1
|
Mirzaei S, Ahangari F, Faramarzi F, Khoshnazar SM, Khormizi FZ, Aghagolzadeh M, Rostami M, Asghariazar V, Alimohammadi M, Rahimzadeh P, Farahani N. MicroRNA-146 family: Molecular insights into their role in regulation of signaling pathways in glioma progression. Pathol Res Pract 2024; 264:155707. [PMID: 39536541 DOI: 10.1016/j.prp.2024.155707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Glioma is a highly lethal brain cancer in humans. Despite advancements in treatment, the prognosis for patients remains unfavorable. Epigenetic factors, along with their interactions and non-coding RNAs (ncRNAs), are crucial in glioma cells' development and aggressive characteristics. MicroRNAs (miRNAs) are a class of small non-coding RNAs (ncRNAs) that modulate the expression of various genes by binding to target mRNA molecules. They play a critical role in regulating essential biological mechanisms such as cell proliferation and differentiation, cell cycle, and apoptosis. MiR-146a/miR-146b is a significant and prevalent miRNA whose expression alterations are linked to various pathological changes in cancer cells, as well as the modulation of several cellular signaling pathways, including NF-κB, TGF-β, PI3K/Akt, and Notch-1. Scientists may identify novel targets in clinical settings by studying the complicated link between Mir-146a/mir-146b, drug resistance, molecular pathways, and pharmacological intervention in gliomas. Additionally, its interactions with other ncRNAs, such as circular RNA and long non-coding RNA, contribute to the pathogenesis of glioma. As well as miR-146 holds potential as both a diagnostic and therapeutic biomarker for patients with this condition. In the current review, we investigate the significance of miRNAs in the context of glioma, with a particular focus on the critical role of Mir-146a/mir-146b in glioma tumors. Additionally, we examined the clinical relevance of this miRNA, highlighting its potential implications for diagnosis and treatment.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Fatemeh Ahangari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Faramarzi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mahboobeh Aghagolzadeh
- Department of Biology, Faculty of Basic Sciences, University of Shahid Chamran of Ahvaz, Ahvaz, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Leili FR, Shali N, Sheibani M, Jafarian MJ, Pashizeh F, Gerami R, Iraj F, Lashkarshekan AA. Detailed pathological role of non-coding RNAs (ncRNAs) in regulating drug resistance of glioblastoma, and update. Pathol Res Pract 2024; 263:155590. [PMID: 39326365 DOI: 10.1016/j.prp.2024.155590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Glioma is a kind of brain tumor that develops in the central nervous system and is classified based on its histology and molecular genetic features. The lifespan of patients does not exceed 22 months. One of the motives for the low effectiveness of glioma treatment is its radioresistance and chemoresistance. Noncoding RNAs (ncRNAs) are a diverse set of transcripts that do not undergo translation to become proteins in glioma. The ncRNAs have been identified as significant regulators of several biological processes in different cell types and tissues, and their abnormal function has been linked to glioma. They are known to impact important occurrences, including carcinogenesis, progression, and enhanced treatment resistance in glioma cells. The ncRNAs control cell proliferation, migration, epithelial-to-mesenchymal transition (EMT), invasion, and drug resistance in glioma cells. The main focus of this study is to inspect the involvement of ncRNAs in the drug resistance of glioma.
Collapse
Affiliation(s)
- Foad Rahmanpour Leili
- Department of Neurology Faculty of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Niloofar Shali
- Department of Clinical Biochemistry, School of Medicine, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Mehrnaz Sheibani
- Division of Pediatric Neurology, University of Tabriz, Tabriz, Iran
| | | | - Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science, Yazd 8916188635, Iran
| | - Reza Gerami
- Department of Radiology, Faculty of Medicine, AJA University of Medical Science, Tehran, Iran.
| | | | | |
Collapse
|
3
|
Abed S, Ebrahimi A, Fattahi F, Kouchakali G, Shekari-Khaniani M, Mansoori-Derakhshan S. The Role of Non-Coding RNAs in Mitochondrial Dysfunction of Alzheimer's Disease. J Mol Neurosci 2024; 74:100. [PMID: 39466447 DOI: 10.1007/s12031-024-02262-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/25/2024] [Indexed: 10/30/2024]
Abstract
Although brain amyloid-β (Aβ) peptide buildup is the main cause of Alzheimer's disease (AD), mitochondrial abnormalities can also contribute to the illness's development, as either a primary or secondary factor, as programmed cell death and efficient energy generation depend on the proper operation of mitochondria. As a result, non-coding RNAs (ncRNAs) may play a crucial role in ensuring that nuclear genes related to mitochondria and mitochondrial genes function normally. Interestingly, a significant number of recent studies have focused on the impact of ncRNAs on the expression of nucleus and mitochondrial genes. Additionally, researchers have proposed some intriguing therapeutic approaches to treat and reduce the severity of AD by adjusting the levels of these ncRNAs. The goal of this work was to consolidate the existing knowledge in this field of study by systematically investigating ncRNAs, with a particular emphasis on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and small nucleolar RNAs (snoRNAs). Therefore, the impact and processes by which ncRNAs govern mitochondrial activity in the onset and progression of AD are thoroughly reviewed in this article. Collectively, the effects of ncRNAs on physiological and molecular mechanisms associated with mitochondrial abnormalities that exacerbate AD are thoroughly reviewed in the current research, while also emphasizing the relationship between disturbed mitophagy in AD and ncRNAs.
Collapse
Affiliation(s)
- Samin Abed
- Department of Genetics, Tabriz University of Medical University, Tabriz, Iran
| | - Amir Ebrahimi
- Department of Genetics, Tabriz University of Medical University, Tabriz, Iran
| | - Fatemeh Fattahi
- Department of Genetics, Tabriz University of Medical University, Tabriz, Iran
| | - Ghazal Kouchakali
- Department of Genetics, Tabriz University of Medical University, Tabriz, Iran
| | | | | |
Collapse
|
4
|
Zakutansky PM, Ku L, Zhang G, Shi L, Li Y, Yao B, Bassell GJ, Read RD, Feng Y. Isoform balance of the long noncoding RNA NEAT1 is regulated by the RNA-binding protein QKI, governs the glioma transcriptome, and impacts cell migration. J Biol Chem 2024; 300:107595. [PMID: 39032650 PMCID: PMC11367543 DOI: 10.1016/j.jbc.2024.107595] [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: 04/19/2024] [Revised: 07/02/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024] Open
Abstract
The long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) is involved in a variety of human cancers. Two overlapping NEAT1 isoforms, NEAT1_1 and NEAT1_2, are produced through mutually exclusive alternative 3' end formation. Previous studies extensively investigated NEAT1 dysregulation in tumors, but often failed to achieve distinct quantification of the two NEAT1 isoforms. Moreover, molecular mechanisms governing the biogenesis of NEAT1 isoforms and the functional impacts of their dysregulation in tumorigenesis remain poorly understood. In this study, we employed an isoform-specific quantification assay and found differential dysregulation of NEAT1 isoforms in patient-derived glioblastoma multiforme cells. We further showed usage of the NEAT1 proximal polyadenylation site (PAS) is a critical mechanism that controls glioma NEAT1 isoform production. CRISPR-Cas9-mediated PAS deletion reduced NEAT1_1 and reciprocally increased NEAT1_2, which enhanced nuclear paraspeckle formation in human glioma cells. Moreover, the utilization of the NEAT1 PAS is facilitated by the RNA-binding protein quaking (QKI), which binds to the proximal QKI recognition elements. Functionally, we identified transcriptomic changes and altered biological pathways caused by NEAT1 isoform imbalance in glioma cells, including the pathway for the regulation of cell migration. Finally, we demonstrated the forced increase of NEAT1_2 upon NEAT1 PAS deletion is responsible for driving glioma cell migration and promoting the expression of genes implicated in the regulation of cell migration. Together, our studies uncovered a novel mechanism that regulates NEAT1 isoforms and their functional impacts on the glioma transcriptome, which affects pathological pathways of glioma, represented by migration.
Collapse
Affiliation(s)
- Paul M Zakutansky
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Graduate Program in Biochemistry, Cell, and Developmental Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, Georgia, USA
| | - Li Ku
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Guannan Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Liang Shi
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Renee D Read
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA; Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Yue Feng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
5
|
Goleij P, Pourali G, Raisi A, Ravaei F, Golestan S, Abed A, Razavi ZS, Zarepour F, Taghavi SP, Ahmadi Asouri S, Rafiei M, Mousavi SM, Hamblin MR, Talei S, Sheida A, Mirzaei H. Role of Non-coding RNAs in the Response of Glioblastoma to Temozolomide. Mol Neurobiol 2024:10.1007/s12035-024-04316-z. [PMID: 39023794 DOI: 10.1007/s12035-024-04316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/16/2024] [Indexed: 07/20/2024]
Abstract
Chemotherapy and radiotherapy are widely used in clinical practice across the globe as cancer treatments. Intrinsic or acquired chemoresistance poses a significant problem for medical practitioners and researchers, causing tumor recurrence and metastasis. The most dangerous kind of malignant brain tumor is called glioblastoma multiforme (GBM) that often recurs following surgery. The most often used medication for treating GBM is temozolomide chemotherapy; however, most patients eventually become resistant. Researchers are studying preclinical models that accurately reflect human disease and can be used to speed up drug development to overcome chemoresistance in GBM. Non-coding RNAs (ncRNAs) have been shown to be substantial in regulating tumor development and facilitating treatment resistance in several cancers, such as GBM. In this work, we mentioned the mechanisms of how different ncRNAs (microRNAs, long non-coding RNAs, circular RNAs) can regulate temozolomide chemosensitivity in GBM. We also address the role of these ncRNAs encapsulated inside secreted exosomes.
Collapse
Affiliation(s)
- Pouya Goleij
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahin Golestan
- Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atena Abed
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Sadat Razavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Sahand Talei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hamed Mirzaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Palizkaran Yazdi M, Barjasteh A, Moghbeli M. MicroRNAs as the pivotal regulators of Temozolomide resistance in glioblastoma. Mol Brain 2024; 17:42. [PMID: 38956588 PMCID: PMC11218189 DOI: 10.1186/s13041-024-01113-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive nervous system tumor with a poor prognosis. Although, surgery, radiation therapy, and chemotherapy are the current standard protocol for GBM patients, there is still a poor prognosis in these patients. Temozolomide (TMZ) as a first-line therapeutic agent in GBM can easily cross from the blood-brain barrier to inhibit tumor cell proliferation. However, there is a high rate of TMZ resistance in GBM patients. Since, there are limited therapeutic choices for GBM patients who develop TMZ resistance; it is required to clarify the molecular mechanisms of chemo resistance to introduce the novel therapeutic targets. MicroRNAs (miRNAs) regulate chemo resistance through regulation of drug metabolism, absorption, DNA repair, apoptosis, and cell cycle. In the present review we discussed the role of miRNAs in TMZ response of GBM cells. It has been reported that miRNAs mainly induced TMZ sensitivity by regulation of signaling pathways and autophagy in GBM cells. Therefore, miRNAs can be used as the reliable diagnostic/prognostic markers in GBM patients. They can also be used as the therapeutic targets to improve the TMZ response in GBM cells.
