1
|
Qu L, Wang F, Wang Y, Li Z. The regulation of LRPs by miRNAs in cancer: influencing cancer characteristics and responses to treatment. Cancer Cell Int 2025; 25:182. [PMID: 40382654 PMCID: PMC12085831 DOI: 10.1186/s12935-025-03804-z] [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: 09/10/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The low-density lipoprotein receptor-related protein (LRP) family is a group of cell surface receptors that participate in a variety of biological processes, including lipid metabolism, Wnt signaling, and bone metabolism. miRNAs are small non-coding RNA molecules that regulate gene expression and play a role in many biological processes, including the occurrence and development of tumors. Accumulating evidence demonstrates that LRP members are modulated by miRNAs across multiple cancer types, influencing key oncogenic processes-including tumor cell proliferation, apoptosis suppression, extracellular matrix remodeling, cell adhesion, and angiogenesis. The LRPs, miRNAs, their upstream lncRNAs, and downstream signaling molecules often form complex signaling pathways to regulate the activity of tumor cells. However, the tissue-specific roles and mechanistic underpinnings of these pathways remain incompletely understood. When examining the emerging concept of the interaction between miRNAs and LRPs, we emphasize the significance of these complex regulatory layers in the initiation and progression of cancer. Collectively, these findings are critical for advancing our understanding of the role of the LRPs family in the occurrence and development of tumors, as well as for the development of new strategies for cancer treatment.
Collapse
Affiliation(s)
- Lianyue Qu
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Fan Wang
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Yuxiang Wang
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Zixuan Li
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China.
- Department of Radiology, The First Hospital of China Medical University, Shenyang, P. R. China.
| |
Collapse
|
2
|
Mastrantuono E, Ghibaudi M, Matias D, Battaglia G. The multifaceted therapeutical role of low-density lipoprotein receptor family in high-grade glioma. Mol Oncol 2024; 18:2966-2976. [PMID: 39276062 PMCID: PMC11619799 DOI: 10.1002/1878-0261.13730] [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: 02/01/2024] [Revised: 07/25/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
The diverse roles of the low-density lipoprotein receptor family (LDLR) have been associated with many processes critical to maintaining central nervous system (CNS) health and contributing to neurological diseases or cancer. In this review, we provide a comprehensive understanding of the LDLR's involvement in common brain tumors, specifically high-grade gliomas, emphasizing the receptors' critical role in the pathophysiology and progression of these tumors due to LDLR's high expression. We delve into LDLR's role in regulating cellular uptake and transport through the brain barrier. Additionally, we highlight LDLR's role in activating several signaling pathways related to tumor proliferation, migration, and invasion, engaging readers with an in-depth understanding of the molecular mechanisms at play. By synthesizing current research findings, this review underscores the significance of LDLR during tumorigenesis and explores its potential as a therapeutic target for high-grade gliomas. The collective insights presented here contribute to a deeper appreciation of LDLR's multifaceted roles and implications for physiological and pathological states, opening new avenues for tumor treatment.
