1
|
Gan L, Zhou L, Chu AL, Sun C, Wang Y, Yang M, Liu Z. GPD1L may inhibit the development of esophageal squamous cell carcinoma through the PI3K/AKT signaling pathway: bioinformatics analysis and experimental exploration. Mol Biol Rep 2024; 51:1149. [PMID: 39535578 DOI: 10.1007/s11033-024-10070-1] [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: 08/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Esophageal squamous carcinoma (ESCC) is the most prevalent pathological subtype of esophageal cancer (EC). It has the characteristics of significant local invasion, quick disease progression, high recurrence rates, and a dismal prognosis for survival. Phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/AKT) is a signaling system whose aberrant activation regulates downstream factors, leading to the promotion of cancer development. This study looks at a protein called Glycerol-3-phosphate dehydrogenase 1-like (GPD1L), which strongly affects the development of several cancers. However, its association with ESCC development and its underlying mechanisms are not clear. METHODS In this paper, we analyzed six ESCC transcriptome data obtained from the GEO database. We utilized bioinformatics technology and immunohistochemistry to differentially analyze GPD1L levels of mRNA and protein expression in ESCC and normal adjacent tissues. Furthermore, we conducted survival, co-expression, enrichment, immune infiltration and drug sensitivity analysis. Moreover, we further investigated the role and mechanism of GPD1L by Western Blot (WB), Cell Counting Kit-8 (CCK8), wound healing assay, Transwell assay, and flow cytometry. Finally, the addition of IGF-1, the activator of PI3K/AKT, could rescue the inhibitory effect of GPD1L on ESCC. RESULTS The findings manifest that the expression of GPD1L was low in ESCC, and functional experiments showed that GPD1L promoted apoptosis in vitro while blocking cell migration, invasion, and proliferation. Based on mechanism research, GPD1L's impact on ESCC could be explained by its suppression of the PI3K/AKT signaling pathway's activation. CONCLUSION To sum up, our findings imply that GPD1L may impede the initiation and advancement of ESCC via modulating the PI3K/AKT signaling pathway. GPD1L is considered to be a promising therapeutic target and biomarker to diagnose and treat ESCC.
Collapse
Affiliation(s)
- LanLan Gan
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
- Tumor Radiotherapy Department, The Second Clinical Medical School of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - Lu Zhou
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
- Tumor Radiotherapy Department, The Second Clinical Medical School of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - ALan Chu
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - Chen Sun
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - YongTai Wang
- Tumor Radiotherapy Department, The Second Clinical Medical School of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - MengLin Yang
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - ZongWen Liu
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China.
| |
Collapse
|
2
|
Liang F, Xu H, Cheng H, Zhao Y, Zhang J. Patient-derived tumor models: a suitable tool for preclinical studies on esophageal cancer. Cancer Gene Ther 2023; 30:1443-1455. [PMID: 37537209 DOI: 10.1038/s41417-023-00652-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
Esophageal cancer (EC) is the tenth most common cancer worldwide and has high morbidity and mortality. Its main subtypes include esophageal squamous cell carcinoma and esophageal adenocarcinoma, which are usually diagnosed during their advanced stages. The biological defects and inability of preclinical models to summarize completely the etiology of multiple factors, the complexity of the tumor microenvironment, and the genetic heterogeneity of tumors severely limit the clinical treatment of EC. Patient-derived models of EC not only retain the tissue structure, cell morphology, and differentiation characteristics of the original tumor, they also retain tumor heterogeneity. Therefore, compared with other preclinical models, they can better predict the efficacy of candidate drugs, explore novel biomarkers, combine with clinical trials, and effectively improve patient prognosis. This review discusses the methods and animals used to establish patient-derived models and genetically engineered mouse models, especially patient-derived xenograft models. It also discusses their advantages, applications, and limitations as preclinical experimental research tools to provide an important reference for the precise personalized treatment of EC and improve the prognosis of patients.
Collapse
Affiliation(s)
- Fan Liang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hongyan Xu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hongwei Cheng
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yabo Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China.
