1
|
Weber AF, Scholl JN, Dias CK, Lima VP, Assmann TS, Anzolin E, Kus WP, Worm PV, Battastini AMO, Figueiró F. In silico, in vitro, and ex vivo analysis reveals miR-27a-3p and miR-155-5p as key microRNAs for glioblastoma progression: Insights into Th1 differentiation and apoptosis induction. FASEB J 2024; 38:e70255. [PMID: 39698937 DOI: 10.1096/fj.202401538r] [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: 07/05/2024] [Revised: 11/22/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Abstract
We explored key microRNAs (miRNAs) related to tumorigenesis and immune modulation in glioblastoma (GBM), employing in silico, in vitro, and ex vivo analysis along with an assessment of the cellular impacts resulting from miRNA inhibition. GBM and T cells miRNA expression profiles from public datasets were used to evaluate differentially expressed miRNAs (DEmiRNAs). Some DEmiRNAs were chosen for validation in GBM cell lines, primary cell cultures, and brain tumor patient samples, using RT-qPCR. Target genes and pathways were identified with bioinformatic analyses. In silico functional enrichment analysis revealed that miR-27a-3p and miR-155-5p modulate immune, metabolic, and GBM-related pathways. A172 cells were transfected with miRNA inhibitors and the effects on cellular processes and immunomodulation were analyzed by co-culture assays and flow cytometry. Upon validation, miR-27a-3p and miR-155-5p miRNAs expressions were consistently increased. Inhibiting these two miRNAs reduced cell viability, but only the inhibition of miR-27a-3p led to apoptosis. Co-culture assays showed an increase in Th1 cells along with elevated Th1/Treg and Th17/Treg ratios, and an increase in Th17 cells exclusively with miR-155-5p inhibition. Immune cells' gene expression modulation induced an antitumor profile, concomitant with an increase in the expression of apoptotic genes in cancer cells after co-culture. This study unveils potential targets for immune and tumor regulation, highlighting overexpressed miRNAs modulation as a novel therapeutic approach for GBM.
Collapse
Affiliation(s)
- Augusto Ferreira Weber
- Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Cancer Immunobiochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Juliete Nathali Scholl
- Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Cancer Immunobiochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Camila Kehl Dias
- Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Cancer Immunobiochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Vinícius Pierdoná Lima
- Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Taís Silveira Assmann
- Molecular and Cellular Biology Laboratory, Endocrinology Division-Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eduardo Anzolin
- Department of Neurosurgery, Hospital Cristo Redentor, Porto Alegre, Brazil
| | | | - Paulo Valdeci Worm
- Department of Neurosurgery, Hospital Cristo Redentor, Porto Alegre, Brazil
| | - Ana Maria Oliveira Battastini
- Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabrício Figueiró
- Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Cancer Immunobiochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
2
|
Behrooz AB, Latifi-Navid H, Nezhadi A, Świat M, Los M, Jamalpoor Z, Ghavami S. Molecular mechanisms of microRNAs in glioblastoma pathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119482. [PMID: 37146725 DOI: 10.1016/j.bbamcr.2023.119482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
Glioblastoma (GBM) is human's most prevalent and severe brain cancer. Epigenetic regulators, micro(mi)RNAs, significantly impact cellular health and disease because of their wide range of targets and functions. The "epigenetic symphony" in which miRNAs perform is responsible for orchestrating the transcription of genetic information. The discovery of regulatory miRNA activities in GBM biology has shown that various miRNAs play a vital role in disease onset and development. Here, we summarize our current understanding of the current state-of-the-art and latest findings regarding the interactions between miRNAs and molecular mechanisms commonly associated with GBM pathogenesis. Moreover, by literature review and reconstruction of the GBM gene regulatory network, we uncovered the connection between miRNAs and critical signaling pathways such as cell proliferation, invasion, and cell death, which provides promising hints for identifying potential therapeutic targets for the treatment of GBM. In addition, the role of miRNAs in GBM patient survival was investigated. The present review, which contains new analyses of the previous literature, may lead to new avenues to explore in the future for the development of multitargeted miRNA-based therapies for GBM.
Collapse
Affiliation(s)
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Akram Nezhadi
- Cognitive Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Maciej Świat
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Marek Los
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
3
|
Lei L, Lu Q, Ma G, Li T, Deng J, Li W. P53 protein and the diseases in central nervous system. Front Genet 2023; 13:1051395. [PMID: 36712862 PMCID: PMC9880595 DOI: 10.3389/fgene.2022.1051395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/08/2022] [Indexed: 01/11/2023] Open
Abstract
P53 protein is the product of P53 gene, which is a well acknowledged tumor suppressor gene. The function of P53 and the relevant mechanisms of anti-neoplasm have raised the interest of researchers since many years ago. It is demonstrated that P53 is a basic cell cycle regulator and a strong inhibitor for versatile cancers in humans. However, most research focuses on other organs and systems instead of the central nervous system (CNS). In fact, in recent years, more and more studies have been suggesting that P53 plays a significant role in multiple CNS tumors and other diseases and disorders such as cerebral stroke and neurodegenerative diseases. In this work, we mainly reviewed the P53's relationship with CNS tumors, cerebral stroke and neurodegenerative diseases, together with the relevant mechanisms, aiming to summarize the research achievements and providing new insight to the future study on diseases in CNS.
