1
|
Vaghari-Tabari M, Qujeq D, Hashemzadeh MS. Long noncoding RNAs as potential targets for overcoming chemoresistance in upper gastrointestinal cancers. Biomed Pharmacother 2024; 179:117368. [PMID: 39214010 DOI: 10.1016/j.biopha.2024.117368] [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: 05/27/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
In the last decade, researchers have paid much attention to the role of noncoding RNA molecules in human diseases. Among the most important of these molecules are LncRNAs, which are RNA molecules with a length of more than 200 nucleotides. LncRNAs can regulate gene expression through various mechanisms, such as binding to DNA sequences and interacting with miRNAs. Studies have shown that LncRNAs may be valuable therapeutic targets in treating various cancers, including upper-gastrointestinal cancers. Upper gastrointestinal cancers, mainly referring to esophageal and gastric cancers, are among the deadliest gastrointestinal cancers. Despite notable advances, traditional chemotherapy remains a common strategy for treating these cancers. However, chemoresistance poses a significant obstacle to the effective treatment of upper gastrointestinal cancers, resulting in a low survival rate. Chemoresistance arises from various events, such as the enhancement of efflux and detoxification of chemotherapy agents, reduction of drug uptake, alteration of drug targeting, reduction of prodrug activation, strengthening of EMT and stemness, and the attenuation of apoptosis in cancerous cells. Tumor microenvironment also plays an important role in chemoresistance. Interestingly, a series of studies have revealed that LncRNAs can influence important mechanisms associated with some of the aforementioned events and may serve as promising targets for mitigating chemoresistance in upper gastrointestinal cancers. In this review paper, following a concise overview of chemoresistance mechanisms in upper gastrointestinal cancers, we will review the most intriguing findings of these investigations in detail.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | | |
Collapse
|
2
|
Liu L, Wang Y, Dong Y, Lin S, Guan W, Song J. Resveratrol as a cardioprotective adjuvant for 5-fluorouracil in the treatment of gastric cancer cells. Braz J Med Biol Res 2024; 57:e13537. [PMID: 39258669 PMCID: PMC11379349 DOI: 10.1590/1414-431x2024e13537] [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: 11/16/2023] [Accepted: 07/17/2024] [Indexed: 09/12/2024] Open
Abstract
The clinical application of 5-fluorouracil (5-Fu), a potent chemotherapeutic agent, is often hindered by its well-documented cardiotoxic effects. Nevertheless, natural polyphenolic compounds like resveratrol (RES), known for their dual anti-tumor and cardioprotective properties, are potential adjunct therapeutic agents. In this investigation, we examined the combined utilization of RES and 5-Fu for the inhibition of gastric cancer using both in vitro and in vivo models, as well as their combined impact on cardiac cytotoxicity. Our study revealed that the co-administration of RES and 5-Fu effectively suppressed MFC cell viability, migration, and invasion, while also reducing tumor weight and volume. Mechanistically, the combined treatment prompted p53-mediated apoptosis and autophagy, leading to a considerable anti-tumor effect. Notably, RES mitigated the heightened oxidative stress induced by 5-Fu in cardiomyocytes, suppressed p53 and Bax expression, and elevated Bcl-2 levels. This favorable influence enhanced primary cardiomyocyte viability, decreased apoptosis and autophagy, and mitigated 5-Fu-induced cardiotoxicity. In summary, our findings suggested that RES holds promise as an adjunct therapy to enhance the efficacy of gastric cancer treatment in combination with 5-Fu, while simultaneously mitigating cardiotoxicity.
Collapse
Affiliation(s)
- Lilong Liu
- Pharmaceutical Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yexin Wang
- Pharmaceutical Department, Qingdao Mental Health Center, Qingdao, China
| | - Yanyan Dong
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Shan Lin
- Pharmaceutical Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenhui Guan
- Pharmaceutical Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jia Song
- Pharmaceutical Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Hu J, Li A, Guo Y, Ma T, Feng S. The relationship between tumor metabolism and 5-fluorouracil resistance. Biochem Pharmacol 2023; 218:115902. [PMID: 37922975 DOI: 10.1016/j.bcp.2023.115902] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Affiliation(s)
- Jingyi Hu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Anqi Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yueyang Guo
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ting Ma
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Siqi Feng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
4
|
Qu Y, Xue S, Zheng Y, Du Y, Zhang G, Huang L, Li H, Li H. Upregulated miR‑378a‑3p expression suppresses energy metabolism and promotes apoptosis by targeting a GLUT‑1/ALDOA/PKM2 axis in esophageal carcinoma. Oncol Lett 2023; 26:421. [PMID: 37664650 PMCID: PMC10472027 DOI: 10.3892/ol.2023.14007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 07/10/2023] [Indexed: 09/05/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive malignancy of the digestive system with increasing incidence and mortality rates. The biological roles of microRNA (miR)-378a-3p in tumor cells remain contested, and the mechanisms underlying the functions, energy metabolism, and cell survival mechanisms in ESCC cells are yet to be fully elucidated. In the present study, miR-378a-3p overexpression and negative control plasmids were transfected into ECA-109 cells using electroporation. Western blotting was used to detect the relative expression of proteins, and flow cytometry was used to detect cell apoptosis. Subsequently, ELISA assays were performed to determine enzyme activity, and an ATP detection kit was used to measure ATP content. Dual-luciferase reporter assays were performed to identify the target genes of miR-378a-3p. The results of the present study demonstrated that miR-378a-3p inhibited the gene expression and enzyme activities of glucose transporter protein 1 (GLUT-1), Aldolase A (ALDOA), and pyruvate kinase M2 (PKM2), all of which are involved in the glycolytic pathway of cells. Energy metabolism was suppressed by miR-378a-3p by reducing ATP content, and this downregulated the expression of Bcl-2 and Survivin. Moreover, increased miR-378a-3p expression promoted cell apoptosis in the early stages by increasing the expression levels and the activity of Bad and Caspase-3, while inhibiting the expression levels of Bcl-2 and Survivin. The results of the present study also demonstrated that GLUT-1/ALDOA/PKM2 were target genes of miR-378a-3p. Notably, miR-378a-3p blocked energy production and promoted the apoptosis of tumor cells via the downregulation of glycolytic enzyme expression and by reducing the mitochondrial membrane potential in ESCC. Bad, Caspase-3, Survivin, and Bcl-2 may be associated with blocking energy production and promoting apoptosis via miR-378a-3p in ESCC cells.
