1
|
Farrokhnazar E, Moghbelinejad S, Najafipour R, Teimoori-Toolabi L. MiR-3664-3p through suppressing ABCG2, CYP3A4, MCL1, and MLH1 increases the sensitivity of colorectal cancer cells to irinotecan. Heliyon 2025; 11:e41933. [PMID: 39931465 PMCID: PMC11808512 DOI: 10.1016/j.heliyon.2025.e41933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 02/13/2025] Open
Abstract
Background Colorectal cancer (CRC) is the third most frequently diagnosed malignancy worldwide. Currently, irinotecan (CPT-11) is used alone or in combination with other drugs to treat patients with advanced CRC. However, the 5-year survival rate for metastatic CRC remains below 10 %, largely due to chemotherapy resistance. Several genes, including ABCG2, CYP3A4, MCL1, and MLH1 contribute to irinotecan resistance. This study aimed to identify microRNAs that simultaneously regulate the expression of these genes in irinotecan-resistant cell lines and study their effect on resistant colorectal cancer cells. Methods Irinotecan-resistant colorectal cancer cell lines were developed by intermittently exposing HCT116 and SW480 cell lines to gradually increasing doses of irinotecan over four generations. These resistant cell lines were designated HCT116-R1, HCT116-R2, HCT116-R3, HCT116-R4 and SW480-R1, SW480-R2, SW480-R3, SW480-R4. The induction of resistance was confirmed using MTT assays, by calculating IC50 values for each generation and comparing them to the parental cells. The expression levels of the ABCG2, CYP3A4, MCL1, and MLH1 genes, along with miR-3664-3p, were initially measured in all resistant and parental cell lines using quantitative real-time PCR. Following transfection of HCT116-R3 and SW480-R3 cells with pre-miR-3664-3p, the expression levels of ABCG2, CYP3A4, MCL1, MLH1, and miR-3664-3p were re-evaluated using real-time PCR. Results In resistant cell lines derived from HCT116 and SW480, increased expression of the ABCG2, CYP3A4, and MCL1 genes was observed. However, a reduction in CYP3A4 expression was noted in the final resistant lines from both cell lines. Additionally, while MLH1 expression increased in HCT116-derived cell lines, no significant increase was observed in SW480-derived lines. A consistent decrease in miR-3664-3p expression was found across all resistant cell lines. When we transfected HCT116-R3 and SW480-R3 cells with pre-miR-3664-3p, there was an increase in miR-3664-3p expression and a reduction in ABCG2, CYP3A4, MCL1, and MLH1 gene expression. This led to increased sensitivity to irinotecan. Conclusion It can be concluded that miR-3664-3p can be considered a regulator of resistance to irinotecan by modulating the expression of ABCG2, CYP3A4, MCL1, and MLH1 genes.
Collapse
Affiliation(s)
- Elham Farrokhnazar
- Research Institute for Prevention of Non-Communicable Diseases, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Iran
- Department of Molecular Medicine, Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sahar Moghbelinejad
- Research Institute for Prevention of Non-Communicable Diseases, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Reza Najafipour
- Genetics Research Center, University of Social Welfare and Rehabilitation Science, Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Iran
| |
Collapse
|
2
|
Hashemi M, Rashidi M, Hushmandi K, Ten Hagen TLM, Salimimoghadam S, Taheriazam A, Entezari M, Falahati M. HMGA2 regulation by miRNAs in cancer: affecting cancer hallmarks and therapy response. Pharmacol Res 2023; 190:106732. [PMID: 36931542 DOI: 10.1016/j.phrs.2023.106732] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