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
3
|
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: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
4
|
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
|
5
|
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
|
6
|
Mucke HA. Drug Repurposing Patent Applications July–September 2022. Assay Drug Dev Technol 2022; 20:359-366. [DOI: 10.1089/adt.2022.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
7
|
Lin X, Zhao Q, Fu B, Xiong Y, Zhang S, Xu S, Wu H. ISOC1 Modulates Inflammatory Responses in Macrophages through the AKT1/PEX11B/Peroxisome Pathway. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27185896. [PMID: 36144632 PMCID: PMC9505204 DOI: 10.3390/molecules27185896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022]
Abstract
Inflammation underlies a variety of physiological and pathological processes and plays an essential role in shaping the ensuing adaptive immune responses and in the control of pathogens. However, its physiological functions are not completely clear. Using a LPS-treated RAW264.7 macrophage inflammation model, we found that the production of inflammatory cytokines in ISOC1-deficient cells was significantly higher than that in the control group. It was further proved that ISOC1 deficiency could activate AKT1, and the overactivation of AKT1 could reduce the stability of PEX11B through protein modification, thereby reducing the peroxisome biogenesis and thus affecting inflammation. In this study, we reported for the first time the role of ISOC1 in innate immunity and elucidated the mechanism by which ISOC1 regulates inflammation through AKT1/PEX11B/peroxisome. Our results defined a new role of ISOC1 in the regulatory mechanism underlying the LPS-induced inflammatory response.
Collapse
|
8
|
Wang Z, Chen Y, Li X, Zhang Y, Zhao X, Zhou H, Lu X, Zhao L, Yuan Q, Shi Y, Zhao J, Dong Z, Jiang Y, Liu K. Tegaserod Maleate Suppresses the Growth of Gastric Cancer In Vivo and In Vitro by Targeting MEK1/2. Cancers (Basel) 2022; 14:cancers14153592. [PMID: 35892850 PMCID: PMC9332868 DOI: 10.3390/cancers14153592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022] Open
Abstract
Gastric cancer (GC) ranks fifth in global incidence and fourth in mortality. The current treatments for GC include surgery, chemotherapy and radiotherapy. Although treatment strategies for GC have been improved over the last decade, the overall five-year survival rate remains less than 30%. Therefore, there is an urgent need to find novel therapeutic or preventive strategies to increase GC patient survival rates. In the current study, we found that tegaserod maleate, an FDA-approved drug, inhibited the proliferation of gastric cancer cells, bound to MEK1/2 and suppressed MEK1/2 kinase activity. Moreover, tegaserod maleate inhibited the progress of gastric cancer by depending on MEK1/2. Notably, we found that tegaserod maleate suppressed tumor growth in the patient-derived gastric xenograft (PDX) model. We further compared the effect between tegaserod maleate and trametinib, which is a clinical MEK1/2 inhibitor, and confirmed that tegaserod maleate has the same effect as trametinib in inhibiting the growth of GC. Our findings suggest that tegaserod maleate inhibited GC proliferation by targeting MEK1/2.
Collapse
Affiliation(s)
- Zitong Wang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
| | - Yingying Chen
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
| | - Xiaoyu Li
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
| | - Yuhan Zhang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450001, China
| | - Xiaokun Zhao
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450001, China
| | - Hao Zhou
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
| | - Xuebo Lu
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450001, China
| | - Lili Zhao
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
| | - Qiang Yuan
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450001, China
| | - Yunshu Shi
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450001, China
| | - Jimin Zhao
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China
- Basic Medicine Research Center, Zhengzhou University, Zhengzhou 450001, China
| | - Ziming Dong
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China
- Basic Medicine Research Center, Zhengzhou University, Zhengzhou 450001, China
| | - Yanan Jiang
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China
- Basic Medicine Research Center, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (Y.J.); (K.L.)
| | - Kangdong Liu
- Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Z.W.); (Y.C.); (X.L.); (Y.Z.); (X.Z.); (H.Z.); (X.L.); (L.Z.); (Q.Y.); (Y.S.); (J.Z.); (Z.D.)
