1
|
Wang M, Wang M, Huang C, Zhu Y, Zhang F, Gao W, Li Z, Peng L, Tian Z, Gao C, Han X. LncRNA U731166 Increases the Accumulation of TGFBR1 by Sponging miR-3607-3p in Esophageal Squamous-Cell Carcinomas (ESCC) to Promote Tumor Metastasis. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3391. [PMID: 39737207 PMCID: PMC11682522 DOI: 10.30498/ijb.2024.343750.3391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/10/2024] [Indexed: 01/01/2025]
Abstract
Background Long non-coding RNA (lncRNA) U731166 and microRNA (miR)-3607-3p are two ncRNAs with critical roles in cancer biology, while their involvement in esophageal squamous-cell carcinomas (ESCC) is unclear. We predicted that U731166 and miR-3607-3p might interact with each other. This study aimed to investigate their role and interaction in ESCC. Objectives This study was therefore conducted to explore the involvement of U731166 and miR-3607-3p in ESCC, with a focus on the interaction between them. Materials and Methods Paired ESCC and non-tumor tissue samples were recruited from 72 ESCC patients. By RT-Qpcr, level of U731166 and miR-3607-3p in paired tissues was measured. By RNA-RNA pulldown assay, the direct interaction between U731166 and miR-3607-3p was detected. U731166 overexpression or miR-3607-3p overexpression was performed to investigate their role in regulating the expression of each other. By RT-qPCR and Western blot analysis, the role of U731166 and miR-3607-3p in regulating the level of TGFBR1 was assessed. By Transwell assays, cell invasion and migration were analyzed. Results Compared to non-tumor tissues, U731166 was highly upregulated in ESCC, while miR-3607-3p was downregulated in ESCC. U731166 and miR-3607-3p directly interacted with each other, but they are not closely correlated and did not regulate the level of each other. Moreover, U731166 reversed the role of miR-3607-3p in downregulating TGFBR1 and inhibiting cancer cell invasion and migration. U731166 and miR-3607-3p were closely associated with patients' tumor metastasis but not tumor size. Conclusion U731166 may upregulate TGFBR1 by sponging miR-3607-3p in ESCC cells to promote tumor metastasis.
Collapse
Affiliation(s)
- Mingbo Wang
- Department of Thoracic Surgery,The Fourth Hospital of Hebei Medical University Shijiazhuang City, Hebei Province, 050000, PR. China
| | - Meng Wang
- Thoracic surgery Department,Tianjin Chest hospital, Tianjin City, 300222, PR. China
| | - Chao Huang
- Department of Thoracic Surgery,The Fourth Hospital of Hebei Medical University Shijiazhuang City, Hebei Province, 050000, PR. China
| | - Yonggang Zhu
- Department of Thoracic Surgery,The Fourth Hospital of Hebei Medical University Shijiazhuang City, Hebei Province, 050000, PR. China
| | - Fan Zhang
- Department of Thoracic Surgery,The Fourth Hospital of Hebei Medical University Shijiazhuang City, Hebei Province, 050000, PR. China
| | - Wenda Gao
- Department of Thoracic Surgery,The Fourth Hospital of Hebei Medical University Shijiazhuang City, Hebei Province, 050000, PR. China
| | - Zhenhua Li
- Department of Thoracic Surgery,The Fourth Hospital of Hebei Medical University Shijiazhuang City, Hebei Province, 050000, PR. China
| | - Liangbiao Peng
- Department of Thoracic Surgery, the fourth hospital of handan, Hebei Province,056200, PR. China
| | - Ziqiang Tian
- Department of Thoracic Surgery,The Fourth Hospital of Hebei Medical University Shijiazhuang City, Hebei Province, 050000, PR. China
| | - Chao Gao
- Department of Radiation Oncology,The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050011, PR. China
| | - Xingpeng Han
- Thoracic surgery Department,Tianjin Chest hospital, Tianjin City, 300222, PR. China
| |
Collapse
|
2
|
Dahl O, Myklebust MP. A study of microRNAs as new prognostic biomarkers in anal cancer patients. Acta Oncol 2024; 63:456-465. [PMID: 38899393 PMCID: PMC11332526 DOI: 10.2340/1651-226x.2024.27976] [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: 11/22/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND MicroRNA (MiR) influences the growth of cancer by regulation of mRNA for 50-60% of all genes. We present as per our knowledge the first global analysis of microRNA expression in anal cancer patients and their prognostic impact. METHODS Twenty-nine patients with T1-4 N0-3 M0 anal cancer treated with curative intent from September 2003 to April 2011 were included in the study. RNA was extracted from fresh frozen tissue and sequenced using NGS. Differentially expressed microRNAs were identified using the R-package DEseq2 and the endpoints were time to progression (TTP) and cancer specific survival (CSS). RESULTS Five microRNAs were significantly associated with 5-year progression free survival (PFS): Low expression of two microRNAs was associated with higher PFS, miR-1246 (100% vs. 55.6%, p = 0.008), and miR-135b-5p (92.9% vs. 59.3%, p = 0.041). On the other hand, high expressions of three microRNAs were associated with higher PFS, miR-148a-3p (93.3% vs. 53.6%, p = 0.025), miR-99a-5p (92.9% vs. 57.1%, p = 0.016), and let-7c-3p (92.9% vs. 57.1%, p = 0.016). Corresponding findings were documented for CSS. INTERPRETATION Our study identified five microRNAs as prognostic markers in anal cancer. MiR-1246 and microRNA-135b-5p were oncoMiRs (miRs with oncogene effects), while miR-148a-3p, miR- 99a-5p, and let-7c-3p acted as tumour suppressors in anal cancer patients.
