linked to tumorigenesis. This study demonstrated that ribonucleotide reductase small subunit M2 (RRM2) is significantly upregulated in ESCC tissue and that its expression is negatively correlated with clinical outcomes. Mechanistically, HuR promotes RRM2 mRNA stabilization by binding to the adenine/uridine (AU)-rich elements (AREs) within the 3'UTR, resulting in persistent overexpression of RRM2. Furthermore, bifonazole is identified as an inhibitor of HuR via computational screening and molecular docking analysis. Bifonazole disrupts HuR-ARE interactions by competitively binding to HuR at F65, R97, I103, and R153 residues, resulting in reduced RRM2 expression. Furthermore, bifonazole exhibited antitumor effects on ESCC patient-derived xenograft (PDX) models by decreasing RRM2 expression and the dNTP pool. In summary, this study reveals the interaction network among HuR, RRM2, and bifonazole and demonstrated that bifonazole is a potential therapeutic compound for ESCC through inhibition of the HuR/RRM2 axis.
Collapse
Affiliation(s)
- Jing Zhang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Qiong Wu
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Yifei Xie
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou 450052, China
| | - Feng Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Huifang Wei
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Yanan Jiang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Yan Qiao
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yinhua Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yanan Sun
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Han Huang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Mengmeng Ge
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Dengyun Zhao
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Zigang Dong
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou 450052, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, China
| | - Kangdong Liu
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou 450052, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, China
| |
Collapse
|
2
|
Yuan Q, Shi Y, Zhang Y, Shi Y, Hussain Z, Zhao J, Jiang Y, Qiao Y, Guo Y, Lu J, Dong Z, Dong Z, Wang J, Liu K. Domperidone inhibits cell proliferation via targeting MEK and CDK4 in esophageal squamous cell carcinoma. Cancer Cell Int 2024; 24:114. [PMID: 38528618 DOI: 10.1186/s12935-024-03291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the leading causes of digestive system tumor related death in the world. Unfortunately, effective chemopreventive agent is lack for patients with ESCC in clinical practice, which leads to the extremely high mortality rate. METHODS A library of prescribed drugs was screened for finding critical anti-tumor properties in ESCC cells. The phosphoproteomics, kinase array, pulldown assay and drug affinity responsive target stabilization assay (DARTS) were applied to explore mechanisms and searched for synergistic targets. Established models of PDX in mice were used to determine the therapeutic effect of domperidone. RESULTS After screening a library of prescribed drugs, we discovered that domperidone has anti-tumor properties. Domperidone, acting as a gastroprokinetic agent, has been widely used in clinic for gastrointestinal motility disorders. Despite limited research, there are indications that domperidone may have anti-tumor properties. In this study, we determined that domperidone significantly inhibited ESCC proliferation in vitro and in vivo. We employed phosphoproteomics to reveal p-ERK, and p-SMAD3 down-regulation upon domperidone treatment. Then, the results of kinase assay and pulldown assay further validated that domperidone directly combined with MEK1/2 and CDK4, leading to the inhibition of their kinase activity. Furthermore, our results revealed that MEK/ERK and CDK4/SMAD3 signal pathway were major pathways in domperidone against ESCC. CONCLUSION Collectively, these findings suggest that domperidone serves as an effective "multi-target" inhibitor of MEK1/2 and CDK4, offering potential benefits for the chemoprevention of ESCC.
Collapse
Affiliation(s)
- Qiang Yuan
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yunshu Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yuhan Zhang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yaqian Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zubair Hussain
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jimin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yan Qiao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yaping Guo
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jing Lu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Ziming Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zigang Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China.
| | - Junyong Wang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
3
|
Ma L, Li M, Gou S, Wang W, Liu K, Zhang Y. Native-compound-Coupled Affinity Matrix (NCAM) in target identification and validation of bioactive compounds: Application, mechanism and outlooks. Bioorg Chem 2023; 140:106828. [PMID: 37690368 DOI: 10.1016/j.bioorg.2023.106828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/02/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023]
Abstract
In drug discovery and development, the direct target identification of bioactive small molecules plays a significant role for understanding the mechanism of action, predicting the side effects, and rationally designing more potent compounds. However, due to the complicated regulatory processes in a cell together with thousands of biomacromolecules, target identification is always the major obstacle. New methods and technologies are continuously invented to tackle this problem. Nevertheless, the mainly used tools possess several disadvantages. High synthetic skills are typically required to laboriously synthesize a probe for protein enrichment. To detect the ligand-protein interaction by analyzing proteins' responses to proteolytic or thermal treatment, costly and precise instruments are always necessary. Therefore, convenient and practical techniques are urgently needed. Over the past decades, a strategy using native compounds without the requirement of chemical modification, also termed Native-compound-Coupled Affinity Matrix (NCAM), is developing continuously. Two practical tactics based on "label-free" compounds have been invented and used, that is Photo-cross-linked Small-molecule Affinity Matrix (PSAM) and Native-compound-Coupled CNBr-activated Beads (NCCB). Presently, we will elucidate the characteristics, coupling mechanism, advantages and disadvantages, and future prospect of NCAM in specific target identification and validation.