Collapse
Affiliation(s)
- Li Lei
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Qixiong Lu
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Guifang Ma
- Department of Ear, Nose and Throat (ENT) and Head and Neck (HN) Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tao Li
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jiahong Deng
- Department of Ear, Nose and Throat (ENT) and Head and Neck (HN) Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China,*Correspondence: Jiahong Deng, ; Weijia Li,
| | - Weijia Li
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China,*Correspondence: Jiahong Deng, ; Weijia Li,
| |
Collapse
|
4
|
Ghafouri-Fard S, Khoshbakht T, Hussen BM, Dong P, Gassler N, Taheri M, Baniahmad A, Dilmaghani NA. A review on the role of cyclin dependent kinases in cancers. Cancer Cell Int 2022; 22:325. [PMID: 36266723 PMCID: PMC9583502 DOI: 10.1186/s12935-022-02747-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
The Cyclin-dependent kinase (CDK) class of serine/threonine kinases has crucial roles in the regulation of cell cycle transition and is mainly involved in the pathogenesis of cancers. The expression of CDKs is controlled by a complex regulatory network comprised of genetic and epigenetic mechanisms, which are dysregulated during the progression of cancer. The abnormal activation of CDKs results in uncontrolled cancer cell proliferation and the induction of cancer stem cell characteristics. The levels of CDKs can be utilized to predict the prognosis and treatment response of cancer patients, and further understanding of the function and underlying mechanisms of CDKs in human tumors would pave the way for future cancer therapies that effectively target CDKs. Defects in the regulation of cell cycle and mutations in the genes coding cell-cycle regulatory proteins lead to unrestrained proliferation of cells leading to formation of tumors. A number of treatment modalities have been designed to combat dysregulation of cell cycle through affecting expression or activity of CDKs. However, effective application of these methods in the clinical settings requires recognition of the role of CDKs in the progression of each type of cancer, their partners, their interactions with signaling pathways and the effects of suppression of these kinases on malignant features. Thus, we designed this literature search to summarize these findings at cellular level, as well as in vivo and clinical levels.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebeh Khoshbakht
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region, Iraq
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Nikolaus Gassler
- Section of Pathology, Institute of Forensic Medicine, Jena University Hospital, Jena, Germany
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| | - Nader Akbari Dilmaghani
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Geng F, Yang F, Liu F, Zhao J, Zhang R, Hu S, Zhang J, Zhang X. A miR-137-XIAP axis contributes to the sensitivity of TRAIL-induced cell death in glioblastoma. Front Oncol 2022; 12:870034. [PMID: 35965517 PMCID: PMC9366219 DOI: 10.3389/fonc.2022.870034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor in the central nervous system with limited therapeutic strategies to prolong the survival rate in clinic. TNF-related apoptosis-inducing ligand (TRAIL)-based strategy has been demonstrated to induce cell death in an extensive spectrum of tumor cells, including GBM, while a considerable proportion of malignant cells are resistant to TRAIL-induced apoptosis. MiR-137 is highly expressed in the brain, but significantly decreases with advanced progression of GBM. However, the functional link between miR-137 and TRAIL-induced apoptosis in GBM cells has not been established. Here, GBM cells were transfected with miR-137, and gene expression levels were examined by qRT-PCR and western blot. Apoptotic cells were measured by Annexin-V staining and TUNEL assay. Our data showed that miR-137 sensitizes GBM cells to the TRAIL-mediated apoptosis. Mechanistically, we identified that XIAP is a bona fide target of miR-137, which is essential for miR-137-regulated sensitivity of TRAIL-induced cell death in GBM cells. Finally, in a xenograft model, combined utilization of miR-137 and TRAIL potently suppresses tumor growth in vivo. Collectively, we demonstrate that a miR-137-XIAP axis is required for the sensitivity of TRAIL-induced cell death and shed a light on the avenue for the treatment of GBM.
Collapse
Affiliation(s)
- Fenghao Geng
- Department of Radiation Medicine, Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Fen Yang
- Department of Neurology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Fang Liu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jianhui Zhao
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Shijie Hu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiao Zhang, ; Jie Zhang, ; Shijie Hu,
| | - Jie Zhang
- Department of Radiation Medicine, Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiao Zhang, ; Jie Zhang, ; Shijie Hu,
| | - Xiao Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Research Office of the Institute of Tropical Medicine, Hainan Hospital of Chinese People's Liberation Army (PLA) General Hospital, Sanya, China
- *Correspondence: Xiao Zhang, ; Jie Zhang, ; Shijie Hu,
| |
Collapse
|
6
|
Liang D, Tian C, Zhang X. lncRNA MNX1‑AS1 promotes prostate cancer progression through regulating miR‑2113/MDM2 axis. Mol Med Rep 2022; 26:231. [PMID: 35616155 PMCID: PMC9178709 DOI: 10.3892/mmr.2022.12747] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/26/2021] [Indexed: 11/17/2022] Open
Abstract
A growing number of dysregulated long non‑coding (lnc)RNAs have been verified to serve an essential role in human prostate cancer. However, the underlying mechanisms of lncRNA MNX1 Antisense RNA 1 (MNX1‑AS1) in prostate cancer has not been explored. Therefore, the present study aimed to explore the function of MNX1‑AS1 in prostate cancer tumorigenesis and investigate the in‑depth mechanism. The expression of MNX1‑AS1, microRNA (miR)‑2113 and murine double min 2 (MDM2) in prostate cancer tissues and corresponding normal tissues were assessed by reverse transcription‑quantitative PCR. The protein expression levels of MDM2 were detected by western blotting. LNCaP and PC‑3 cells were transfected with short hairpin (sh)‑MNX1‑AS1, miR‑2113 mimics, miR‑2113 inhibitor and pCDH‑MDM2 vector using Lipofectamine® 3000. Cell proliferation, migration and invasion abilities were assessed by CCK‑8 assay, colony formation and Transwell assay, respectively. Dual luciferase reporter assay was carried out to confirm the putative targets of MNX1‑AS1 and miR‑2113. Tumor formation experiment in nude mice was applied to evaluate the tumor growth effect of MNX1‑AS1 in vivo. The expression of MNX1‑AS1 was significantly upregulated in the prostate cancer tissues and cell lines. MNX1‑AS1 knockdown suppressed the abilities of cell viability and migration and invasion in vitro and inhibited tumor growth in vivo. Additionally, luciferase reporter assay revealed that MNX1‑AS1 could target miR‑2113 and negatively interacted with miR‑2113 in prostate cancer cells. miR‑2113 directly targeted to MDM2 and negatively modulated the expression of MDM2. Rescue assays suggested that the viability, migration and invasion of impaired cells triggered by transfection with sh‑MNX1‑AS1 alone could be recovered by co‑transfection with sh‑MNX1‑AS1 + miR‑2113 inhibitor or sh‑MNX1‑AS1 + pCDH‑ MDM2 vector. The present study demonstrated that MNX1‑AS1 promoted prostate cancer progression through regulating miR‑2113/ MDM2 axis.
Collapse
Affiliation(s)
- Dong Liang
- Department of Urology Surgery, Binhai County Hospital of TCM, Yancheng, Jiangsu 224500, P.R. China
| | - Chuanjie Tian
- Department of Urology Surgery, Heqiao Hospital, Heqiao, Yixing, Jiangsu 214200, P.R. China
| | - Xiaowen Zhang
- Department of Urology Surgery, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
7
|
Wang H, Zeng Z, Yi R, Luo J, Chen J, Lou J. MicroRNA-3200-3p targeting CAMK2A modulates the proliferation and metastasis of glioma in vitro. Bioengineered 2022; 13:7785-7797. [PMID: 35287547 PMCID: PMC9208524 DOI: 10.1080/21655979.2022.2048995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
MicroRNA (miRNA) is strongly interrelated with the pathogenesis of glioma. However, its potential biological effect and underlying mechanism of miR-3200-3p in human glioma remain elusive. In the current study, we checked the level of miR-3200-3p in different glioma cells. Then, its biological functions on glioma cell proliferation metastasis was investigated using the miR-3200-3p mimic and inhibitor. The direct target of miR-3200-3p was tested in these cells. Results demonstrated that miR-3200-3p is remarkably downregulated in human glioma cells. The relative level of miR-3200-3p is strongly associated with biological features, including proliferation, colony formation, and metastasis. Additionally, Ca2+/calmodulin dependent kinase 2a (CAMK2A) might be the direct target gene of miR-3200-3p, and CAMK2A overexpression reversed the anticancer roles of miR-3200-3p on glioma cellular function. Importantly, these results further showed that miR-3200-3p downregulated the proliferation and metastasis by suppressing the expression of CAMK2A, thus regulating the Ras/Raf/MEK/ERK pathway. This study provided provided insights into the biological role of miR-3200-3p, which might function as a potential biomarker in glioma therapy.