Collapse
Affiliation(s)
- Yuan Qu
- Department of Labour Hygiene and Sanitary Science, College of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Shan Xue
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Yujian Zheng
- Department of Labour Hygiene and Sanitary Science, College of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Yajing Du
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Guoping Zhang
- Tumor Department, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Liting Huang
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Hui Li
- Central Laboratory of Xinjiang Medical University, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Huiwu Li
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| |
Collapse
|
5
|
Zhang S, Liu S, Ren J, Zhang H, Chen S, Chen Y, Zhang S, Chen W, Xu C, Zhong S, Liu S, Lin C. Tumor-derived extracellular vesicles confer 5-fluorouracil resistance in esophageal cancer via long noncoding RNA AC116025.2 delivery. Mol Carcinog 2022; 61:1177-1190. [PMID: 36239547 DOI: 10.1002/mc.23469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023]
Abstract
5-Fluorouracil (5-FU) resistance is one of the main causes for treatment failure in esophageal cancer (EC). Here, we intended to elucidate the mechanism of tumor-derived extracellular vesicles (TEVs)-encapsulated long noncoding RNAs (lncRNAs) AC116025.2 in 5-FU resistance in EC. EVs were isolated from the serum samples of EC patients and HEEC, TE-1, and TE-1/5-FU cells, followed by RT-qPCR detection of AC116025.2 expression in EVs. The relationship among AC116025.2, microRNA (miR)-4496, and SEMA5A was evaluated. Next, EC cells were cocultured with EVs, followed by lentivirus transduction and plasmid transfection for studying the role of TEVs-AC116025.2 in EC cells in relation to miR-4496 and SEMA5A. Tumor formation in nude mice was applied for in vivo confirmation. Elevated AC116025.2 expression was seen in the EVs from the serum of 5-FU insensitive patients and from 5-FU-resistant EC cells. Mechanistically, AC116025.2 bound to miR-4496 that inversely targeted SEMA5A in EC cells. EVs-oe-AC116025.2 augmented EC cell viability, colony formation, and 5-FU resistance, but diminished their apoptosis through miR-4496-mediated SEMA5A. Furthermore, EVs-oe-AC116025.2 augmented tumor formation and 5-FU resistance of EC cells in vivo. Conclusively, our data offered evidence of the promoting mechanism of TEVs in the 5-FU resistance of EC by delivering AC116025.2.
Collapse
Affiliation(s)
- Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Shaojie Liu
- Department of Gastrointestinal Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Jingqing Ren
- Department of Gastrointestinal Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Hanshuo Zhang
- Department of Gastrointestinal Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Song Chen
- Department of Medical Imaging, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Shengqi Zhang
- Dafeng Hospital of Chaoyang District in Shantou City, Shantou, China
- Department of Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Wang Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Chengcheng Xu
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Jinan University, Guangzhou, China
| | - Shilong Zhong
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Sulin Liu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chaoxian Lin
- Shantou Chaonan Minsheng Hospital, Shantou, China
| |
Collapse
|
6
|
Tseng YC, Shu CW, Chang HM, Lin YH, Tseng YH, Hsu HS, Goan YG, Tseng CJ. Assessment of Early Growth Response 1 in Tumor Suppression of Esophageal Squamous Cell Carcinoma. J Clin Med 2022; 11:jcm11195792. [PMID: 36233659 PMCID: PMC9572560 DOI: 10.3390/jcm11195792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/08/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Esophageal squamous cell carcinoma (ESCC) is associated with poor survival despite surgical resection, and its pathogenesis has been broadly investigated in the past decade. Early growth response 1 (EGR-1) could involve regulating tumor development in ESCC cells. Methods: An attempt was made to examine the molecular and cellular influence of EGR-1 in esophageal cancer cells by RNA extraction, real-time PCR (qRT-PCR), cell culture, small interfering RNA (siRNA) knockdown, western blot, migration assay, and cell viability assay. One hundred and forty-four samples of ESCC were collected from our hospital and analyzed. Significantly higher EGR-1 expression was noted in tumor-adjacent normal tissue compared with tumor lesions. Results: The univariate analysis showed no significant impacts of EGR-1 expression on patients’ survival. However, after adjusting for the pathological stage, patients with EGR-1 expression > 68th percentile had lower risks of cancer-related death. Moreover, knockdown of EGR-1 significantly enhanced cell migration, invasion, and resistance to chemotherapeutic agents in two ESCC cell lines. Conclusions: EGR-1 plays a key role in tumor suppression involving tumor viability suppression and reflects the treatment effect of current chemotherapy for ESCC.