Collapse
Affiliation(s)
- Olav Dahl
- Department of Oncology, Haukeland University Hospital, Bergen, Norway; University of Bergen, Bergen Norway.
| | | |
Collapse
|
3
|
Baili E, Gazouli M, Lazaris AC, Kanavidis P, Boura M, Michalinos A, Charalabopoulos A, Liakakos T, Alexandrou A. Associations of long non-coding RNAs HOTAIR, LINC00951, POLR2E and HULC polymorphisms with the risk of esophageal and esophagogastric junction cancer in a western population: a case-control study. Mol Biol Rep 2024; 51:249. [PMID: 38300349 PMCID: PMC10834655 DOI: 10.1007/s11033-024-09206-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND The incidence of single-nucleotide-polymorphisms with malignant potential in esophageal cancer tissues has only been sparsely investigated in the west. Hence, we explored the contribution of four long non-coding RNAs' polymorphisms HOTAIR rs920778, LINC00951 rs11752942, POLR2E rs3787016 and HULC rs7763881 in esophageal cancer susceptibility. METHODS AND RESULTS Formalin-fixed paraffin-embedded tissue specimens from 95 consecutive patients operated for esophageal/esophagogastric junction carcinoma during 25/03/2014-25/09/2018 were processed. Demographic data, histopathological parameters, surgical and oncological outcomes were collected. DNA findings of the abovementioned population were compared with 121 healthy community controls. Both populations were of European/Greek ancestry. Sixty-seven patients underwent Ivor Lewis/McKeown esophagectomy for either squamous cell esophageal carcinoma (N = 6) or esophageal/esophagogastric junction Siewert I or II adenocarcinoma (N = 61). Twenty-eight patients were subjected to extended total gastrectomy for esophagogastric junction Siewert III adenocarcinoma. Neither LINC00951 rs11752942 nor HULC rs7763881 polymorphisms were detected more frequently in esophageal cancer patients compared with healthy community subjects. A significantly higher presence of HOTAIR rs920778 TT genotype in esophagogastric junction Siewert I/II adenocarcinoma was identified. POLR2E rs3787016 C allele and CC genotypes were overrepresented in the control group, and when found in esophageal cancer carriers were associated with earlier disease stages, as well as with minor lymph node involvement and lesser metastatic potential. CONCLUSIONS HOTAIR rs920778 may serve as a potential therapeutic suppression target, while POLR2E rs3787016 may represent a valuable biomarker to evaluate esophageal cancer predisposition and predict treatment response and prognosis. Clinical implications of these findings need to be verified with further prospective studies with larger sample-size.
Collapse
Affiliation(s)
- Efstratia Baili
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece.
- King's Health Partners, London, UK.
| | - Maria Gazouli
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas C Lazaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Prodromos Kanavidis
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| | - Maria Boura
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| | - Adamantios Michalinos
- Department of General Surgery/Anatomy, School of Medicine, European University of Cyprus, Nicosia, Cyprus
| | - Alexandros Charalabopoulos
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| | - Theodore Liakakos
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| | - Andreas Alexandrou
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Agiou Thoma 17, Athens, 11527, Greece
| |
Collapse
|
4
|
Huang T, You Q, Huang D, Zhang Y, He Z, Shen X, Li F, Shen Q, Onyebuchi IC, Wu C, Liu F, Zhu S. A positive feedback between PDIA3P1 and OCT4 promotes the cancer stem cell properties of esophageal squamous cell carcinoma. Cell Commun Signal 2024; 22:60. [PMID: 38254118 PMCID: PMC10801955 DOI: 10.1186/s12964-024-01475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Increasing evidence has indicated that long non-coding RNAs (lncRNAs) have been proven to regulate esophageal cancer progression. The lncRNA protein disulfide isomerase family A member 3 pseudogene 1 (PDIA3P1) has been shown to promote cancer stem cell properties; however, its mechanism of action remains unclear. In this study, we investigated the regulation of esophageal cancer stem cell properties by the interaction of PDIA3P1 with proteins. METHODS The GEPIA2 and Gene Expression Omnibus databases were used to analyze gene expression. PDIA3P1 expression in human esophageal squamous cell carcinoma (ESCC) tissues and cell lines was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Loss-of-function experiments were performed to determine the effects of PDIA3P1 on ESCC cell proliferation, migration, and invasion. The sphere formation assay, number of side population cells, and CD271 + /CD44 + cells were detected by flow cytometry to identify the cancer stem cell properties. RNA immunoprecipitation (RIP), RNA pull-down, co-immunoprecipitation (co-IP), dual luciferase reporter, and cleavage under targets and tagmentation (CUT&Tag) assays were performed to elucidate the underlying molecular mechanisms. RESULTS PDIA3P1 expression was upregulated in ESCC cell lines and tissues. Functionally, higher PDIA3P1 expression promoted cell proliferation, invasion, and metastasis and inhibited apoptosis in esophageal cancer. Importantly, PDIA3P1 promoted cancer stem cell properties in ESCC. Mechanistically, PDIA3P1 interacted with and stabilized octamer-binding transcription factor 4 (OCT4) by eliminating its ubiquitination by the ubiquitinating enzyme WW domain-containing protein 2 (WWP2). Moreover, as a transcription factor, OCT4 bound to the PDIA3P1 promoter and promoted its transcription. CONCLUSIONS Our research revealed a novel mechanism by which a positive feedback loop exists between PDIA3P1 and OCT4. It also demonstrated that the PDIA3P1-WWP2-OCT4 loop is beneficial for promoting the cancer stem cell properties of ESCC. Owing to this regulatory relationship, the PDIA3P1-WWP2-OCT4-positive feedback loop might be used in the diagnosis and prognosis, as well as in the development of novel therapeutics for esophageal cancer.