Collapse
Affiliation(s)
- Elisa Mastrantuono
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de MedicinaUniversidade de LisboaPortugal
- Institute for Bioengineering of CataloniaBarcelona Institute of Science and TechnologySpain
| | - Matilde Ghibaudi
- Institute for Bioengineering of CataloniaBarcelona Institute of Science and TechnologySpain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER‐BBN)MadridSpain
| | - Diana Matias
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de MedicinaUniversidade de LisboaPortugal
| | - Giuseppe Battaglia
- Institute for Bioengineering of CataloniaBarcelona Institute of Science and TechnologySpain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER‐BBN)MadridSpain
- Catalan Institution for Research and Advanced StudiesPasseig de Lluís CompanysBarcelonaSpain
| |
Collapse
|
3
|
Liang J, Xie J, He J, Li Y, Wei D, Zhou R, Wei G, Liu X, Chen Q, Li D. Inhibiting lncRNA NEAT1 Increases Glioblastoma Response to TMZ by Reducing Connexin 43 Expression. Cancer Rep (Hoboken) 2024; 7:e70031. [PMID: 39453684 PMCID: PMC11505515 DOI: 10.1002/cnr2.70031] [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: 05/21/2024] [Revised: 08/13/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVES Glioblastoma multiforme (GBM) is considered the most assailant subtype of gliomas, presenting a formidable obstacle because of its inherent resistance to temozolomide (TMZ). This study aimed to characterize the function of lncRNA NEAT1 in facilitating the advancement of gliomas. METHODS The expression level of NEAT1 in glioma tissues and cells was detected by qRT-PCR. RNA interference experiment, cell proliferation assay, FITC/PI detection assay, immunoblotting, bioinformatics prediction, a double luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay, SLDT assay and correlation analysis of clinical samples were performed to explore the regulatory effects of NEAT1, miR-454-3p and Cx43 and their role in malignant progression of GBM. The role of NEAT1 in vivo was investigated by an intracranial tumor formation experiment in mice. RESULTS The results showed that recurring gliomas displayed elevated levels of NEAT1 compared to primary gliomas. The suppression of NEAT1 led to a restoration of sensitivity in GBM cells to TMZ. NEAT1 functioned as a competitive endogenous RNA against miR-454-3p. Connexin 43 was identified as a miR-454-3p target. NEAT1 was found to regulate gap junctional intercellular communication by modulating Connexin 43, thereby impacting the response of GBM cells to TMZ chemotherapy. Downregulation of NEAT1 resulted in enhanced chemosensitivity to TMZ and extended the survival of mice. CONCLUSIONS Overall, these results indicated that the NEAT1/miR-454-3p/Connexin 43 pathway influences GBM cell response to TMZ and could offer a potential new strategy for treating GBM.
Collapse
Affiliation(s)
- Jinxing Liang
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
- Pharmaceutical CollegeGuangxi Medical UniversityNanningChina
| | - Jia‐xiu Xie
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
| | - Junhui He
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
| | - Yi Li
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
| | - Dongmei Wei
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
| | - Rongfei Zhou
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
- Pharmaceutical CollegeGuangxi Medical UniversityNanningChina
| | - Guining Wei
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
| | - Xuehua Liu
- Department of CardiologySir Run Run Hospital of Nanjing Medical UniversityNanjingChina
| | - Qiudan Chen
- Department of Clinical Laboratory, Central Laboratory, Jing'an District Center Hospital of ShanghaiFudan UniversityShanghaiChina
| | - Dongmei Li
- Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards, Research Center of Traditional Chinese Medicine and Ethnic MedicineGuangxi Institute of Chinese Medicine and Pharmaceutical ScienceNanningChina
- School of Chemistry & Pharmaceutical Sciences, State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal ResourcesGuangxi Normal UniversityGuilinChina
| |
Collapse
|
4
|
Sandhanam K, Tamilanban T, Manasa K, Bhattacharjee B. Unlocking novel therapeutic avenues in glioblastoma: Harnessing 4-amino cyanine and miRNA synergy for next-gen treatment convergence. Neuroscience 2024; 553:1-18. [PMID: 38944146 DOI: 10.1016/j.neuroscience.2024.06.032] [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: 03/15/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/01/2024]
Abstract
Glioblastoma (GBM) poses a formidable challenge in oncology due to its aggressive nature and dismal prognosis, with average survival rates around 15 months despite conventional treatments. This review proposes a novel therapeutic strategy for GBM by integrating microRNA (miRNA) therapy with 4-amino cyanine molecules possessing near-infrared (NIR) properties. miRNA holds promise in regulating gene expression, particularly in GBM, making it an attractive therapeutic target. 4-amino cyanine molecules, especially those with NIR properties, have shown efficacy in targeted tumor cell degradation. The combined approach addresses gene expression regulation and precise tumor cell degradation, offering a breakthrough in GBM treatment. Additionally, the review explores noncoding RNAs classification and characteristics, highlighting their role in GBM pathogenesis. Advanced technologies such as antisense oligonucleotides (ASOs), locked nucleic acids (LNAs), and peptide nucleic acids (PNAs) show potential in targeting noncoding RNAs therapeutically, paving the way for precision medicine in GBM. This synergistic combination presents an innovative approach with the potential to advance cancer therapy in the challenging landscape of GBM.