High mobility group A 2 (HMGA2) is a protein that modulates the structure of chromatin in the nucleus. Importantly, aberrant expression of HMGA2 occurs during carcinogenesis, and this protein is an upstream mediator of cancer hallmarks including evasion of apoptosis, proliferation, invasion, metastasis, and therapy resistance. HMGA2 targets critical signaling pathways such as Wnt/β-catenin and mTOR in cancer cells. Therefore, suppression of HMGA2 function notably decreases cancer progression and improves outcome in patients. As HMGA2 is mainly oncogenic, targeting expression by non-coding RNAs (ncRNAs) is crucial to take into consideration since it affects HMGA2 function. MicroRNAs (miRNAs) belong to ncRNAs and are master regulators of vital cell processes, which affect all aspects of cancer hallmarks. Long ncRNAs (lncRNAs) and circular RNAs (circRNAs), other members of ncRNAs, are upstream mediators of miRNAs. The current review intends to discuss the importance of the miRNA/HMGA2 axis in modulation of various types of cancer, and mentions lncRNAs and circRNAs, which regulate this axis as upstream mediators. Finally, we discuss the effect of miRNAs and HMGA2 interactions on the response of cancer cells to therapy. Regarding the critical role of HMGA2 in regulation of critical signaling pathways in cancer cells, and considering the confirmed interaction between HMGA2 and one of the master regulators of cancer, miRNAs, targeting miRNA/HMGA2 axis in cancer therapy is promising and this could be the subject of future clinical trial experiments.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Shuai SY, Liu SS, Liu XJ, Zhang GS, Zheng Q, Yue PF, Yang M, Hu PY. Essential oil of Ligusticum chuanxiong Hort. Regulated P-gp protein and tight junction protein to change pharmacokinetic parameters of temozolomide in blood, brain and tumor. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115646. [PMID: 36031103 DOI: 10.1016/j.jep.2022.115646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/08/2022] [Accepted: 08/13/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The existence of the blood-brain barrier/blood tumor barrier (BBB/BTB) severely restricts the effectiveness of anti-tumor drugs, thus glioma is still an incurable disease with a high fatality rate. Chuanxiong (Ligusticum chuanxiong Hort., Umbelliferae) was used as a messenger drug to increase the distribution of drugs in brain tissue, and its application in Chinese herbal formula for treating glioma was also the highest. AIM OF THE STUDY Our previous researches showed that essential oil (EO) of chuanxiong could promote temozolomide (TMZ) entry into glioma cells in vitro and enhance TMZ-induced anticancer efficiency in vivo, and therefore, the aim of this study was to investigate whether EO could increase the concentration accumulation of TMZ in brain or tumor of C6 glioma rats and the related mechanisms. MATERIALS AND METHODS The pharmacokinetics were conducted in C6 glioma rats by administering either TMZ alone or combined with EO through oral routes. TMZ concentration in blood, brain and tumor was detected using liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) and then pharmacokinetic parameters were calculated. The changed expressions of P-gp protein, tight junction occludin, claudin-5 and zonula occludens-1 (ZO-1) in brain of glioma rats were studied by Western blot to clarify the mechanism. Finally, the chemical composition of EO was analyzed by gas chromatography-massspectrometry (GC-MS). RESULTS The results showed that EO significantly affected the pharmacokinetic parameters such as Tmax, Cmax and CL (p < 0.01), but did not significantly change the AUC(0→∞) of TMZ in blood (p > 0.05). However, EO markedly improved the AUC(0→∞)of TMZ in brain and tumor (p < 0.01). The calculate drug targeting index was greater than 1, indicating that EO could promote the distribution of TMZ to the brain and tumor. Western blot analysis showed that EO significantly inhibited the expression of P-gp, tight junction protein claudin-5, occludin and ZO-1. And meanwhile, the expressions of P-gp, claudin-5 and occludin also markedly down-regulated in EO-TMZ co-administration treatment. GC-MS analysis of the TIC component of EO was (E)-Ligustilide (36.93%), Terpinolene (7.245%), gamma-terpinene (7.225%) etc. CONCLUSION: EO could promote the distribution of TMZ in the brain and tumor of C6 glioma rats, which may attribute to down-regulate the expression of P-gp, claudin-5 and occludin.
