1
|
Barbato A, Piscopo F, Salati M, Pollastro C, Evangelista L, Ferrante L, Limongello D, Brillante S, Iuliano A, Reggiani-Bonetti L, Salatiello M, Iaccarino A, Pisapia P, Malapelle U, Troncone G, Indrieri A, Dominici M, Franco B, Carotenuto P. A MiR181/Sirtuin1 regulatory circuit modulates drug response in biliary cancers. Clin Exp Med 2024; 24:74. [PMID: 38598008 PMCID: PMC11006774 DOI: 10.1007/s10238-024-01332-0] [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: 01/25/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024]
Abstract
Despite recent advances, biliary tract cancer (BTC) remains one of the most lethal tumor worldwide due to late diagnosis, limited therapeutic strategies and resistance to conventional therapies. In recent years, high-throughput technologies have enabled extensive genome, and transcriptome sequencing unveiling, among others, the regulatory potential of microRNAs (miRNAs). Compelling evidence shown that miRNA are attractive therapeutic targets and promising candidates as biomarkers for various therapy-resistant tumors. The analysis of miRNA profile successfully identified miR-181c and -181d as significantly downregulated in BTC patients. Low miR-181c and -181d expression levels were correlated with worse prognosis and poor treatment efficacy. In fact, progression-free survival analysis indicated poor survival rates in miR-181c and -181d low expressing patients. The expression profile of miR-181c and -181d in BTC cell lines revealed that both miRNAs were dysregulated. Functional in vitro experiments in BTC cell lines showed that overexpression of miR-181c and -181d affected cell viability and increased sensitivity to chemotherapy compared to controls. In addition, by using bioinformatic tools we showed that the miR-181c/d functional role is determined by binding to their target SIRT1 (Sirtuin 1). Moreover, BTC patients expressing high levels of miR-181 and low SIRT1 shown an improved survival and treatment response. An integrative network analysis demonstrated that, miR-181/SIRT1 circuit had a regulatory effect on several important metabolic tumor-related processes. Our study demonstrated that miR-181c and -181d act as tumor suppressor miRNA in BTC, suggesting the potential use as therapeutic strategy in resistant cancers and as predictive biomarker in the precision medicine of BTC.
Collapse
Affiliation(s)
- Anna Barbato
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
| | - Fabiola Piscopo
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
| | - Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41125, Modena, Italy
| | - Carla Pollastro
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
| | - Lorenzo Evangelista
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Luigi Ferrante
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Davide Limongello
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Simona Brillante
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- IRGB, Institute for Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Antonella Iuliano
- Department of Mathematics, Computer Science and Economics (DIMIE), University of Basilicata, 85100, Potenza, Italy
| | - Luca Reggiani-Bonetti
- Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, 41125, Modena, Italy
| | - Maria Salatiello
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Antonino Iaccarino
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Universita' degli Studi di Napoli-AOU Federico II, 80131, Naples, Italy
| | - Alessia Indrieri
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- IRGB, Institute for Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41125, Modena, Italy
| | - Brunella Franco
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, 80078, Naples, Italy
| | - Pietro Carotenuto
- TIGEM, Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy.
- Department of Translational Medical Science, Medical Genetics, University of Naples "Federico II", 80131, Naples, Italy.
| |
Collapse
|
2
|
Dai P, Xiong L, Wei Y, Wei X, Zhou X, Zhao J, Tang H. A pancancer analysis of the oncogenic role of cyclin B1 (CCNB1) in human tumors. Sci Rep 2023; 13:16226. [PMID: 37758792 PMCID: PMC10533567 DOI: 10.1038/s41598-023-42801-y] [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: 07/31/2022] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Aberrant levels of the G2/M cyclin cyclin B1 (gene CCNB1) have been associated with multiple cancers; however, the literature lacks a focused and comprehensive analysis of the regulation of this important regulator of cell proliferation in cancer. Through this work, we performed a pancancer analysis of the levels of CCNB1 and dissected aspects of regulation and how this correlates with cancer prognosis. We comprehensively evaluated the expression and promoter methylation of CCNB1 across 38 cancers based on RNA sequencing data obtained from the Cancer Genome Atlas (TCGA). The correlation of CCNB1 with prognosis and the tumor microenvironment was explored. Using lung adenocarcinoma data, we studied the potential upstream noncoding RNAs involved in the regulation of CCNB1 and validated the protein levels and prognostic value of CCNB1 for this disease site. CCNB1 was highly expressed, and promoter methylation was reduced in most cancers. Gene expression of CCNB1 correlated positively with poor prognosis of tumor patients, and these results were confirmed at the protein level using lung adenocarcinoma. CCNB1 expression was associated with the infiltration of T helper cells, and this further correlated with poor prognosis for certain cancers, including renal clear cell carcinoma and lung adenocarcinoma. Subsequently, we identified a specific upstream noncoding RNA contributing to CCNB1 overexpression in lung adenocarcinoma through correlation analysis, expression analysis and survival analysis. This study provides a comprehensive analysis of the expression and methylation status of CCNB1 across several forms of cancer and provides further insight into the mechanistic pathways regulating Cyclin B1 in the tumorigenesis process.