Collapse
Affiliation(s)
- Tao Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Qi You
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Dengjun Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Yan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Zhijie He
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Xuguang Shen
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Fei Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Qiang Shen
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Ifeanyi Christian Onyebuchi
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Chengwei Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China
| | - Feng Liu
- Department of Thoracic Surgery, Lishui Branch, Zhongda Hospital Affiliated to Southeast University, Nanjing, 211200, China.
| | - Shaojin Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, China.
| |
Collapse
|
5
|
Zhang ZD, Hou XR, Cao XL, Wang XP. Long non‑coding RNAs, lipid metabolism and cancer (Review). Exp Ther Med 2023; 26:470. [PMID: 37664674 PMCID: PMC10468807 DOI: 10.3892/etm.2023.12169] [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: 03/06/2023] [Accepted: 07/14/2023] [Indexed: 09/05/2023] Open
Abstract
Cancer has emerged as the most common cause of death in China. The change in lipid metabolism has been confirmed to have a role in several tumor types, such as esophageal, gastric, colorectal and liver cancer. Cancer cells use lipid metabolism for energy and then rapidly proliferate, invade and migrate. The main pathway by which cancer cell lipid metabolism influences cancer progression is increased fatty acid synthesis. Long non-coding (lnc)RNAs are important ncRNAs that were indicated to have significant roles in the development of human tumors. They are considered potential tumor biomarkers. Increased lipid synthesis or uptake due to deregulation of lncRNAs contributes to rapid tumor growth. In the present review, current studies on the relationship between lncRNAs, lipid metabolism and the occurrence and development of tumors were collated and summarized, and their mechanism of action was discussed. The review is expected to provide a theoretical basis for tumor treatment and prognosis evaluation based on the effective regulation of lncRNAs and lipid metabolism.
Collapse
Affiliation(s)
- Zhen-Dong Zhang
- Graduate School, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- Key Laboratory of High-Altitude Hypoxia Environment and Life Health, Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan Medicine, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| | - Xin-Rui Hou
- Graduate School, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- Key Laboratory of High-Altitude Hypoxia Environment and Life Health, Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan Medicine, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| | - Xiao-Lan Cao
- Graduate School, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- Key Laboratory of High-Altitude Hypoxia Environment and Life Health, Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan Medicine, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| | - Xiao-Ping Wang
- Key Laboratory of High-Altitude Hypoxia Environment and Life Health, Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan Medicine, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- School of Medicine, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| |
Collapse
|
6
|
Zhang S, Zhong J, Guo D, Zhang S, Huang G, Chen Y, Xu C, Chen W, Zhang Q, Zhao C, Liu S, Luo Z, Lin C. MIAT shuttled by tumor-secreted exosomes promotes paclitaxel resistance in esophageal cancer cells by activating the TAF1/SREBF1 axis. J Biochem Mol Toxicol 2023; 37:e23380. [PMID: 37132394 DOI: 10.1002/jbt.23380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 02/23/2023] [Accepted: 04/14/2023] [Indexed: 05/04/2023]
Abstract
Chemoresistance remains a major obstacle to the treatment of esophageal cancer (EC). Exosome-mediated transfer of long noncoding RNAs (lncRNAs) has recently been unveiled to correlate with the regulation of drug resistance in EC. This study aimed to investigate the physiological mechanisms by which exosome-encapsulated lncRNA myocardial infarction-associated transcript (MIAT) derived from tumor cells might mediate the paclitaxel (PTX) resistance of EC cells. First, MIAT was experimentally determined to be upregulated in PTX nonresponders and PTX-resistant EC cells. Silencing of MIAT in PTX-resistant EC cells decreased cell viability and enhanced apoptosis, corresponding to a reduced half-maximal inhibitory concentration (IC50 ) value. Next, exosomes were isolated from EC109 and EC109/T cells, and EC109 cells were cocultured with EC109/T-cell-derived exosomes. Accordingly, MIAT was revealed to be transmitted through exosomes from EC109/T cells to EC109 cells. Tumor-derived exosomes carrying MIAT increased the IC50 value of PTX and suppressed apoptosis in EC109 cells to promote PTX resistance. Furthermore, MIAT promoted the enrichment of TATA-box binding protein-associated Factor 1 (TAF1) in the promoter region of sterol regulatory element binding transcription factor 1 (SREBF1), as shown by a chromatin immunoprecipitation assay. This might be the mechanism by which MIAT could promote PTX resistance. Finally, in vivo experiments further confirmed that the knockdown of MIAT attenuated the resistance of EC cells to PTX. Collectively, these results indicate that tumor-derived exosome-loaded MIAT activates the TAF1/SREBF1 axis to induce PTX resistance in EC cells, providing a potential therapeutic target for overcoming PTX resistance in EC.