Collapse
Affiliation(s)
- Shu-Yuan Shuai
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Shan-Shan Liu
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Xiao-Jin Liu
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Guo-Song Zhang
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Qin Zheng
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Peng-Fei Yue
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Ming Yang
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Peng-Yi Hu
- Key Lab of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| |
Collapse
|
4
|
Wang X, Jiao B, Wu J, Yang J, Hu Y, Cui K. Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance. CNS Neurosci Ther 2022; 28:2319-2330. [PMID: 36184801 PMCID: PMC9627370 DOI: 10.1111/cns.13981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/09/2022] [Accepted: 07/16/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS We aimed to investigate the role of receptor-interacting protein 2 (RIP2) in regulation of stemness of glioma cells and chemotherapy resistance. METHODS Plasmid transfection was used to overexpress RIP2. Chemical inhibitors were used to inhibit RIP2 or NF-κB activity. Cancer stemness of glioma cells was investigated by sphere formation assays, clone formation assays, and xenograft tumor formation assays. The expression of RIP2, p-NF-κB, IκBα, CD133, or SOX-2 was detected by Western blotting and immunofluorescence. Apoptosis was detected by flow cytometry. Immunohistochemical staining was used to detect the expression of RIP2, CD133, and SOX-2 in xenograft tumor tissue. The effect of the RIP2/NF-κB pathway on temozolomide (TMZ) resistance was evaluated by xenograft tumor assay. RESULTS Transfection with RIP2 plasmid enhanced the sphere formation capability of U251 cells, clone formation capability, and xenograft tumor formation capability. RIP2 could mediate TMZ resistance by upregulating the expression of CD133 and SOX-2 by activating the NF-κB pathway. Both RIP2 inhibitor GSK583 and the NF-κB inhibitor SC75741 could reverse the resistance of U251 cells to TMZ. CONCLUSION RIP2 mediates TMZ resistance by regulating the maintenance of stemness in glioma cells through NF-κB. Interventions targeting the RIP2/NF-κB pathway may be a new strategy for TMZ-resistant gliomas.
Collapse
Affiliation(s)
- Xiao‐liang Wang
- Department of NeurosurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Bao‐hua Jiao
- Department of NeurosurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Jian‐liang Wu
- Department of NeurosurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Jian‐kai Yang
- Department of NeurosurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yu‐hua Hu
- Department of NeurosurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Kai Cui
- Department of NeurosurgeryThe Fourth Hospital of Hebei Medcial UniversityShijiazhuangChina
| |
Collapse
|
5
|
Xiao Q, Zhao W, Wu C, Wang X, Chen J, Shi X, Sha S, Li J, Liang X, Yang Y, Guo H, Wang Y, Fan J. Lemon-Derived Extracellular Vesicles Nanodrugs Enable to Efficiently Overcome Cancer Multidrug Resistance by Endocytosis-Triggered Energy Dissipation and Energy Production Reduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105274. [PMID: 35187842 PMCID: PMC9284146 DOI: 10.1002/advs.202105274] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Indexed: 05/20/2023]
Abstract
Multidrug resistance remains a great challenge for cancer chemotherapy. Herein, a biomimetic drug delivery system based on lemon-derived extracellular vesicles (EVs) nanodrugs (marked with heparin-cRGD-EVs-doxorubicin (HRED)) is demonstrated, achieving highly efficient overcoming cancer multidrug resistance. The HRED is fabricated by modifying functional heparin-cRGD (HR) onto the surface of EVs and then by loading with doxorubicin (DOX). The obtained HRED enable to effectively enter DOX-resistant cancer cells by caveolin-mediated endocytosis (main), macropinocytosis (secondary), and clathrin-mediated endocytosis (last), exhibiting excellent cellular uptake capacity. The diversified endocytosis capacity of HRED can efficiently dissipate intracellular energy and meanwhile trigger downstream production reduction of adenosine triphosphate (ATP), leading to a significant reduction of drug efflux. Consequently, they show excellent anti-proliferation capacities to DOX-resistant ovarian cancer, ensuring the efficiently overcoming ovarian cancer multidrug resistance in vivo. The authors believe this strategy provides a new strategy by endocytosis triggered-energy dissipation and ATP production reduction to design drug delivery system for overcoming cancer multidrug resistance.