Collapse
Affiliation(s)
- Peng Dai
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lecai Xiong
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yanhong Wei
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiaoyan Wei
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Xuefeng Zhou
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinping Zhao
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
3
|
Zanon MF, Scapulatempo-Neto C, Gama RR, Marques MMC, Reis RM, Evangelista AF. Identification of MicroRNA Expression Profiles Related to the Aggressiveness of Salivary Gland Adenoid Cystic Carcinomas. Genes (Basel) 2023; 14:1220. [PMID: 37372400 DOI: 10.3390/genes14061220] [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: 03/21/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Adenoid cystic carcinoma (ACC) has been reported as the second most common carcinoma of the salivary glands. Few studies have associated miRNA expression with ACC aggressiveness. In this study, we evaluated the miRNA profile of formalin-fixed, paraffin-embedded (FFPE) samples of salivary gland ACC patients using the NanoString platform. We studied the miRNA expression levels associated with the solid growth pattern, the more aggressive histologic feature of ACCs, compared with the tubular and cribriform growth patterns. Moreover, the perineural invasion status, a common clinicopathological feature of the disease that is frequently associated with the clinical progression of ACC, was investigated. The miRNAs showing significant differences between the study groups were selected for target prediction and functional enrichment, which included associations with the disease according to dedicated databases. We observed decreased expression of miR-181d, miR-23b, miR-455, miR-154-5p, and miR-409 in the solid growth pattern compared with tubular and cribriform growth patterns. In contrast, miR-29c, miR-140, miR-195, miR-24, miR-143, and miR-21 were overexpressed in patients with perineural invasion. Several target genes of the miRNAs identified have been associated with molecular processes involved in cell proliferation, apoptosis, and tumor progression. Together, these findings allowed the characterization of miRNAs potentially associated with aggressiveness in salivary gland adenoid cystic carcinoma. Our results highlight important new miRNA expression profiles involved in ACC carcinogenesis that could be associated with the aggressive behavior of this tumor type.
Collapse
Affiliation(s)
- Maicon Fernando Zanon
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil
| | | | - Ricardo Ribeiro Gama
- Department of Head and Neck Surgery, Barretos Cancer Hospital, Barretos 14784-400, Brazil
| | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Adriane Feijó Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil
- Sergio Arouca National School of Public Health, Oswaldo Cruz Foundation, Manguinhos, Rio de Janeiro 21040-361, Brazil
| |
Collapse
|
4
|
Gómez-Romero L, Alvarez-Suarez DE, Hernández-Lemus E, Ponce-Castañeda MV, Tovar H. The regulatory landscape of retinoblastoma: a pathway analysis perspective. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220031. [PMID: 35620002 PMCID: PMC9114937 DOI: 10.1098/rsos.220031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/13/2022] [Indexed: 05/03/2023]
Abstract
Retinoblastoma (Rb) is a rare intraocular tumour in early childhood, with an approximate incidence of 1 in 18 000 live births. Experimental studies for Rb are complex due to the challenges associated with obtaining a normal retina to contrast with diseased tissue. In this work, we reanalyse a dataset that contains normal retina samples. We identified the individual genes whose expression is different in Rb in contrast with normal tissue, determined the pathways whose global expression pattern is more distant from the global expression observed in normal tissue, and finally, we identified which transcription factors regulate the highest number of differentially expressed genes (DEGs) and proposed as transcriptional master regulators (TMRs). The enrichment of DEGs in the phototransduction and retrograde endocannabinoid signalling pathways could be associated with abnormal behaviour of the processes leading to cellular differentiation and cellular proliferation. On the other hand, the TMRs nuclear receptor subfamily 5 group A member 2 and hepatocyte nuclear factor 4 gamma are involved in hepatocyte differentiation. Therefore, the enrichment of aberrant expression in these transcription factors could suggest an abnormal retina development that could be involved in Rb origin and progression.