Collapse
Affiliation(s)
- K Sandhanam
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India
| | - T Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India.
| | - K Manasa
- Department of Pharmacology, MNR College of Pharmacy, Sangareddy 502294, Telangana, India
| | - Bedanta Bhattacharjee
- Department of Pharmacology, Girijananda Chowdhury University-Tezpur Campus, 784501 Assam, India
| |
Collapse
|
5
|
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
|
6
|
Xue W, Zhu B, Zhao K, Huang Q, Luo H, Shou Y, Huang Z, Guo H. Targeting LRP6: A new strategy for cancer therapy. Pharmacol Res 2024; 204:107200. [PMID: 38710241 DOI: 10.1016/j.phrs.2024.107200] [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: 12/29/2023] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Targeting specific molecular drivers of tumor growth is a key approach in cancer therapy. Among these targets, the low-density lipoprotein receptor-related protein 6 (LRP6), a vital component of the Wnt signaling pathway, has emerged as an intriguing candidate. As a cell-surface receptor and vital co-receptor, LRP6 is frequently overexpressed in various cancer types, implicating its pivotal role in driving tumor progression. The pursuit of LRP6 as a target for cancer treatment has gained substantial traction, offering a promising avenue for therapeutic intervention. Here, this comprehensive review explores recent breakthroughs in our understanding of LRP6's functions and underlying molecular mechanisms, providing a profound discussion of its involvement in cancer pathogenesis and drug resistance. Importantly, we go beyond discussing LRP6's role in cancer by discussing diverse potential therapeutic approaches targeting this enigmatic protein. These approaches encompass a wide spectrum, including pharmacological agents, natural compounds, non-coding RNAs, epigenetic factors, proteins, and peptides that modulate LRP6 expression or disrupt its interactions. In addition, also discussed the challenges associated with developing LRP6 inhibitors and their advantages over Wnt inhibitors, as well as the drugs that have entered phase II clinical trials. By shedding light on these innovative strategies, we aim to underscore LRP6's significance as a valuable and multifaceted target for cancer treatment, igniting enthusiasm for further research and facilitating translation into clinical applications.
Collapse
Affiliation(s)
- Wei Xue
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China; Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Bo Zhu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning 530021, China
| | - Kaili Zhao
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Qiuju Huang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Hua Luo
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region of China
| | - Yiwen Shou
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Zhaoquan Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules&College of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
7
|
Elgebaly SA, Peacock WF, Christenson RH, Kreutzer DL, Faraag AHI, Sarguos AMM, El-Khazragy N. Integrated Bioinformatics Analysis Confirms the Diagnostic Value of Nourin-Dependent miR-137 and miR-106b in Unstable Angina Patients. Int J Mol Sci 2023; 24:14783. [PMID: 37834231 PMCID: PMC10573268 DOI: 10.3390/ijms241914783] [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: 08/27/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The challenge of rapidly diagnosing myocardial ischemia in unstable angina (UA) patients presenting to the Emergency Department (ED) is due to a lack of sensitive blood biomarkers. This has prompted an investigation into microRNAs (miRNAs) related to cardiac-derived Nourin for potential diagnostic application. The Nourin protein is rapidly expressed in patients with acute coronary syndrome (ACS) (UA and acute myocardial infarction (AMI)). MicroRNAs regulate gene expression through mRNA binding and, thus, may represent potential biomarkers. We initially identified miR-137 and miR-106b and conducted a clinical validation, which demonstrated that they were highly upregulated in ACS patients, but not in healthy subjects and non-ACS controls. Using integrated comprehensive bioinformatics analysis, the present study confirms that the Nourin protein targets miR-137 and miR-106b, which are linked to myocardial ischemia and inflammation associated with ACS. Molecular docking demonstrated robust interactions between the Nourin protein and miR137/hsa-miR-106b, involving hydrogen bonds and hydrophobic interactions, with -10 kcal/mol binding energy. I-TASSER generated Nourin analogs, with the top 10 chosen for structural insights. Antigenic regions and MHCII epitopes within the Nourin SPGADGNGGEAMPGG sequence showed strong binding to HLA-DR/DQ alleles. The Cytoscape network revealed interactions of -miR137/hsa-miR--106b and Phosphatase and tensin homolog (PTEN) in myocardial ischemia. RNA Composer predicted the secondary structure of miR-106b. Schrödinger software identified key Nourin-RNA interactions critical for complex stability. The study identifies miR-137 and miR-106b as potential ACS diagnostic and therapeutic targets. This research underscores the potential of miRNAs targeting Nourin for precision ACS intervention. The analysis leverages RNA Composer, Schrödinger, and I-TASSER tools to explore interactions and structural insights. Robust Nourin-miRNA interactions are established, bolstering the case for miRNA-based interventions in ischemic injury. In conclusion, the study contributes to UA and AMI diagnosis strategies through bioinformatics-guided exploration of Nourin-targeting miRNAs. Supported by comprehensive molecular analysis, the hypoxia-induced miR-137 for cell apoptosis (a marker of cell damage) and the inflammation-induced miR-106b (a marker of inflammation) confirmed their potential clinical use as diagnostic biomarkers. This research reinforces the growing role of miR-137/hsa-miR-106b in the early diagnosis of myocardial ischemia in unstable angina patients.