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China
- Basic Medicine Research Center, Zhengzhou University, Zhengzhou 450001, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450001, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450001, China
- Correspondence: (Y.J.); (K.L.)
| |
Collapse
|
9
|
Kumbhar P, Kole K, Yadav T, Bhavar A, Waghmare P, Bhokare R, Manjappa A, Jha NK, Chellappan DK, Shinde S, Singh SK, Dua K, Salawi A, Disouza J, Patravale V. Drug repurposing: An emerging strategy in alleviating skin cancer. Eur J Pharmacol 2022; 926:175031. [PMID: 35580707 DOI: 10.1016/j.ejphar.2022.175031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/22/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Skin cancer is one of the most common forms of cancer. Several million people are estimated to have affected with this condition worldwide. Skin cancer generally includes melanoma and non-melanoma with the former being the most dangerous. Chemotherapy has been one of the key therapeutic strategies employed in the treatment of skin cancer, especially in advanced stages of the disease. It could be also used as an adjuvant with other treatment modalities depending on the type of skin cancer. However, there are several shortfalls associated with the use of chemotherapy such as non-selectivity, tumour resistance, life-threatening toxicities, and the exorbitant cost of medicines. Furthermore, new drug discovery is a lengthy and costly process with minimal likelihood of success. Thus, drug repurposing (DR) has emerged as a new avenue where the drug approved formerly for the treatment of one disease can be used for the treatment of another disease like cancer. This approach is greatly beneficial over the de novo approach in terms of time and cost. Moreover, there is minimal risk of failure of repurposed therapeutics in clinical trials. There are a considerable number of studies that have reported on drugs repurposed for the treatment of skin cancer. Thus, the present manuscript offers a comprehensive overview of drugs that have been investigated as repurposing candidates for the efficient treatment of skin cancers mainly melanoma and its oncogenic subtypes, and non-melanoma. The prospects of repurposing phytochemicals against skin cancer are also discussed. Furthermore, repurposed drug delivery via topical route and repurposed drugs in clinical trials are briefed. Based on the findings from the reported studies discussed in this manuscript, drug repurposing emerges to be a promising approach and thus is expected to offer efficient treatment at a reasonable cost in devitalizing skin cancer.
Collapse
Affiliation(s)
- Popat Kumbhar
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Kapil Kole
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Tejashree Yadav
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Ashwini Bhavar
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Pramod Waghmare
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Rajdeep Bhokare
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Arehalli Manjappa
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, 201310, Uttar Pradesh, India; Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sunita Shinde
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, India
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - John Disouza
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur Maharashtra, 416113, India.
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, Maharashtra, 400019, India.
| |
Collapse
|
10
|
Tegaserod Maleate Inhibits Breast Cancer Progression and Enhances the Sensitivity of Immunotherapy. JOURNAL OF ONCOLOGY 2022; 2022:5320421. [PMID: 35154317 PMCID: PMC8831063 DOI: 10.1155/2022/5320421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/08/2022] [Indexed: 01/22/2023]
Abstract
Background. Breast cancer (BC) is the most commonly diagnosed cancer in women worldwide. The challenge in managing this heterogeneous malignancy is that BC is highly aggressive and is always associated with chemical resistance, radiation resistance, hormone therapy resistance, and targeted therapy resistance. Therefore, there is an urgent need to find effective drugs to treat BC. Methods. Based on the Selleck drug library approved by FDA, we screened 800 drugs for anti-BC cells and found that tegaserod maleate (TM), a 5-hydroxytryptamine 4-receptor (HTR4) partial agonist had the best anti-BC effect, which was further verified. The effects of different concentrations of TM on cell proliferation, invasion, and migration were evaluated in vitro using CCK8, plate cloning, transwell, and scratch assays. The UALCAN database, Kaplan–Meier Plotter database, Human Protein Atlas, and GEPIA2 were used to explore the correlation between HTR4 expression and BC patients’ clinicopathological data as well as immune response. In vivo experiments demonstrated the effect of the TM and immunotherapy drug (anti-PD1/anti-TIGIT) combination on BC tumor growth in mice. Results. TM significantly inhibited the proliferation, invasion, and migration of BC cells, and the higher the concentration, the better the inhibition effect. HTR4 was significantly downregulated in BC tissues compared to paracancerous tissues. The downregulation of HTR4 was correlated with clinicopathological data and positively correlated with BC prognosis. Interestingly, the GEPIA2 database suggested that there was a strong positive correlation between the expression of HTR4 and effector T cells, effector memory T cells, and exhausted T cells. In vitro experiments showed that TM, anti-PD1, and anti-TIGIT could all inhibit the growth and weight of BC tumors as compared with the control group. However, when anti-PD1 or anti-TIGIT was used simultaneously with TM, the inhibition of tumors significantly exceeded that in the control group. Moreover, the combination of anti-TIGIT and TM has the best inhibitory effect. Conclusion. TM inhibited the progression of breast cancer, and its combination with anti-TIGIT could effectively inhibit tumor growth and improve the sensitivity of immunotherapy in breast cancer.