Collapse
Affiliation(s)
- Laura Gómez-Romero
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| | - Diana E. Alvarez-Suarez
- Medical Research Unit in Infectious Diseases, Hospital de Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Pharmacology Department, CINVESTAV, Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
- Center for Complexity Sciences, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - M. Verónica Ponce-Castañeda
- Medical Research Unit in Infectious Diseases, Hospital de Pediatría, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Hugo Tovar
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
| |
Collapse
|
5
|
Jhan JH, Hsu WC, Lee YC, Li WM, Huang AM, Lin HH, Wang CS, Wu YR, Li CC, Wu WJ, Ke HL. MicroRNA-375-3p Suppresses Upper Tract Urothelial Carcinoma Cell Migration and Invasion via Targeting Derlin-1. Cancers (Basel) 2022; 14:cancers14040880. [PMID: 35205628 PMCID: PMC8869792 DOI: 10.3390/cancers14040880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Little is known regarding the molecular characterization of upper tract urothelial carcinoma (UTUC). Novel therapeutic targets and prognostic predictors are imminent. In the present study, we aim to examine the oncogenic function and molecular mechanism of Derlin-1 in UTUC. Derlin-1 overexpression is significantly associated with poor prognosis in patients with UTUC. In vitro, knockdown or over-expression of Derlin-1 markedly regulated UTUC cell invasion and migration. We further discovered miR-375-3p suppresses cell invasion and migration by inversely regulating Derlin-1 and blocking EMT in UTUC cells. Taking this together, miR-375-3p functions as a tumor suppressive microRNA by directly targeting Derlin-1 and blocking epithelial-mesenchymal transition (EMT) in UTUC.
Collapse
Affiliation(s)
- Jhen-Hao Jhan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-H.J.); (W.-C.H.); (W.-M.L.); (H.-H.L.); (C.-C.L.); (W.-J.W.)
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
- Department of Urology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 81267, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Wei-Chi Hsu
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-H.J.); (W.-C.H.); (W.-M.L.); (H.-H.L.); (C.-C.L.); (W.-J.W.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yi-Chen Lee
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Wei-Ming Li
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-H.J.); (W.-C.H.); (W.-M.L.); (H.-H.L.); (C.-C.L.); (W.-J.W.)
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
- Department of Urology, Ministry of Health and Welfare, Pingtung Hospital, Pingtung 90054, Taiwan
| | - A-Mei Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hui-Hui Lin
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-H.J.); (W.-C.H.); (W.-M.L.); (H.-H.L.); (C.-C.L.); (W.-J.W.)
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
| | - Chien-Sheng Wang
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yi-Ru Wu
- General Division, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Chia Li
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-H.J.); (W.-C.H.); (W.-M.L.); (H.-H.L.); (C.-C.L.); (W.-J.W.)
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
| | - Wen-Jeng Wu
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-H.J.); (W.-C.H.); (W.-M.L.); (H.-H.L.); (C.-C.L.); (W.-J.W.)
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hung-Lung Ke
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-H.J.); (W.-C.H.); (W.-M.L.); (H.-H.L.); (C.-C.L.); (W.-J.W.)
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80145, Taiwan
- Correspondence: ; Tel.: +886-07-3121101 (ext. 6694); Fax: +886-07-3211033
| |
Collapse
|
6
|
Christoph S, Alexander Q, Fritz T, Walter SS, Steffi U, Ralf K, Joachim O. MiRNA-181d expression correlates in tumor versus plasma of glioblastoma patients - the base of a preoperative stratification tool for local carmustine wafer use. World Neurosurg 2021; 159:e324-e333. [PMID: 34942386 DOI: 10.1016/j.wneu.2021.12.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Patients with low micro-RNA-181d level in glioblastoma tissue benefit most of carmustine wafer use. The study compares preoperative miRNA-181d plasma and tumor expression. This may form the base to decide, from a preoperative blood test, if carmustine wafer implantation is recommendable. METHODS A total of 60 patients suffering from glioblastoma treated between 2018 and 2020 were enrolled prospectively. Preoperatively, blood was drawn; and the plasma was isolated. Tumor specimens were collected. Blood samples from 30 healthy individuals served as a reference. Micro-RNA-181d expression in plasma and tumor were acquired as fold change, using qRT-PCR. Results were correlated with relevant demographic, clinical and histopathologic aspects of the cohort. Further factors like tumour volume as well as blood panel results were considered. A TCGA analysis was performed to investigate specific miRNA-181d - protein interactions to elude how miRNA-181 impact therapy response to carmustine. RESULTS Patients with glioblastoma showed a significant overexpression of miRNA-181d compared to healthy individuals (p = 0.029). There was a significant correlation between miRNA-181d expression in tumor tissue and plasma (p = 0.001, R = 0.51). Sensitivity of low miRNA-181d expression in plasma predicting low miRNA-181d tumor expression was 76.6%. Tumor volume, preoperative medication as well as items of blood panel analysis did not influence the prognostic value of plasma miRNA-181d expression. TCGA analysis revealed eight potential protein targets to be regulated by miRNA-181d. CONCLUSION miRNA-181d seems to be a potential molecular marker which can reliably be detected in blood samples of glioblastoma patients. It should therefore prospectively be evaluated as a potential preoperative prognostic marker regarding carmustine wafer implantation.