Collapse
Affiliation(s)
- Yen-Chiang Tseng
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Chih-Wen Shu
- Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (C.-W.S.); (C.-J.T.); Tel.: +886-7-3422121 (ext. 1505) (C.-J.T.); Fax: +886-7-3422288 (C.-J.T.)
| | - Hui-Min Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
| | - Yi-Hsuan Lin
- Department of Family Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei 100, Taiwan
| | - Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Han-Shui Hsu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yih-Gang Goan
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ching-Jiunn Tseng
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Correspondence: (C.-W.S.); (C.-J.T.); Tel.: +886-7-3422121 (ext. 1505) (C.-J.T.); Fax: +886-7-3422288 (C.-J.T.)
| |
Collapse
|
7
|
Nicolás-Morala J, Portillo-Esnaola M, Terrén S, Gutiérrez-Pérez M, Gilaberte Y, González S, Juarranz Á. In vitro 5-Fluorouracil resistance produces enhanced photodynamic therapy damage in SCC and tumor resistance in BCC. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 233:112483. [PMID: 35679749 DOI: 10.1016/j.jphotobiol.2022.112483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Non-melanoma skin cancer (NMSC) is the most common malignancy worldwide, with rising incidence in the recent years. It includes basal cell carcinoma (BCC), and squamous cell carcinoma (SCC). Several non-invasive therapies have been developed for its treatment such as topical 5-Fluorouracil (5FU) and photodynamic therapy (PDT), among others. Despite both are appropriated for NMSC treatment, recurrence cases have been reported. To prevent this, in this work we explore the potential of the combination of PDT and 5FU to treat SCC and BCC. First we evaluate the efficacy of PDT in cells resistant to 5FU. For this purpose, we use SCC-13 and CSZ-1 cells, obtained from a human SCC and a murine BCC, respectively. We first induced 5FU resistance in these cell lines by repeated treatments with the drug and then, the efficacy to PDT was evaluated. The results obtained indicated that SCC-5FU resistant cells were sensible to PDT administration, whereas BCC-5FU resistant cells were also resistant to PDT. The observed responses in both cell lines are in concordance to Protoporphyrin IX (PpIX) and reactive oxygen species (ROS) levels produced after the incubation with MAL and subsequent light exposure. The obtained data support the fact that PDT seems to be an appropriate therapeutic option to be administered after 5FU resistance in SCC. However, PDT would not be a choice therapy for resistant BCC cells to 5FU.
Collapse
Affiliation(s)
- Jimena Nicolás-Morala
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain.
| | - Mikel Portillo-Esnaola
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Samuel Terrén
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain
| | - María Gutiérrez-Pérez
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain
| | | | - Salvador González
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Medicine and Medical Specialties, Alcalá de Henares University, 28805 Madrid, Spain.
| | - Ángeles Juarranz
- Department of Biology, Faculty of Sciences, Autónoma University of Madrid (UAM), Madrid 28049, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain.
| |
Collapse
|
8
|
Tang X, Li R, Wu D, Wang Y, Zhao F, Lv R, Wen X. Development and Validation of an ADME-Related Gene Signature for Survival, Treatment Outcome and Immune Cell Infiltration in Head and Neck Squamous Cell Carcinoma. Front Immunol 2022; 13:905635. [PMID: 35874705 PMCID: PMC9304892 DOI: 10.3389/fimmu.2022.905635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
ADME genes are a set of genes which are involved in drug absorption, distribution, metabolism, and excretion (ADME). However, prognostic value and function of ADME genes in head and neck squamous cell carcinoma (HNSCC) remain largely unclear. In this study, we established an ADME-related prognostic model through the least absolute shrinkage and selection operator (LASSO) analysis in the Cancer Genome Atla (TCGA) training cohort and its robustness was validated by TCGA internal validation cohort and a Gene Expression Omnibus (GEO) external cohort. The 14-gene signature stratified patients into high- or low-risk groups. Patients with high-risk scores exhibited significantly poorer overall survival (OS) and disease-free survival (DFS) than those with low-risk scores. Receiver operating characteristic (ROC) curve analysis was used to confirm the signature’s predictive efficacy for OS and DFS. Furthermore, gene ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analyses showed that immune-related functions and pathways were enriched, such as lymphocyte activation, leukocyte cell-cell adhesion and T-helper cell differentiation. The Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and other analyses revealed that immune cell (especially B cell and T cell) infiltration levels were significantly higher in the low-risk group. Moreover, patients with low-risk scores were significantly associated with immunotherapy and chemotherapy treatment benefit. In conclusion, we constructed a novel ADME-related prognostic and therapeutic biomarker associated with immune cell infiltration of HNSCC patients.