Collapse
Affiliation(s)
- Qian Xiao
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Wei Zhao
- Division of Vascular and Interventional RadiologyDepartment of General Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515P. R. China
| | - Chentian Wu
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Xuejiao Wang
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Jianping Chen
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Xiubo Shi
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Suinan Sha
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Jinheng Li
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Xiaomei Liang
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Yulu Yang
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Haoyan Guo
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Ying Wang
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
| | - Jun‐Bing Fan
- Cancer Research InstituteExperimental Education/Administration CenterSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515P. R. China
- Department of Hepatobiliary Surgery IIZhujiang HospitalSouthern Medical UniversityGuangzhou510515P. R. China
| |
Collapse
|
6
|
Mahinfar P, Mansoori B, Rostamzadeh D, Baradaran B, Cho WC, Mansoori B. The Role of microRNAs in Multidrug Resistance of Glioblastoma. Cancers (Basel) 2022; 14:3217. [PMID: 35804989 PMCID: PMC9265057 DOI: 10.3390/cancers14133217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor that develops from neuroglial stem cells and represents a highly heterogeneous group of neoplasms. These tumors are predominantly correlated with a dismal prognosis and poor quality of life. In spite of major advances in developing novel and effective therapeutic strategies for patients with glioblastoma, multidrug resistance (MDR) is considered to be the major reason for treatment failure. Several mechanisms contribute to MDR in GBM, including upregulation of MDR transporters, alterations in the metabolism of drugs, dysregulation of apoptosis, defects in DNA repair, cancer stem cells, and epithelial-mesenchymal transition. MicroRNAs (miRNAs) are a large class of endogenous RNAs that participate in various cell events, including the mechanisms causing MDR in glioblastoma. In this review, we discuss the role of miRNAs in the regulation of the underlying mechanisms in MDR glioblastoma which will open up new avenues of inquiry for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Parvaneh Mahinfar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - Behnaz Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 175-14115, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran;
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Behzad Mansoori
- The Wistar Institute, Molecular & Cellular Oncogenesis Program, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Zeng Z, Chen Y, Geng X, Zhang Y, Wen X, Yan Q, Wang T, Ling C, Xu Y, Duan J, Zheng K, Sun Z. NcRNAs: Multi‑angle participation in the regulation of glioma chemotherapy resistance (Review). Int J Oncol 2022; 60:76. [PMID: 35506469 PMCID: PMC9083885 DOI: 10.3892/ijo.2022.5366] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/13/2022] [Indexed: 11/23/2022] Open
Abstract
As the most common primary tumour of the central nervous system, gliomas have a high recurrence rate after surgical resection and are resistant to chemotherapy, particularly high‑grade gliomas dominated by glioblastoma multiforme (GBM). The prognosis of GBM remains poor despite improvements in treatment modalities, posing a serious threat to human health. At present, although drugs such as temozolomide, cisplatin and bevacizumab, are effective in improving the overall survival of patients with GBM, most patients eventually develop drug resistance, leading to poor clinical prognosis. The development of multidrug resistance has therefore become a major obstacle to improving the effectiveness of chemotherapy for GBM. The ability to fully understand the underlying mechanisms of chemotherapy resistance and to develop novel therapeutic targets to overcome resistance is critical to improving the prognosis of patients with GBM. Of note, growing evidence indicates that a large number of abnormally expressed noncoding RNAs (ncRNAs) have a central role in glioma chemoresistance and may target various mechanisms to modulate chemosensitivity. In the present review, the roles and molecular mechanisms of ncRNAs in glioma drug resistance were systematically summarized, the potential of ncRNAs as drug resistance markers and novel therapeutic targets of glioma were discussed and prospects for glioma treatment were outlined. ncRNAs are a research direction for tumor drug resistance mechanisms and targeted therapies, which not only provides novel perspectives for reversing glioma drug resistance but may also promote the development of precision medicine for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Zhaomu Zeng