Collapse
Affiliation(s)
- Sippl Christoph
- Department of Neurosurgery, Saarland University, Faculty of Medicine, Homburg/Saar, Germany.
| | - Quiring Alexander
- Department of Neurosurgery, Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Teping Fritz
- Department of Neurosurgery, Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Schulz-Schaeffer Walter
- Institute of Neuropathology, Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Urbschat Steffi
- Department of Neurosurgery, Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Ketter Ralf
- Department of Neurosurgery, Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Oertel Joachim
- Department of Neurosurgery, Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| |
Collapse
|
7
|
Yao C, Li Y, Luo L, Xiong Q, Zhong X, Xie F, Feng P. Identification of miRNAs and genes for predicting Barrett's esophagus progressing to esophageal adenocarcinoma using miRNA-mRNA integrated analysis. PLoS One 2021; 16:e0260353. [PMID: 34818353 PMCID: PMC8612537 DOI: 10.1371/journal.pone.0260353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
Barrett's esophagus (BE) is defined as any metaplastic columnar epithelium in the distal esophagus, which predisposes to esophageal adenocarcinoma (EAC). Yet, the mechanism through which BE develops to EAC still remain unclear. Moreover, the miRNA-mRNA regulatory network in distinguishing BE from EAC still remains poorly understood. To identify differentially expressed miRNAs (DEMs) and genes (DEGs) between EAC and BE from tissue samples, gene expression microarray datasets GSE13898, GSE26886, GSE1420 and miRNA microarray datasets GSE16456, GSE20099 were downloaded from Gene Expression Omnibus (GEO) database. GEO2R was used to screen the DEMs and DEGs. Pathway and functional enrichment analysis were performed by DAVID database. The protein-protein interaction (PPI) network was constructed by STRING and been visualized by Cytoscape software. Finnal, survival analysis was performed basing TCGA database. A total of 21 DEMs were identified. The enriched functions and pathways analysis inclued Epstein-Barr virus infection, herpesvirus infection and TRP channels. GART, TNFSF11, GTSE1, NEK2, ICAM1, PSMD12, CTNNB1, CDH1, PSEN1, IL1B, CTNND1, JAG1, CDH17, ITCH, CALM1 and ITGA6 were considered as the hub-genes. Hsa-miR-143 and hsa-miR-133b were the highest connectivity target gene. JAG1 was predicted as the largest number of target miRNAs. The expression of hsa-miR-181d, hsa-miR-185, hsa-miR-15b, hsa-miR-214 and hsa-miR-496 was significantly different between normal tissue and EAC. CDH1, GART, GTSE1, NEK2 and hsa-miR-496, hsa-miR-214, hsa-miR-15b were found to be correlated with survival.
Collapse
Affiliation(s)
- Chengjiao Yao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Department of Geriatrics of the Affiliated Hospital, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yilin Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lihong Luo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qin Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaowu Zhong
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- * E-mail: (PF); (XZ)
| | - Fengjiao Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Peimin Feng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- * E-mail: (PF); (XZ)
| |
Collapse
|
8
|
Xia Y, Tang G, Wang C, Zhong J, Chen Y, Hua L, Li Y, Liu H, Zhu B. Functionalized selenium nanoparticles for targeted siRNA delivery silence Derlin1 and promote antitumor efficacy against cervical cancer. Drug Deliv 2020; 27:15-25. [PMID: 31830840 PMCID: PMC6968560 DOI: 10.1080/10717544.2019.1667452] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Small interfering RNA (siRNA) exhibits great potential as a novel therapeutic option due to its highly sequence-specific ability to silence genes. However, efficient and safe delivery carriers are required for developing novel therapeutic paradigms. Thus, the successful development of efficient delivery platforms for siRNA is a crucial issue for the development of siRNA-based drugs in cancer treatments. In this study, biocompatible selenium nanoparticles (SeNPs) were loaded with RGDfC peptide to fabricate tumor-targeting gene delivery vehicle RGDfC-SeNPs. Subsequently, RGDfC-SeNPs were loaded with Derlin1-siRNA to fabricate RGDfC-Se@siRNA, which are functionalized selenium nanoparticles. RGDfC-Se@siRNA showed greater uptake in HeLa cervical cancer cells in comparison with that in human umbilical vein endothelial cells (HUVECs), verifying the RGDfC-mediated specific uptake of RGDfC-Se@siRNA. RGDfC-Se@siRNA was capable of entering HeLa cells via clathrin-associated endocytosis, and showed faster siRNA release in a cancer cell microenvironment in comparison with a normal physiological environment. qPCR and western blotting assays both indicated that RGDfC-Se@siRNA exhibited an obvious gene silencing efficacy in HeLa cells. RGDfC-Se@siRNA suppressed the invasion, migration and the proliferation of HeLa cells, and triggered HeLa cell apoptosis. Moreover, RGDfC-Se@siRNA induced the disruption of mitochondrial membrane potentials. Meanwhile, RGDfC-Se@siRNA enhanced the generation of reactive oxygen species (ROS) in HeLa cell, suggesting that mitochondrial dysfunction mediated by ROS might play a significant role in RGDfC-Se@siRNA-induced apoptosis. Interestingly, RGDfC-SeNPs@siRNA exhibited significant antitumor activity in a HeLa tumor-bearing mouse model. Additionally, RGDfC-SeNPs@siRNA is nontoxic to main organ of mouse. The above results indicate that RGDfC-Se@siRNA provides a promising potential for cervical cancer therapy.