Collapse
Affiliation(s)
- Xinran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dehua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yikai Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Zhao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruxue Lv
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Wen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xin Wen,
| |
Collapse
|
9
|
Li RY, Zheng ZY, Li ZM, Heng JH, Zheng YQ, Deng DX, Xu XE, Liao LD, Lin W, Xu HY, Huang HC, Li EM, Xu LY. Cisplatin-induced pyroptosis is mediated via the CAPN1/CAPN2-BAK/BAX-caspase-9-caspase-3-GSDME axis in esophageal cancer. Chem Biol Interact 2022; 361:109967. [PMID: 35525317 DOI: 10.1016/j.cbi.2022.109967] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/15/2022] [Accepted: 04/29/2022] [Indexed: 02/05/2023]
Abstract
Esophageal cancer is the seventh most common cancer globally. Chemotherapy resistance remains a significant challenge in the treatment of esophageal cancer patients. Cisplatin can damage tumor cells by inducing pyroptosis. However, the underlying molecular mechanisms remain unclear. In this work, we aim to investigate pyroptosis-dependent molecular mechanisms underlying cisplatin sensitivity and find potential biomarkers to predict response to cisplatin-based chemotherapy for esophageal cancer patients. Pyroptosis-associated proteins were screened via proteomics for esophageal cancer (n = 124) and bioinformatics analysis. We observed that high calpain-1 (CAPN1) and calpain-2 (CAPN2) expression were associated with favorable clinical outcomes and prolonged survival in esophageal cancer patients. We employed immunohistochemistry to evaluate the expression of CAPN1 and CAPN2 in pretreatment tumor biopsies from 108 patients with esophageal cancer who received concurrent chemoradiotherapy (CCRT). These results suggested that esophageal cancer patients with high expression of both CAPN1 and CAPN2 are likely to experience a complete response to CCRT and have significantly better survival. Western blotting, LDH release, calpain activity and cell viability assays indicated that cisplatin could activate calpain activity, while calpain inhibition or knockout suppressed cisplatin-induced pyroptosis. Mechanistically, we uncovered a novel mechanism whereby cisplatin induced pyroptosis via activation of a CAPN1/CAPN2-BAK/BAX-caspase-9-caspase-3-GSDME signaling axis in esophageal cancer cells. Collectively, this study is the first to explore the effects of calpain on cisplatin-induced pyroptosis in esophageal cancer cells. Further, our findings also imply that the combination of CAPN1 and CAPN2 could be considered as a promising biomarker of cisplatin sensitivity and prognosis in patients with esophageal cancer, providing a possibility to guide individualized treatment.
Collapse
Affiliation(s)
- Rong-Yao Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhen-Yuan Zheng
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zhi-Mao Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Jing-Hua Heng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Ya-Qi Zheng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Dan-Xia Deng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xiu-E Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Lian-Di Liao
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Wan Lin
- Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Hong-Yao Xu
- Department of Radiation Oncology, Shantou Central Hospital, Shantou, 515041, China
| | - He-Cheng Huang
- Department of Radiation Oncology, Shantou Central Hospital, Shantou, 515041, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Li-Yan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
- Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou, 515041, Guangdong, China
| |
Collapse
|
10
|
Chen D, Wei X, Yang K, Liu X, Song Y, Bai F, Jiang Y, Guo Y, Jha RK. Piperlongumine combined with vitamin C as a new adjuvant therapy against gastric cancer regulates the ROS-STAT3 pathway. J Int Med Res 2022; 50:3000605221093308. [PMID: 35481419 PMCID: PMC9087272 DOI: 10.1177/03000605221093308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/21/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE To investigate the effects of piperlongumine (PL) and vitamin C (VC) on signal transducer and activator of transcription 3 (STAT3) signalling in gastric cancer cell lines. METHODS In vivo tumour xenograft anticancer assays were undertaken to confirm the anticancer activity of PL. Cell viability, flow cytometry and Western blot assays were undertaken to evaluate the anticancer effects of PL, VC and combinations of PL and VC in AGS and KATO III cells. RESULTS Both PL and VC induced apoptosis and inhibited cell proliferation in AGS and KATO III cells. These effects were dependent on reactive oxygen species (ROS). PL effectively suppressed STAT3 activation while VC caused abnormal activation of STAT3. The combination of PL and VC exhibited a stronger apoptotic effect compared with either agent alone. PL reversed the abnormal activation of STAT3 by VC, which could be a key to their synergistic effect. CONCLUSIONS PL combined with VC exhibited a stronger anticancer effect by regulating the ROS-STAT3 pathway, suggesting that this combination might be a potential adjuvant therapy for gastric cancer.