Collapse
Affiliation(s)
- Haibin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Zhaobin Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Renhui Yi
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Jun Luo
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Jinming Chen
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, China
| | - Jianyun Lou
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, Jiangxi, 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
|
Casciati A, Tanori M, Gianlorenzi I, Rampazzo E, Persano L, Viola G, Cani A, Bresolin S, Marino C, Mancuso M, Merla C. Effects of Ultra-Short Pulsed Electric Field Exposure on Glioblastoma Cells. Int J Mol Sci 2022; 23:ijms23063001. [PMID: 35328420 PMCID: PMC8950115 DOI: 10.3390/ijms23063001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common brain cancer in adults. GBM starts from a small fraction of poorly differentiated and aggressive cancer stem cells (CSCs) responsible for aberrant proliferation and invasion. Due to extreme tumor heterogeneity, actual therapies provide poor positive outcomes, and cancers usually recur. Therefore, alternative approaches, possibly targeting CSCs, are necessary against GBM. Among emerging therapies, high intensity ultra-short pulsed electric fields (PEFs) are considered extremely promising and our previous results demonstrated the ability of a specific electric pulse protocol to selectively affect medulloblastoma CSCs preserving normal cells. Here, we tested the same exposure protocol to investigate the response of U87 GBM cells and U87-derived neurospheres. By analyzing different in vitro biological endpoints and taking advantage of transcriptomic and bioinformatics analyses, we found that, independent of CSC content, PEF exposure affected cell proliferation and differentially regulated hypoxia, inflammation and P53/cell cycle checkpoints. PEF exposure also significantly reduced the ability to form new neurospheres and inhibited the invasion potential. Importantly, exclusively in U87 neurospheres, PEF exposure changed the expression of stem-ness/differentiation genes. Our results confirm this physical stimulus as a promising treatment to destabilize GBM, opening up the possibility of developing effective PEF-mediated therapies.
Collapse
Affiliation(s)
- Arianna Casciati
- Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Division of Health Protection Technologies, Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (M.T.); (C.M.)
| | - Mirella Tanori
- Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Division of Health Protection Technologies, Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (M.T.); (C.M.)
| | - Isabella Gianlorenzi
- Department of Ecological and Biological Sciences, University of Tuscia, Largo dell’Università, snc, 01100 Viterbo, Italy;
| | - Elena Rampazzo
- Department of Women’s and Children’s Health (SDB), University of Padova, via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Division of Pediatric Hematology, Oncology and Hematopoietic Cell & Gene Therapy, Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Luca Persano
- Department of Women’s and Children’s Health (SDB), University of Padova, via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Division of Pediatric Hematology, Oncology and Hematopoietic Cell & Gene Therapy, Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Giampietro Viola
- Department of Women’s and Children’s Health (SDB), University of Padova, via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Division of Pediatric Hematology, Oncology and Hematopoietic Cell & Gene Therapy, Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Alice Cani
- Department of Women’s and Children’s Health (SDB), University of Padova, via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Division of Pediatric Hematology, Oncology and Hematopoietic Cell & Gene Therapy, Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Silvia Bresolin
- Department of Women’s and Children’s Health (SDB), University of Padova, via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Division of Pediatric Hematology, Oncology and Hematopoietic Cell & Gene Therapy, Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Carmela Marino
- Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Division of Health Protection Technologies, Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (M.T.); (C.M.)
| | - Mariateresa Mancuso
- Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Division of Health Protection Technologies, Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (M.T.); (C.M.)
- Correspondence: (M.M.); (C.M.)
| | - Caterina Merla
- Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Division of Health Protection Technologies, Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (M.T.); (C.M.)
- Correspondence: (M.M.); (C.M.)
| |
Collapse
|
10
|
Zhou L, Zhi Z, Chen P, Du C, Wang B, Fang X, Tang W, Li H. LncRNA-RMST Functions as a Transcriptional Co-regulator of SOX2 to Regulate miR-1251 in the Progression of Hirschsprung's Disease. Front Pediatr 2022; 10:749107. [PMID: 35321017 PMCID: PMC8936393 DOI: 10.3389/fped.2022.749107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/12/2022] [Indexed: 11/30/2022] Open
Abstract
Hirschsprung's disease (HSCR) is a congenital disorder characterized by the absence of enteric neural crest cells (ENCCs). LncRNA rhabdomyosarcoma 2-associated transcript (RMST) is essential for the growth and development of neuron. This study aimed to reveal the role of RMST in the pathogenesis of HSCR. The expression level of RMST, miR-1251, SOX2, and AHNAK was evaluated with qRT-PCR or western blot. CCK-8 and transwell assays were applied to detect cell proliferation and migration. CHIP and RIP assays were applied to determine the combination relationship between SOX2 and promoter region of miR-1251 or RMST and SOX2, respectively. Dual-luciferase reporter assay was performed to confirm miR-1251 targeted AHNAK. As results have shown, RMST was downregulated in the aganglionic colon of HSCR patients. The knockdown of RMST attenuated cell proliferation and migration significantly. MiR-1251, the intronic miRNA of RMST, was also low expressed in HSCR, but RMST did not alter the expression of miR-1251 directly. Furthermore, SOX2 was found to regulate the expression of miR-1251 via binding to the promoter region of miR-1251, and RMST strengthened this function by interacting with SOX2. Moreover, AHNAK was the target gene of miR-1251, which was co-regulated by RMST and SOX2. In conclusion, our study demonstrated that RMST functioned as a transcriptional co-regulator of SOX2 to regulate miR-1251 and resulted in the upregulation of AHNAK, leading to the occurrence of HSCR. The novel RMST/SOX2/miR-1251/AHNAK axis provided potential targets for the diagnosis and treatment of HSCR during embryonic stage.
Collapse
Affiliation(s)
- Lingling Zhou
- General Surgery Department, Children's Hospital of Wujiang District, Suzhou, China
| | - Zhengke Zhi
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Pingfa Chen
- Intensive Care Unit, The Fourth Affiliated Hospital of Nantong University, The First People's Hospital of Yancheng, Yancheng, China
| | - Chunxia Du
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Binyu Wang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Fang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hongxing Li
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Bao X, Peng Y, Shen J, Yang L. Sevoflurane inhibits progression of glioma via regulating the HMMR antisense RNA 1/microRNA-7/cyclin dependent kinase 4 axis. Bioengineered 2021; 12:7893-7906. [PMID: 34719318 PMCID: PMC8806593 DOI: 10.1080/21655979.2021.1976712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 11/04/2022] Open
Abstract
Sevoflurane (Sev) is a volatile anesthetic that can inhibit tumor malignancy. Glioma is a main brain problem, but the mechanism of Sev in glioma progression is largely unclear. This study aims to explore a potential regulatory network of long noncoding RNA (lncRNA)/microRNA (miRNA)/mRNA associated with the function of Sev in glioma progression. LncRNA HMMR antisense RNA 1 (HMMR-AS1), miR-7 and cyclin-dependent kinase 4 (CDK4) abundances were examined via quantitative reverse transcription polymerase chain reaction and western blot. Cell viability, invasion, and colony formation ability were analyzed via cell counting kit-8, transwell analysis, and colony formation. The target association was analyzed via dual-luciferase reporter analysis and RNA pull-down. The in vivo function of Sev was investigated by xenograft model. HMMR-AS1 abundance was increased in glioma tissues and cells, and reduced via Sev. Sev constrained cell viability, invasion, and colony formation ability via decreasing HMMR-AS1 in glioma cells. miR-7 expression was decreased in glioma, and was targeted via HMMR-AS1. HMMR-AS1 silence restrained cell viability, invasion, and colony formation ability by up-regulating miR-7 in glioma cells. Sev increases miR-7 abundance via decreasing HMMR-AS1. CDK4 was targeted via miR-7, and highly expressed in glioma. miR-7 overexpression inhibited cell viability, invasion, and colony formation ability via reducing CDK4 in glioma cells. CDK4 expression was reduced by Sev via HMMR-AS1/miR-7 axis. Sev suppressed cell growth in glioma by regulating HMMR-AS1. Sev represses glioma cell progression by regulating HMMR-AS1/miR-7/CDK4 axis.