Collapse
Affiliation(s)
- Yu Xia
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Guoyi Tang
- Department of Obstetrics Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Changbing Wang
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiayu Zhong
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yi Chen
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Liang Hua
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yinghua Li
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hongsheng Liu
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Bing Zhu
- Central Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Zhang Y, Li C, Guan C, Zhou B, Wang L, Yang C, Zhen L, Dai J, Zhao L, Jiang W, Xu Y. MiR-181d-5p Targets KLF6 to Improve Ischemia/Reperfusion-Induced AKI Through Effects on Renal Function, Apoptosis, and Inflammation. Front Physiol 2020; 11:510. [PMID: 32581828 PMCID: PMC7295155 DOI: 10.3389/fphys.2020.00510] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Renal tubular epithelial cell (RTEC) death and renal interstitial inflammation are the most crucial pathophysiological changes in acute kidney ischemia/reperfusion injury (IRI). The microRNA (miR)-181d family plays diverse roles in cell proliferation, apoptosis and inflammation, but its renal target and potential role in IRI are unknown. Here, we showed that the expression of miR-181d-5p decreased and Krueppel-like factor 6 (KLF6) increased in a renal cell (HK-2) model of hypoxia/reoxygenation (H/R) injury and a mouse model of renal IRI. They were mainly distributed in the renal tubules. After renal IRI, miR-181d-5p overexpression significantly inhibited inflammatory mediators, reduced apoptosis and further improved renal function. KLF6 exacerbated RTEC damage and acted as a NF-κB co-activator to aggravate the renal IRI inflammatory response. Mechanistically, KLF6 was predicted as a new potential target gene of miR-181d-5p through bioinformatic analysis and luciferase reporter assay verification. After overexpressing miR-181d-5p and inhibiting KLF6, the role of miR-181d-5p was weakened on the renal damage improvement. In conclusion, miR-181d-5p upregulation produced protective antiapoptotic and anti-inflammatory effects against IRI in kidneys in vivo and H/R injury in HK-2 cells in vitro, and these effects were achieved by targeted inhibition of KLF6. Thus, our results provide novel insights into the molecular mechanisms associated with IRI and a potential novel therapeutic target.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenyu Li
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Nephrologisches Zentrum, Ludwig Maximilian University of Munich, Munich, Germany
| | - Chen Guan
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Zhou
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Wang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengyu Yang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Zhen
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jie Dai
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Long Zhao
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Jiang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Xu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
10
|
Liu HW, Lee PM, Bamodu OA, Su YK, Fong IH, Yeh CT, Chien MH, Kan IH, Lin CM. Enhanced Hsa-miR-181d/p-STAT3 and Hsa-miR-181d/p-STAT5A Ratios Mediate the Anticancer Effect of Garcinol in STAT3/5A-Addicted Glioblastoma. Cancers (Basel) 2019; 11:cancers11121888. [PMID: 31783691 PMCID: PMC6966688 DOI: 10.3390/cancers11121888] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/16/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Glioblastoma (GBM), a malignant grade IV tumor, is the most malignant brain tumor due to its hyper-proliferative and apoptosis-evading characteristics. The signal transducer and activators of transcription (STAT) family genes, including STAT3 and STAT5A, have been indicated to play important roles in GBM progression. Increasing number of reports suggest that garcinol, a polyisoprenylated benzophenone and major bioactive component of Garcinia indica contains potent anti-cancer activities. Material and Methods: The present study investigated the anti-GBM effects of garcinol, focusing on the STAT3/STAT5A activation, using a combination of bioinformatics, in vitro, and ex vivo assays. Results: Our bioinformatics analysis of The Cancer Genome Atlas (TCGA)–GBM cohort (n = 173) showed that STAT3 and STAT5A are preferentially elevated in primary and recurrent GBM, compared to non-tumor brain tissues, and is significantly correlated with reduced overall survival. In support, our immunohistochemical staining of a GBM cohort (n = 45) showed an estimated 5.3-fold (p < 0.001) elevation in STAT3 and STAT5A protein expression in primary and recurrent GBM versus the non-tumor group. In vitro, garcinol treatment significantly suppressed the proliferative, invasive, and migratory potential of U87MG or GBM8401 cells, dose-dependently. In addition, garcinol anticancer effect significantly attenuated the GBM stem cell-like phenotypes, as reflected by diminished ability of U87MG or GBM8401 to form colonies and tumorspheres and suppressed expression of OCT4 and SOX2. Furthermore, analysis on GBM transcriptome revealed an inverse correlation between the level of STAT3/5A and hsa-miR-181d. Garcinol-mediated anti-GBM effects were associated with an increased hsa-miR-181d/STAT3 and hsa-miR-181d/5A ratio. The results were further verified in vivo using U87MG mouse xenograft model where administration of garcinol significantly inhibited tumor growth. Conclusions: We present evidence of anti-GBM efficacy of garcinol mediated by enhancing the hsa-miR-181d/STAT3 and hsa-miR-181d/5A ratios in GBM cells. Our findings suggest a potential new therapeutic agent for combating aggressive GBM.