- Department of Surgery, School of Clinical Medicine, Hebei University, Baoding, Hebei 071000, P.R. China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Xiuchao Geng
- Department of Nursing, School of Medicine, Taizhou University, Jiaojiang, Zhejiang 318000, P.R. China
| | - Yuhao Zhang
- Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated to Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Xichao Wen
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Qingyu Yan
- Office of Academic Research, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Tingting Wang
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Chen Ling
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Yan Xu
- Clinical Laboratory, Affiliated Hospital of Jinggangshan University, Ji'an, Jiangxi 343100, P.R. China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Kebin Zheng
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Zhiwei Sun
- Department of Surgery, School of Clinical Medicine, Hebei University, Baoding, Hebei 071000, P.R. China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
8
|
Ghaemi S, Fekrirad Z, Zamani N, Rahmani R, Arefian E. Non-coding RNAs Enhance the Apoptosis Efficacy of Therapeutic Agents Used for the Treatment of Glioblastoma Multiform. J Drug Target 2022; 30:589-602. [DOI: 10.1080/1061186x.2022.2047191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Shokoofeh Ghaemi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Zahra Fekrirad
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Nina Zamani
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Rana Rahmani
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Zhong C, Tao B, Yang F, Xia K, Yang X, Chen L, Peng T, Xia X, Li X, Peng L. Histone demethylase JMJD1C promotes the polarization of M1 macrophages to prevent glioma by upregulating miR-302a. Clin Transl Med 2021; 11:e424. [PMID: 34586733 PMCID: PMC8473479 DOI: 10.1002/ctm2.424] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/26/2021] [Accepted: 05/01/2021] [Indexed: 12/23/2022] Open
Abstract
Glioma is regarded as an aggressive lethal primary brain tumor. Jumonji domain containing 1C (JMJD1C) is a H3K9 demethylase which participates in the progression of various tumors, but its specific function and underlying mechanism in glioma development remain undefined, which is the purpose of our work. We initially assessed JMJD1C expression in glioma tissues and cells using the assays of RT-qPCR and immunohistochemistry. Meanwhile, the H3K9 level at the microRNA (miR)-302a promoter region was measured by chromatin immunoprecipitation assay, while luciferase-based reporter assay was performed for validation of the binding affinity between miR-302a and methyltransferase-like 3 (METTL3). The effect of METTL3 on suppressor of cytokine signaling 2 (SOCS2) was subsequently analyzed by MeRIP-RT-qPCR. Finally, a xenograft tumor model was established in nude mice, followed by measurement of tumor-associated macrophages using flow cytometry. JMJD1C was poorly expressed in glioma tissues. Furthermore, JMJD1C increased miR-302a expression through promoting H3K9me1 demethylation at the miR-302a promoter region. miR-302a was identified to target METTL3, which could inhibit SOCS2 expression via m6A modification. JMJD1C promoted M1 macrophage polarization and suppressed the growth of glioma xenografts through the miR-302a/METTL3/SOCS2 axis both in vivo and in vitro. In conclusion, JMJD1C could enhance M1 macrophage polarization to inhibit the onset of glioma, bringing a new insight into the contribution of JMJD1C to the pathobiology of glioma, with possible implications for targeted therapeutic method.