Collapse
Affiliation(s)
- Xi’an Bao
- Department of Anesthesiology, The Affiliated Nanchang Hospital of SUN YAT-SEN University, Nanchang, 330006, China
| | - Yibo Peng
- Department of Anesthesiology, Chinese Medicine Hospital of Yangxin County, Huangshi, China
| | - Jun Shen
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Longqiu Yang
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Footprints of microRNAs in Cancer Biology. Biomedicines 2021; 9:biomedicines9101494. [PMID: 34680611 PMCID: PMC8533183 DOI: 10.3390/biomedicines9101494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs involved in post-transcriptional gene regulation. Over the past years, various studies have demonstrated the role of aberrant miRNA expression in the onset of cancer. The mechanisms by which miRNA exerts its cancer-promoting or inhibitory effects are apparent through the various cancer hallmarks, which include selective proliferative advantage, altered stress response, vascularization, invasion and metastasis, metabolic rewiring, the tumor microenvironment and immune modulation; therefore, this review aims to highlight the association between miRNAs and the various cancer hallmarks by dissecting the mechanisms of miRNA regulation in each hallmark separately. It is hoped that the information presented herein will provide further insights regarding the role of cancer and serve as a guideline to evaluate the potential of microRNAs to be utilized as biomarkers and therapeutic targets on a larger scale in cancer research.
Collapse
|
13
|
A Preclinical Investigation of GBM-N019 as a Potential Inhibitor of Glioblastoma via Exosomal mTOR/CDK6/STAT3 Signaling. Cells 2021; 10:cells10092391. [PMID: 34572040 PMCID: PMC8471927 DOI: 10.3390/cells10092391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive brain malignancies with high incidences of developing treatment resistance, resulting in poor prognoses. Glioma stem cell (GSC)-derived exosomes are important players that contribute to GBM tumorigenesis and aggressive properties. Herein, we investigated the inhibitory roles of GBM-N019, a novel small molecule on the transfer of aggressive and invasive properties through the delivery of oncogene-loaded exosomes from GSCs to naïve and non-GSCs. Our results indicated that GBM-N019 significantly downregulated the expressions of the mammalian target of rapamycin (mTOR), signal transducer and activator of transcription 3 (STAT3), and cyclin-dependent kinase 6 (CDK6) signaling networks with concomitant inhibitory activities against viability, clonogenicity, and migratory abilities of U251 and U87MG cells. Treatments with GBM-N019 halted the exosomal transfer of protein kinase B (Akt), mTOR, p-mTOR, and Ras-related protein RAB27A to the naïve U251 and U87MG cells, and rescued the cells from invasive and stemness properties that were associated with activation of these oncogenes. GBM-N019 also synergized with and enhanced the anti-GBM activities of palbociclib in vitro and in vivo. In conclusion, our results suggested that GBM-N019 possesses good translational relevance as a potential anti-glioblastoma drug candidate worthy of consideration for clinical trials against recurrent glioblastomas.
Collapse
|
14
|
Gheidari F, Arefian E, Adegani FJ, Kalhori MR, Seyedjafari E, Kabiri M, Teimoori-Toolabi L, Soleimani M. miR-424 induces apoptosis in glioblastoma cells and targets AKT1 and RAF1 oncogenes from the ERBB signaling pathway. Eur J Pharmacol 2021; 906:174273. [PMID: 34153339 DOI: 10.1016/j.ejphar.2021.174273] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma is a lethal and incurable cancer. Tumor suppressor miRNAs are promising gene therapy tools for cancer treatment. In silico, we predicted miR-424 as a tumor suppressor. It had several target genes from the epidermal growth factor receptor (ERBB) signaling pathway that are overactive in most glioblastoma cases. We overexpressed miR-424 by lentiviral transduction of U-251 and U-87 glioblastoma cells confirmed with fluorescent microscopy and real-time quantitative PCR (qRT-PCR). Then the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) proliferation assay and scratch wound migration assay were performed to investigate the miR-424 tumor suppressor effect in glioblastoma. miR-424's effect on glioblastoma apoptosis and cell-cycle arrest was verified using Annexin V- phosphatidylethanolamine (PE) and 7-minoactinomycin D (7-AAD) apoptosis assay and cell-cycle assay. miR-424 predicted target genes mRNA and protein level were measured after miR-424 overexpression in comparison to the control group by qRT-PCR and western blotting, respectively. We confirmed miR-424 direct target genes by dual-luciferase reporter assay. miR-424 overexpression significantly suppressed cell proliferation and migration rate in glioblastoma cells based on the MTT and scratch assays. Flow cytometry results confirmed that miR-424 promotes apoptosis and cell-cycle arrest in glioblastoma cells. Predicted target genes of miR-424 from the ERBB pathway were downregulated by miR-424 overexpression. qRT-PCR and western blotting showed that KRAS, RAF1, MAP2K1, EGFR, PDGFRA, AKT1, and mTOR mRNA expression levels and KRAS, RAF1, MAP2K1, EGFR, and AKT1 protein level, respectively, had significantly decreased as a result of miR-424 overexpression in comparison to the control group. Dual-luciferase reporter assay confirmed that miR-424 directly targets RAF1 and AKT1 oncogenes. Overall, miR-424 acts as tumor suppressor miRNA in glioblastoma cells.