Collapse
Affiliation(s)
- Di Chen
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumour
Immunity, Xi’an Medical University, Xi’an, Shaanxi Province, China
| | - Xinyue Wei
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| | - Ke Yang
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| | - Xinyue Liu
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| | - Yujin Song
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| | - Futing Bai
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| | - Yi Jiang
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| | - Yuhang Guo
- Institute of Basic Medical Sciences, Xi’an Medical University,
Xi’an, Shaanxi Province, China
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| | - Rajiv Kumar Jha
- China-Nepal Friendship Medical Research Centre of Rajiv Kumar
Jha, School of Clinical Medicine, Xi’an Medical University, Xi’an, Shaanxi
Province, China
| |
Collapse
|
11
|
Li Q, Qin X, Kou X, Li J, Li Z, Chen C. Anagliptin promotes apoptosis in mouse colon carcinoma cells via MCT-4/lactate-mediated intracellular acidosis. Exp Ther Med 2022; 23:282. [PMID: 35317435 PMCID: PMC8908463 DOI: 10.3892/etm.2022.11211] [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: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer cells frequently exhibit an acidic extracellular microenvironment, where inversion of the transmembrane pH gradient is associated with tumor proliferation and metastasis. To elucidate a new therapeutic target against cancer, the current study aimed to determine the mechanism by which the dipeptidyl peptidase-4 inhibitor anagliptin regulates the cellular pH gradient and concomitant extracellular acidosis during cancer progression. A total of 5x105 CT-26 cells (resuspended in phosphate buffer saline) were injected subcutaneously in the right flank of male BALB/c mice (weighing 25-28 g). The tumor samples were harvested, and lactate was detected using a lactate assay kit. Immunohistochemistry was used to detect the Ki67 and PCNA. MTT assay and flow cytometric were used to detect cell viability. Intracellular pH was detected by fluorescence pH indicator. The results revealed that anagliptin effectively reduced tumor growth, but did not affect the body weight of treated mice. Anagliptin reduced the accumulation of lactate in tumor sample. Treatment with anagliptin stimulated the apoptosis of CT-26 cells. And lactate excretion inhibition is accompanied by an increase in extracellular pH (pHe) after treatment with anagliptin. Furthermore, anagliptin induced intracellular acidification and reversed the low pHe gradient via monocarboxylate transporter-4 (MCT-4)-mediated lactate excretion. Additionally, anagliptin reversed the aberrant transmembrane extracellular/intracellular pH gradient by suppressing MCT-4-mediated lactate excretion, while also reducing mitochondrial membrane potential and inducing apoptosis. These data revealed a novel function of anagliptin in regulating lactate excretion from cancer cells, suggesting that anagliptin may be used as a potential treatment for cancer.
Collapse
Affiliation(s)
- Qi Li
- The Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150086, P.R. China
| | - Xiaoling Qin
- The Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150086, P.R. China
| | - Xiaotong Kou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jingyu Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Zhongsha Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Chang Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
12
|
He S, Xu J, Liu X, Zhen Y. Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B 2021; 11:3379-3392. [PMID: 34900524 PMCID: PMC8642427 DOI: 10.1016/j.apsb.2021.03.008] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/24/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Esophageal cancer (EC) is one of the most common cancers with high morbidity and mortality rates. EC includes two histological subtypes, namely esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). ESCC primarily occurs in East Asia, whereas EAC occurs in Western countries. The currently available treatment strategies for EC include surgery, chemotherapy, radiation therapy, molecular targeted therapy, and combinations thereof. However, the prognosis remains poor, and the overall five-year survival rate is very low. Therefore, achieving the goal of effective treatment remains challenging. In this review, we discuss the latest developments in chemotherapy and molecular targeted therapy for EC, and comprehensively analyze the application prospects and existing problems of immunotherapy. Collectively, this review aims to provide a better understanding of the currently available drugs through in-depth analysis, promote the development of new therapeutic agents, and eventually improve the treatment outcomes of patients with EC.