Collapse
|
11
|
Oxethazaine inhibits esophageal squamous cell carcinoma proliferation and metastasis by targeting aurora kinase A. Cell Death Dis 2022; 13:189. [PMID: 35217647 PMCID: PMC8881465 DOI: 10.1038/s41419-022-04642-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 01/28/2022] [Accepted: 02/07/2022] [Indexed: 11/09/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC), a malignant neoplasm with high incidence, is a severe global public health threat. The current modalities used for treating ESCC include surgery, chemotherapy, and radiotherapy. Although ESCC management and treatment strategies have improved over the last decade, the overall 5-year survival rate remains <20%. Therefore, the identification of novel therapeutic strategies that can increase ESCC patient survival rates is urgently needed. Oxethazaine, an amino-amide anesthetic agent, is mainly prescribed in combination with antacids to relieve esophagitis, dyspepsia, and other gastric disorders. In the present study, we found that oxethazaine inhibited the proliferation and migration of esophageal cancer cells. According to the results of in vitro screening and binding assays, oxethazaine binds directly to AURKA, suppresses AURKA activity, and inhibits the downstream effectors of AURKA. Notably, we found that oxethazaine suppressed tumor growth in three patient-derived esophageal xenograft mouse models and tumor metastasis in vivo. Our findings suggest that oxethazaine can inhibit ESCC proliferation and metastasis in vitro and in vivo by targeting AURKA.
Collapse
|
12
|
Active Targeted Nanoemulsions for Repurposing of Tegaserod in Alzheimer's Disease Treatment. Pharmaceutics 2021; 13:pharmaceutics13101626. [PMID: 34683919 PMCID: PMC8540544 DOI: 10.3390/pharmaceutics13101626] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose: The activation of 5-HT4 receptors with agonists has emerged as a valuable therapeutic strategy to treat Alzheimer’s disease (AD) by enhancing the nonamyloidogenic pathway. Here, the potential therapeutic effects of tegaserod, an effective agent for irritable bowel syndrome, were assessed for AD treatment. To envisage its efficient repurposing, tegaserod-loaded nanoemulsions were developed and functionalized by a blood–brain barrier shuttle peptide. Results: The butyrylcholinesterase inhibitory activity of tegaserod and its neuroprotective cellular effects were highlighted, confirming the interest of this pleiotropic drug for AD treatment. In regard to its drugability profile, and in order to limit its peripheral distribution after IV administration, its encapsulation into monodisperse lipid nanoemulsions (Tg-NEs) of about 50 nm, and with neutral zeta potential characteristics, was performed. The stability of the formulation in stock conditions at 4 °C and in blood biomimetic medium was established. The adsorption on Tg-NEs of peptide-22 was realized. The functionalized NEs were characterized by chromatographic methods (SEC and C18/HPLC) and isothermal titration calorimetry, attesting the efficiency of the adsorption. From in vitro assays, these nanocarriers appeared suitable for enabling tegaserod controlled release without hemolytic properties. Conclusion: The developed peptide-22 functionalized Tg-NEs appear as a valuable tool to allow exploration of the repurposed tegaserod in AD treatment in further preclinical studies.
Collapse
|