Collapse
Affiliation(s)
- Heng-Wei Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; (H.-W.L.); (Y.-K.S.); (C.-T.Y.); (M.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan;
| | - Peter Mingjui Lee
- Department of Clinical Oncology, College of Medicine, California North State University, Elk Grove, CA 95757, USA;
| | - Oluwaseun Adebayo Bamodu
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Hematology and Oncology, Cancer Center, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department of Medical Research and Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Yu-Kai Su
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; (H.-W.L.); (Y.-K.S.); (C.-T.Y.); (M.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan;
| | - Iat-Hang Fong
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan;
| | - Chi-Tai Yeh
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; (H.-W.L.); (Y.-K.S.); (C.-T.Y.); (M.-H.C.)
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Hematology and Oncology, Cancer Center, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department of Medical Research and Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; (H.-W.L.); (Y.-K.S.); (C.-T.Y.); (M.-H.C.)
| | - I-Hung Kan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; (H.-W.L.); (Y.-K.S.); (C.-T.Y.); (M.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan;
- Correspondence: (I.-H.K.); (C.-M.L.); Tel.: +886-2-2490088 (ext. 8881) (I.-H.K.)
| | - Chien-Min Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; (H.-W.L.); (Y.-K.S.); (C.-T.Y.); (M.-H.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Division of Neurosurgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan;
- Correspondence: (I.-H.K.); (C.-M.L.); Tel.: +886-2-2490088 (ext. 8881) (I.-H.K.)
| |
Collapse
|
11
|
Zeng J, Tian Q, Zeng Z, Cai J, Ye M, Liu Y, Liu A. Derlin-1 exhibits oncogenic activities and indicates an unfavorable prognosis in breast cancer. Cell Biol Int 2019; 44:593-602. [PMID: 31670413 DOI: 10.1002/cbin.11259] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/26/2019] [Indexed: 01/22/2023]
Abstract
Derlin-1 is involved in the elimination of misfolded proteins and has been implicated in the progression of human cancers. However, its prognostic value and biological function in breast cancer remain unknown. Here, we show that Derlin-1 is overexpressed in breast cancer and exhibits oncogenic activities via interaction with UBE2C. Increased expression of Derlin-1 is correlated with lymph node metastasis, advanced clinical stage, and unfavorable overall survival in two cohorts containing over 1,000 patients. Multivariate analyses by the Cox regression model suggest Derlin-1 is an independent factor for poor prognosis. In vitro experiments demonstrate that Derlin-1 expression is transcriptionally upregulated by c-Myc. Ectopic expression of Derlin-1 promotes breast cancer cell proliferation and migration, whereas the knockdown of Derlin-1 results in the opposite phenotypes. Mechanistically, Derlin-1 directly binds to UBE2C to increase the phosphorylation of AKT and ERK. The treatment of UBE2C siRNA markedly attenuates Derlin-1-mediated cell growth and migration. Collectively, our data suggest Derlin-1 is a potential prognostic factor and functions as an oncogene in breast cancer.