Collapse
Affiliation(s)
- Fatemeh Gheidari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran; Stem Cell Technology Research Center, 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.
| | - Fatemeh Jamshidi Adegani
- Laboratory for Stem Cell & Regenerative Medicine, Natural and Medicinal Sciences Research Center, University of Nizwa, Nizwa, Oman.
| | - Mohammad Reza Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | - Mahboubeh Kabiri
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | - Ladan Teimoori-Toolabi
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
15
|
Miles X, Vandevoorde C, Hunter A, Bolcaen J. MDM2/X Inhibitors as Radiosensitizers for Glioblastoma Targeted Therapy. Front Oncol 2021; 11:703442. [PMID: 34307171 PMCID: PMC8296304 DOI: 10.3389/fonc.2021.703442] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Inhibition of the MDM2/X-p53 interaction is recognized as a potential anti-cancer strategy, including the treatment of glioblastoma (GB). In response to cellular stressors, such as DNA damage, the tumor suppression protein p53 is activated and responds by mediating cellular damage through DNA repair, cell cycle arrest and apoptosis. Hence, p53 activation plays a central role in cell survival and the effectiveness of cancer therapies. Alterations and reduced activity of p53 occur in 25-30% of primary GB tumors, but this number increases drastically to 60-70% in secondary GB. As a result, reactivating p53 is suggested as a treatment strategy, either by using targeted molecules to convert the mutant p53 back to its wild type form or by using MDM2 and MDMX (also known as MDM4) inhibitors. MDM2 down regulates p53 activity via ubiquitin-dependent degradation and is amplified or overexpressed in 14% of GB cases. Thus, suppression of MDM2 offers an opportunity for urgently needed new therapeutic interventions for GB. Numerous small molecule MDM2 inhibitors are currently undergoing clinical evaluation, either as monotherapy or in combination with chemotherapy and/or other targeted agents. In addition, considering the major role of both p53 and MDM2 in the downstream signaling response to radiation-induced DNA damage, the combination of MDM2 inhibitors with radiation may offer a valuable therapeutic radiosensitizing approach for GB therapy. This review covers the role of MDM2/X in cancer and more specifically in GB, followed by the rationale for the potential radiosensitizing effect of MDM2 inhibition. Finally, the current status of MDM2/X inhibition and p53 activation for the treatment of GB is given.
Collapse
Affiliation(s)
- Xanthene Miles
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Alistair Hunter
- Radiobiology Section, Division of Radiation Oncology, Department of Radiation Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| |
Collapse
|
16
|
Xu H, Wang F, Wang L. Suppression of miR-106a-5p expression inhibits tumorigenesis via increasing CELF-2 expression in spinal cord glioma. Oncol Lett 2021; 22:627. [PMID: 34267819 PMCID: PMC8258617 DOI: 10.3892/ol.2021.12888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 05/13/2021] [Indexed: 12/26/2022] Open
Abstract
Spinal cord glioma is a tumor characterized by high recurrence and mortality rates, and its treatment remains a major challenge. It has been reported that abnormal expression of microRNAs (miRNAs/miRs) is associated with tumor progression. Therefore, the current study aimed to identify novel miRNAs associated with spinal cord glioma. Herein, the expression levels of several miRNAs were determined in human spinal cord glioma and adjacent non-cancerous tissues by reverse transcription-quantitative (RT-qPCR). The results revealed that miR-106a-5p expression was markedly upregulated in spinal cord glioma tissues compared with in non-cancerous tissues. Furthermore, the biological effects of miR-106a-5p on spinal cord glioma cells were evaluated by MTT, Transwell and flow cytometric assays. In 0231SCG cells transfected with miR-106a-5p inhibitor, cell proliferation, migration and invasion were attenuated, whereas apoptosis was enhanced. A search of the TargetScan database revealed that miR-106a-5p directly targeted CUGBP Elav-like family member 2 (CELF-2). Western blot and RT-qPCR experiments further confirmed the association between miR-106a-5p and CELF-2 expression in spinal cord glioma tissues. The current results demonstrated that CELF-2 was a direct target of miR-106a-5p, and that the expression levels of CELF-2 were negatively associated with those of miR-106a-5p. In addition, overexpression of CELF-2 in spinal cord glioma cells reversed the tumor-promoting effects of miR-106a-5p both in vitro and in vivo. Overall, the aforementioned findings indicated that miR-106a-5p, which was highly expressed in spinal cord glioma tissues, may affect the proliferation, migration, invasion and apoptosis of spinal cord glioma cells via targeting CELF-2, thus indicating a potential approach to the future clinical management of spinal cord glioma.
Collapse
Affiliation(s)
- Hao Xu
- Department of Neurosurgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Lin Wang
- Department of Neurosurgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| |
Collapse
|
17
|
Wang G, Zhou H, Tian L, Yan T, Han X, Chen P, Li H, Wang W, Xiao Z, Hou L, Xue X. A Prognostic DNA Damage Repair Genes Signature and Its Impact on Immune Cell Infiltration in Glioma. Front Oncol 2021; 11:682932. [PMID: 34123852 PMCID: PMC8193723 DOI: 10.3389/fonc.2021.682932] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
Objective Glioma is the most frequent type of malignant cerebral tumors. DNA damage repair genes (DDRGs) play a crucial role in the development of cancer. In this study, we constructed a DDRGs signature and investigated the potential mechanisms involved in this disease. Methods RNA sequence data, microarray data, and corresponding clinical information of gliomas were downloaded from The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), and Gene Expression Omnibus (GEO). Subsequently, we identified candidate genes by differential analysis and Cox regression analysis. The least absolute shrinkage and selection operator Cox regression model was utilized to construct a DDRGs signature using TCGA training dataset. According to this signature, patients with glioma were divided into low- and high-risk groups. The predictive ability of the signature was validated by prognostic analysis, receiver operating characteristic curves, principal component analysis, and stratification analysis in TCGA testing and CGGA verification datasets. CIBERSORT and single-sample gene set enrichment analysis (ssGSEA) were used to evaluate the immune microenvironment of glioma. Moreover, we conducted GSEA to determine the functions and pathways in the low- and high-risk groups. Finally, a nomogram was constructed by combining the signature and other clinical features. Results A total of 1,431 samples of glioma (592 from TCGA, 686 from the CGGA, and 153 from the GEO) and 23 samples of normal brain tissue from the GEO were analyzed in this study. There were 51 prognostic differentially expressed DDRGs. Additionally, five DDRGs (CDK4、HMGB2、WEE1、SMC3 and GADD45G) were selected to construct a DDRGs signature for glioma, stratifying patients into low- and high-risk groups. The survival analysis showed that the DDRGs signature could differentiate the outcome of the low- and high-risk groups, showing that high-risk gliomas were associated with shorter overall survival. The immune microenvironment analysis revealed that more immunosuppressive cells, such as tumor associated macrophages and regulatory T cells, were recruited in the high-risk group. GSEA also showed that high-risk glioma was correlated with the immune and extracellular matrix pathways. Conclusion The five DDRGs signature and its impact on the infiltration of immunosuppressive cells could precisely predict the prognosis and provide guidance on the treatment of glioma.