Collapse
Affiliation(s)
- Mahsa Palizkaran Yazdi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Barjasteh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Hazra R, Debnath R, Tuppad A. Glioblastoma stem cell long non-coding RNAs: therapeutic perspectives and opportunities. Front Genet 2024; 15:1416772. [PMID: 39015773 PMCID: PMC11249581 DOI: 10.3389/fgene.2024.1416772] [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: 04/13/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
Glioblastoma poses a formidable challenge among primary brain tumors: its tumorigenic stem cells, capable of self-renewal, proliferation, and differentiation, contribute substantially to tumor initiation and therapy resistance. These glioblastoma stem cells (GSCs), resembling conventional stem and progenitor cells, adopt pathways critical for tissue development and repair, promoting uninterrupted tumor expansion. Long non-coding RNAs (lncRNAs), a substantial component of the human transcriptome, have garnered considerable interest for their pivotal roles in normal physiological processes and cancer pathogenesis. They display cell- or tissue-specific expression patterns, and extensive investigations have highlighted their impact on regulating GSC properties and cellular differentiation, thus offering promising avenues for therapeutic interventions. Consequently, lncRNAs, with their ability to exert regulatory control over tumor initiation and progression, have emerged as promising targets for innovative glioblastoma therapies. This review explores notable examples of GSC-associated lncRNAs and elucidates their functional roles in driving glioblastoma progression. Additionally, we delved deeper into utilizing a 3D in vitro model for investigating GSC biology and elucidated four primary methodologies for targeting lncRNAs as potential therapeutics in managing glioblastoma.
Collapse
Affiliation(s)
- Rasmani Hazra
- University of New Haven, Biology and Environmental Science Department, West Haven, CT, United States
| | - Rinku Debnath
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Arati Tuppad
- University of New Haven, Biology and Environmental Science Department, West Haven, CT, United States
| |
Collapse
|
8
|
Kucinska M, Pospieszna J, Tang J, Lisiak N, Toton E, Rubis B, Murias M. The combination therapy using tyrosine kinase receptors inhibitors and repurposed drugs to target patient-derived glioblastoma stem cells. Biomed Pharmacother 2024; 176:116892. [PMID: 38876048 DOI: 10.1016/j.biopha.2024.116892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
The lesson from many studies investigating the efficacy of targeted therapy in glioblastoma (GBM) showed that a future perspective should be focused on combining multiple target treatments. Our research aimed to assess the efficacy of drug combinations against glioblastoma stem cells (GSCs). Patient-derived cells U3042, U3009, and U3039 were obtained from the Human Glioblastoma Cell Culture resource. Additionally, the study was conducted on a GBM commercial U251 cell line. Gene expression analysis related to receptor tyrosine kinases (RTKs), stem cell markers and genes associated with significant molecular targets was performed, and selected proteins encoded by these genes were assessed using the immunofluorescence and flow cytometry methods. The cytotoxicity studies were preceded by analyzing the expression of specific proteins that serve as targets for selected drugs. The cytotoxicity study using the MTS assay was conducted to evaluate the effects of selected drugs/candidates in monotherapy and combinations. The most cytotoxic compounds for U3042 cells were Disulfiram combined with Copper gluconate (DSF/Cu), Dacomitinib, and Foretinib with IC50 values of 52.37 nM, 4.38 µM, and 4.54 µM after 24 h incubation, respectively. Interactions were assessed using SynergyFinder Plus software. The analysis enabled the identification of the most effective drug combinations against patient-derived GSCs. Our findings indicate that the most promising drug combinations are Dacomitinib and Foretinib, Dacomitinib and DSF/Cu, and Foretinib and AZD3759. Since most tested combinations have not been previously examined against glioblastoma stem-like cells, these results can shed new light on designing the therapeutic approach to target the GSC population.
Collapse
Affiliation(s)
- Malgorzata Kucinska
- Department of Toxicology, Poznan University of Medical Sciences 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Julia Pospieszna
- Department of Toxicology, Poznan University of Medical Sciences 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland.
| | - Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Ewa Toton
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Blazej Rubis
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences 3 Rokietnicka Street, Poznan 60-806, Poland.
| |
Collapse
|
9
|
Pokorná M, Černá M, Boussios S, Ovsepian SV, O’Leary VB. lncRNA Biomarkers of Glioblastoma Multiforme. Biomedicines 2024; 12:932. [PMID: 38790894 PMCID: PMC11117901 DOI: 10.3390/biomedicines12050932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNA molecules of 200 nucleotides or more in length that are not translated into proteins. Their expression is tissue-specific, with the vast majority involved in the regulation of cellular processes and functions. Many human diseases, including cancer, have been shown to be associated with deregulated lncRNAs, rendering them potential therapeutic targets and biomarkers for differential diagnosis. The expression of lncRNAs in the nervous system varies in different cell types, implicated in mechanisms of neurons and glia, with effects on the development and functioning of the brain. Reports have also shown a link between changes in lncRNA molecules and the etiopathogenesis of brain neoplasia, including glioblastoma multiforme (GBM). GBM is an aggressive variant of brain cancer with an unfavourable prognosis and a median survival of 14-16 months. It is considered a brain-specific disease with the highly invasive malignant cells spreading throughout the neural tissue, impeding the complete resection, and leading to post-surgery recurrences, which are the prime cause of mortality. The early diagnosis of GBM could improve the treatment and extend survival, with the lncRNA profiling of biological fluids promising the detection of neoplastic changes at their initial stages and more effective therapeutic interventions. This review presents a systematic overview of GBM-associated deregulation of lncRNAs with a focus on lncRNA fingerprints in patients' blood.
Collapse
Affiliation(s)
- Markéta Pokorná
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, Vinohrady, 10000 Prague, Czech Republic; (M.Č.); (V.B.O.)
| | - Marie Černá
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, Vinohrady, 10000 Prague, Czech Republic; (M.Č.); (V.B.O.)
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK;
- Faculty of Medicine, Health, and Social Care, Canterbury Christ Church University, Canterbury CT2 7PB, UK
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
| | - Saak V. Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent ME4 4TB, UK;
- Faculty of Medicine, Tbilisi State University, Tbilisi 0177, Georgia
| | - Valerie Bríd O’Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, Vinohrady, 10000 Prague, Czech Republic; (M.Č.); (V.B.O.)
| |
Collapse
|
10
|
E Y, Zhang X, Ma H, Dong F. Long Non-coding RNA Prader Willi/Angelman Region RNA 6 Suppresses Glioma Development by Modulating MicroRNA-106a-5p. Biochem Genet 2024; 62:1365-1378. [PMID: 37610693 DOI: 10.1007/s10528-023-10479-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023]
Abstract
As one of the most frequent intracranial tumors, glioma showed invasive development and poor prognosis. lncRNAs have been illustrated to serve as biomarkers in various cancers. Whether the long non-coding RNA Prader Willi/Angelman region RNA 6 (PWAR6) was involved in glioma development and the underlying mechanism was investigated. PWAR6 in glioma was evaluated by polymerase chain reaction and its clinical significance was assessed with a series of statistical analyses. The biological function of PWAR6 was investigated with the cell counting kit 8 and Transwell assay. The potential underlying mechanism was studied with the luciferase reporter assay. The significant downregulation of PWAR6 was observed in glioma, which showed a close relationship with the major clinicopathological features and poor prognosis of patients. PWAR6 restrained cell growth, migration and invasion of glioma, which was alleviated by the overexpression of microRNA-106a-5p (miR-106a-5p). PWAR6 functioned as a prognostic biomarker and tumor suppressor of glioma through regulating miR-106a-5p.
Collapse
Affiliation(s)
- Yongjun E
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, 121000, Liaoning, China
| | - Xianglin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, 121000, Liaoning, China.
| | - Heji Ma
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, 121000, Liaoning, China
| | - Furen Dong
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, 121000, Liaoning, China
| |
Collapse
|
11
|
Singh RR, Mondal I, Janjua T, Popat A, Kulshreshtha R. Engineered smart materials for RNA based molecular therapy to treat Glioblastoma. Bioact Mater 2024; 33:396-423. [PMID: 38059120 PMCID: PMC10696434 DOI: 10.1016/j.bioactmat.2023.11.007] [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: 04/25/2023] [Revised: 10/19/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive malignancy of the central nervous system (CNS) that remains incurable despite the multitude of improvements in cancer therapeutics. The conventional chemo and radiotherapy post-surgery have only been able to improve the prognosis slightly; however, the development of resistance and/or tumor recurrence is almost inevitable. There is a pressing need for adjuvant molecular therapies that can successfully and efficiently block tumor progression. During the last few decades, non-coding RNAs (ncRNAs) have emerged as key players in regulating various hallmarks of cancer including that of GBM. The levels of many ncRNAs are dysregulated in cancer, and ectopic modulation of their levels by delivering antagonists or overexpression constructs could serve as an attractive option for cancer therapy. The therapeutic potential of several types of ncRNAs, including miRNAs, lncRNAs, and circRNAs, has been validated in both in vitro and in vivo models of GBM. However, the delivery of these RNA-based therapeutics is highly challenging, especially to the tumors of the brain as the blood-brain barrier (BBB) poses as a major obstacle, among others. Also, since RNA is extremely fragile in nature, careful considerations must be met while designing a delivery agent. In this review we have shed light on how ncRNA therapy can overcome the limitations of its predecessor conventional therapy with an emphasis on smart nanomaterials that can aide in the safe and targeted delivery of nucleic acids to treat GBM. Additionally, critical gaps that currently exist for successful transition from viral to non-viral vector delivery systems have been identified. Finally, we have provided a perspective on the future directions, potential pathways, and target areas for achieving rapid clinical translation of, RNA-based macromolecular therapy to advance the effective treatment of GBM and other related diseases.