Collapse
Affiliation(s)
- Salwa A. Elgebaly
- Research & Development, Nour Heart, Inc., Vienna, VA 22180, USA
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA;
| | - W. Frank Peacock
- Department of Emergency Medicine, Baylor College of Medicine, Houston, TX 77057, USA;
| | - Robert H. Christenson
- Department of Pathology, School of Medicine, University of Maryland, Baltimore, MD 2120, USA;
| | - Donald L. Kreutzer
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA;
| | - Ahmed Hassan Ibrahim Faraag
- Department of Botany and Microbiology, Faculty of Science Helwan University, Cairo 11795, Egypt;
- School of Biotechnology, Badr University, Cairo 11829, Egypt
| | | | - Nashwa El-Khazragy
- Department of Clinical Pathology-Hematology, Ain Shams Medical Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
- Department of Genetics and Molecular Biology, Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo11599, Egypt
| |
Collapse
|
8
|
Elshaer SS, Abulsoud AI, Fathi D, Abdelmaksoud NM, Zaki MB, El-Mahdy HA, Ismail A, Elsakka EGE, Abd-Elmawla MA, Abulsoud LA, Doghish AS. miRNAs role in glioblastoma pathogenesis and targeted therapy: Signaling pathways interplay. Pathol Res Pract 2023; 246:154511. [PMID: 37178618 DOI: 10.1016/j.prp.2023.154511] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/28/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
High mortality and morbidity rates and variable clinical behavior are hallmarks of glioblastoma (GBM), the most common and aggressive primary malignant brain tumor. Patients with GBM often have a dismal outlook, even after undergoing surgery, postoperative radiation, and chemotherapy, which has fueled the search for specific targets to provide new insights into the development of contemporary therapies. The ability of microRNAs (miRNAs/miRs) to posttranscriptionally regulate the expression of various genes and silence many target genes involved in cell proliferation, cell cycle, apoptosis, invasion, angiogenesis, stem cell behavior and chemo- and radiotherapy resistance makes them promising candidates as prognostic biomarkers and therapeutic targets or factors to advance GBM therapeutics. Hence, this review is like a crash course in GBM and how miRNAs related to GBM. Here, we will outline the miRNAs whose role in the development of GBM has been established by recent in vitro or in vivo research. Moreover, we will provide a summary of the state of knowledge regarding oncomiRs and tumor suppressor (TS) miRNAs in relation to GBM with an emphasis on their potential applications as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Shereen Saeid Elshaer
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11823, Egypt; Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Doaa Fathi
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Logyna A Abulsoud
- Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
9
|
Sharpe MA, Baskin DS, Johnson RD, Baskin AM. Acquisition of Immune Privilege in GBM Tumors: Role of Prostaglandins and Bile Salts. Int J Mol Sci 2023; 24:3198. [PMID: 36834607 PMCID: PMC9958596 DOI: 10.3390/ijms24043198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Based on the postulate that glioblastoma (GBM) tumors generate anti-inflammatory prostaglandins and bile salts to gain immune privilege, we analyzed 712 tumors in-silico from three GBM transcriptome databases for prostaglandin and bile synthesis/signaling enzyme-transcript markers. A pan-database correlation analysis was performed to identify cell-specific signal generation and downstream effects. The tumors were stratified by their ability to generate prostaglandins, their competency in bile salt synthesis, and the presence of bile acid receptors nuclear receptor subfamily 1, group H, member 4 (NR1H4) and G protein-coupled bile acid receptor 1 (GPBAR1). The survival analysis indicates that tumors capable of prostaglandin and/or bile salt synthesis are linked to poor outcomes. Tumor prostaglandin D2 and F2 syntheses are derived from infiltrating microglia, whereas prostaglandin E2 synthesis is derived from neutrophils. GBMs drive the microglial synthesis of PGD2/F2 by releasing/activating complement system component C3a. GBM expression of sperm-associated heat-shock proteins appears to stimulate neutrophilic PGE2 synthesis. The tumors that generate bile and express high levels of bile receptor NR1H4 have a fetal liver phenotype and a RORC-Treg infiltration signature. The bile-generating tumors that express high levels of GPBAR1 are infiltrated with immunosuppressive microglia/macrophage/myeloid-derived suppressor cells. These findings provide insight into how GBMs generate immune privilege and may explain the failure of checkpoint inhibitor therapy and provide novel targets for treatment.