Collapse
Affiliation(s)
- Jinsheng Zeng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi, 330006, China
| | - Qiuhong Tian
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 330006, Jiangxi, China
| | - Zhimin Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, 330006, Jiangxi, China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, 330006, Jiangxi, China
| | - Meiyu Ye
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi, 330006, China
| | - Yaxiong Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi, 330006, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, 330006, Jiangxi, China
| |
Collapse
|
12
|
Li Z, Guo Q, Lu Y, Tian T. Increased expression of miR-181d is associated with poor prognosis and tumor progression of gastric cancer. Cancer Biomark 2019; 26:353-360. [PMID: 31524144 DOI: 10.3233/cbm-190091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Aberrant expression of miR-181d has been noted in multiple human cancers, but its role in gastric cancer (GC) remains unclear. The aim of this study was to investigate the expression, clinical significance and functional role of miR-181d in GC. We applied quantitative real-time polymerase chain reaction (qRT-PCR) to quantify the expression of miR-181d in 131 GC tissues, as well as in GC cell lines. The correlation of miR-181d expression with overall survival of GC patients was analyzed using the Kaplan-Meier survival method. Cox regression analysis was conducted to further determine the prognostic value of miR-181d in GC. Cellular functional experiments were carried out to calculate the effect that miR-181d had on GC behaviors. MiR-181d expression was significantly up-regulated in both GC tissues and cells (all P< 0.001), and correlated with TNM stage and lymph node metastasis (all P< 0.05). GC patients in the high miR-181d expression group had shorter survival time than those in the low miR-181d expression group (log-rank P< 0.001). Multivariate Cox regression analysis demonstrated that miR-181d expression and TNM stage were two independent prognostic markers for GC. Overexpression of miR-181d significantly promoted the NCI-N87 and MGC-803 cells proliferation, migration and invasion (all P< 0.05). MiR-181d serves a role as an oncogene in GC by promoting tumor progression. And miR-181d might be a novel predictive marker for the prognosis of GC patients.
Collapse
Affiliation(s)
- Zhichun Li
- Department of General Surgery, Caoxian people's Hospital, Heze, Shandong, China.,Department of General Surgery, Caoxian people's Hospital, Heze, Shandong, China
| | - Qingbo Guo
- Department of Laboratory Medicine, Yidu Central Hospital of Weifang, Shandong, China.,Department of General Surgery, Caoxian people's Hospital, Heze, Shandong, China
| | - Yugang Lu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tian Tian
- Department of General Medicine, Shengli Oilfield Central Hospital, Dongying, China
| |
Collapse
|
13
|
Li L, Liu M, Zhang Z, Zhang W, Liu N, Sheng X, Wei P. Derlin1 functions as an oncogene in cervical cancer via AKT/mTOR signaling pathway. Biol Res 2019; 52:8. [PMID: 30808417 PMCID: PMC6391802 DOI: 10.1186/s40659-019-0215-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/01/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Cervical cancer (CC) ranks third in the morbidity and mortality of female cancer around the world. Derlin1 has been found to be overexpressed in several human cancers. However, it is still unclear about its roles in CC. The research aims to explore the relationship between Derlin1 and CC. METHODS We purchased a human CC tissues microarray, which contained CC tissues and corresponding para-cancerous tissues from 93 patients with primary cervical squamous cell carcinoma. Immunohistochemical staining was used to confirm the expression of Derlin1 in these tissues. And we detected the differential expression of Derlin1 in cervical cancer cell lines and normal cervical epithelial cells (H8). Further, the cervical cancer cell lines SiHa and C33A were used as an in vitro model, which was down-regulated the expression of Derlin1 using siRNA interference technology. The effects of Derlin1 down-regulating in CC cell lines on cell proliferation and migration were detected by CCK8 assay and transwell assay, respectively. The effect of Derlin1 down-regulating on apoptosis was analyzed by flow cytometry, and apoptosis-related proteins were detected using western blotting. In-depth mechanisms were studied using western blotting. In addition, the effects of Derlin1 up-regulating in normal cervical epithelial cells also were exposed. RESULTS Derlin1 was significantly elevated in CC tissues (81.7%, 76/93), and the expression of Derlin1 was positively correlated with the tumor size, pathological grade, and lymph node metastasis in CC patients. And Derlin1 was high expressed in cervical cancer cell lines compared to H8 cells. Knockdown of Derlin1 in cervical cancer cell lines inhibited cell proliferation and migration. Moreover, knockdown of Derlin1 induced apoptosis and affected the expression of apoptosis-related proteins, including Bcl-2, Bax, Bim, caspase3 and caspase9. Further experiments showed that AKT/mTOR signal pathway might be involve in this processes that knockdown of Derlin1 inhibited the expression of p-AKT and p-mTOR. Over-expression of Derlin1 in H8 cells promoted cell proliferation and migration via up-regulated the expression of p-AKT and p-mTOR. CONCLUSION Derlin1 is an oncogene in CC via AKT/mTOR pathway. It might be a potential therapeutic target for CC.