Collapse
Affiliation(s)
- Ravi Raj Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- University of Queensland –IIT Delhi Academy of Research (UQIDAR)
| | - Indranil Mondal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Taskeen Janjua
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
12
|
Wang D, Wang Z, Dai X, Zhang L, Li M. Apigenin and Temozolomide Synergistically Inhibit Glioma Growth Through the PI3K/ AKT Pathway. Cancer Biother Radiopharm 2024; 39:125-132. [PMID: 33471569 DOI: 10.1089/cbr.2020.4283] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Glioma is a devastating disease with the worst prognosis among human malignant tumors. Although temozolomide (TMZ) improves the overall survival of glioma patients, there are still many glioma patients who are resistant to TMZ. In this study, we focused on the effect of apigenin (API) and TMZ on glioma cells in vitro and in vivo, and we studied the underlying molecular mechanisms. Materials and Methods: To investigate the effect of API on glioblastoma cell proliferation, cell viability was assessed after glioma cells were incubated with various concentrations of API with or without TMZ using MTT assays. Then, we explored the synergistic effect of API and TMZ on glioma cell cycle, apoptosis, and migration. To investigate the molecular mechanism behind the synergism of API and TMZ, we examined the related genes of the major signaling pathways involved in glioma pathogenesis by Western blotting. Results: In this study, we found that API significantly suppressed the proliferation of glioma cells in a dose- and time-dependent manner. Combining API and TMZ significantly induced glioma cells arrest at the G2 phase and inhibited glioma cells proliferation compared with API or TMZ alone. In addition, API promoted the ability of TMZ to induce glioma cells apoptosis and inhibit glioma cells invasion. Furthermore, compared with treatment with individual agents, the combination of API and TMZ significantly inhibited the growth of subcutaneous tumors in mice. These results implied that API could synergistically suppress the growth of glioma cells when combined with TMZ. Combining API and TMZ significantly inhibited the protein expression of p-AKT, cyclin D1, Bcl-2, Matrix Metallopeptidase 2, and Matrix Metallopeptidase 9. Conclusion: API and TMZ synergistically inhibited glioma growth through the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Dong Wang
- Department of Neurosurgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhijun Wang
- Clinical Medicine, Weifang Medical University, Weifang, People's Republic of China
| | - Xuedong Dai
- Department of Neurosurgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, People's Republic of China
| | - Liang Zhang
- Department of Neurosurgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, People's Republic of China
| | - Min Li
- Department of Neurosurgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
13
|
Qiao W, Wang Y, Luo C, Wu J, Qin G, Zhang J, Yao Y. Development of preoperative and postoperative models to predict recurrence in postoperative glioma patients: a longitudinal cohort study. BMC Cancer 2024; 24:274. [PMID: 38418976 PMCID: PMC10900633 DOI: 10.1186/s12885-024-11996-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Glioma recurrence, subsequent to maximal safe resection, remains a pivotal challenge. This study aimed to identify key clinical predictors influencing recurrence and develop predictive models to enhance neurological diagnostics and therapeutic strategies. METHODS This longitudinal cohort study with a substantial sample size (n = 2825) included patients with non-recurrent glioma who were pathologically diagnosed and had undergone initial surgical resection between 2010 and 2018. Logistic regression models and stratified Cox proportional hazards models were established with the top 15 clinical variables significantly influencing outcomes screened by the least absolute shrinkage and selection operator (LASSO) method. Preoperative and postoperative models predicting short-term (within 6 months) postoperative recurrence in glioma patients were developed to explore the risk factors associated with short- and long-term recurrence in glioma patients. RESULTS Preoperative and postoperative logistic models predicting short-term recurrence had accuracies of 0.78 and 0.87, respectively. A range of biological and early symptomatic characteristics linked to short- and long-term recurrence have been pinpointed. Age, headache, muscle weakness, tumor location and Karnofsky score represented significant odd ratios (t > 2.65, p < 0.01) in the preoperative model, while age, WHO grade 4 and chemotherapy or radiotherapy treatments (t > 4.12, p < 0.0001) were most significant in the postoperative period. Postoperative predictive models specifically targeting the glioblastoma and IDH wildtype subgroups were also performed, with an AUC of 0.76 and 0.80, respectively. The 50 combinations of distinct risk factors accommodate diverse recurrence risks among glioma patients, and the nomograms visualizes the results for clinical practice. A stratified Cox model identified many prognostic factors for long-term recurrence, thereby facilitating the enhanced formulation of perioperative care plans for patients, and glioblastoma patients displayed a median progression-free survival (PFS) of only 11 months. CONCLUSION The constructed preoperative and postoperative models reliably predicted short-term postoperative glioma recurrence in a substantial patient cohort. The combinations risk factors and nomograms enhance the operability of personalized therapeutic strategies and care regimens. Particular emphasis should be placed on patients with recurrence within six months post-surgery, and the corresponding treatment strategies require comprehensive clinical investigation.
Collapse
Affiliation(s)
- Wanyu Qiao
- Department of Biostatistics, School of Public Health & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Wang
- Department of Tumor Screening and Prevention, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Luo
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Guoyou Qin
- Department of Biostatistics, School of Public Health & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Jie Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- Neurosurgical Institute, Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
| | - Ye Yao
- Department of Biostatistics, School of Public Health & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Lu X, Zhang D. Expression of lncRNAs in glioma: A lighthouse for patients with glioma. Heliyon 2024; 10:e24799. [PMID: 38322836 PMCID: PMC10844031 DOI: 10.1016/j.heliyon.2024.e24799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Glioma is the most common malignant tumour in the central nervous system, accounting for approximately 30 % of the primary tumours of this system. The World Health Organization grades for glioma include: Grade I (pilocytic astrocytoma), Grade II (astrocytoma, oligodastoma, etc.), Grade III (anaplastic astrocytoma, anaplastic oligodastoma, etc.) and Grade IV (glioblastoma). With grade increases, the proliferation, invasion and other malignant biological properties of the glioma are enhanced, and the treatment results are less satisfactory. The overall survival of patients with glioblastoma is less than 15 months. Recent research has focused on the roles of long non-coding RNAs, previously regarded as "transcriptional noise", in diseases, leading to a new understanding of these roles. Therefore, we conducted this review to explore the progress of research regarding the expression and mechanism of long non-coding RNAs in glioma.
Collapse
Affiliation(s)
- Xiaolin Lu
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dongzhi Zhang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
15
|
Davidson CL, Vengoji R, Jain M, Batra SK, Shonka N. Biological, diagnostic and therapeutic implications of exosomes in glioma. Cancer Lett 2024; 582:216592. [PMID: 38092145 PMCID: PMC10832613 DOI: 10.1016/j.canlet.2023.216592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/11/2023] [Accepted: 12/05/2023] [Indexed: 01/04/2024]
Abstract
Despite therapeutic advances, overall survival in glioblastoma is dismal. To optimize progress, a more detailed understanding of glioma's molecular, cellular, and intercellular pathophysiology is needed. Recent investigation has revealed a vital role for exosomes in inter-cellular signaling, tumor cell support, and regulation of the tumor microenvironment. Exosomes carry miRNAs, lncRNAs, mRNAs, proteins, immune regulatory molecules, nucleic acids, and lipids; however, the composition of exosome cargo is variable depending on the cell of origin. Specific exosomal miRNA contents such as miR-21, miR-301a, miR-151a, miR-148a, and miR-5096 are altered in high-grade glioma. Unique proteomic, genomic, and miRNA signatures of tumor exosomes have been associated with disease pathobiology, temozolomide resistance, immunosuppression, and tumor proliferation. Exosomes hold promise for tissue diagnostic glioma diagnosis and monitoring response to therapy. This review summarizes the current understanding of exosomes, their crucial role in glioma pathology, and future directions for their use in diagnosis and treatment. METHODS: The MEDLINE/PubMed database was reviewed for papers written in English and publication dates of 1981-2023, using the search string "Exosome", "Extracellular vesicles", "Glioma", "Exosomes in glioma".
Collapse
Affiliation(s)
- Caroline L Davidson
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
| |
Collapse
|
16
|
Fattahi M, Alamdari-Palangi V, Rahimi Jaberi K, Ehtiati S, Ojaghi S, Rahimi-Jaberi A, Samavarchi Tehrani S, Dang P, Movahedpour A, Hossein Khatami S. Exosomal long non-coding RNAs in glioblastoma. Clin Chim Acta 2024; 553:117705. [PMID: 38086498 DOI: 10.1016/j.cca.2023.117705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent primary tumor found in the central nervous system, accounting for 70% of all adult brain tumors. The median overall survival rate is one year post-diagnosis with treatment, and only four months without treatment. Current GBM diagnostic methods, such as magnetic resonance imaging (MRI), surgery, and brain biopsies, have limitations. These include difficulty distinguishing between tumor recurrence and post-surgical necrotic regions, and operative risks associated with obtaining histological samples through direct surgery or biopsies. Consequently, there is a need for rapid, inexpensive, and minimally invasive techniques for early diagnosis and improved subsequent treatment. Research has shown that tumor-derived exosomes containing various long non-coding RNAs (lncRNAs) play critical regulatory roles in immunomodulation, cancer metastasis, cancer development, and drug resistance in GBM. They regulate genes that enhance cancer growth and progression and alter the expression of several key signaling pathways. Due to the specificity and sensitivity of exosomal lncRNAs, they have the potential to be used as biomarkers for early diagnosis and prognosis, as well as to monitor a patient's response to chemotherapy for GBM. In this review, we discuss the role of exosomal lncRNAs in the pathogenesis of GBM and their potential clinical applications for early diagnosis.
Collapse
Affiliation(s)
- Mehdi Fattahi
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam; School of Engineering & Technology, Duy Tan University, Da Nang, Viet Nam
| | - Vahab Alamdari-Palangi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Khojaste Rahimi Jaberi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajad Ehtiati
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Ojaghi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Rahimi-Jaberi
- Department of Neurology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sadra Samavarchi Tehrani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran
| | - Phuyen Dang
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam; School of Engineering & Technology, Duy Tan University, Da Nang, Viet Nam
| | | | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Gareev I, Encarnacion Ramirez MDJ, Nurmukhametov R, Ivliev D, Shumadalova A, Ilyasova T, Beilerli A, Wang C. The role and clinical relevance of long non-coding RNAs in glioma. Noncoding RNA Res 2023; 8:562-570. [PMID: 37602320 PMCID: PMC10432901 DOI: 10.1016/j.ncrna.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 08/22/2023] Open
Abstract
Glioma represents a complex and heterogeneous disease, posing significant challenges to both clinicians and researchers. Despite notable advancements in glioma treatment, the overall survival rate for most glioma patients remains dishearteningly low. Hence, there is an urgent necessity to discover novel biomarkers and therapeutic targets specifically tailored for glioma. In recent years, long non-coding RNAs (lncRNAs) have emerged as pivotal regulators of gene expression and have garnered attention for their involvement in the development and progression of various cancers, including glioma. The dysregulation of lncRNAs plays a critical role in glioma pathogenesis and influences clinical outcomes. Consequently, there is growing interest in exploring the potential of lncRNAs as diagnostic and prognostic biomarkers, as well as therapeutic targets. By understanding the functions and dysregulation of lncRNAs in glioma, researchers aim to unlock new avenues for the development of innovative treatment strategies catered to glioma patients. The identification and thorough characterization of lncRNAs hold the promise of novel therapeutic approaches that could potentially improve patient outcomes and enhance the management of glioma, ultimately striving for better prospects and enhanced quality of life for those affected by this challenging disease. The primary objective of this paper is to comprehensively review the current state of knowledge regarding lncRNA biology and their intricate roles in glioma. It also delves into the potential of lncRNAs as valuable diagnostic and prognostic indicators and explores their feasibility as promising targets for therapeutic interventions.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Manuel de Jesus Encarnacion Ramirez
- Department of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Renat Nurmukhametov
- Division of Spine Surgery, Central Clinical Hospital of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Denis Ivliev
- Department of Neurosurgery, Smolensk State Medical University of the Ministry of Health of the Russian Federation, Smolensk, Russia
| | - Alina Shumadalova
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Tatiana Ilyasova
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, Tyumen, Russia
| | - Chunlei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
18
|
Hussain MS, Gupta G, Afzal M, Alqahtani SM, Samuel VP, Hassan Almalki W, Kazmi I, Alzarea SI, Saleem S, Dureja H, Singh SK, Dua K, Thangavelu L. Exploring the role of lncrna neat1 knockdown in regulating apoptosis across multiple cancer types: A review. Pathol Res Pract 2023; 252:154908. [PMID: 37950931 DOI: 10.1016/j.prp.2023.154908] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as pivotal regulators of gene expression, contributing significantly to a diverse range of cellular processes, including apoptosis. One such lncRNA is NEAT1, which is elevated in several types of cancer and aid in cancer growth. However, recent studies have also demonstrated that the knockdown of NEAT1 can inhibit cancer cells proliferation, movement, and infiltration while enhancing apoptosis. This article explores the function of lncRNA NEAT1 knockdown in regulating apoptosis across multiple cancer types. We explore the existing understanding of NEAT1's involvement in the progression of malignant conditions, including its structure and functions. Additionally, we investigate the molecular mechanisms by which NEAT1 modulates the cell cycle, cellular proliferation, apoptosis, movement, and infiltration in diverse cancer types, including acute myeloid leukemia, breast cancer, cervical cancer, colorectal cancer, esophageal squamous cell carcinoma, glioma, non-small cell lung cancer, ovarian cancer, prostate cancer, and retinoblastoma. Furthermore, we review the recent studies investigating the therapeutic potential of NEAT1 knockdown in cancer treatment. Targeting the lncRNA NEAT1 presents a promising therapeutic approach for treating cancer. It has shown the ability to suppress cancer cell proliferation, migration, and invasion while promoting apoptosis in various cancer types.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017 Jaipur, Rajasthan, India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Safar M Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK Medical & Health Sciences University, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah, United Arab Emirates
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Kamal Dua
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
19
|
Liao Y, Qiu X, Liu J, Zhang Z, Liu B, Jin C. The role of m6A-modified CircEPHB4 in glioma pathogenesis: Insights into cancer stemness metastasis. Ann Clin Transl Neurol 2023; 10:1749-1767. [PMID: 37614011 PMCID: PMC10578901 DOI: 10.1002/acn3.51864] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/27/2023] [Accepted: 07/17/2023] [Indexed: 08/25/2023] Open
Abstract
OBJECTIVE While existing literature has provided insights into involvement of circEPHB4, SOX2 in glioma, their precise molecular mechanisms and synergistic implications in glioma pathogenesis still dim. This study aims to investigate significance and underlying mechanism of m6A-modified circEPHB4 in regulating SOX2/PHLDB2 axis in gliomas. METHODS The mRNA and protein expression were tested by qRT-PCR and Western blot, respectively. ChIP assay was performed to detect SOX2 enrichment on the PHLDB2 promoter. Cell sphere-forming assay to detect self-renewal ability, flow cytometry to determine positivity of CD133 expressions, Malignant behavior of glioma cells were detected by CCK-8, plate colony formation, scratch, and transwell assays. Glioma xenograft models were constructed to investigate effects of CircEPHB4 in tumor development in vivo. RESULTS Methyltransferase MELLT3 upregulated m6A modification of CircEPHB4, and YTHDC1 promoted cytoplasmic localization of m6A-modified CircEPHB4. Overexpression of wild-type CircEPHB4 enhanced glioma cells' stemness, metastasis, and proliferation. Cytoplasmic CircEPHB4 increased SOX2 mRNA stability by binding to IGF2BP2, and the effects observed by SOX2 knockdown were reversed by CircEPHB4 in glioma cells. SOX2 promoted transcriptional expression of PHLDB2 by enriching the PHLDB2 promoter region. SOX2 reversed the inhibition of PHLDB2 knockdown on stemness of glioma, cell proliferation, and metastasis. In vivo experiments also revealed that CircEPHB4 upregulated PHLDB2 expression by stabilizing SOX2 mRNA, which promoted in vivo tumor growth and accelerated stemness of glioma cells and metastasis. CONCLUSION This study reveals functional interaction and molecular mechanisms of m6A-modified circEPHB4 in regulating SOX2/PHLDB2 axis, highlighting their importance in glioma pathogenesis and potential as therapeutic targets.