Collapse
Affiliation(s)
- Martyn A. Sharpe
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
| | - David S. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ryan D. Johnson
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
| | - Alexandra M. Baskin
- Department of Natural Science, Marine Science, Hawaii Pacific University, Honolulu, HI 96801, USA
| |
Collapse
|
10
|
Geng F, Yang F, Liu F, Zhao J, Zhang R, Hu S, Zhang J, Zhang X. A miR-137-XIAP axis contributes to the sensitivity of TRAIL-induced cell death in glioblastoma. Front Oncol 2022; 12:870034. [PMID: 35965517 PMCID: PMC9366219 DOI: 10.3389/fonc.2022.870034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor in the central nervous system with limited therapeutic strategies to prolong the survival rate in clinic. TNF-related apoptosis-inducing ligand (TRAIL)-based strategy has been demonstrated to induce cell death in an extensive spectrum of tumor cells, including GBM, while a considerable proportion of malignant cells are resistant to TRAIL-induced apoptosis. MiR-137 is highly expressed in the brain, but significantly decreases with advanced progression of GBM. However, the functional link between miR-137 and TRAIL-induced apoptosis in GBM cells has not been established. Here, GBM cells were transfected with miR-137, and gene expression levels were examined by qRT-PCR and western blot. Apoptotic cells were measured by Annexin-V staining and TUNEL assay. Our data showed that miR-137 sensitizes GBM cells to the TRAIL-mediated apoptosis. Mechanistically, we identified that XIAP is a bona fide target of miR-137, which is essential for miR-137-regulated sensitivity of TRAIL-induced cell death in GBM cells. Finally, in a xenograft model, combined utilization of miR-137 and TRAIL potently suppresses tumor growth in vivo. Collectively, we demonstrate that a miR-137-XIAP axis is required for the sensitivity of TRAIL-induced cell death and shed a light on the avenue for the treatment of GBM.
Collapse
Affiliation(s)
- Fenghao Geng
- Department of Radiation Medicine, Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Fen Yang
- Department of Neurology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Fang Liu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jianhui Zhao
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Shijie Hu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiao Zhang, ; Jie Zhang, ; Shijie Hu,
| | - Jie Zhang
- Department of Radiation Medicine, Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiao Zhang, ; Jie Zhang, ; Shijie Hu,
| | - Xiao Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Research Office of the Institute of Tropical Medicine, Hainan Hospital of Chinese People's Liberation Army (PLA) General Hospital, Sanya, China
- *Correspondence: Xiao Zhang, ; Jie Zhang, ; Shijie Hu,
| |
Collapse
|
11
|
Mahinfar P, Mansoori B, Rostamzadeh D, Baradaran B, Cho WC, Mansoori B. The Role of microRNAs in Multidrug Resistance of Glioblastoma. Cancers (Basel) 2022; 14:3217. [PMID: 35804989 PMCID: PMC9265057 DOI: 10.3390/cancers14133217] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor that develops from neuroglial stem cells and represents a highly heterogeneous group of neoplasms. These tumors are predominantly correlated with a dismal prognosis and poor quality of life. In spite of major advances in developing novel and effective therapeutic strategies for patients with glioblastoma, multidrug resistance (MDR) is considered to be the major reason for treatment failure. Several mechanisms contribute to MDR in GBM, including upregulation of MDR transporters, alterations in the metabolism of drugs, dysregulation of apoptosis, defects in DNA repair, cancer stem cells, and epithelial-mesenchymal transition. MicroRNAs (miRNAs) are a large class of endogenous RNAs that participate in various cell events, including the mechanisms causing MDR in glioblastoma. In this review, we discuss the role of miRNAs in the regulation of the underlying mechanisms in MDR glioblastoma which will open up new avenues of inquiry for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Parvaneh Mahinfar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - Behnaz Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 175-14115, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran;
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj 7591994799, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (P.M.); (B.M.); (B.B.)