Collapse
Affiliation(s)
- Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, P. R. China
- Department of Pharmacology, Shantou University Medical College, Shantou, P. R. China
| | - Junyong Zhong
- Department of Oncology, Longgang District Central Hospital of Shenzhen, Shenzhen, P. R. China
| | - Dainian Guo
- Good Clinical Practice, Cancer Hospital of Shantou University Medical College, Shantou, P. R. China
| | - Shengqi Zhang
- Dafeng Hospital of Chaoyang District in Shantou City, Shantou, P. R. China
- Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, P. R. China
| | - Guifeng Huang
- Dafeng Hospital of Chaoyang District in Shantou City, Shantou, P. R. China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, P. R. China
| | - Chengcheng Xu
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, P. R. China
- Department of Pharmacology, Shantou University Medical College, Shantou, P. R. China
| | - Wang Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, P. R. China
| | - Qiuzhen Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou, P. R. China
| | - Chengkuan Zhao
- Department of Pharmacy, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, P. R. China
- Department of Pharmacology, Shantou University Medical College, Shantou, P. R. China
| | - Sulin Liu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, P. R. China
| | - Zebin Luo
- Dafeng Hospital of Chaoyang District in Shantou City, Shantou, P. R. China
| | - Chaoxian Lin
- The First Affiliated Hospital of Shantou University Medical College, Shantou, P. R. China
- Shantou Chaonan Minsheng Hospital, Shantou, P. R. China
| |
Collapse
|
7
|
Macedo-Silva C, Constâncio V, Miranda-Gonçalves V, Leite-Silva P, Salta S, Lobo J, Guimarães R, Carvalho-Maia C, Gigliano D, Farinha M, Sousa O, Henrique R, Jerónimo C. DNA methylation biomarkers accurately detect esophageal cancer prior and post neoadjuvant chemoradiation. Cancer Med 2023; 12:8777-8788. [PMID: 36670548 PMCID: PMC10134363 DOI: 10.1002/cam4.5623] [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: 09/20/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Esophageal cancer (ECa) is associated with high mortality, mostly due to late diagnosis, precluding curativeintent surgery. Hence, neoadjuvant chemoradiation (ChRT) is recommended in most patients regardless of histological subtype. A proportion of these patients, however, achieve complete disease remission and might be spared of radical surgery. The lack of reliable, minimally invasive biomarkers able to detect post-ChRT disease persistence is, nonetheless, a major drawback. We have previously shown that miRNA promotor methylation enables accurate cancer detection in tissues and liquid biopsies but has been seldom explored in ECa patients. AIMS Herein, we sought to unveil and validate novel candidate biomarkers able to detect ECa prior and post ChRT. MATERIALS AND METHODS Promoter methylation of miR129-2, miR124-3 and ZNF569 was assessed, using quantitative methylation-specific PCR (qMSP), in tissue samples from normal esophagus, treatment-naïve and post-ChRT ECa, as well as in liquid biopsies from ECa patients. RESULTS All genes disclosed significantly different promoter methylation levels between ECa and normal esophagus, accurately detecting post-ChRT disease, especially for adenocarcinoma. Remarkably, miR129-2me /ZNF569me methylation panel identified ECa in liquid samples with 53% sensitivity and 87% specificity. DISCUSSION MiR129-2me , miR124-3me and ZNF569me accurately discriminate ECa, either pre- or post-ChRT, from normal tissue, enabling ECa detection. Furthermore, circulalting methylation-based biomarkers are promising minimally invasive tools to detect post-ChRT residual ECa. CONCLUSION Overall, our results encourage the use of miRNA methylation biomarkers as accurate ECa detection tools as a novel approach for ChRT response monitoring.
Collapse
Affiliation(s)
- Catarina Macedo-Silva
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
| | - Vera Constâncio
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
| | - Vera Miranda-Gonçalves
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Pedro Leite-Silva
- Cancer Epidemiology Group, Research Center & Department of Epidemiology of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
| | - Sofia Salta
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
| | - João Lobo
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Rita Guimarães
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Carina Carvalho-Maia
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
| | - Davide Gigliano
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Mónica Farinha
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Olga Sousa
- Department of Radiation Oncology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Rui Henrique
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
8
|
He H, Zhang P, Li F, Zeng C, Liu D, Wu K. Predicting the prognosis of esophageal cancer based on extensive analysis of new inflammatory response‐related signature. J Biochem Mol Toxicol 2022; 37:e23291. [PMID: 36536508 DOI: 10.1002/jbt.23291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/25/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
The prognosis of esophageal cancer (ESCA) is very poor, with a 5-year survival rate of less than 20%. On the other hand, inflammation is the characteristic hallmark of ESCA; however, the prognostic relationship between inflammatory response-related genes and ESCA has not been clarified yet. Therefore, in the present manuscript, we intend to investigate the correlation and specific signature of inflammation for the prediction of the prognosis of ESCA. A total of 173 samples were obtained from The Cancer Genome Atlas (TCGA) database, including 162 tumors and 11 normal specimens. The prognostic signature was established by least absolute shrinkage and selection operator Cox regression analysis. The transcription factor regulatory network with genes of the prognostic signature was analyzed from the transcriptional regulatory relationships unravelled by sentence-based text-mining database. Chemotherapy sensitivity and immunotherapy analysis were also performed. Multivariate Cox analysis showed that the signature was an independent prognostic risk factor. The low-risk group had poorer outcomes than the high-risk group. In the high-risk group, the infiltration of most immune cells was high and strongly correlated with the riskScore. In chemotherapeutic drug sensitivity analysis, OSM, AHR, and BTG2 were significantly correlated with the current chemotherapeutic drugs of ESCA. We have demonstrated a valid prognostic signature of inflammatory response-related genes and found strong associations with immune cells, targeted genes, and chemotherapeutic agents.