Collapse
Affiliation(s)
- Yuxiang Liao
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangsha410008HunanP.R. China
| | - Xiaohui Qiu
- Department of Plastic Surgery, The Third Xiangya HospitalCentral South UniversityChangsha410013HunanP.R. China
| | - Jingping Liu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangsha410008HunanP.R. China
| | - Zhiping Zhang
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangsha410008HunanP.R. China
| | - Bo Liu
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangsha410008HunanP.R. China
| | - Chen Jin
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangsha410008HunanP.R. China
| |
Collapse
|
20
|
Mekala JR, Adusumilli K, Chamarthy S, Angirekula HSR. Novel sights on therapeutic, prognostic, and diagnostics aspects of non-coding RNAs in glioblastoma multiforme. Metab Brain Dis 2023; 38:1801-1829. [PMID: 37249862 PMCID: PMC10227410 DOI: 10.1007/s11011-023-01234-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 05/31/2023]
Abstract
Glioblastoma Multiforme (GBM) is the primary brain tumor and accounts for 200,000 deaths each year worldwide. The standard therapy includes surgical resection followed by temozolomide (TMZ)-based chemotherapy and radiotherapy. The survival period of GBM patients is only 12-15 months. Therefore, novel treatment modalities for GBM treatment are urgently needed. Mounting evidence reveals that non-coding RNAs (ncRNAs) were involved in regulating gene expression, the pathophysiology of GBM, and enhancing therapeutic outcomes. The combinatory use of ncRNAs, chemotherapeutic drugs, and tumor suppressor gene expression induction might provide an innovative, alternative therapeutic approach for managing GBM. Studies have highlighted the role of Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) in prognosis and diagnosis. Dysregulation of ncRNAs is observed in virtually all tumor types, including GBMs. Studies have also indicated the blood-brain barrier (BBB) as a crucial factor that hinders chemotherapy. Although several nanoparticle-mediated drug deliveries were degrading effectively against GBM in vitro conditions. However, the potential to cross the BBB and optimum delivery of oligonucleotide RNA into GBM cells in the brain is currently under intense clinical trials. Despite several advances in molecular pathogenesis, GBM remains resistant to chemo and radiotherapy. Targeted therapies have less clinical benefit due to high genetic heterogeneity and activation of alternative pathways. Thus, identifying GBM-specific prognostic pathways, essential genes, and genomic aberrations provide several potential benefits as subtypes of GBM. Also, these approaches will provide insights into new strategies to overcome the heterogenous nature of GBM, which will eventually lead to successful therapeutic interventions toward precision medicine and precision oncology.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India.
| | - Kowsalya Adusumilli
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India
| | - Sahiti Chamarthy
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India
| | - Hari Sai Ram Angirekula
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India
| |
Collapse
|
21
|
Li R, Chen H, Li C, Qi Y, Zhao K, Wang J, You C, Huang H. The prognostic value and immune landscaps of m6A/m5C-related lncRNAs signature in the low grade glioma. BMC Bioinformatics 2023; 24:274. [PMID: 37403043 DOI: 10.1186/s12859-023-05386-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/14/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) and 5-methylcytosine (m5C) are the main RNA methylation modifications involved in the oncogenesis of cancer. However, it remains obscure whether m6A/m5C-related long non-coding RNAs (lncRNAs) affect the development and progression of low grade gliomas (LGG). METHODS We summarized 926 LGG tumor samples with RNA-seq data and clinical information from The Cancer Genome Atlas and Chinese Glioma Genome Atlas. 105 normal brain samples with RNA-seq data from the Genotype Tissue Expression project were collected for control. We obtained a molecular classification cluster from the expression pattern of sreened lncRNAs. The least absolute shrinkage and selection operator Cox regression was employed to construct a m6A/m5C-related lncRNAs prognostic signature of LGG. In vitro experiments were employed to validate the biological functions of lncRNAs in our risk model. RESULTS The expression pattern of 14 sreened highly correlated lncRNAs could cluster samples into two groups, in which various clinicopathological features and the tumor immune microenvironment were significantly distinct. The survival time of cluster 1 was significantly reduced compared with cluster 2. This prognostic signature is based on 8 m6A/m5C-related lncRNAs (GDNF-AS1, HOXA-AS3, LINC00346, LINC00664, LINC00665, MIR155HG, NEAT1, RHPN1-AS1). Patients in the high-risk group harbored shorter survival times. Immunity microenvironment analysis showed B cells, CD4 + T cells, macrophages, and myeloid-derived DC cells were significantly increased in the high-risk group. Patients in high-risk group had the worse overall survival time regardless of followed TMZ therapy or radiotherapy. All observed results from the TCGA-LGG cohort could be validated in CGGA cohort. Afterwards, LINC00664 was found to promote cell viability, invasion and migration ability of glioma cells in vitro. CONCLUSION Our study elucidated a prognostic prediction model of LGG by 8 m6A/m5C methylated lncRNAs and a critical lncRNA regulation function involved in LGG progression. High-risk patients have shorter survival times and a pro-tumor immune microenvironment.
Collapse
Affiliation(s)
- Ran Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haiyan Chen
- Department of Ophthalmology, General Hospital of Central Theatre Command of People's Liberation Arm, Wuhan, 430070, China
| | - Chaoxi Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yiwei Qi
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kai Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Junwen Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Haohao Huang
- Department of Neurosurgery, General Hospital of Central Theatre Command of People's Liberation Arm, Wuhan, 430070, China.
- General Hospital Of Central Theater Command and Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, China.
| |
Collapse
|
22
|
Villa GR, Chiocca EA. The Role of Long Noncoding Ribonucleic Acids in Glioblastoma: What the Neurosurgeon Should Know. Neurosurgery 2023; 92:1104-1111. [PMID: 36880757 DOI: 10.1227/neu.0000000000002449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/12/2023] [Indexed: 03/08/2023] Open
Abstract
A significant proportion of the human transcriptome, long noncoding RNAs (lncRNAs) play pivotal roles in several aspects of glioblastoma (GBM) pathophysiology including proliferation, invasion, radiation and temozolomide resistance, and immune modulation. The majority of lncRNAs exhibit tissue- and tumor-specific expression, lending them to be attractive targets for therapeutic translation. In recent years, unprecedented progress has been made toward our understanding of lncRNA in GBM. In this review, we discuss the function of lncRNAs, including specific lncRNAs that have critical roles in key aspects of GBM pathophysiology, and potential clinical relevance of lncRNAs for patients with GBM.
Collapse
Affiliation(s)
- Genaro Rodriguez Villa
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston , Massachusetts , USA
| | | |
Collapse
|
23
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
24
|
Li Z, Yang Z, Liu W, Zhu W, Yin L, Han Z, Xian Y, Wen J, Tang H, Lin X, Yang Y, Wang J, Zhang K. Disheveled3 enhanced EMT and cancer stem-like cells properties via Wnt/β-catenin/c-Myc/SOX2 pathway in colorectal cancer. J Transl Med 2023; 21:302. [PMID: 37147666 PMCID: PMC10161491 DOI: 10.1186/s12967-023-04120-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/09/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Epithelial-to-mesenchymal transition (EMT) and cancer stem-like cells (CSLCs) play crucial role in tumor metastasis and drug-resistance. Disheveled3 (DVL3) is involved in malignant behaviors of cancer. However, the role and potential mechanism of DVL3 remain elusive in EMT and CSLCs of colorectal cancer (CRC). METHODS UALCAN and PrognoScan databases were employed to evaluate DVL3 expression in CRC tissues and its correlation with CRC prognosis, respectively. Transwell, sphere formation and CCK8 assay were used to assess metastasis, stemness and drug sensitivity of CRC cells, respectively. Western blotting and dual luciferase assay were performed to analyze the protein expression and Wnt/β-catenin activation, respectively. Lentiviral transfection was used to construct the stable cell lines. Animal studies were performed to analyze the effect of silencing DVL3 on tumorigenicity and metastasis of CRC cells in vivo. RESULTS DVL3 was overexpressed in CRC tissues and several CRC cell lines. DVL3 expression was also higher in CRC tissues with lymph node metastasis than tumor tissues without metastasis, and correlated with poor prognosis of CRC patients. DVL3 positively regulated the abilities of migration, invasion and EMT-like molecular changes in CRC cells. Moreover, DVL3 promoted CSLCs properties and multidrug resistance. We further identified that Wnt/β-catenin was crucial for DVL3-mediated EMT, stemness and SOX2 expression, while silencing SOX2 inhibited DVL3-mediated EMT and stemness. Furthermore, c-Myc, a direct target gene of Wnt/β-catenin, was required for SOX2 expression and strengthened EMT and stemness via SOX2 in CRC cells. Finally, knockdown of DVL3 suppressed tumorigenicity and lung metastasis of CRC cells in nude mice. CONCLUSION DVL3 promoted EMT and CSLCs properties of CRC via Wnt/β-catenin/c-Myc/SOX2 axis, providing a new strategy for successful CRC treatment.