Collapse
Affiliation(s)
- Lu Ma
- Basic Medical Research Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglong Li
- Basic Medical Research Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Shanshan Gou
- Basic Medical Research Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Wei Wang
- Departments of Pharmacology & Toxicology and Chemistry & Biochemistry, and BIO5 Institute, University of Arizona, Tucson, AZ 85721, United States
| | - Kangdong Liu
- Basic Medical Research Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yueteng Zhang
- Basic Medical Research Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
4
|
Cheng M, Xin Q, Ma S, Ge M, Wang F, Yan X, Jiang B. Advances in the Theranostics of Oesophageal Squamous Carcinoma. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202200251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Indexed: 01/04/2025]
Abstract
AbstractOesophageal squamous carcinoma (ESCC) is one of the most lethal human malignancies, and it is a more aggressive form of oesophageal cancer (EC) that comprises over 90% of all EC cases in China compared with oesophageal adenocarcinoma (EAC). The high mortality of ESCC is attributed to the late‐stage diagnosis, chemoradiotherapy resistance, and lack of appropriate therapeutic targets and corresponding therapeutic formulations. Recently, emerging clinical and translational investigations have involved genome analyses, diagnostic biomarkers, and targeted therapy for ESCC, and these studies provide a new horizon for improving the clinical outcomes of patients with ESCC. Here, the latest research advances in the theranostics of ESCC are reviewed and the unique features of ESCC (including differences from EAC, genomic alterations, and microbe infections), tissue and circulating biomarkers, chemoradiotherapy resistance, clinical targeted therapy for ESCC, identification of novel therapeutic targets, and designation of nanotherapeutic systems for ESCC are particularly focused on. Finally, the perspectives for future clinical and translational theranostic research of ESCC are discussed and the obstacles that must be overcome in ESCC theranostics are described.
Collapse
Affiliation(s)
- Miaomiao Cheng
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Qi Xin
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Saiyu Ma
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Mengyue Ge
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Feng Wang
- Oncology Department The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450000 China
| | - Xiyun Yan
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Protein and Peptide Pharmaceuticals Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| | - Bing Jiang
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
| |
Collapse
|
5
|
Wu W, Xu J, Gao D, Xie Z, Chen W, Li W, Yuan Q, Duan L, Zhang Y, Yang X, Chen Y, Dong Z, Liu K, Jiang Y. TOPK promotes the growth of esophageal cancer in vitro and in vivo by enhancing YB1/eEF1A1 signal pathway. Cell Death Dis 2023; 14:364. [PMID: 37328464 PMCID: PMC10276051 DOI: 10.1038/s41419-023-05883-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/30/2023] [Accepted: 06/08/2023] [Indexed: 06/18/2023]
Abstract
T-LAK-originated protein kinase (TOPK), a dual specificity serine/threonine kinase, is up-regulated and related to poor prognosis in many types of cancers. Y-box binding protein 1 (YB1) is a DNA/RNA binding protein and serves important roles in multiple cellular processes. Here, we reported that TOPK and YB1 were both highly expressed in esophageal cancer (EC) and correlated with poor prognosis. TOPK knockout effectively suppressed EC cell proliferation and these effects were reversible by rescuing YB1 expression. Notably, TOPK phosphorylated YB1 at Thr 89 (T89) and Ser 209 (S209) amino acid residues, then the phosphorylated YB1 bound with the promoter of the eukaryotic translation elongation factor 1 alpha 1 (eEF1A1) to activate its transcription. Consequently, the AKT/mTOR signal pathway was activated by up-regulated eEF1A1 protein. Importantly, TOPK inhibitor HI-TOPK-032 suppressed the EC cell proliferation and tumor growth by TOPK/YB1/eEF1A1 signal pathway in vitro and in vivo. Taken together, our study reveals that TOPK and YB1 are essential for the growth of EC, and TOPK inhibitors may be applied to retard cell proliferation in EC. This study highlights the promising therapeutic potential of TOPK as a target for treatment of EC.