Collapse
Affiliation(s)
- Ling Li
- Department of Oncology, People's Hospital of Tengzhou City, Tengzhou, 277599, Shandong, China
| | - Ming Liu
- Department of Gynecologic Oncology, Shandong Tumor Hospital and Institute, No. 440, Yan Ji Road, Jinan, 250117, Shandong, China
| | - Zhihu Zhang
- Shandong Academy of Occupational Health and Medicine, Jinan, 250062, Shandong, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong, China
| | - Naifu Liu
- Department of Gynecologic Oncology, Shandong Tumor Hospital and Institute, No. 440, Yan Ji Road, Jinan, 250117, Shandong, China
| | - Xiugui Sheng
- Department of Gynecologic Oncology, Tumor Hospital of Chinese Academy of Medical Sciences, Shenzhen, 518116, China
| | - Ping Wei
- Department of Gynecologic Oncology, Shandong Tumor Hospital and Institute, No. 440, Yan Ji Road, Jinan, 250117, Shandong, China.
| |
Collapse
|
14
|
Chen Y, Thomas PS, Kumar RK, Herbert C. The role of noncoding RNAs in regulating epithelial responses in COPD. Am J Physiol Lung Cell Mol Physiol 2018; 315:L184-L192. [PMID: 29722561 DOI: 10.1152/ajplung.00063.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD), one of the leading causes of death in the world, is a chronic inflammatory disease of the airways usually caused by long-term exposure to inhaled irritants. Airway epithelial cells (AECs) play a key role in initializing COPD and driving the exacerbation of this disease through the release of various cytokines. This AEC-derived cytokine response is tightly regulated possibly through the regulatory effects of noncoding RNAs (ncRNAs). Although the importance of ncRNAs in pulmonary diseases has been increasingly realized, little is known about the role of ncRNA in the regulation of inflammatory responses in COPD. This review outlines the features of AEC-derived cytokine responses in COPD and how ncRNAs regulate these inflammatory responses.
Collapse
Affiliation(s)
- Yifan Chen
- Department of Pathology, School of Medical Sciences, University of New South Wales Australia , Sydney , Australia
| | - Paul S Thomas
- Department of Pathology, School of Medical Sciences, University of New South Wales Australia , Sydney , Australia.,Department of Respiratory Medicine, Prince of Wales Hospital , Sydney , Australia
| | - Rakesh K Kumar
- Department of Pathology, School of Medical Sciences, University of New South Wales Australia , Sydney , Australia
| | - Cristan Herbert
- Department of Pathology, School of Medical Sciences, University of New South Wales Australia , Sydney , Australia
| |
Collapse
|
15
|
The role of microRNAs in the occurrence and development of esophageal squamous cell carcinoma. ACTA ACUST UNITED AC 2017. [DOI: 10.31491/csrc.2017.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Dong Q, Fu L, Zhao Y, Liu Y, Li Q, Qiu X, Wang E. Derlin-1 is a target to improve radiotherapy effect of esophageal squamous cell carcinoma. Oncotarget 2017; 8:55135-55146. [PMID: 28903408 PMCID: PMC5589647 DOI: 10.18632/oncotarget.19069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/18/2017] [Indexed: 01/12/2023] Open
Abstract
Radiotherapy is widely used for treatment of esophageal squamous cell carcinoma (ESCC). This study aimed to explore the role of Derlin-1 on the sensitivity of ESCC to radiotherapy and its underlying mechanism. We examined the clinical significance of Derlin-1 in 125 ESCC tissues. We found that Derlin-1 protein was higher in ESCC tissues than that in normal esophageal epithelial tissues. Derlin-1 overexpression was correlated with chemoradiotherapy resistance in ESCC patients and served an independent predictor for short overall survival. siRNA knockdown and plasmid transfection were carried out in ESCC cell lines. Derlin-1 depletion inhibited cell growth while its overexpression facilitated cell growth. Derlin-1 overexpression in Eca-109 cells dramatically enhanced its resistance to radiotherapy with decreased apoptosis rate. On the contrary, Derlin-1 depletion in TE-1 cell line showed the opposite effects. In addition, radioresistance conferred by Derlin-1 was attributed to its role of activating AKT/Bcl-2 signaling pathway and reducing caspase3 cleavage. Blockage of AKT signaling attenuated the role of Derlin-1 on radioresistance. Furthermore, Derlin-1 could interact with PI3K p110α in ESCC cell lines. Taken together, Our data demonstrate that Derlin-1 overexpression predicts poor prognosis and protects ESCC from irradiation induced apoptosis through PI3K/AKT/Bcl-2 signaling pathway. Derlin-1 may serve as a novel predictor for radiosentivity and a molecular target for ESCC.
Collapse
Affiliation(s)
- Qianze Dong
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Lin Fu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yue Zhao
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yang Liu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Qingchang Li
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xueshan Qiu
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Enhua Wang
- Department of Pathology, College of Basic Medical Sciences and The First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|