Collapse
Affiliation(s)
- Zhengguang Li
- Oncology Department of Chengdu Seventh People's Hospital, Chengdu, China.
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China.
| | - Zhirong Yang
- Pathology Department of Deyang People's Hospital, Deyang, 618000, China
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Wei Liu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 61051, China
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Wanglong Zhu
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Lan Yin
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Zhenyu Han
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Yu Xian
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Jie Wen
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Hualong Tang
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Xinyue Lin
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Yuhan Yang
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China
| | - Jingyi Wang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 61051, China.
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China.
| | - Kun Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 61051, China.
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China.
| |
Collapse
|
25
|
Ding LN, Yu YY, Ma CJ, Lei CJ, Zhang HB. SOX2-associated signaling pathways regulate biological phenotypes of cancers. Biomed Pharmacother 2023; 160:114336. [PMID: 36738502 DOI: 10.1016/j.biopha.2023.114336] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
SOX2 is a transcription factor involved in multiple stages of embryonic development. In related reports, SOX2 was found to be abnormally expressed in tumor tissues and correlated with clinical features such as TNM staging, tumor grade, and prognosis in patients with various cancer types. In most cancer types, SOX2 is a tumor-promoting factor that regulates tumor progression and metastasis primarily by maintaining the stemness of cancer cells. In addition, SOX2 also regulates the proliferation, apoptosis, invasion, migration, ferroptosis and drug resistance of cancer cells. However, SOX2 acts as a tumor suppressor in some cases in certain cancer types, such as gastric and lung cancer. These key regulatory functions of SOX2 involve complex regulatory networks, including protein-protein and protein-nucleic acid interactions through signaling pathways and noncoding RNA interactions, modulating SOX2 expression may be a potential therapeutic strategy for clinical cancer patients. Therefore, we sorted out the phenotypes related to SOX2 in cancer, hoping to provide a basis for further clinical translation.
Collapse
Affiliation(s)
- L N Ding
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Y Y Yu
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - C J Ma
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - C J Lei
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - H B Zhang
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
26
|
Stevanovic M, Kovacevic-Grujicic N, Petrovic I, Drakulic D, Milivojevic M, Mojsin M. Crosstalk between SOX Genes and Long Non-Coding RNAs in Glioblastoma. Int J Mol Sci 2023; 24:ijms24076392. [PMID: 37047365 PMCID: PMC10094781 DOI: 10.3390/ijms24076392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
Glioblastoma (GBM) continues to be the most devastating primary brain malignancy. Despite significant advancements in understanding basic GBM biology and enormous efforts in developing new therapeutic approaches, the prognosis for most GBM patients remains poor with a median survival time of 15 months. Recently, the interplay between the SOX (SRY-related HMG-box) genes and lncRNAs (long non-coding RNAs) has become the focus of GBM research. Both classes of molecules have an aberrant expression in GBM and play essential roles in tumor initiation, progression, therapy resistance, and recurrence. In GBM, SOX and lncRNAs crosstalk through numerous functional axes, some of which are part of the complex transcriptional and epigenetic regulatory mechanisms. This review provides a systematic summary of current literature data on the complex interplay between SOX genes and lncRNAs and represents an effort to underscore the effects of SOX/lncRNA crosstalk on the malignant properties of GBM cells. Furthermore, we highlight the significance of this crosstalk in searching for new biomarkers and therapeutic approaches in GBM treatment.
Collapse
|
27
|
An Updated Review of Contribution of Long Noncoding RNA-NEAT1 to the Progression of Human Cancers. Pathol Res Pract 2023; 245:154380. [PMID: 37043964 DOI: 10.1016/j.prp.2023.154380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023]
Abstract
Long non-coding RNAs (lncRNAs) present pivotal roles in cancer tumorigenesis and progression. Recently, nuclear paraspeckle assembly transcript 1 (NEAT1) as a lncRNA has been shown to mediate cell proliferation, migration, and EMT in tumor cells. NEAT1 by targeting several miRNAs/mRNA axes could regulate cancer cell behavior. Therefore, NEAT1 may function as a potent biomarker for the prediction and treatment of some human cancers. In this review, we summarized various NEAT1-related signaling pathways that are critical in cancer initiation and progression.
Collapse
|
28
|
Vishnubalaji R, Shaath H, Al-Alwan M, Abdelalim EM, Alajez NM. Reciprocal interplays between MicroRNAs and pluripotency transcription factors in dictating stemness features in human cancers. Semin Cancer Biol 2022; 87:1-16. [PMID: 36354097 DOI: 10.1016/j.semcancer.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
The interplay between microRNAs (miRNAs) and pluripotency transcription factors (TFs) orchestrates the acquisition of cancer stem cell (CSC) features during the course of malignant transformation, rendering them essential cancer cell dependencies and therapeutic vulnerabilities. In this review, we discuss emerging themes in tumor heterogeneity, including the clonal evolution and the CSC models and their implications in resistance to cancer therapies, and then provide thorough coverage on the roles played by key TFs in maintaining normal and malignant stem cell pluripotency and plasticity. In addition, we discuss the reciprocal interactions between miRNAs and MYC, OCT4, NANOG, SOX2, and KLF4 pluripotency TFs and their contributions to tumorigenesis. We provide our view on the potential to interfere with key miRNA-TF networks through the use of RNA-based therapeutics as single agents or in combination with other therapeutic strategies, to abrogate the CSC state and render tumor cells more responsive to standard and targeted therapies.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Hibah Shaath
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Essam M Abdelalim
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M Alajez
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
| |
Collapse
|
29
|
Mukherjee S, Kundu U, Desai D, Pillai PP. Particulate Matters Affecting lncRNA Dysregulation and Glioblastoma Invasiveness: In Silico Applications and Current Insights. J Mol Neurosci 2022; 72:2188-2206. [PMID: 36370303 DOI: 10.1007/s12031-022-02069-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/14/2022] [Indexed: 11/15/2022]
Abstract
With a reported rise in global air pollution, more than 50% of the population remains exposed to toxic air pollutants in the form of particulate matters (PMs). PMs, from various sources and of varying sizes, have a significant impact on health as long-time exposure to them has seen a correlation with various health hazards and have also been determined to be carcinogenic. In addition to disrupting known cellular pathways, PMs have also been associated with lncRNA dysregulation-a factor that increases predisposition towards the onset or progression of cancer. lncRNA dysregulation is further seen to mediate glioblastoma multiforme (GBM) progression. The vast array of information regarding cancer types including GBM and its various precursors can easily be obtained via innovative in silico approaches in the form of databases such as GEO and TCGA; however, a need to obtain selective and specific information correlating anthropogenic factors and disease progression-in the case of GBM-can serve as a critical tool to filter down and target specific PMs and lncRNAs responsible for regulating key cancer hallmarks in glioblastoma. The current review article proposes an in silico approach in the form of a database that reviews current updates on correlation of PMs with lncRNA dysregulation leading to GBM progression.
Collapse
Affiliation(s)
- Swagatama Mukherjee
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Uma Kundu
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Dhwani Desai
- Integrated Microbiome Resource, Department of Pharmacology and Marine Microbial Genomics and Biogeochemistry lab, Department of Biology, Dalhousie University, Halifix, Canada
| | - Prakash P Pillai
- Division of Neurobiology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, 390 002, Gujarat, India.
| |
Collapse
|
30
|
The Effect of lncRNA-PVT1 on Liver Cancer Rats by Regulating the Expression of MMP9. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4126839. [PMID: 36164442 PMCID: PMC9509214 DOI: 10.1155/2022/4126839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/12/2022] [Accepted: 08/13/2022] [Indexed: 11/26/2022]
Abstract
As a malignant tumor, liver cancer has a high lethality rate. The research on the pathogenesis of liver cancer is the key to the treatment of liver cancer. The latest research claims that lncRNA-PVT1 as a tumor gene participates in the generation and development of tumors by regulating matrix metalloproteinase MMP9. The purpose of this article is to explore the specific effect and mechanism of lncRNA-PVT1 on liver cancer rats by regulating the expression of MMP9. In this article, 50 rats are used as experimental subjects, the rats are divided into control group and observation group, and the liver cancer cell line HepG2 is cultured. The transplanted tumor liver cancer model was constructed by transfection of hair, the expression of lncRNA-PVT1 in the observation group was reduced by knockdown method, the expression levels and changes of lncRNA-PVT1 and MMP9 in the two groups were detected by PCR fluorescence method, and the difference between lncRNA-PVT1 and MMP9 was analyzed. The MTT method was used to detect the proliferation, migration, and invasion capabilities of the two groups of liver cancer cells (LCC) and to explore the effect of lncRNA-PVT1 on rat LCC by regulating the expression of MMP9. The results of the study showed that after knocking down the expression of lncRNA-PVT1 in the observation group, the expression of MMP9 also decreased. At the same time, the migration rate of LCC HepG2 decreased by 27.4%, the level of invasion ability decreased by 21.6%, and the proliferation rate of LCC decreased by 17.8%. Therefore, it can be seen that lncRNA-PVT1 plays a positive regulatory role on the expression of MMP9, and the expression of MMP9 promotes the proliferation, migration, and invasion of rat LCC.
Collapse
|
31
|
Goenka A, Tiek DM, Song X, Iglesia RP, Lu M, Hu B, Cheng SY. The Role of Non-Coding RNAs in Glioma. Biomedicines 2022; 10:2031. [PMID: 36009578 PMCID: PMC9405925 DOI: 10.3390/biomedicines10082031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022] Open
Abstract
For decades, research in cancer biology has been focused on the protein-coding fraction of the human genome. However, with the discovery of non-coding RNAs (ncRNAs), it has become known that these entities not only function in numerous fundamental life processes such as growth, differentiation, and development, but also play critical roles in a wide spectrum of human diseases, including cancer. Dysregulated ncRNA expression is found to affect cancer initiation, progression, and therapy resistance, through transcriptional, post-transcriptional, or epigenetic processes in the cell. In this review, we focus on the recent development and advances in ncRNA biology that are pertinent to their role in glioma tumorigenesis and therapy response. Gliomas are common, and are the most aggressive type of primary tumors, which account for ~30% of central nervous system (CNS) tumors. Of these, glioblastoma (GBM), which are grade IV tumors, are the most lethal brain tumors. Only 5% of GBM patients survive beyond five years upon diagnosis. Hence, a deeper understanding of the cellular non-coding transcriptome might help identify biomarkers and therapeutic agents for a better treatment of glioma. Here, we delve into the functional roles of microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA) in glioma tumorigenesis, discuss the function of their extracellular counterparts, and highlight their potential as biomarkers and therapeutic agents in glioma.