Collapse
Affiliation(s)
- Hongbo He
- Department of Thoracic Surgery The First Affiliated Hospital of Zhengzhou University Henan Zhengzhou P. R. China
| | - Peng Zhang
- Department of Thoracic Surgery The First Affiliated Hospital of Zhengzhou University Henan Zhengzhou P. R. China
| | - Feng Li
- Department of Thoracic Surgery The First Affiliated Hospital of Zhengzhou University Henan Zhengzhou P. R. China
| | - Cheng Zeng
- Department of Thoracic Surgery The First Affiliated Hospital of Zhengzhou University Henan Zhengzhou P. R. China
| | - Donglei Liu
- Department of Thoracic Surgery The First Affiliated Hospital of Zhengzhou University Henan Zhengzhou P. R. China
| | - Kai Wu
- Department of Thoracic Surgery The First Affiliated Hospital of Zhengzhou University Henan Zhengzhou P. R. China
| |
Collapse
|
9
|
Zhu JL, Xue WB, Jiang ZB, Feng W, Liu YC, Nie XY, Jin LY. Long noncoding RNA CDKN2B-AS1 silencing protects against esophageal cancer cell invasion and migration by inactivating the TFAP2A/FSCN1 axis. Kaohsiung J Med Sci 2022; 38:1144-1154. [PMID: 36161699 DOI: 10.1002/kjm2.12596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Esophageal cancer (EC) is the most aggressive malignancy in the gastrointestinal tract. Long noncoding RNA cyclin-dependent kinase inhibitor 2 B antisense RNA 1 (CDKN2B-AS1) is implicated in EC development. However, the specific mechanisms involved remain poorly defined. Therefore, this research aimed to explore the mechanism of action of CDKN2B-AS1 in EC. Quantitative real-time polymerase chain reaction was conducted to measure CDKN2B-AS1 expression in EC cells and western blotting was utilized to evaluate transcription factor AP-2 alpha (TFAP2A) and fascin actin-bundling protein 1 (FSCN1) expression. After gain-of-function and loss-of-function assays, cell proliferation, migration, invasion, apoptosis, and apoptosis-related protein expression were assessed using cell counting kit-8, scratch tests, Transwell assays, flow cytometry, and western blotting, respectively. The binding relationship between CDKN2B-AS1 and TFAP2A was assessed by RNA immunoprecipitation and RNA pull-down assays. The binding relationship between TFAP2A and FSCN1 was evaluated using dual-luciferase reporter and chromatin immunoprecipitation assays. Tumor xenografts from nude mice were used for in vivo verification. CDKN2B-AS1, TFAP2A, and FSCN1 were upregulated in EC cells. Mechanistically, CDKN2B-AS1 transcriptionally activated FSCN1 by recruiting TFAP2A to the FSCN1 promoter. Silencing CDKN2B-AS1 or TFAP2A suppressed EC cell proliferative, migrating, and invasive properties and augmented apoptosis. TFAP2A was bound to CDKN2B-AS1 and the FSCN1 promoter. Overexpression of TFAP2A or FSCN1 abolished the effects of CDKN2B-AS1-silencing on EC cell function. CDKN2B-AS1 silencing curtailed tumorigenesis in nude mice, which was nullified by the upregulation of TFAP2A or FSCN1. Our findings demonstrated the antioncogenic effects of silencing CDKN2B-AS1 in EC through inactivation of the TFAP2A/FSCN1 axis.
Collapse
Affiliation(s)
- Jia-Liang Zhu
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Wen-Bo Xue
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhi-Bin Jiang
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Wei Feng
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yi-Cai Liu
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xiong-Ying Nie
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Long-Yu Jin
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
10
|
Huang T, Wu Z, Zhu S. The roles and mechanisms of the lncRNA-miRNA axis in the progression of esophageal cancer: a narrative review. J Thorac Dis 2022; 14:4545-4559. [PMID: 36524088 PMCID: PMC9745524 DOI: 10.21037/jtd-22-1449] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/08/2022] [Indexed: 12/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Esophageal cancer is one of the most common malignant digestive tract tumors. Despite various treatment methods, the prognosis of patients remains unsatisfactory, largely due to an insufficient understanding of the mechanisms involved in the pathogenesis and progression of esophageal cancer. More than 98% of the nucleotide sequences in the human genome do not encode proteins, and their transcription products are noncoding RNAs (ncRNAs), mainly long noncoding RNAs (lncRNAs) and microRNAs (miRNAs). Experiments have shown that lncRNAs and miRNAs play crucial roles in the occurrence and progression of various human malignancies. These ncRNAs influence the progression of esophageal cancer through an intricate regulatory network. We herein summarized the roles and mechanisms of the lncRNA-miRNA axis in esophageal cancer cell proliferation, apoptosis, epithelial-mesenchymal transition (EMT), invasion and metastasis, drug resistance, radiotherapy resistance, and angiogenesis. This review provides a rationale for anticancer therapy that targets the lncRNA-miRNA axis in esophageal cancer. METHODS Related articles published in the PubMed database between 05/30/2008 to 09/10/2022 were identified using the following terms: "lncRNA AND miRNA AND esophageal cancer", "lncRNA AND miRNA AND cell proliferation", "lncRNA AND miRNA AND apoptosis", "lncRNA AND miRNA AND EMT", "lncRNA AND miRNA AND invasion and metastasis", "lncRNA AND miRNA AND drug resistance", and "lncRNA AND miRNA AND radiotherapy resistance". Published articles written in English available to readers were considered. KEY CONTENT AND FINDINGS We summarized the roles of the lncRNA-miRNA axis in the progression of esophageal cancer, including cell proliferation, apoptosis, EMT, invasion and metastasis, drug resistance, radio resistance, and other progressions, and determined that the lncRNA-miRNA axis may serve as a potential clinical treatment target for esophageal cancer. CONCLUSIONS The lncRNA-miRNA axis is closely related to the progression of esophageal cancer and may act as a potential biological target for the clinical treatment of patients with esophageal cancer.