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Behzad Mansoori
- The Wistar Institute, Molecular & Cellular Oncogenesis Program, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Drljača J, Popović A, Bulajić D, Stilinović N, Vidičević Novaković S, Sekulić S, Milenković I, Ninković S, Ljubković M, Čapo I. Diazepam diminishes temozolomide efficacy in the treatment of U87 glioblastoma cell line. CNS Neurosci Ther 2022; 28:1447-1457. [PMID: 35703405 PMCID: PMC9344091 DOI: 10.1111/cns.13889] [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: 02/22/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 11/26/2022] Open
Abstract
AIMS Many patients with glioblastoma (GBM) suffer from comorbid neurological/psychiatric disorders and, therefore, are treated with psychopharmacological agents. Diazepam (DIA) is widely adopted to treat status epilepticus, alleviate anxiety, and inhibit chemotherapy-associated delayed emesis in GBM patients. Even though temozolomide (TMZ) and DIA could be found as possible combination therapy in clinical practice, there are no reports of their combined effects in GBM. Hence, it may be of interest to investigate whether DIA enhances the antitumor efficacy of TMZ in GBM cells. METHODS U87 human GBM was used to examine the effects of combined TMZ and DIA on cell viability, and the oxygen consumption within the cells, in order to evaluate mitochondrial bioenergetic response upon the treatment. RESULTS The cooperative index showed the presence of antagonism between TMZ and DIA, which was confirmed on long-term observation. Moreover, the level of apoptosis after the TMZ treatment was significantly decreased when administered with DIA (p < 0.001). Concomitant use of TMZ and DIA increased the basal cell respiration rate, the oxidative phosphorylation rate, and maximal capacity of mitochondrial electron transport chain, as well as the activities of complexes I and II, vs. TMZ alone (p < 0.001). CONCLUSION Comparing our results with data reported that DIA elicits cell cycle arrest in the G0/G1 phase and favors senescence reveals that DIA diminishes TMZ efficacy in concomitant use in the treatment of GBM. However, due to its great potency to hinder GBM proliferation and metabolism, it could be considered using DIA as maintenance therapy after TMZ cycles.
Collapse
Affiliation(s)
- Jovana Drljača
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Aleksandra Popović
- Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Department of Physiology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Dragica Bulajić
- Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Nebojša Stilinović
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Sašenka Vidičević Novaković
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Slobodan Sekulić
- Department of Neurology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Ivan Milenković
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Srđan Ninković
- Department of Surgery, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Marko Ljubković
- Department of Physiology, University of Split School of Medicine, Split, Croatia
| | - Ivan Čapo
- Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Department of Histology and Embryology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
13
|
Balihodzic A, Prinz F, Dengler MA, Calin GA, Jost PJ, Pichler M. Non-coding RNAs and ferroptosis: potential implications for cancer therapy. Cell Death Differ 2022; 29:1094-1106. [PMID: 35422492 PMCID: PMC9177660 DOI: 10.1038/s41418-022-00998-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/15/2022] Open
Abstract
Ferroptosis is a recently defined form of regulated cell death, which is biochemically and morphologically distinct from traditional forms of programmed cell death such as apoptosis or necrosis. It is driven by iron, reactive oxygen species, and phospholipids that are oxidatively damaged, ultimately resulting in mitochondrial damage and breakdown of membrane integrity. Numerous cellular signaling pathways and molecules are involved in the regulation of ferroptosis, including enzymes that control the cellular redox status. Alterations in the ferroptosis-regulating network can contribute to the development of various diseases, including cancer. Evidence suggests that ferroptosis is commonly suppressed in cancer cells, allowing them to survive and progress. However, cancer cells which are resistant to common chemotherapeutic drugs seem to be highly susceptible to ferroptosis inducers, highlighting the great potential of pharmacologic modulation of ferroptosis for cancer treatment. Non-coding RNAs (ncRNAs) are considered master regulators of various cellular processes, particularly in cancer where they have been implicated in all hallmarks of cancer. Recent work also demonstrated their involvement in the molecular control of ferroptosis. Hence, ncRNA-based therapeutics represent an exciting alternative to modulate ferroptosis for cancer therapy. This review summarizes the ncRNAs implicated in the regulation of ferroptosis in cancer and highlights their underlying molecular mechanisms in the light of potential therapeutic applications.