Collapse
Affiliation(s)
- Shiming He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Jian Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
13
|
Kong D, Long D, Liu B, Pei D, Cao N, Zhang G, Xia Z, Luo M. Downregulation of long non-coding RNA LOC101928477 correlates with tumor progression by regulating the epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Thorac Cancer 2021; 12:1303-1311. [PMID: 33713583 PMCID: PMC8088935 DOI: 10.1111/1759-7714.13858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/09/2021] [Accepted: 01/09/2021] [Indexed: 02/07/2023] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is one of the deadliest malignancies. There is a growing body of evidence showing that long non‐coding RNAs (lncRNAs) play critical roles in ESCC oncogenesis. The present study aimed to explore the role of LOC101928477, a newly discovered lncRNA, in the development and metastasis of ESCC. Methods In this study, real‐time PCR, western blotting, cell counting kit‐8 (CCK‐8), flow cytometry, colony formation, wound healing, transwell migration/invasion assay, immunofluorescence, and immunohistochemistry were used. We also applied an in situ xenograft mouse model and a lung metastasis mouse model to verify our findings. Results We determined that LOC101928477 expression was inhibited in ESCC tissue and ESCC cell lines when compared with controls. Moreover, forced expression of LOC101928477 effectively inhibited ESCC cell proliferation, colony formation, migration, and invasion via suppression of epithelial‐mesenchymal transition (EMT). Furthermore, LOC101928477 overexpression inhibited in situ tumor growth and lung metastasis in a mouse model. Conclusions Together, our results suggested that LOC101928477 could be a novel suppressor gene involved in ESCC progression.
Collapse
Affiliation(s)
- Demiao Kong
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Dali Long
- Department of Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| | - Bo Liu
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Dengke Pei
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Na Cao
- Department of Logistics, Guizhou Provincial People's Hospital, Guizhou, Guiyang, China
| | - Guihua Zhang
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Zhenkun Xia
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Meng Luo
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
14
|
Dzobo K, Ganz C, Thomford NE, Senthebane DA. Cancer Stem Cell Markers in Relation to Patient Survival Outcomes: Lessons for Integrative Diagnostics and Next-Generation Anticancer Drug Development. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 25:81-92. [PMID: 33170084 DOI: 10.1089/omi.2020.0185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Solid tumors display a complex biology that requires a multipronged treatment strategy. Most anticancer interventions, including chemotherapy, are currently unable to prevent treatment resistance and relapse. In general, therapeutics target cancer cells and overlook the tumor microenvironment (TME) and the presence of cancer stem cells (CSCs) with self-renewal and tumorigenic abilities. CSCs have been postulated to play key roles in tumor initiation, progression, therapy resistance, and metastasis. Hence, CSC markers have been suggested as diagnostics to forecast cancer prognosis as well as molecular targets for new-generation cancer treatments, especially in resistant disease. We report here original findings on expression and prognostic significance of CSC markers in several cancers. We examined and compared the transcriptional expression of CSC markers (ABCB1, ABCG2, ALDH1A1, CD24, CD44, CD90, CD133, CXCR4, EPCAM, ICAM1, and NES) in tumor tissues versus the adjacent normal tissues using publicly available databases, The Cancer Genome Atlas and Gene Expression Profiling Interactive Analysis. We found that CSC transcriptional markers were, to a large extent, expressed in higher abundance in solid tumors such as colon, lung, pancreatic, and esophageal cancers. On the other hand, no CSC marker in our analysis was expressed in the same pattern in all cancers, while individual CSC marker expression, alone, was not significantly associated with overall patient survival. Innovation in next-generation cancer therapeutics and diagnostics ought to combine CSC markers as well as integrative diagnostics that pool knowledge from CSCs and other TME components and cancer cells.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Chelene Ganz
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nicholas Ekow Thomford
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute for Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Medical Biochemistry, School of Medical Sciences, College of Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Dimakatso Alice Senthebane
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
15
|
Metabolic Reprogramming of Chemoresistant Cancer Cells and the Potential Significance of Metabolic Regulation in the Reversal of Cancer Chemoresistance. Metabolites 2020; 10:metabo10070289. [PMID: 32708822 PMCID: PMC7408410 DOI: 10.3390/metabo10070289] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/15/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of tumors. Alterations of cellular metabolism not only contribute to tumor development, but also mediate the resistance of tumor cells to antitumor drugs. The metabolic response of tumor cells to various chemotherapy drugs can be analyzed by metabolomics. Although cancer cells have experienced metabolic reprogramming, the metabolism of drug resistant cancer cells has been further modified. Metabolic adaptations of drug resistant cells to chemotherapeutics involve redox, lipid metabolism, bioenergetics, glycolysis, polyamine synthesis and so on. The proposed metabolic mechanisms of drug resistance include the increase of glucose and glutamine demand, active pathways of glutaminolysis and glycolysis, promotion of NADPH from the pentose phosphate pathway, adaptive mitochondrial reprogramming, activation of fatty acid oxidation, and up-regulation of ornithine decarboxylase for polyamine production. Several genes are associated with metabolic reprogramming and drug resistance. Intervening regulatory points described above or targeting key genes in several important metabolic pathways may restore cell sensitivity to chemotherapy. This paper reviews the metabolic changes of tumor cells during the development of chemoresistance and discusses the potential of reversing chemoresistance by metabolic regulation.