Collapse
Affiliation(s)
- Anshika Goenka
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Deanna Marie Tiek
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiao Song
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rebeca Piatniczka Iglesia
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Minghui Lu
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Master of Biotechnology Program, Northwestern University, Evanston, IL 60208, USA
| | - Bo Hu
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shi-Yuan Cheng
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
32
|
Molecular Interactions of the Long Noncoding RNA NEAT1 in Cancer. Cancers (Basel) 2022; 14:cancers14164009. [PMID: 36011001 PMCID: PMC9406559 DOI: 10.3390/cancers14164009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
As one of the best-studied long noncoding RNAs, nuclear paraspeckle assembly transcript 1 (NEAT1) plays a pivotal role in the progression of cancers. NEAT1, especially its isoform NEAT1-1, facilitates the growth and metastasis of various cancers, excluding acute promyelocytic leukemia. NEAT1 can be elevated via transcriptional activation or stability alteration in cancers changing the aggressive phenotype of cancer cells. NEAT1 can also be secreted from other cells and be delivered to cancer cells through exosomes. Hence, elucidating the molecular interaction of NEAT1 may shed light on the future treatment of cancer. Herein, we review the molecular function of NEAT1 in cancer progression, and explain how NEAT1 interacts with RNAs, proteins, and DNA promoter regions to upregulate tumorigenic factors.
Collapse
|
33
|
Oxidative stress-induced endothelial cells-derived exosomes accelerate skin flap survival through Lnc NEAT1-mediated promotion of endothelial progenitor cell function. Stem Cell Res Ther 2022; 13:325. [PMID: 35850692 PMCID: PMC9290268 DOI: 10.1186/s13287-022-03013-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/08/2022] [Indexed: 12/14/2022] Open
Abstract
Background Flap transplantation is commonly used in reconstructive surgery. A prerequisite for skin flap survival is sufficient blood supply. However, such approaches remain unclear. This study aimed to explore the underlying mechanisms of exosomes derived from human umbilical vascular endothelial cells (HUVECs) exposed to oxidative stress on endothelial progenitor cells (EPCs) and their subsequent influence on the survival of skin flaps. Methods HUVECs were treated with various concentrations of H2O2 to establish an oxidative stress model. To investigate the effects of H2O2-HUVEC-Exos and HUVEC-Exos, Cell Counting Kit-8, tube formation, invasion assays, and quantitative real-time polymerase chain reaction (qRT-PCR) were performed in EPCs. Microarray analysis was used to reveal the differentially expressed long non-coding RNAs (lncRNAs) in the H2O2-HUVEC-Exos and HUVEC-Exos. In addition, gene silencing and western blotting were employed to determine the mechanism behind lncRNA nuclear enrichment enriched transcript 1 (Lnc NEAT1) in EPCs. Further, a rat skin flap model was used to determine the role of the exosomes in skin flap survival in vivo. Results HUVECs were stimulated with 100 μmol/L H2O2 for 12 h to establish an oxidative stress model. H2O2-HUVEC-Exos promoted the proliferation, tube formation, and invasion of EPCs and remarkably increased skin flap survival compared to the HUVEC-Exos and control groups. Sequencing of exosome RNAs revealed that the Lnc NEAT1 level was dramatically increased in the H2O2-HUVEC-Exos, leading to activation of the Wnt/β-catenin signaling pathway. Comparatively, knockdown of Lnc NEAT1 in HUVEC-Exos and H2O2-HUVEC-Exos significantly inhibits the angiogenic capacity of EPCs, reduced the survival area of skin flap and downregulated the expression levels of Wnt/β-catenin signaling pathway proteins, whereas Wnt agonist partly reversed the negative effect of NEAT1 downregulation on EPCs through the Wnt/β-catenin signaling pathway. Conclusions Exosomes derived from HUVECs stimulated by oxidative stress significantly promoted the pro-angiogenic ability of EPCs through the Wnt/β-catenin signaling pathway mediated by Lnc NEAT1 and hence enhanced random flap survival in vivo. Therefore, the application of H2O2-HUVEC-Exos may serve as an alternative therapy for improving random skin flap survival. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03013-9.
Collapse
|
34
|
LINC01426 aggravates the malignant progression of glioma through miR-661/Mdm2 axis. Brain Res Bull 2022; 188:110-121. [PMID: 35772605 DOI: 10.1016/j.brainresbull.2022.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/20/2022] [Accepted: 06/26/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Long intergenic non-protein coding RNA 1426 (LINC01426) is up-regulated in glioma and functions as a tumor promoter. However, the role of LINC01426 in glioma required further exploration. Therefore, this article mainly studied the role and possible mechanism of LINC01426 in glioma. METHODS The area under the receiver operating characteristic curve was used to determine the diagnostic value of LINC01426. The effect of LINC01426 on tumor growth was analyzed by tumorigenesis assay and immunohistochemical analysis. Bioinformatics analysis, dual-luciferase assay, RNA pull-down, Pearson test, and real-time quantitative PCR (RT-qPCR) were applied to verify the relationship between target genes. The expressions and effects of LINC01426, miR-661 and MDM2 proto-oncogene (Mdm2) in glioma were examined by bioinformatics analysis combined with molecular and functional experiments (RT-qRCR, 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide, clone formation, BrdU, flow cytometry). The expressions of proliferation and apoptosis-related proteins were determined by Western blot. RESULTS LINC01426, which was high-expressed in glioma and was related to poor prognosis, could be used as a diagnostic marker for glioma. SiLINC01426 inhibited the malignant phenotype of glioma cells in vitro and attenuated tumor growth and PCNA expression in vivo, while the effects of LINC01426 were the opposite. LINC01426 targeted and inversely correlated with miR-661, which was low-expressed in glioma. MiR-661 inhibitor evidently overturned the effect of siLINC01426 on biological functions, proliferation, and apoptosis-related proteins of glioma cells. Mdm2 bound to miR-661. Moreover, siMdm2 reversed the effects of miR-661 inhibitor on the biological characteristics and Mdm2/p53/p21 expression of glioma cells. CONCLUSION LINC01426 aggravated the malignant progression of glioma through miR-661/Mdm2 axis.
Collapse
|
35
|
Liao G, Huang Z, Gan T, Wu C, Wang X, Li D. Long non-coding RNA nuclear enriched abundant transcript 1 (NEAT1) modulates inhibitor of DNA binding 1 (ID1) to facilitate papillary thyroid carcinoma development by sponging microRNA-524-5p. Bioengineered 2022; 13:13201-13212. [PMID: 35635748 PMCID: PMC9275871 DOI: 10.1080/21655979.2022.2076498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Long non-coding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) exerts a pro-oncogenic role in several cancers, whereas its underlying regulatory mechanism in papillary thyroid carcinoma (PTC) progression remains unknown. This research mainly explored the roles of NEAT1 in PTC development. Quantitative real-time polymerase-chain reaction (qRT-PCR) was applied to measure NEAT1, miR-524-5p, and inhibitor of DNA binding 1 (ID1) expression in PTC tissues and cells. Western blot was conducted for detecting the protein levels. MTT, transwell, and flow cytometry assays were applied to assess cell proliferation, metastasis, and apoptosis in PTC cells in vitro. The PTC xenograft tumor model was used for investigating the role of NEAT1 in vivo. Dual-luciferase reporter assay was utilized for confirming the interaction between miR-524-5p and NEAT1 or ID1. In PTC tissues and cells, NEAT1 was significantly up-regulated. NEAT1 silencing blocked cell proliferation, metastasis, and facilitated apoptosis in vitro and impeded xenograft tumor growth in vivo. Bioinformatics prediction revealed the existence of binding sites between NEAT1 and miR-524-5p. Besides, ID1 was confirmed as a direct target to miR-524-5p, and the enhancement of ID1 reversed the regulation of miR-524-5p upregulation on cell progression. In addition, NEAT1 promoted PTC development by regulating ID1 expression via sponging miR-524-5p in PTC. In summary, we demonstrate that NEAT1 advanced the process of PTC by miR-524-5p/ID1 axis, which may enhance our comprehension of PTC pathogenesis.
Collapse
Affiliation(s)
- Guansheng Liao
- Department of Thyroid Surgery, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Zhuoya Huang
- Department of Pathology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Tianyu Gan
- Department of Thyroid Surgery, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Cong Wu
- Department of Thyroid Surgery, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Xiaolong Wang
- Department of Thyroid Surgery, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Dexiang Li
- Department of Thyroid Surgery, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| |
Collapse
|
36
|
Huang L, Sun C, Peng R, Liu Z. The Effect of LncRNA H19 on Human Dental Pulp Cells Through Tumor Growth Factor- β1 (TGF- β1)/Smad Signaling Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The pulp tissue is located in the pulp cavity of tooth and has the ability of nutrition, feeling, and defense. Human dental pulp cells (hDPCs) are the main cells of pulp with the ability to self-renew and multi-directional differentiation. LncRNA H19 is involved in the regulation of
human dental pulp stem cells, but the specific mechanism has not been elucidated. hDPCs were isolated and cultured in vitro to measure vimentin expression by immunohistochemistry (IHC). hDPCs were randomly assigned into control group, negative control (NC) group and lncRNA H19 overexpression
group followed by measuring lncRNA H19, DSPP, and DMP-1 mRNA expression, cell proliferation, ALP activity, BMP-2 expression by ELISA, TGF-β1, Smad2, and Smad4 expression by Western blot. hDPCs were positive for vimentin staining and confirmed to be derived from mesenchymal. Transfection
of pcDNA3.1-LncRNA H19 plasmid significantly increased LncRNA H19 expression, promoted cell proliferation, enhanced ALP activity, upregulated DSPP and DMP-1, elevated BMP-2 expression in cell supernatant, as well as promoted TGF-β1, Smad2, and Smad4 expressions compared with control
(P < 0.05). In conclusion, lncRNA H19 facilitates hDPCs differentiation into odontoblasts by promoting cell proliferation and increasing BMP-2 secretion via regulating TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Li Huang
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Chuankong Sun
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Ruobing Peng
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Zhiming Liu
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| |
Collapse
|
37
|
Wu X, Yang L, Wang J, Hao Y, Wang C, Lu Z. The Involvement of Long Non-Coding RNAs in Glioma: From Early Detection to Immunotherapy. Front Immunol 2022; 13:897754. [PMID: 35619711 PMCID: PMC9127066 DOI: 10.3389/fimmu.2022.897754] [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: 03/16/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Glioma is a brain tumor that arises in the central nervous system and is categorized according to histology and molecular genetic characteristics. Long non-coding RNAs (lncRNAs) are RNAs longer than 200 nucleotides in length. They have been reported to influence significant events such as carcinogenesis, progression, and increased treatment resistance on glioma cells. Long non-coding RNAs promote cell proliferation, migration, epithelial-to-mesenchymal transition and invasion in glioma cells. Various significant advancements in transcriptomic profiling studies have enabled the identification of immune-related long non-coding RNAs as immune cell-specific gene expression regulators that mediates both stimulatory and suppressive immune responses, implying lncRNAs as potential candidates for improving immunotherapy efficacy against tumors and due to the lack of different diagnostic and treatments for glioma, lncRNAs are potential candidates to be used as future diagnostic, prognostic biomarker and treatment tools for glioma. This review’s primary purpose is to concentrate on the role of long non-coding RNAs in early glioma identification, treatment, and immunotherapy.