Collapse
Affiliation(s)
- Amar Balihodzic
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria.,Research Unit "Non-Coding RNAs and Genome Editing in Cancer", Division of Oncology, Medical University of Graz, 8036, Graz, Austria
| | - Felix Prinz
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria.,Research Unit "Non-Coding RNAs and Genome Editing in Cancer", Division of Oncology, Medical University of Graz, 8036, Graz, Austria
| | - Michael A Dengler
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Philipp J Jost
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria.,Medical Department III for Hematology and Oncology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Martin Pichler
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria. .,Research Unit "Non-Coding RNAs and Genome Editing in Cancer", Division of Oncology, Medical University of Graz, 8036, Graz, Austria. .,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Wang J, Huo C, Yin J, Tian L, Ma L, Wang D. Hypermethylation of the Promoter of miR-338-5p Mediates Aberrant Expression of ETS-1 and Is Correlated With Disease Severity Of Astrocytoma Patients. Front Oncol 2021; 11:773644. [PMID: 34858853 PMCID: PMC8632532 DOI: 10.3389/fonc.2021.773644] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
The pro-oncogene ETS-1 (E26 transformation-specific sequence 1) is a key regulator of the proliferation and invasion of cancer cells. The present work examined the correlation of the aberrant expression of ETS-1 with histological or clinical classification of astrocytoma: grade I (pilocytic astrocytoma), grade II (diffuse astrocytoma), grade III (anaplastic astrocytoma), and grade IV (glioblastoma multiforme). MicroRNA, miR-338-5p, was predicted by an online tool (miRDB) to potentially target the 3' untranslated region of ETS-1; this was confirmed by multi-assays, including western blot experiments or the point mutation of the targeting sites of miR-338-5p in ETS-1's 3'untralation region (3'UTR). The expression of miR-338-5p was negatively associated with that of ETS-1 in astrocytoma, and deficiency of miR-338-5p would mediate aberrant expression of ETS-1 in astrocytoma. Mechanistically, hypermethylation of miR-338-5p by DNA methyltransferase 1 (DNMT1) resulted in repression of miR-338-5p expression and the aberrant expression of ETS-1. Knockdown or deactivation of DNMT1 decreased the methylation rate of the miR-338-5p promoter, increased the expression of miR-338-5p, and repressed the expression of ETS-1 in astrocytoma cell lines U251 and U87. These results indicate that hypermethylation of the miR-338-5p promoter by DNMT1 mediates the aberrant expression of ETS-1 related to disease severity of patients with astrocytoma.