Collapse
|
16
|
Lee MC, Chen YK, Hsu YJ, Lin BR. Niclosamide inhibits the cell proliferation and enhances the responsiveness of esophageal cancer cells to chemotherapeutic agents. Oncol Rep 2019; 43:549-561. [PMID: 31894334 PMCID: PMC6967135 DOI: 10.3892/or.2019.7449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Niclosamide is an FDA-approved anthelmintic drug, and may elicit antineoplastic effects through direct STAT3 inhibition, which has been revealed in numerous human cancer cells. Chemotherapy is the standard treatment for advanced esophageal cancers, but also causes severe systemic side effects. The present study represents the first study evaluating the anticancer efficacy of niclosamide in esophageal cancers. Through western blot assay, it was demonstrated that niclosamide suppressed the STAT3 signaling pathway in esophageal adenocarcinoma cells (BE3) and esophageal squamous cell carcinoma cells (CE48T and CE81T). In addition, niclosamide inhibited cell proliferation as determined by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay and soft agar colony forming assay, and induced cell apoptosis as determined by Annexin V and PI staining. The induction of p21 and G1 arrest of the cell cycle also was revealed in niclosamide-treated CE81T cells by qPCR and flow cytometric assays, respectively. Furthermore, in the combination analysis of niclosamide and chemotherapeutic agents by MTS assay, low IC50 values were detected in cells co-treated with niclosamide, with the exception of cisplatin-treated CE81T cells. To confirm the results using an apoptosis assay, the apoptotic enhancement of niclosamide was only demonstrated in CE48T cells co-treated with 5-FU, cisplatin, or paclitaxel, and in BE3 cells co-treated with paclitaxel, but not in CE81T cells. These findings indicate a future clinical application of niclosamide in esophageal cancers.
Collapse
Affiliation(s)
- Ming-Cheng Lee
- Department of Research and Development, DrSignal BioTechnology Ltd., New Taipei City 23143, Taiwan, R.O.C
| | - Yin-Kai Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan, R.O.C
| | - Yih-Jen Hsu
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei 10051, Taiwan, R.O.C
| | - Bor-Ru Lin
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei 10051, Taiwan, R.O.C
| |
Collapse
|
17
|
Zhao W, Shan B, He D, Cheng Y, Li B, Zhang C, Duan C. Recent Progress in Characterizing Long Noncoding RNAs in Cancer Drug Resistance. J Cancer 2019; 10:6693-6702. [PMID: 31777598 PMCID: PMC6856905 DOI: 10.7150/jca.30877] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
Drug resistance is an important cause of failure in cancer chemotherapies. A large number of long noncoding RNAs (lncRNAs) have been found to be related to drug resistance in cancers. Therefore, lncRNAs provide potential targets for cancer therapies. The lncRNAs involved in cancer drug resistance are attracting interest from an increasing number of researchers. This review summarizes the latest research on the mechanisms and functions of lncRNAs in cancer drug resistance and envisages their future developments and therapeutic applications. This research suggests that lncRNAs regulate drug resistance through multiple mechanisms. LncRNAs do not affect drug resistance directly; usually, they do so by regulating the expression of some intermediate regulatory factors. In addition, lncRNAs exhibit a diversity of functions in cancer drug resistance. The overexpression of most lncRNAs promotes drug resistance, while a few lncRNAs have inhibitory effects.
Collapse
Affiliation(s)
- Wenyuan Zhao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Bin Shan
- College of Medical Sciences, Washington State University Spokane, WA, USA
| | - Dan He
- Hunan Cancer Hospital, The Affiliated Tumor Hospital of Xiangya Medical College, Central South University, Changsha, PR China
| | - Yuanda Cheng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, PR China
| | - Bin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, PR China
| | - Chaojun Duan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| |
Collapse
|
18
|
Redox-Mediated Mechanism of Chemoresistance in Cancer Cells. Antioxidants (Basel) 2019; 8:antiox8100471. [PMID: 31658599 PMCID: PMC6826977 DOI: 10.3390/antiox8100471] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cellular reactive oxygen species (ROS) status is stabilized by a balance of ROS generation and elimination called redox homeostasis. ROS is increased by activation of endoplasmic reticulum stress, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family members and adenosine triphosphate (ATP) synthesis of mitochondria. Increased ROS is detoxified by superoxide dismutase, catalase, and peroxiredoxins. ROS has a role as a secondary messenger in signal transduction. Cancer cells induce fluctuations of redox homeostasis by variation of ROS regulated machinery, leading to increased tumorigenesis and chemoresistance. Redox-mediated mechanisms of chemoresistance include endoplasmic reticulum stress-mediated autophagy, increased cell cycle progression, and increased conversion to metastasis or cancer stem-like cells. This review discusses changes of the redox state in tumorigenesis and redox-mediated mechanisms involved in tolerance to chemotherapeutic drugs in cancer.