Collapse
Affiliation(s)
- Xiaoben Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lei Yang
- Department of Medical Engineering, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yingying Hao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Changyin Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
38
|
Zhu R, Wang Z. Inhibitory Effect of MicroRNA-376b-Overexpressing Bone Marrow Mesenchymal Stem Cells (BMSCs) on Malignant Characteristics of Glioma Cells Through Targeting Forkhead Box Protein P2 (FOXP2). J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study investigated the impact of microRNA (miR)-376b derived from BMSCs on glioma progression. BMSCs were transfected with miR-376b mimic, miR-376b inhibitor or NC and then cocultured with glioma cells followed by measuring cell behaviors by MTT assay, Transwell assay and flow
cytometry, FOXP2 and miR-376b expression by Western blot and RT-qPCR. After confirming the inhibitory and mimicking activity of transfection, we found that overexpression of miR-376b in BMSCs decreased glioma cell invasion, migration and proliferation but promoted cell apoptosis within 24
h and 48 h after transfection along with reduced number of cells in S-phase. Mechanically, miR-376b targeted miR-376b and up-regulation of miR-376b caused down-regulation of FOXP2 (p < 0.05). Overexpression of miR-376b in BMSCs decelerated glioma cell cycle and inhibitedmalignant
behaviors of glioma cells by targeting FOXP2 expression. These evidence unveils the potential role of FOXP2 as a biomarker for the treatment of gliomas.
Collapse
Affiliation(s)
- Ruoyu Zhu
- Faculty of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jianan, Shandong, 250014, China
| | - Zhonglin Wang
- Department of Neurology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| |
Collapse
|
39
|
LncRNA MNX1-AS1: A novel oncogenic propellant in cancers. Biomed Pharmacother 2022; 149:112801. [PMID: 35290890 DOI: 10.1016/j.biopha.2022.112801] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
To date, recent studies have shown that long non-coding RNAs (lncRNAs) are key players in gene regulation processes involved in cancer pathogenesis. In general, Motor neuron and pancreas homeobox 1-antisense RNA1 (MNX1-AS1) is highly expressed in all cancers as reported so far and exerts oncogenic effects through different mechanisms. In this review, we comprehensively summarize the detailed mechanisms of potential functions of MNX1-AS1 in different cancer types as well as the latest knowledge highlighting the potential of MNX1-AS1 as a therapeutic target for cancer. Aberrant expression of MNX1-AS1 closely correlates with clinicopathological parameters. such as lymphatic metastasis, tumor size, tumor stage, OS and DFS. Thus, MNX1-AS1 can be used as a diagnostic and prognostic biomarker or even a therapeutic prognostic target. This article reviews its function, molecular mechanism and clinical prognosis in various malignancies.
Collapse
|
40
|
Liu K, Chen H, Wang Y, Jiang L, Li Y. Evolving Insights Into the Biological Function and Clinical Significance of Long Noncoding RNA in Glioblastoma. Front Cell Dev Biol 2022; 10:846864. [PMID: 35531099 PMCID: PMC9068894 DOI: 10.3389/fcell.2022.846864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is one of the most prevalent and aggressive cancers worldwide. The overall survival period of GBM patients is only 15 months even with standard combination therapy. The absence of validated biomarkers for early diagnosis mainly accounts for worse clinical outcomes of GBM patients. Thus, there is an urgent requirement to characterize more biomarkers for the early diagnosis of GBM patients. In addition, the detailed molecular basis during GBM pathogenesis and oncogenesis is not fully understood, highlighting that it is of great significance to elucidate the molecular mechanisms of GBM initiation and development. Recently, accumulated pieces of evidence have revealed the central roles of long noncoding RNAs (lncRNAs) in the tumorigenesis and progression of GBM by binding with DNA, RNA, or protein. Targeting those oncogenic lncRNAs in GBM may be promising to develop more effective therapeutics. Furthermore, a better understanding of the biological function and underlying molecular basis of dysregulated lncRNAs in GBM initiation and development will offer new insights into GBM early diagnosis and develop novel treatments for GBM patients. Herein, this review builds on previous studies to summarize the dysregulated lncRNAs in GBM and their unique biological functions during GBM tumorigenesis and progression. In addition, new insights and challenges of lncRNA-based diagnostic and therapeutic potentials for GBM patients were also introduced.
Collapse
Affiliation(s)
- Kun Liu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
| | - Yuanyuan Wang
- Department of Pathology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
| | - Liping Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Yi Li, ; Liping Jiang,
| | - Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Yi Li, ; Liping Jiang,
| |
Collapse
|
41
|
Serum-derived extracellular vesicles facilitate temozolomide resistance in glioblastoma through a HOTAIR-dependent mechanism. Cell Death Dis 2022; 13:344. [PMID: 35418162 PMCID: PMC9008004 DOI: 10.1038/s41419-022-04699-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 02/08/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022]
Abstract
Extracellular vesicle (EV)-mediated transfer of long non-coding RNAs (lncRNAs) has been reported to regulate chemoresistance in various cancers. We herein investigate the therapeutic potential of bioinformatically identified HOTAIR transferred by serum-derived EVs (serum-EVs) in temozolomide (TMZ) resistance of glioblastoma (GBM) and the downstream mechanisms. EVs were isolated from the serum of GBM patients. Expression of HOTAIR was examined in the clinical tissue samples and serum-EVs of GBM patients. The downstream miRNAs of HOTAIR and its target genes were predicted in silico. The effects of the HOTAIR transmitted by serum-EVs in malignant phenotypes, tumor growth, and TMZ resistance were assessed in vitro and in vivo. HOTAIR expression was upregulated in clinical tissues, cells, and serum-EVs of GBM. Co-culture data showed that GBM-serum-EVs facilitated GBM cell proliferative and invasive phenotypes and TMZ resistance by elevating HOTAIR. In GBM cells, HOTAIR competitively bound to miR-526b-3p and weakened miR-526b-3p’s binding ability to EVA1, thus increasing the expression of EVA1. Furthermore, HOTAIR carried by serum-EVs promoted tumor growth and TMZ resistance in vivo by suppressing miR-526b-3p-mediated EVA1 inhibition. GBM-serum-EV-enclosed HOTAIR may augment GBM progression and chemoresistance through miR-526b-3p downregulation and EVA1 upregulation. These results provide a strategy to reduce TMZ resistance in GBM treatment.
Collapse
|
42
|
Liu B, Qiang L, Guan B, Ji Z. Targeting kinesin family member 21B by miR-132-3p represses cell proliferation, migration and invasion in gastric cancer. Bioengineered 2022; 13:9006-9018. [PMID: 35341446 PMCID: PMC9161970 DOI: 10.1080/21655979.2022.2054755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Recently, kinesin family member 21B (KIF21B) has been reported to be an oncogene in non-small cell lung cancer and hepatocellular carcinoma. However, the functional role of KIF21B and related molecular mechanisms in gastric cancer (GC) remain largely uncovered. In this study, online bioinformatics analysis showed that KIF21B was overexpression in GC and predicted poor prognosis. Consistently, we found that the protein expression of KIF21B was upregulated in GC tissues compared with adjacent tissues by immunohistochemistry. Knockdown of KIF21B significantly suppressed cell proliferation, migration and invasion in GC cell lines (AGS and SNU-5) using Cell counting kit‑8 (CCK-8) assay, colony formation and transwell assay. KIF21B was confirmed as the target of miR-132-3p in GC cells by luciferase reporter assay. Moreover, miR-132-3p was down-regulated and KIF21B expression was upregulated in GC tissues. Overexpression of KIF21B reversed the miR-132-3p-mediated suppressive effects on GC cell proliferation, migration and invasion. Furthermore, miR-132-3p overexpression downregulated the protein levels of Wnt1, c-Myc, β-catenin, proliferating cell nuclear antigen (PCNA) and N-cadherin, and upregulated E-cadherin expression in GC cells, which were all alleviated after KIF21B overexpression. In conclusion, our findings indicate that down-regulation of KIF21B by miR-132-3p suppresses cellular functions in GC, which might be linked to reduced Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Bingtian Liu
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ling Qiang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Bingxin Guan
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhipeng Ji
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
43
|
Long noncoding RNA NEAT1 promotes ferroptosis by modulating the miR-362-3p/MIOX axis as a ceRNA. Cell Death Differ 2022; 29:1850-1863. [PMID: 35338333 PMCID: PMC9433379 DOI: 10.1038/s41418-022-00970-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
Ferroptosis, a novel form of regulated cell death induced by iron-dependent lipid peroxidation, plays an essential role in the development and drug resistance of tumors. Long noncoding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) has been reported to be involved in the regulation of cell cycle, proliferation, apoptosis, and migration of tumor cells. However, the function and molecular mechanism of NEAT1 in regulating ferroptosis in tumors remain unclear. Here, we found that ferroptosis inducers erastin and RSL3 increased NEAT1 expression by promoting the binding of p53 to the NEAT1 promoter. Induced NEAT1 promoted the expression of MIOX by competitively binding to miR-362-3p. MIOX increased ROS production and decreased the intracellular levels of NADPH and GSH, resulting in enhanced erastin- and RSL3-induced ferroptosis. Importantly, overexpression of NEAT1 increased the anti-tumor activity of erastin and RSL3 by enhancing ferroptosis both in vitro and in vivo. Collectively, these data suggest that NEAT1 plays a novel and indispensable role in ferroptosis by regulating miR-362-3p and MIOX. Considering the clinical findings that HCC patients are insensitive to chemotherapy and immunotherapy, ferroptosis induction may be a promising therapeutic strategy for HCC patients with high NEAT1 expression.
Collapse
|
44
|
Hepatoblastoma: Derived Exosomal LncRNA NEAT1 Induces BMSCs Differentiation into Tumor-Supporting Myofibroblasts via Modulating the miR-132/MMP9 Axis. JOURNAL OF ONCOLOGY 2022; 2022:7630698. [PMID: 35300348 PMCID: PMC8923764 DOI: 10.1155/2022/7630698] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/24/2022] [Accepted: 02/17/2022] [Indexed: 01/10/2023]
Abstract
Background Hepatoblastoma (HB) is the most common malignant tumor of the liver. MMP9 plays an essential role in HB. The purpose of our study was to screen for differentially expressed lncRNAs and miRNAs that targeted MMP9. Based on this, the role of lncRNA NEAT1/miR-132/MMP9 in HB and the mechanisms involved were discussed. Methods Bioinformatics analysis was used to screen the differentially expressed lncRNAs and miRNAs targeting MMP9. Exosomes were extracted from HB cells and normal liver cells for characterization and identification. Exosome uptake assay was conducted to determine whether exosomes were absorbed by bone marrow stromal cells (BMSCs). α-SMA, fibronectin, and s-100 expressions in tissues and cells were detected by IHC and ICC. lncRNA XIST, lncRNA NEAT1, miR-132, and MMP9 expressions were characterized by qRT-PCR. Western blot was performed to measure MMP9, α-SMA, and s-100 expressions. Flow cytometry was used to stain α-SMA, s-100. Bioinformatics and dual-luciferase reporter assay were applied to verify the interaction between lncRNA NEAT1 and miR-132, and miR-132 and MMP9. The effect of lncRNA NEAT1 on the development of HB in nude mice was studied. Results Differentially expressed lncRNA NEAT1/miR-132/MMP9 was obtained through bioinformatics analysis and cell verification. HB-derived exosomal lncRNA NEAT1 regulated miR-132 and MMP9 expression in BMSCs. In addition, HB-derived exosomal lncRNA NEAT1 promoted BMSCs differentiation toward invasive myofibroblast via miR-132/MMP9 axis. LncRNA NEAT1 regulated MMP9 through miR-132. Tumor formation experiments in nude mice showed that HB-derived exosomal lncRNA NEAT1 could affect the development of HB. Conclusion HB-derived exosomal lncRNA NEAT1 induced BMSCs differentiation into tumor-supporting myofibroblasts via modulating miR-132/MMP9 axis, which provided a new target for HB treatment.