Collapse
Affiliation(s)
- Tao Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, China
| | - Zhihao Wu
- Research Laboratory of Tumor Microenvironment, Wannan Medical College, Wuhu, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, China
| | - Shaojin Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| |
Collapse
|
11
|
Chen X, Yu Y, Wu H, Qiu J, Ke D, Wu Y, Lin M, Liu T, Zheng Q, Zheng H, Yang J, Wang Z, Li H, Liu L, Yao Q, Li J, Cheng W. A Novel Model Combining Tumor Length, Tumor Thickness, TNM_Stage, Nutritional Index, and Inflammatory Index Might Be Superior to the 8th TNM Staging Criteria in Predicting the Prognosis of Esophageal Squamous Cell Carcinoma Patients Treated With Definitive Chemoradiotherapy. Front Oncol 2022; 12:896788. [PMID: 35719969 PMCID: PMC9198351 DOI: 10.3389/fonc.2022.896788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/29/2022] [Indexed: 01/14/2023] Open
Abstract
Background We aimed to determine whether the tumor length and tumor thickness should be used as prognostic factors for esophageal squamous cell carcinoma (ESCC) patients treated with definitive chemoradiotherapy (dCRT). Methods A retrospective analysis consists of 902 non-operative ESCC patients received dCRT. The nomogram was used to predict the survival. Besides, Restricted Cubic Splines (RCS) was used to examine the relationship between prognostic factors and survival outcomes. Finally, the prognostic index (PI) scores were constructed according to the tumor length and tumor thickness, and the patients were divided into the low-, medium-, and high-risk groups. Results The median follow-up of overall survival (OS) and progression-free survival (PFS) were 23.0 months and 17.5 months. Multivariate Cox regression analysis showed that tumor length and tumor thickness were independent prognostic factors associated with survival. Our novel nomograms for OS and PFS were superior to the TNM classification (p < 0.001). Besides, RCS analysis demonstrated that the death hazard of tumor length and tumor thickness sharply increased at 7.7 cm and 1.6 cm (p < 0.001). Finally, there were significant differences for ESCC patients with clinical TNM stage group of the OS and PFS in different risk groups. The higher risk group was significantly associated with shorter OS and PFS in ESCC patients (both p < 0.001 for all). Conclusion The study results suggest that the novel models integrating tumor length and tumor thickness may provide a simple and widely available method for evaluating the prognosis of non-operative ESCC patients. The tumor length and tumor thickness should be considered as prognostic factors for ESCC.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Thoracic Surgery, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Graduate School of Fujian Medical University , Fuzhou, China
| | - Yilin Yu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Haishan Wu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jianjian Qiu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Dongmei Ke
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Yahua Wu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Mingqiang Lin
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Tianxiu Liu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Qunhao Zheng
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Hongying Zheng
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jun Yang
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Zhiping Wang
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Hui Li
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Lingyun Liu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Qiwei Yao
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Graduate School of Fujian Medical University , Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- *Correspondence: Wenfang Cheng, ; Jiancheng Li, ; Qiwei Yao,
| | - Jiancheng Li
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Graduate School of Fujian Medical University , Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- *Correspondence: Wenfang Cheng, ; Jiancheng Li, ; Qiwei Yao,
| | - Wenfang Cheng
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
- Graduate School of Fujian Medical University , Fuzhou, China
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- *Correspondence: Wenfang Cheng, ; Jiancheng Li, ; Qiwei Yao,
| |
Collapse
|
12
|
Liu D, Jia H, Lu J, Zou X, Qian T, Peng F, Cao G, Wang M, Liu S. Expression of CircATXN7 in esophageal cancer tissues and its effect on cell proliferation and invasion. J Gastrointest Oncol 2022; 13:912-922. [PMID: 35837168 DOI: 10.21037/jgo-22-481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/20/2022] [Indexed: 11/06/2022] Open
Abstract
Background The goal of the current research was to investigate circATXN7 expression in esophageal cancer (EC) and its impact on the proliferation, migration, and invasion of EC cells. Methods Determination of circATXN7 expression in esophageal cancer tissues and adjacent tissueswas carried out using quantitative reverse transcription polymerase chain reaction (qRT-PCR), and we further analyzed the correlation between patients' clinical characteristics and circATXN7 expression. EC cell lines (EC-9706, Eca-109, TE-1, KYSE-30, and KYSE-150) and normal esophageal cell line (HET-1A) were cultured, and circATXN7 expression was detected by qRT-PCR. The lowest circATXN7-containing Eca-109 cells were selected to be transfected with an overexpressing lentiviral vector (circATXN7). EC-9706 cells with the highest expression of circATXN7 were selected for transfection with knockdown vectors [short hairpin RNA (shRNA)#1 and shRNA#2] of the circATXN7 sequence. Cell proliferation was determined via MTT assay. The formation of cell clones was investigated via colony formation assay. Transwell migration assay was utilized to determine cell migration and invasion ability. Results Significantly higher levels of circATXN7 were observed in EC tissues compared with paracancerous tissues (P<0.01), and circATXN7 expression level showed a significant correlation with the tumor/lymph nodes/metastasis (TNM) stage and metastasis of lymph nodes (P<0.05). Among all esophageal cell lines, EC-9706 had the highest expression level and Eca-109 had the lowest expression level. The MTT assay revealed that circATXN7 overexpression could significantly promote the proliferation of Eca-109 cells, while circATXN7 knockdown was capable of significantly inhibiting EC cell proliferation. The colony formation experiments revealed a significant increase in the number of clones in the circATXN7 overexpression model and a significant decrease in the circATXN7 knockdown model. The results of transwell migration experiments suggested that circATXN7 overexpression could promote EC cell invasion and migration, while knockdown of circATXN7 expression was associated with significant inhibition of the invasion and migration of these cells. Conclusions CircATXN7 exerted a critical role in the incidence and progression of EC. This study identified a novel molecular target and established a theoretical basis for the early detection and treatment of EC.