Collapse
Affiliation(s)
- Wenjie Wu
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Jialuo Xu
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Dan Gao
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenliang Xie
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjing Chen
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjing Li
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Qiang Yuan
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Lina Duan
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Yuhan Zhang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Xiaoxiao Yang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Yingying Chen
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ziming Dong
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Kangdong Liu
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Research Center of Basic Medical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, 450052, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, 450000, China.
| | - Yanan Jiang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Research Center of Basic Medical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
6
|
Li M, Duan L, Wu W, Li W, Zhao L, Li A, Lu X, He X, Dong Z, Liu K, Jiang Y. Vortioxetine hydrobromide inhibits the growth of gastric cancer cells in vivo and in vitro by targeting JAK2 and SRC. Oncogenesis 2023; 12:24. [PMID: 37147297 PMCID: PMC10163056 DOI: 10.1038/s41389-023-00472-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/07/2023] Open
Abstract
Gastric cancer is the fourth leading cause of cancer deaths worldwide. Most patients are diagnosed in the advanced stage. Inadequate therapeutic strategies and the high recurrence rate lead to the poor 5-year survival rate. Therefore, effective chemopreventive drugs for gastric cancer are urgently needed. Repurposing clinical drugs is an effective strategy for discovering cancer chemopreventive drugs. In this study, we find that vortioxetine hydrobromide, an FDA-approved drug, is a dual JAK2/SRC inhibitor, and has inhibitory effects on cell proliferation of gastric cancer. Computational docking analysis, pull-down assay, cellular thermal shift assay (CETSA) and in vitro kinase assays are used to illustrate vortioxetine hydrobromide directly binds to JAK2 and SRC kinases and inhibits their kinase activities. The results of non-reducing SDS-PAGE and Western blotting indicate that vortioxetine hydrobromide suppresses STAT3 dimerization and nuclear translocation activity. Furthermore, vortioxetine hydrobromide inhibits the cell proliferation dependent on JAK2 and SRC and suppresses the growth of gastric cancer PDX model in vivo. These data demonstrate that vortioxetine hydrobromide, as a novel dual JAK2/SRC inhibitor, curbs the growth of gastric cancer in vitro and in vivo by JAK2/SRC-STAT3 signaling pathways. Our results highlight that vortioxetine hydrobromide has the potential application in the chemoprevention of gastric cancer.
Collapse
Affiliation(s)
- Mingzhu Li
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Lina Duan
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Wenjie Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Wenjing Li
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Lili Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Ang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Xuebo Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Xinyu He
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China.
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China.
- Center for Basic Medical Research, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Yanan Jiang
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
7
|
Liu Y, Sun X, Liu J, Liu W, Jin J, Liu Y. Baicalein Inhibits the Growth of Transplanted Esophageal Cancer in Mice and the Effect on the Expression of PAK4. Bull Exp Biol Med 2023; 174:478-481. [PMID: 36899200 DOI: 10.1007/s10517-023-05733-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Indexed: 03/12/2023]
Abstract
We studied the mechanism underlying the effect of baicalein on the growth of transplanted esophageal cancer in NOG mice and its effect on the expression of PAK4. For that purpose, we developed a new model of transplanted esophageal cancer (human esophageal cancer OE19 cells (107 cells/ml) were inoculated to NOG mice). Three experimental groups with transplanted esophageal cancer cells received baicalein in different doses (1, 1.5, and 2 mg/kg). In 32 days, the tumors were resected, and the expression of PAK4 and the level of activated PAK4 were assayed by reverse transcription PCR and Western blotting, respectively. The results showed a dose-depending anti-tumor effect of baicalein on the transplanted esophageal cancer in NOG mice: this effect of baicalein (determined by the size and weight of the tumor) increased with increasing the dose of the substance. Furthermore, the anti-tumor effect of baicalein was also confirmed by reduction of PAK4 expression. Thus, baicalein can inhibit tumor growth by inhibiting activation of PAK4. Therefore, our results showed that baicalein could inhibit the growth of esophageal cancer cells by inhibiting the activity of PAK4, which can be an important mechanism of its antitumor effect.
Collapse
Affiliation(s)
- Yao Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University/Hebei Tumor Hospital, Shijiazhuang, China
| | - X Sun
- Department of Pathology, The Fourth Hospital of Hebei Medical University/Hebei Tumor Hospital, Shijiazhuang, China
| | - J Liu
- Health Examination Center, Langfang TCM Hospital, Langfang, China
| | - W Liu
- Department of Anorectal Surgery, Langfang TCM Hospital, Langfang, China
| | - J Jin
- Scientific Research Center, The Fourth Hospital of Hebei Medical University/Hebei Tumor Hospital, Shijiazhuang, China
| | - Yu Liu
- Department of Gastrointestinal Surgery, The Fourth Hospital of Hebei Medical University/Hebei Tumor Hospital, Shijiazhuang, China.