Collapse
|
45
|
Mukherjee S, Pillai PP. Current insights on extracellular vesicle-mediated glioblastoma progression: Implications in drug resistance and epithelial-mesenchymal transition. Biochim Biophys Acta Gen Subj 2022; 1866:130065. [PMID: 34902452 DOI: 10.1016/j.bbagen.2021.130065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is one of the most fatal tumors of the central nervous system with high rate of disease progression, diagnosis, prognosis and low survival rate. Therapeutic approaches that relied on surgical resection and chemotherapy have been unable to curb the disease progression and subsequently leading to increase in incidences of GBM reoccurrence. SCOPE OF THE REVIEW In the recent times, membrane-bound extracellular vesicles (EVs) have been observed as one of the key reasons for the uncontrolled growth of GBM. EVs are shown to have the potential to contribute to the disease progression via mediating drug resistance and epithelial-mesenchymal transition. The GBM-derived EVs (GDEVs) with its cargo contents act as the biological trojan horse and lead to disease progression after being received by the recipient target cells. This review article highlights the biophysical, biochemical properties of EVs, its cargo contents and its potential role in the growth and progression of GBM by altering tumour microenvironment. MAJOR CONCLUSIONS EVs are being explored for serving as novel disease biomarkers in a variety of cancer types such as adenocarcinoma, pancreatic cancer, color rectal cancer, gliomas and glioblastomas. Improvement in the EV isolation protocols, polymer-based separation techniques and transcriptomics, have made EVs a key diagnostic marker to unravel the progression and early GBM diagnosis. GDEVs role in tumour progression is under extensive investigations. GENERAL SIGNIFICANCE Attempts have been also made to discuss and compare the usage of EVs as potential therapeutic targets versus existing therapies targeting drug resistance and EMT.
Collapse
Affiliation(s)
- Swagatama Mukherjee
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Prakash P Pillai
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India.
| |
Collapse
|
46
|
Wang X, Wang Y, Xie F, Song ZT, Zhang ZQ, Zhao Y, Wang SD, Hu H, Zhang YS, Qian LJ. Norepinephrine promotes glioma cell migration through up-regulating the expression of Twist1. BMC Cancer 2022; 22:213. [PMID: 35219305 PMCID: PMC8882280 DOI: 10.1186/s12885-022-09330-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 02/17/2022] [Indexed: 02/08/2023] Open
Abstract
Background Glioma cells are characterized by high migration ability, resulting in aggressive growth of the tumors and poor prognosis of patients. It has been reported that the stress-induced hormone norepinephrine (NE) contributes to tumor progression through mediating a number of important biological processes in various cancers. However, the role of NE in the regulation of glioma migration is still unclear. Epithelial-to-mesenchymal transition (EMT) is one of the most important steps for tumor migration and metastasis. Twist1, as a key regulator of EMT, has been found to be elevated during glioma migration. But it is still unknown whether Twist1 is involved in the effect of NE on the migration of glioma cells. Methods Wound healing assay and transwell assay were conducted to evaluate the migration of glioma cells upon different treatments. The mesenchymal-like phenotype and the expression of Twist1 after NE treatment were assessed by cell diameters, real-time PCR, western blot and immunofluorescence staining. The gain-and loss-of-function experiments were carried out to investigate the biological function of Twist1 in the migration induced by NE. Finally, the clinical significance of Twist1 was explored among three public glioma datasets. Results In this study, our finding revealed a facilitative effect of NE on glioma cell migration in a β-adrenergic receptor (ADRB)-dependent way. Mechanistically, NE induced mesenchymal-like phenotype and the expression of Twist1. Twist1 overexpression promoted glioma cells migration, while knockdown of Twist1 abolished the discrepancy in the migration ability between NE treated glioma cells and control cells. In addition, the clinical analysis demonstrated that Twist1 was up-regulated in malignant gliomas and recurrent gliomas, and predicted a poor prognosis of glioma patients. Conclusions NE enhanced the migration ability of glioma cells through elevating the expression of Twist1. Our finding may provide potential therapeutic target for protecting patients with glioma from the detrimental effects of stress biology on the tumor progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09330-9.
Collapse
|
47
|
Pei Y, Zhang H, Lu K, Tang X, Li J, Zhang E, Zhang J, Huang Y, Yang Z, Lu Z, Li Y, Zhang H, Dong L. Circular RNA circRNA_0067934 promotes glioma development by modulating the microRNA miR-7/ Wnt/β-catenin axis. Bioengineered 2022; 13:5792-5802. [PMID: 35213267 PMCID: PMC8973834 DOI: 10.1080/21655979.2022.2033382] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Glioma, one of the most prevalent malignant tumors, is well-known for its poor prognosis and low survival rate among patients. As a type of non-coding RNA, circular RNAs (circRNAs) play a significant role in tumor progression. However, the function and role of circRNAs in glioma development remain unclarified. In our experiments, the relative expression level of circRNA_0067934 and miR-7 in glioma tissue was detected by qRT-PCR, and specific gene knockdown was mediated by siRNA and miRNA-inhibitor. Dual-luciferase reporter assay was carried out to determine whether miR-7 successfully targeted circRNA_0067934. Also, CCK-8 and Transwell were performed to evaluate the malignant behaviors of glioma tissues. Western blotting and immunofluorescence were used to evaluate relative protein expression levels. The results of qRT-PCR indicated that circRNA_0067934 was over-expressed in glioma tissues, and down regulation of circRNA_0067934 reduced the tumor progression by inhibiting cell proliferation, invasion, and migration. The relative expression level of miR-7 was significantly reduced in glioma tissues, which showed a negative association with the expression of circRNA_0067934. CircRNA_0067934 could tagete the miR-7 to regulate progression of glioma cell. In addition, the Wnt/β-catenin signaling pathway might involve in down stream regulation of circRNA_0067934 and miR-7. In conclusion, our results revealed that circRNA_0067934 regulates glioma cells progression by targeting miR-7/ Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Yunlong Pei
- Department of Clinical Medicine, Dalian Medical University, Dalian, Liaoning, China.,Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Hongying Zhang
- Department of Image, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Kongye Lu
- Department of Clinical Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaojia Tang
- Department of Rehabilitation, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Jialing Li
- Department of clinical science, Ross University School of Medicine (RUSM), Bridgetown, Barbados
| | - Enpeng Zhang
- Department of Clinical Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Jun Zhang
- Department of Neurosurgery, Fudan University, Yangzhou, Jiangsu, China
| | - Yujia Huang
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhijie Yang
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhenggang Lu
- Department of Clinical Medicine, Dalian Medical University, Dalian, Liaoning, China.,Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuping Li
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Hengzhu Zhang
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| | - Lun Dong
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
48
|
Zhou H, Wang Y, Liu Z, Zhang Z, Xiong L, Wen Y. Recent advances of NEAT1-miRNA interactions in cancer. Acta Biochim Biophys Sin (Shanghai) 2022; 54:153-162. [PMID: 35538025 PMCID: PMC9827865 DOI: 10.3724/abbs.2021022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
With high incidence rate, cancer is the main cause of death in humans. Non-coding RNAs, as novel master regulators, especially long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), play important roles in the regulation of tumorigenesis. lncRNA NEAT1 has recently gained much attention, as it is dysregulated in a broad spectrum of cancers, where it acts as either an oncogene or a tumor suppressor gene. Accumulating evidence shows that NEAT1 is correlated with the process of carcinogenesis, including proliferation, invasion, survival, drug resistance, and metastasis. NEAT1 is considered to be a biomarker and a novel therapeutic target for the diagnosis and prognosis of different cancer types. The mechanisms by which NEAT1 plays a critical role in cancers are mainly via interactions with miRNAs. NEAT1-miRNA regulatory networks play significant roles in tumorigenesis, which has attracted much attention from researchers around the world. In this review, we summarize the interaction of NEAT1 with miRNAs in the regulation of protein-coding genes in cancer. A better understanding of the NEAT1-miRNA interactions in cancer will help develop new diagnostic biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Wen
- Correspondence address. Tel: +86-731-85294099; E-mail:
| |
Collapse
|
49
|
Liang J, Liu C, Xu D, Xie K, Li A. LncRNA NEAT1 facilitates glioma progression via stabilizing PGK1. J Transl Med 2022; 20:80. [PMID: 35123484 PMCID: PMC8818213 DOI: 10.1186/s12967-022-03273-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/23/2022] [Indexed: 01/09/2023] Open
Abstract
Background Long noncoding RNA NEAT1 has been implicated in glioma progression. However, the effect of NEAT1 on glycolysis of glioma cell and the potential mechanism remain unclear. Methods In vitro experiments, including CCK-8, colony formation, ECAR, and lactate detection assays were performed to evaluate the effect of NEAT1 on proliferation and glycolysis of glioma cell. RNA pulldown and RIP assays were performed to identify the interaction between NEAT1 and PGK1. Truncated mutation of NEAT1 and PGK1 was used to confirm the specific interactive domains between NEAT1 and PGK1. Animal studies were performed to analyze the effect of NEAT1/PGK1 on glioma progression. Results NEAT1 knockdown significantly suppressed the proliferation and glycolysis of glioma cells. NEAT1 could specifically interact with PGK1, which promotes PGK1 stability. Hairpin A of NEAT1 is essential for interaction with M1 domain of PGK1. Depletion of NEAT1 markedly inhibited tumor growth in mice, while PGK1 could reverse this effect. Higher expression of NEAT1 was associated with poor overall survival of GBM patients. Conclusions NEAT1 over expression promotes glioma progression through stabilizing PGK1. NEAT1/PGK1 axis is a candidate therapeutic target for glioma treatment.
Collapse
|
50
|
Tumor Cell Infiltration into the Brain in Glioblastoma: From Mechanisms to Clinical Perspectives. Cancers (Basel) 2022; 14:cancers14020443. [PMID: 35053605 PMCID: PMC8773542 DOI: 10.3390/cancers14020443] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most common and malignant primary brain tumor, defined by its highly aggressive nature. Despite the advances in diagnostic and surgical techniques, and the development of novel therapies in the last decade, the prognosis for glioblastoma is still extremely poor. One major factor for the failure of existing therapeutic approaches is the highly invasive nature of glioblastomas. The extreme infiltrating capacity of tumor cells into the brain parenchyma makes complete surgical removal difficult; glioblastomas almost inevitably recur in a more therapy-resistant state, sometimes at distant sites in the brain. Therefore, there are major efforts to understand the molecular mechanisms underpinning glioblastoma invasion; however, there is no approved therapy directed against the invasive phenotype as of now. Here, we review the major molecular mechanisms of glioblastoma cell invasion, including the routes followed by glioblastoma cells, the interaction of tumor cells within the brain environment and the extracellular matrix components, and the roles of tumor cell adhesion and extracellular matrix remodeling. We also include a perspective of high-throughput approaches utilized to discover novel players for invasion and clinical targeting of invasive glioblastoma cells.
Collapse
|