Collapse
Affiliation(s)
- Delin Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Department of Oncology, No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Jia
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jianwei Lu
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xi Zou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Ting Qian
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Fanyu Peng
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Department of Radiotherapy, The Fourth Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Guochun Cao
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of the Pain Management, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Shenlin Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
13
|
Li Y, Wang Z, Fu R, Wang S, Zhang T, Tian X, Yang D. Clinical Utility of Circulating Tumor Cells in Patients With Esophageal Cancer. Front Oncol 2022; 12:828368. [PMID: 35387131 PMCID: PMC8977550 DOI: 10.3389/fonc.2022.828368] [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: 12/03/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background As one of the most aggressive gastrointestinal tract cancers, esophageal carcinoma (EC) had the tenth morbidity and sixth mortality rate globally in 2020. This study was conducted to investigate whether circulating tumor cells (CTCs) could be used as diagnostic and prognostic tools for patients with EC. Methods Peripheral blood samples were collected from 129 patients newly diagnosed with EC, 17 individuals with benign diseases, and 75 healthy donors for CTC analysis using the negative enrichment-fluorescence in situ hybridization (NE-FISH) approach. The relationship between CTCs (counts and karyotypes) and clinicopathological features was then investigated. Moreover, overall survival (OS) and progression-free survival (PFS) were analyzed to evaluate the predictive value of CTCs. Results The detection of CTCs using the NE-FISH approach helped in differentiating patients with EC from benign or healthy controls at a threshold of 2 per 3.2 ml peripheral blood with a sensitivity and specificity of 70.54% and 96.74%, respectively (area under the curve = 0.826, 95% CI 0.770–0.874, p < 0.001). The CTC count was associated with tumor depth (p = 0.012), but there was no correlation with other clinicopathological characteristics. Furthermore, the proportion of CTCs with chromosome 7 triploidy was linked to distant metastasis (p = 0.033) and TNM stage (p = 0.002). The OS was significantly shorter for patients with CTCs ≥ 3 than for those with CTCs < 3. Univariate analysis revealed that sex, vascular invasion, distant metastasis, tumor depth, lymph node metastasis, and TNM stage were the significant prognostic factors for patients with EC. Multivariate analysis demonstrated that distant metastasis (hazard ratio (HR) 3.262, 95% CI 1.671–6.369, p = 0.001 for PFS; HR 3.759, 95% CI 1.867–7.571, p < 0.001 for OS) was a significant prognostic factor for patients with EC. Conclusions Detection of CTCs using NE-FISH could be helpful in the diagnosis of EC. The proportion of CTCs with chromosome 7 triploidy was related to distant metastasis and TNM stage. Patients with CTCs ≥ 3 had short OS, while distant metastasis was an independent factor indicating a poor prognosis for patients with EC.