Collapse
Affiliation(s)
- Junping Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Sinopharm Tongmei General Hospital, Datong, China
| | - Cheng Huo
- Department of Neurosurgery, The Sinopharm Tongmei General Hospital, Datong, China
| | - Jinzhu Yin
- Department of Neurosurgery, The Sinopharm Tongmei General Hospital, Datong, China
| | - Lixia Tian
- Department of Neurosurgery, The Sinopharm Tongmei General Hospital, Datong, China
| | - Lili Ma
- Department of Neurology, The Yantaishan Hospital, Yantai, China
| | - Dongsheng Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
15
|
MicroRNA-137 Inhibited Hypoxia-Induced Proliferation of Pulmonary Artery Smooth Muscle Cells by Targeting Calpain-2. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2202888. [PMID: 34513987 PMCID: PMC8426064 DOI: 10.1155/2021/2202888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 01/22/2023]
Abstract
The proliferation of pulmonary artery smooth muscle cells (PASMCs) is an important cause of pulmonary vascular remodeling in pulmonary hypertension (PH). It has been reported that miR-137 inhibits the proliferation of tumor cells. However, whether miR-137 is involved in PH remains unclear. In this study, male Sprague-Dawley rats were subjected to 10% O2 for 3 weeks to establish PH, and rat primary PASMCs were treated with hypoxia (3% O2) for 48 h to induce cell proliferation. The effect of miR-137 on PASMC proliferation and calpain-2 expression was assessed by transfecting miR-137 mimic and inhibitor. The effect of calpain-2 on PASMC proliferation was assessed by transfecting calpain-2 siRNA. The present study found for the first time that miR-137 was downregulated in pulmonary arteries of hypoxic PH rats and in hypoxia-treated PASMCs. miR-137 mimic inhibited hypoxia-induced PASMC proliferation and upregulation of calpain-2 expression in PASMCs. Furthermore, miR-137 inhibitor induced the proliferation of PASMCs under normoxia, and knockdown of calpain-2 mRNA by siRNA significantly inhibited hypoxia-induced proliferation of PASMCs. Our study demonstrated that hypoxia-induced downregulation of miR-137 expression promoted the proliferation of PASMCs by targeting calpain-2, thereby potentially resulting in pulmonary vascular remodeling in hypoxic PH.
Collapse
|
16
|
He X, Sun H, Jiang Q, Chai Y, Li X, Wang Z, Zhu B, You S, Li B, Hao J, Xin S. Hsa-miR-4277 Decelerates the Metabolism or Clearance of Sorafenib in HCC Cells and Enhances the Sensitivity of HCC Cells to Sorafenib by Targeting cyp3a4. Front Oncol 2021; 11:735447. [PMID: 34381736 PMCID: PMC8350395 DOI: 10.3389/fonc.2021.735447] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/12/2021] [Indexed: 12/18/2022] Open
Abstract
Increasing evidence has shown that the metabolism and clearance of molecular targeted agents, such as sorafenib, plays an important role in mediating the resistance of HCC cells to these agents. Metabolism of sorafenib is performed by oxidative metabolism, which is initially mediated by CYP3A4. Thus, targeting CYP3A4 is a promising approach to enhance the sensitivity of HCC cells to chemotherapeutic agents. In the present work, we examined the association between CYP3A4 and the prognosis of HCC patients receiving sorafenib. Using the online tool miRDB, we predicted that has-microRNA-4277 (miR-4277), an online miRNA targets the 3’UTR of the transcript of cyp3a4. Furthermore, overexpression of miR-4277 in HCC cells repressed the expression of CYP3A4 and reduced the elimination of sorafenib in HCC cells. Moreover, miR-4277 enhanced the sensitivity of HCC cells to sorafenib in vitro and in vivo. Therefore, our results not only expand our understanding of CYP3A4 regulation in HCC, but also provide evidence for the use of miR-4277 as a potential therapeutic in advanced HCC.
Collapse
Affiliation(s)
- Xi He
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Department of Liver Disease of Chinese PLA General Hospital, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Huiwei Sun
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Institute of Infectious Disease, Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qiyu Jiang
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Institute of Infectious Disease, Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yantao Chai
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Institute of Infectious Disease, Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaojuan Li
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Institute of Infectious Disease, Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhijie Wang
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Institute of Infectious Disease, Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Bing Zhu
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Department of Liver Disease of Chinese PLA General Hospital, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shaoli You
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Department of Liver Disease of Chinese PLA General Hospital, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Boan Li
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Department of Clinical Laboratory, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junfeng Hao
- Department of Nephrology, Jin Qiu Hospital of Liaoning Province/Geriatric Hospital of Liaoning Province, Shenyang, China
| | - Shaojie Xin
- Chinese People's Liberation Army (PLA) Medical School, Beijing, China.,Department of Liver Disease of Chinese PLA General Hospital, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|