Collapse
Affiliation(s)
- Guohui Wang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Radiation Oncology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Huandi Zhou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Tian
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tianfang Yan
- Department of Neurological Diagnosis and Restoration, Osaka University Graduate School of Medicine, Suita, Japan
| | - Xuetao Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pengyu Chen
- Department of Neurosurgery, Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Haonan Li
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenyan Wang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhiqing Xiao
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Liubing Hou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoying Xue
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
18
|
Li ZK, Gao LF, Zhu XA, Xiang DK. LncRNA HOXA-AS3 Promotes the Progression of Pulmonary Arterial Hypertension through Mediation of miR-675-3p/PDE5A Axis. Biochem Genet 2021; 59:1158-1172. [PMID: 33687636 DOI: 10.1007/s10528-021-10053-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 02/19/2021] [Indexed: 11/25/2022]
Abstract
Pulmonary arterial hypertension (PAH) seriously threatens the elder people. Long non-coding RNAs (lncRNAs) are involved in multiple diseases. However, the study of the lncRNAs in the occurrence of PAH is just beginning. For this, we sought to explore the biological function of lncRNA HOXA cluster antisense RNA 3 (HOXA-AS3) in PAH. Hypoxia (HYP) was used to mimic in vitro model of PAH. Gene and protein expressions in cells were detected by q-PCR and Western blotting, respectively. In addition, cell proliferation and viability were tested by CCK-8 and MTT assay. Cell apoptosis was measured by flow cytometry. Wound healing was used to detect cell migration. Furthermore, the connection of HOXA-AS3, miR-675-3p, and phosphodiesterase 5A (PDE5A) was verified by dual-luciferase report assay. HOXA-AS3 and PDE5A were upregulated in human pulmonary artery smooth muscle cells (HPASMCs) in the presence of HYP, while miR-675-3p was downregulated. Moreover, knockdown of HOXA-AS3 suppressed the growth and migration of HPASMCs, but induced the apoptosis. Overexpression of miR-675-3p achieved the same effect. MiR-675-3p inhibitor or overexpression of PDE5A notably reversed the inhibitory effect of HOXA-AS3 knockdown on PAH. Finally, HOXA-AS3 could sponge miR-675-3p, and PDE5A was directly targeted by miR-675-3p. HOXA-AS3 increased the development of PAH via regulation of miR-675-3p/PDE5 axis, which could be the potential biomarker for treatment of PAH.
Collapse
Affiliation(s)
- Zhong-Kui Li
- Department of Cardiovascular Surgery, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, 550002, Guizhou Province, People's Republic of China
| | - Lu-Fang Gao
- Department of Cardiovascular Surgery, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, 550002, Guizhou Province, People's Republic of China
| | - Xi-An Zhu
- Department of Cardiovascular Surgery, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, 550002, Guizhou Province, People's Republic of China
| | - Dao-Kang Xiang
- Department of Cardiovascular Surgery, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, 550002, Guizhou Province, People's Republic of China.
| |
Collapse
|
19
|
Yao X, Shen H, Peng Q, Yu J. TP53/miR-129/MDM2/4/TP53 feedback loop modulates cell proliferation and apoptosis in retinoblastoma. Cell Cycle 2021; 20:603-615. [PMID: 33678118 DOI: 10.1080/15384101.2021.1892321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Retinoblastoma (RB) is commonly-seen cancer in children. The p53 pathway dysfunction, which can lead to elevated MDM2 or MDM4 (p53 antagonists) protein expression, is frequently observed in almost all human cancers, including RB. The present study attempted to investigate the underlying mechanism from the perspective of non-coding RNA regulation. Here, we demonstrated that p53 and miR-129 were positively correlated with each other in RB. miR-129 directly targeted MDM2/4 to inhibit expression, therefore counteracting MDM2/4-mediated p53 signaling suppression and modulating RB cell proliferation and apoptosis. Moreover, p53 could activate the transcription of miR-129 via binding to the miR-129 promoter region, therefore forming a regulatory loop with MDM2/4 to affect RB progression. Altogether, the p53/miR-129/MDM2/4/p53 regulatory loop can modulate RB cell growth. We provide a solid experimental basis for developing novel therapies for RB.
Collapse
Affiliation(s)
- Xiaolei Yao
- Department of Ophthalmology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China.,Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.,Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Eye, Ear, Nose and Throat Diseases in Hunan Province, Changsha, China
| | - Hui Shen
- Department of Ophthalmology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Qinghua Peng
- Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.,Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Eye, Ear, Nose and Throat Diseases in Hunan Province, Changsha, China.,First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jingsheng Yu
- Department of Ophthalmology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
20
|
Zepecki JP, Karambizi D, Fajardo JE, Snyder KM, Guetta-Terrier C, Tang OY, Chen JS, Sarkar A, Fiser A, Toms SA, Tapinos N. miRNA-mediated loss of m6A increases nascent translation in glioblastoma. PLoS Genet 2021; 17:e1009086. [PMID: 33684100 PMCID: PMC7971852 DOI: 10.1371/journal.pgen.1009086] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 03/18/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Within the glioblastoma cellular niche, glioma stem cells (GSCs) can give rise to differentiated glioma cells (DGCs) and, when necessary, DGCs can reciprocally give rise to GSCs to maintain the cellular equilibrium necessary for optimal tumor growth. Here, using ribosome profiling, transcriptome and m6A RNA sequencing, we show that GSCs from patients with different subtypes of glioblastoma share a set of transcripts, which exhibit a pattern of m6A loss and increased protein translation during differentiation. The target sequences of a group of miRNAs overlap the canonical RRACH m6A motifs of these transcripts, many of which confer a survival advantage in glioblastoma. Ectopic expression of the RRACH-binding miR-145 induces loss of m6A, formation of FTO/AGO1/ILF3/miR-145 complexes on a clinically relevant tumor suppressor gene (CLIP3) and significant increase in its nascent translation. Inhibition of miR-145 maintains RRACH m6A levels of CLIP3 and inhibits its nascent translation. This study highlights a critical role of miRNAs in assembling complexes for m6A demethylation and induction of protein translation during GSC state transition.