Collapse
Affiliation(s)
- Yang Li
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, China
| | - Zhenxing Wang
- Department of Thoracic Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Rao Fu
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, China
| | - Shuang Wang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, China
| | - Tingting Zhang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, China
| | - Xudong Tian
- Department of Thoracic Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
14
|
Zhang W, Zhang L, Cai XJ, Li D, Cao FJ, Zuo ZG, Song Y, Yu XJ, Liu S. Dexmedetomidine inhibits the growth and metastasis of esophageal cancer cells by down-regulation of lncRNA MALAT1. Kaohsiung J Med Sci 2022; 38:585-593. [PMID: 35199933 DOI: 10.1002/kjm2.12506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/19/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
This study aims to evaluate the effect of dexmedetomidine (DEX)-on esophageal cancer (EC) via regulating long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). The effect of DEX on MALAT1 expression and EC cell viability was detected. EC cells were divided into Blank, DEX, scrambled/MALAT1 siRNA, and DEX + control/MALAT1 groups, followed by a series of experiments including quantitative reverse-transcription polymerase chain reaction (qRT-PCR), western blotting, 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT), Annexin V-FITC/PI staining, wound healing, and Transwell assays. Additionally, mice were subjected to the subcutaneous injection of Eca109 cells transfected by control/MALAT1 activation lentiviral vector to construct EC models with the DEX treatment, and then the tumor volume and the expression of Ki-67 and active caspase-3 were determined. DEX reduced the expression of MALAT1 in EC cells in a dose-dependent manner. DEX inhibited the viability of EC cells, but increased the cell apoptosis, which, however, was reversed by MALAT1 overexpression. Moreover, MALAT1 overexpression abolished the inhibitory effect of DEX on the epithelial-mesenchymal transition (EMT) of EC cells, with enhanced migration and invasion. Furthermore, DEX succeeded in decreasing the tumor volume with the down-regulation of MALAT1. In comparison with the DEX group, mice in the DEX + MALAT1 group had larger tumors, with the up-regulation of Ki-67 and the down-regulation of active caspase-3. DEX can reduce the expression of MALAT1 in EC cells, thereby inhibiting the proliferation, invasion and migration, as well as EMT, and promoting the apoptosis of EC cells.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Zhang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiao-Jun Cai
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Dong Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Feng-Jun Cao
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Zhi-Gang Zuo
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Ying Song
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiong-Jie Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Shan Liu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
15
|
Yang B, Ma H, Bian Y. LINC00261 Inhibits Esophageal Cancer Radioresistance by Down-Regulating microRNA-552-3p and Promoting DIRAS1. Cancer Manag Res 2021; 13:8559-8573. [PMID: 34803403 PMCID: PMC8597985 DOI: 10.2147/cmar.s332640] [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: 08/04/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Esophageal cancer (EC) represents a life-threatening tumor with an ever-increasing incidence rate. Long intergenic non-protein coding RNAs (LINCs) have also become a topic of interest in EC. In a similar light, the current study aimed to investigate the role of LINC00261 in EC radioresistance. METHODS Firstly, radioresistant EC cell lines TE-1-R and TE-5-R were established using TE-1 and TE-5 cells. Subsequently, LINC00261, microRNA (miR)-552-3p, and DIRAS1 expression patterns in EC tissues and adjacent normal tissues and EC cells were evaluated. In addition, survival fraction (SF), colony formation, apoptosis, and γ-H2AX levels were analyzed, followed by the detection of the binding relation between LINC00261 and miR-552-3p and between miR-552-3p and DIRAS1. Lastly, xenograft transplantation was carried out to confirm the effects of LINC00261 on EC radioresistance in vivo. RESULTS LINC00261 and DIRAS1 were poorly-expressed in EC tissues and cells, but miR-552-3p was over-expressed. In EC cells with X-ray radiation, over-expression of LINC00261 reduced SF and cell viability, strengthened γ-H2AX levels, and promoted apoptosis, while all these trends were counteracted by miR-522-3p over-expression or DIRAS1 silencing. Mechanistic investigation further validated the binding relation between LINC00261 and miR-552-3p, and between miR-552-3p and DIRAS1. Moreover, LINC00261 over-expression suppressed tumor growth and reduced EC radioresistance in vivo. CONCLUSION Altogether, our findings indicated that LINC00261 exerts a suppressive effect on EC radioresistance via the competing endogenous RNA network to sponge miR-552-3p and up-regulate DIRAS1 transcription.
Collapse
Affiliation(s)
- Baolong Yang
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Xi ‘an Jiaotong University, Xi ‘an, Shanxi Province, 710004, People’s Republic of China
| | - Hongbing Ma
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Xi ‘an Jiaotong University, Xi ‘an, Shanxi Province, 710004, People’s Republic of China
| | - Yan Bian
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Xi ‘an Jiaotong University, Xi ‘an, Shanxi Province, 710004, People’s Republic of China
| |
Collapse
|
16
|
New Trends in Esophageal Cancer Management. Cancers (Basel) 2021; 13:cancers13123030. [PMID: 34204314 PMCID: PMC8235022 DOI: 10.3390/cancers13123030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/10/2021] [Accepted: 06/13/2021] [Indexed: 12/08/2022] Open
|
17
|
Li S, Wang X. The potential roles of exosomal noncoding RNAs in osteosarcoma. J Cell Physiol 2020; 236:3354-3365. [PMID: 33044018 DOI: 10.1002/jcp.30101] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
Abstract
Clinically, it is difficult to efficaciously screen and diagnose osteosarcoma (OS) in advance due to the low sensitivity and poor specificity of the existing tumor markers. Exosomes (Exos) are nanoscale vesicles containing RNAs, lipids, and proteins with a diameter of 30-100 nm. They are multivesicular bodies formed during the invagination of lysosomal particles in cells and released extracellularly after fusing with cell membranes. Besides, Exos are important carriers of cell-to-cell communication signals and genetic materials in the tumor microenvironment. During tumorigenesis, the tumor cells interplay with immune cells, endothelial cells, and related fibroblasts through Exos and boost cancer development. After altering the surrounding microenvironment, the Exos drive tumor cells to proliferate, speed up angiogenesis, and boost cancers to develop along with body fluid transportation. Currently, Exos are becoming novel noninvasive tumor diagnostic markers with high sensitivity, exerting pivotal impacts in fundamental research and clinical applications. Here, we review the existing literature on the roles of exosomal noncoding RNAs in OS progression and their potential clinical applications as novel biomarkers and therapeutics.
Collapse
Affiliation(s)
- Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China.,School of Fundamental Sciences, Center of 3D Printing and Organ Manufacturing, China Medical University (CMU), Shenyang, China
| | - Xiaohong Wang
- School of Fundamental Sciences, Center of 3D Printing and Organ Manufacturing, China Medical University (CMU), Shenyang, China.,Department of Mechanical Engineering, Center of Organ Manufacturing, Tsinghua University, Beijing, China
| |
Collapse
|