Collapse
Affiliation(s)
- Chuanhong Zhong
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| | - Bei Tao
- Department of Rheumatologythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
| | - Feilong Yang
- Neurosurgery Departmentthe Affiliated Santai Hospital of North Sichuan Medical CollegeMianyang621100P. R. China
| | - Kaiguo Xia
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| | - Xiaobo Yang
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| | - Ligang Chen
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| | - Tangming Peng
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| | - Xiangguo Xia
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| | - Xianglong Li
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| | - Lilei Peng
- Department of Neurosurgerythe Affiliated Hospital of Southwest Medical UniversityLuzhouP. R. China
- Sichuan Clinical Research Center for NeurosurgeryLuzhouP. R. China
| |
Collapse
|
10
|
Gao K, Wang T, Qiao Y, Cui B. miR-23b-5p promotes the chemosensitivity of temozolomide via negatively regulating TLR4 in glioma. Acta Biochim Biophys Sin (Shanghai) 2021; 53:979-987. [PMID: 34110366 DOI: 10.1093/abbs/gmab066] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Indexed: 11/13/2022] Open
Abstract
Glioma is the most common malignancy in the brain, with poor survival and often highly resistant to chemotherapy and radiotherapy. Temozolomide (TMZ) is an alkylating agent widely used for glioma treatment. However, resistance to TMZ results in treatment failure, while the underlying mechanisms remain unclear. Mounting evidence suggests that dysregulated microRNA (miRNA) expression plays a critical function in glioma development and resistance to TMZ treatment. In this study, we found that miR-23b-5p was downregulated in glioma tissues and cell lines. Overexpression of miR-23b-5p inhibited cell proliferation and promoted cell apoptosis in glioma cells, while miR-23b-5p enhanced the chemosensitivity of TMZ in glioma cells. Furthermore, we identified that Toll-like receptor 4 (TLR4) is a direct target of miR-23b-5p in glioma cells. Knockdown of TLR4 suppressed cell proliferation and enhanced cell apoptosis and promoted chemosensitivity to TMZ treatment in glioma cells. In addition, we demonstrated that overexpression of TLR4 abrogated the regulatory function of miR-23b-5p in glioma cells on cell proliferation, cell apoptosis, and the chemosensitivity of TMZ treatment. In summary, our data suggest that miR-23b-5p promotes the chemosensitivity of TMZ via negatively regulating TLR4 in glioma, which provides a new therapeutic strategy for TMZ-resistant glioma treatment.
Collapse
Affiliation(s)
- Ke Gao
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Tuo Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yuan Qiao
- Department of Endocrinology, Shaanxi Provincial People’s Hospital, Xi’an 710068, China
| | - Bo Cui
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
11
|
Zhang Y, Meng H, Guo K. Inhibition of MicroRNA-302c on Stemness of Colon Cancer Stem Cells via the CARF/Wnt/β-Catenin Axis. Dig Dis Sci 2021; 66:1906-1915. [PMID: 32617772 DOI: 10.1007/s10620-020-06435-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/21/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Even though the relevance of microRNA (miR)-302c has been studied, little is known about its involvement in colon cancer (CC). AIMS Our aim here was to investigate the role of miR-302c in the cancer stem cells (CSCs) of CC. METHODS Firstly, the CSCs were screened out from cultured SW1116 and SW480 cells by flow cytometry, and the differentially expressed miRNAs in cell were obtained by microarray analysis. The expression of miR-302c, collaborator of ARF (CARF), and Wnt/β-catenin-related genes in CSCs was determined by means of RT-qPCR and Western blot. A dual-luciferase reporter assay was conducted to authenticate the binding relationship between miR-302c and CARF. Proliferation, migration, invasion, sphere formation as well as apoptosis of CSCs were assessed by cell counting kit-8, Transwell assay, sphere formation assay as well as flow cytometric analysis, respectively. The roles of miR-302c and CARF in tumor growth were determined in vivo. RESULTS The expression of miR-302c in CC cells was reduced versus that in normal cells. The overexpression of miR-302c weakened the stemness, proliferation, invasion, and migration abilities while induced apoptosis of CSCs in CC. Also, miR-302c reduced tumor size and weight in mice, accompanied with lowered CARF expression. The mechanistic analysis manifested that miR-302c bound to CARF and suppressed its expression and disrupted the Wnt/β-catenin signaling. CONCLUSION This study offers a novel characterization of miR-302c function in CSCs in CC, which may be beneficial to the development of capable therapeutic options for CC.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Gastroenterology, Caoxian People's Hospital, Development Zone, Fumin Avenue, Caoxian, 274400, Shandong, People's Republic of China
| | - Hua Meng
- Department of Gastroenterology, Caoxian People's Hospital, Development Zone, Fumin Avenue, Caoxian, 274400, Shandong, People's Republic of China
| | - Kun Guo
- Department of Gastroenterology, Caoxian People's Hospital, Development Zone, Fumin Avenue, Caoxian, 274400, Shandong, People's Republic of China.
| |
Collapse
|