Collapse
Affiliation(s)
- John P. Zepecki
- Laboratory of Cancer Epigenetics and Plasticity, Brown University, Rhode Island Hospital, Providence Rhode Island, United States of America
| | - David Karambizi
- Laboratory of Cancer Epigenetics and Plasticity, Brown University, Rhode Island Hospital, Providence Rhode Island, United States of America
| | - J. Eduardo Fajardo
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Kristin M. Snyder
- University of Minnesota, College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Charlotte Guetta-Terrier
- Laboratory of Cancer Epigenetics and Plasticity, Brown University, Rhode Island Hospital, Providence Rhode Island, United States of America
| | - Oliver Y. Tang
- Laboratory of Cancer Epigenetics and Plasticity, Brown University, Rhode Island Hospital, Providence Rhode Island, United States of America
| | - Jia-Shu Chen
- Laboratory of Cancer Epigenetics and Plasticity, Brown University, Rhode Island Hospital, Providence Rhode Island, United States of America
| | - Atom Sarkar
- Department of Neurosurgery, Drexel Neuroscience Institute, Philadelphia Pennsylvania, United States of America
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Steven A. Toms
- Department of Neurosurgery, Brown University, Providence Rhode Island, United States of America
| | - Nikos Tapinos
- Laboratory of Cancer Epigenetics and Plasticity, Brown University, Rhode Island Hospital, Providence Rhode Island, United States of America
- Department of Neurosurgery, Brown University, Providence Rhode Island, United States of America
- Cancer Biology Program, Brown University, Lifespan Cancer Institute, Providence RI, USA
- Carney Institute for Brain Science, Brown University, Providence, RI, USA
| |
Collapse
|
21
|
Guan Q, Yuan L, Lin A, Lin H, Huang X, Ruan J, Zhuo Z. KRAS gene polymorphisms are associated with the risk of glioma: a two-center case-control study. Transl Pediatr 2021; 10:579-586. [PMID: 33850816 PMCID: PMC8039792 DOI: 10.21037/tp-20-359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Glioma, also known as neuroglioma, is the most common primary tumors of the central nervous system. Many previous studies have reported associations between RAS gene polymorphisms and multiple tumors. However, the role of RAS gene polymorphisms on glioma risk has not been investigated. METHODS We conducted a two-center case-control study to investigate whether the RAS gene polymorphisms predispose individuals to gliomas in 248 healthy controls and 191 glioma patients. RAS gene polymorphisms (rs12587 G>T, rs7973450 A>G, rs7312175 G>A in KRAS, rs2273267 A>T in NRAS) were genotyped by the TaqMan assay. The relationship between RAS gene functional single nucleotide polymorphisms (SNPs) and the risk of glioma was evaluated based on odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Individuals with KRAS rs7312175 GA genotype were more likely to develop glioma than those with GG genotype (adjusted OR =1.66, 95% CI: 1.05-2.64, P=0.030). However, the other three SNPs could not affect glioma risk. In stratified analysis of age, gender, subtypes, and clinical stages, rs7312175 GA carriers were more likely to develop glioma in the following subgroups: children less than 60 months, tumor derived from the astrocytic tumors, and clinical stages I. CONCLUSIONS The study showed that polymorphism rs7312175 GA in the KRAS gene was associated with increased glioma susceptibility. Further investigation is warranted to confirm these findings and to better elucidate the involved biological pathways.
Collapse
Affiliation(s)
- Qian Guan
- School of Medicine, South China University of Technology, Guangzhou, China.,Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Li Yuan
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Ao Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Huiran Lin
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Xiaokai Huang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jichen Ruan
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zhenjian Zhuo
- School of Medicine, South China University of Technology, Guangzhou, China.,Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou, China
| |
Collapse
|
22
|
Abstract
This review considers glioma molecular markers in brain tissues and body fluids, shows the pathways of their formation, and describes traditional methods of analysis. The most important optical properties of glioma markers in the terahertz (THz) frequency range are also presented. New metamaterial-based technologies for molecular marker detection at THz frequencies are discussed. A variety of machine learning methods, which allow the marker detection sensitivity and differentiation of healthy and tumor tissues to be improved with the aid of THz tools, are considered. The actual results on the application of THz techniques in the intraoperative diagnosis of brain gliomas are shown. THz technologies’ potential in molecular marker detection and defining the boundaries of the glioma’s tissue is discussed.
Collapse
|
23
|
Gheidari F, Arefian E, Jamshidi Adegani F, Fallah Atanaki F, Soleimani M. The miR-142 Suppresses U-87 Glioblastoma Cell Growth by Targeting EGFR Oncogenic Signaling Pathway. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:202-212. [PMID: 35194440 PMCID: PMC8842594 DOI: 10.22037/ijpr.2021.115089.15193] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Glioblastoma is the most lethal malignancy of the brain and is resistant to conventional cancer treatments. Gene-therapy approaches like using tumor suppressor miRNAs are promising in the treatment of glioblastoma. They control the expression of oncogenes and influence tumor features and behaviors. Therefore, in the present study, it was predicted that miR-142 regulates oncogenic epidermal growth factor receptor (EGFR) signaling pathway via TargetScan and miRWalk online tools. Its differential expression level was reduced in glioblastoma according to the previous microarray results, and its predicted target genes were upregulated, as shown by the Expression Atlas. The miR-142 was overexpressed in U-87 glioblastoma cells via lentiviral transduction, and the way it influences proliferation and migration of cells was investigated through MTT assay and wound healing assay. Apoptosis rate was also measured via the Annexin V assay, and cell-cycle analysis was done. Then, real-time polymerase chain reaction (real-time PCR) and western blotting were performed to assess fold changes in mRNA and protein levels of the miR-142 predicted targets. Direct target genes of miR-142 were confirmed through a dual-luciferase reporter assay. The miR-142 significantly suppressed cell proliferation and migration and induced apoptosis and cell-cycle arrest in U-87 glioblastoma cells. This was accompanied by a decrease in expression of SHC adaptor protein 4 (SHC4), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), v-akt murine thymoma viral oncogene homolog 1 (AKT1), Kirsten rat sarcoma viral oncogene homolog (KRAS), and mitogen-activated protein kinase 8 (MAPK8) oncogenes at mRNA and protein levels in glioblastoma cells. Also, AKT1 was demonstrated as a direct target of miR-142. Overall, miR-424 acts as tumor suppressor miRNA in glioblastoma cells.
Collapse
Affiliation(s)
- Fatemeh Gheidari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
- Stem Cell Technology Research Center, 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.
| | - Fatemeh Jamshidi Adegani
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medicinal Sciences Research Center, University of Nizwa, Nizwa, P. O. Box: 33, PC 616, Oman.