| |
Collapse
|
8
|
Qian H. Azelnidipine may be a valuable drug for chemoprevention of ESCC with high MEK1/2 levels. Mol Ther Oncolytics 2022; 27:203. [DOI: 10.1016/j.omto.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
9
|
Bennett AN, Huang RX, He Q, Lee NP, Sung WK, Chan KHK. Drug repositioning for esophageal squamous cell carcinoma. Front Genet 2022; 13:991842. [PMID: 36246638 PMCID: PMC9554346 DOI: 10.3389/fgene.2022.991842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Esophageal cancer (EC) remains a significant challenge globally, having the 8th highest incidence and 6th highest mortality worldwide. Esophageal squamous cell carcinoma (ESCC) is the most common form of EC in Asia. Crucially, more than 90% of EC cases in China are ESCC. The high mortality rate of EC is likely due to the limited number of effective therapeutic options. To increase patient survival, novel therapeutic strategies for EC patients must be devised. Unfortunately, the development of novel drugs also presents its own significant challenges as most novel drugs do not make it to market due to lack of efficacy or safety concerns. A more time and cost-effective strategy is to identify existing drugs, that have already been approved for treatment of other diseases, which can be repurposed to treat EC patients, with drug repositioning. This can be achieved by comparing the gene expression profiles of disease-states with the effect on gene-expression by a given drug. In our analysis, we used previously published microarray data and identified 167 differentially expressed genes (DEGs). Using weighted key driver analysis, 39 key driver genes were then identified. These driver genes were then used in Overlap Analysis and Network Analysis in Pharmomics. By extracting drugs common to both analyses, 24 drugs are predicted to demonstrate therapeutic effect in EC patients. Several of which have already been shown to demonstrate a therapeutic effect in EC, most notably Doxorubicin, which is commonly used to treat EC patients, and Ixazomib, which was recently shown to induce apoptosis and supress growth of EC cell lines. Additionally, our analysis predicts multiple psychiatric drugs, including Venlafaxine, as repositioned drugs. This is in line with recent research which suggests that psychiatric drugs should be investigated for use in gastrointestinal cancers such as EC. Our study shows that a drug repositioning approach is a feasible strategy for identifying novel ESCC therapies and can also improve the understanding of the mechanisms underlying the drug targets.
Collapse
Affiliation(s)
- Adam N. Bennett
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Rui Xuan Huang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qian He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Nikki P. Lee
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wing-Kin Sung
- Department of Computer Sciences, National University of Singapore, Singapore, Singapore
| | - Kei Hang Katie Chan
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Epidemiology, Centre for Global Cardiometabolic Health, Brown University, Providence, RI, United States
| |
Collapse
|
10
|
Li Y, Lu Q, Xie C, Yu Y, Zhang A. Recent advances on development of p21-activated kinase 4 inhibitors as anti-tumor agents. Front Pharmacol 2022; 13:956220. [PMID: 36105226 PMCID: PMC9465411 DOI: 10.3389/fphar.2022.956220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 12/05/2022] Open
Abstract
The p21-activated kinase 4 (PAK4) is a member of the PAKs family. It is overexpressed in multiple tumor tissues. Pharmacological inhibition of PAK4 attenuates proliferation, migration, and invasion of cancer cells. Recent studies revealed that inhibition of PAK4 sensitizes immunotherapy which has been extensively exploited as a new strategy to treat cancer. In the past few years, a large number of PAK4 inhibitors have been reported. Of note, the allosteric inhibitor KPT-9274 has been tested in phase Ⅰ clinic trials. Herein, we provide an update on recent research progress on the PAK4 mediated signaling pathway and highlight the development of the PAK4 small molecular inhibitors in recent 5 years. Meanwhile, challenges, limitations, and future developmental directions will be discussed as well.
Collapse
Affiliation(s)
- Yang Li
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Lu
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Chenghu Xie
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yiming Yu
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ao Zhang
- Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Ao Zhang,
| |
Collapse
|
11
|
Brown JS. Treatment of cancer with antipsychotic medications: Pushing the boundaries of schizophrenia and cancer. Neurosci Biobehav Rev 2022; 141:104809. [PMID: 35970416 DOI: 10.1016/j.neubiorev.2022.104809] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 10/15/2022]
Abstract
Over a century ago, the phenothiazine dye, methylene blue, was discovered to have both antipsychotic and anti-cancer effects. In the 20th-century, the first phenothiazine antipsychotic, chlorpromazine, was found to inhibit cancer. During the years of elucidating the pharmacology of the phenothiazines, reserpine, an antipsychotic with a long historical background, was likewise discovered to have anti-cancer properties. Research on the effects of antipsychotics on cancer continued slowly until the 21st century when efforts to repurpose antipsychotics for cancer treatment accelerated. This review examines the history of these developments, and identifies which antipsychotics might treat cancer, and which cancers might be treated by antipsychotics. The review also describes the molecular mechanisms through which antipsychotics may inhibit cancer. Although the overlap of molecular pathways between schizophrenia and cancer have been known or suspected for many years, no comprehensive review of the subject has appeared in the psychiatric literature to assess the significance of these similarities. This review fills that gap and discusses what, if any, significance the similarities have regarding the etiology of schizophrenia.
Collapse
|