Collapse
|
19
|
Drug metabolizing enzymes and their inhibitors' role in cancer resistance. Biomed Pharmacother 2018; 105:53-65. [PMID: 29843045 DOI: 10.1016/j.biopha.2018.05.117] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 12/15/2022] Open
Abstract
Despite continuous research on chemotherapeutic agents, different mechanisms of resistance have become a major pitfall in cancer chemotherapy. Although, exhaustive efforts are being made by several researchers to target resistance against chemotherapeutic agents, there is another class of resistance mechanism which is almost carrying on unattended. This class of resistance includes pharmacokinetics resistance such as efflux by ABC transporters and drug metabolizing enzymes. ABC transporters are the membrane bound proteins which are responsible for the movement of substrates through the cell membrane. Drug metabolizing enzymes are an integral part of phase-II metabolism that helps in the detoxification of exogenous, endogenous and xenobiotics substrates. These include uridine diphospho-glucuronosyltransferases (UGTs), glutathione-S-transferases (GSTs), dihydropyrimidine dehydrogenases (DPDs) and thiopurine methyltransferases (TPMTs). These enzymes may affect the role of drugs in both positive as well negative manner, depending upon the type of tissue and cells present and when present in tumors, can result in drug resistance. However, the underlying mechanism of resistance by drug metabolizing enzymes is still not clear. Here, we have tried to cover various aspects of these enzymes in relation to anticancer drugs.
Collapse
|
20
|
Lin S, Li Y, Zamyatnin AA, Werner J, Bazhin AV. Reactive oxygen species and colorectal cancer. J Cell Physiol 2018; 233:5119-5132. [PMID: 29215746 DOI: 10.1002/jcp.26356] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/20/2017] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) has become the fourth leading cause of cancer-related death in the worldwide. It is urgent to find more effective therapeutic strategies for it. Reactive oxygen species (ROS) play multiple roles in normal cellular physiology processes. Thus, a certain level of ROS is essential to keep normal cellular function. However, the accumulation of ROS shows dual roles for cells, which is mainly dependent on the concentration of ROS, the origin of the cancer cell and the activated signaling pathways during tumor progression. In general, moderate level of ROS leads to cell damage, DNA mutation and inflammation, which promotes the initiation and development of cancer. Excessive high level of ROS induces cancer cell death, showing an anti-cancer role. ROS are commonly higher in CRC cells than their normal counterpart cells. Therefore, it is possible that ROS induce cell death in cancer cells while not affecting the normal cells, demonstrating lower side effects. Besides, ROS also play a role in tumor microenvironment and drug resistance. These multiple roles of ROS make them a promising therapeutic target for cancer. To explore potential ROS-target therapies against CRC, it is worth to comprehensively understanding the role of ROS in CRC and therapy. In this review, we mainly discuss the strategies of ROS in CRC therapy, including direct CRC cell target and indirect tumor environment target. In addition, the influences of ROS in drug resistance will also been discussed.
Collapse
Affiliation(s)
- Sisi Lin
- Department of Pathophysiology, Institute of Digestive Disease, Tongji University School of Medicine, Shanghai, China.,Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yongyu Li
- Department of Pathophysiology, Institute of Digestive Disease, Tongji University School of Medicine, Shanghai, China
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
21
|
Liu L, Ju Y, Wang J, Zhou R. Epigallocatechin-3-gallate promotes apoptosis and reversal of multidrug resistance in esophageal cancer cells. Pathol Res Pract 2017; 213:1242-1250. [PMID: 28964574 DOI: 10.1016/j.prp.2017.09.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/14/2017] [Accepted: 09/05/2017] [Indexed: 02/07/2023]
Abstract
Evidence for demonstrating the role of the green tea component epigallocatechin-3-gallate (EGCG) in esophageal squamous cell carcinoma cells is limited. In this study, we investigated apoptosis induced by EGCG and the underlying molecular mechanisms in human esophageal squamous cell carcinoma cells. The growth-inhibitory effects of EGCG on esophageal cancer cell (Eca109 and Ec9706) were detected by MTT. Using flow cytometry, we determined the cellular apoptosis, bcl-2, bax and caspase-3 protein expression in Eca109 and Ec9706 cells following treatment with EGCG for 24h. After treatment of Eca109/ABCG2 (an esophageal cancer multidrug resistance cell line) cells with adriamycin (ADM) combined with EGCG for 24h, the cellular apoptosis, mitochondrial membrane potential, ADM concentration in cells and ABCG2 protein expression were detected by flow cytometry. EGCG inhibited the growth of Eca109 and Ec9706 cells in a dose- and time- dependent manner. EGCG induced apoptosis, decreased the bcl-2 protein expression and increased the expression of bax and caspase-3 protein. The rate of apoptosis and ADM concentration in the Eca109/ABCG2 cells following treatment with ADM and EGCG were higher than that with ADM treatment alone, although the mitochondrial membrane potential was significantly lower (P<0.01). EGCG reduced the ABCG2 expression of Eca109/ABCG2 cells. Our data indicated that EGCG inhibited cell growth and induced esophageal cancer cell apoptosis. It reduced the bcl-2 protein expression and increased the bax and caspase-3 protein expression. EGCG reversed multi-drug resistance by reducing ABCG2 expression and increasing the anticancer drug concentration in cancer cells.
Collapse
Affiliation(s)
- Liang Liu
- Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, PR China.
| | - Yingchao Ju
- Animal Experimental Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, PR China
| | - Jing Wang
- Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, PR China
| | - Rongmiao Zhou
- Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, PR China
| |
Collapse
|