| | - Fereshteh Fallah Atanaki
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
24
|
Uddin MS, Mamun AA, Alghamdi BS, Tewari D, Jeandet P, Sarwar MS, Ashraf GM. Epigenetics of glioblastoma multiforme: From molecular mechanisms to therapeutic approaches. Semin Cancer Biol 2020; 83:100-120. [PMID: 33370605 DOI: 10.1016/j.semcancer.2020.12.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common form of brain cancer and one of the most aggressive cancers found in humans. Most of the signs and symptoms of GBM can be mild and slowly aggravated, although other symptoms might demonstrate it as an acute ailment. However, the precise mechanisms of the development of GBM remain unknown. Due to the improvement of molecular pathology, current researches have reported that glioma progression is strongly connected with different types of epigenetic phenomena, such as histone modifications, DNA methylation, chromatin remodeling, and aberrant microRNA. Furthermore, the genes and the proteins that control these alterations have become novel targets for treating glioma because of the reversibility of epigenetic modifications. In some cases, gene mutations including P16, TP53, and EGFR, have been observed in GBM. In contrast, monosomies, including removals of chromosome 10, particularly q23 and q25-26, are considered the standard markers for determining the development and aggressiveness of GBM. Recently, amid the epigenetic therapies, histone deacetylase inhibitors (HDACIs) and DNA methyltransferase inhibitors have been used for treating tumors, either single or combined. Specifically, HDACIs are served as a good choice and deliver a novel pathway to treat GBM. In this review, we focus on the epigenetics of GBM and the consequence of its mutations. We also highlight various treatment approaches, namely gene editing, epigenetic drugs, and microRNAs to combat GBM.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region
| | - Badrah S Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687, Reims Cedex 2, France
| | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
25
|
Zhao L, Tian C, Xiao E, Du J, Liang J, Chen X, Chi W. Clinical significance and potential mechanisms of miR-223-3p and miR-204-5p in squamous cell carcinoma of head and neck: a study based on TCGA and GEO. Open Med (Wars) 2020; 15:728-738. [PMID: 33336030 PMCID: PMC7712329 DOI: 10.1515/med-2020-0146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/10/2020] [Accepted: 06/21/2020] [Indexed: 01/13/2023] Open
Abstract
Objective To explore the clinical significance and mechanisms of altered miRNAs in squamous cell carcinoma of head and neck (SCCHN) and provide references for SCCHN diagnosis and prognosis. Method Differential expressed miRNAs (DEMs) in SCCHN were screened through gene expression omnibus (GEO) DataSets and verified by the cancer genome atlas (TCGA) database. Next, the overall survival analysis, receiver operating characteristics, and clinical correlation analysis were adopted to filter the miRNAs with diagnostic and prognostic values. Finally, functional enrichment analyses were conducted for inquiring into the mechanisms of miRNAs. Results Total 103 DEMs (p < 0.05, fold change ≥ 2) in SCCHN were screened out from GSE124566. Partly, the expression levels of the selected (12/17) miRNAs were verified by TCGA. Followed, of the 12 miRNAs, two miRNA expression levels were associated with the overall survival, and five miRNAs showed diagnostic values (AUC ≥ 0.85). Besides, miR-223-3p and miR-204-5p expression levels were correlated to certain clinical features. Epithelial–mesenchymal transition (EMT) related biological process and energy metabolism controlling related AMPK signaling pathway might mediate the roles of miR-223-3p and miR-204-5p, respectively. Conclusion With diagnostic and prognostic values, miR-223-3p and miR-204-5p may be involved in the progression of SCCHN through EMT-related biological process and energy balance related AMPK signaling pathway, respectively.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Otorhinolaryngology, The Affiliated Hospital of Hebei University, Baoding, No. 212 Yuhua Road, Hebei Province, 071000, China
| | - Congzhe Tian
- Department of Otorhinolaryngology, The Affiliated Hospital of Hebei University, Baoding, No. 212 Yuhua Road, Hebei Province, 071000, China
| | - Erbin Xiao
- Department of Otorhinolaryngology, The Affiliated Hospital of Hebei University, Baoding, No. 212 Yuhua Road, Hebei Province, 071000, China
| | - Jinduo Du
- Department of Otorhinolaryngology, The Affiliated Hospital of Hebei University, Baoding, No. 212 Yuhua Road, Hebei Province, 071000, China
| | - Jingwei Liang
- Department of Otorhinolaryngology, The Affiliated Hospital of Hebei University, Baoding, No. 212 Yuhua Road, Hebei Province, 071000, China
| | - Xianghong Chen
- Department of Otorhinolaryngology, The Affiliated Hospital of Hebei University, Baoding, No. 212 Yuhua Road, Hebei Province, 071000, China
| | - Weiwei Chi
- Department of Otorhinolaryngology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050031, China
| |
Collapse
|
26
|
Singh P, Singh A, Shah S, Vataliya J, Mittal A, Chitkara D. RNA Interference Nanotherapeutics for Treatment of Glioblastoma Multiforme. Mol Pharm 2020; 17:4040-4066. [PMID: 32902291 DOI: 10.1021/acs.molpharmaceut.0c00709] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nucleic acid therapeutics for RNA interference (RNAi) are gaining attention in the treatment and management of several kinds of the so-called "undruggable" tumors via targeting specific molecular pathways or oncogenes. Synthetic ribonucleic acid (RNAs) oligonucleotides like siRNA, miRNA, shRNA, and lncRNA have shown potential as novel therapeutics. However, the delivery of such oligonucleotides is significantly hampered by their physiochemical (such as hydrophilicity, negative charge, and instability) and biopharmaceutical features (in vivo serum stability, fast renal clearance, interaction with extracellular proteins, and hindrance in cellular internalization) that markedly reduce their biological activity. Recently, several nanocarriers have evolved as suitable non-viral vectors for oligonucleotide delivery, which are known to either complex or conjugate with these oligonucleotides efficiently and also overcome the extracellular and intracellular barriers, thereby allowing access to the tumoral micro-environment for the better and desired outcome in glioblastoma multiforme (GBM). This Review focuses on the up-to-date advancements in the field of RNAi nanotherapeutics utilized for GBM treatment.
Collapse
Affiliation(s)
- Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Aditi Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Shruti Shah
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Jalpa Vataliya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| |
Collapse
|
27
|
Mao H, Nian J, Wang Z, Li X, Huang C. KDELR2 is an unfavorable prognostic biomarker and regulates CCND1 to promote tumor progression in glioma. Pathol Res Pract 2020; 216:152996. [PMID: 32534703 DOI: 10.1016/j.prp.2020.152996] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/07/2020] [Accepted: 04/22/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND The KDEL receptor is a seven-transmembrane-domain protein, which plays a key role in ER quality control and in the ER stress response, KDELR2 involved in regulation of cellular functions, including cell proliferation, survival, promotes glioblastoma tumorigenesis. The aim of this study was to investigate the clinicpathological value and biological role of KDELR2 in glioma. METHODS We studied the expression of KEDLR2 and its association with the prognosis through the TCGA, CGGA, and GSE16011 database. To explore the role of KDELR2 in glioma, KDELR2 siRNA was constructed and transfected into U87 glioma cells. CCK-8, colony formation and Transwell assays were used to investigate the roles of KDELR2 on GBM cell proliferation. We further studied the effect of KDELR2 on tumorigenesis in animal model. Additionally, flow cytometry was used to monitor the changes in the cell cycle and apoptosis following transfection with KDELR2 siRNA. We applied GeneChip primeview expression array to analysis the differential gene expression profiling. Ingenuity Pathway Analysis to show that KDELR2 has a significant impact in canonical pathway in cell cycle regulation and participate in multiple pathways. And we detected the cell cycle proteins CCND1 expression by Western blot analysis. RESULTS Our results showed that KDELR2 was up-regulated in glioma tissue and cell lines. Knockdown KDELR2 was able to reduce cell viability, promote cell cycle arrest at the G1 phase, and induce apoptotic cell death. Moreover, our results suggested that KDELR2 regulated the cellular functions of U87 cells by targeting CCND1. Therefore, we demonstrated that KDELR2 is a novel biomarker in glioma. CONCLUSIONS KDELR2 is highly expressed in human glioma tissues and cell lines, a higher expression of KDELR2 is associated with a poor prognosis of glioma patients. Moreover, KDELR2 regulated the cellular functions of U87 cells by targeting CCND1. The KDELR2/CCND1 axis may provide a new therapeutic target for the treatment of glioma and deepen our understanding of glioma mechanisms.
Collapse
Affiliation(s)
- Hui Mao
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China
| | - Jiang Nian
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhao Wang
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China
| | - XueJun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - ChunHai Huang
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China.
| |
Collapse
|