1
|
Xue J, Zhang J, Zhu J. Unraveling molecular signatures and prognostic biomarkers in glioblastoma: a comprehensive study on treatment resistance and personalized strategies. Discov Oncol 2024; 15:743. [PMID: 39630160 PMCID: PMC11618281 DOI: 10.1007/s12672-024-01649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive primary brain tumor with limited treatment success and poor prognosis. Despite surgical resection and adjuvant therapies, GBM often recurs, and resistance to radiotherapy and temozolomide presents significant challenges. This study aimed to elucidate molecular signatures associated with treatment responses, identify potential biomarkers, and enhance personalized treatment strategies for GBM. METHODS We conducted a comprehensive analysis using the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. The GEO dataset (GSE206225) was used to identify differentially expressed genes (DEGs) between radiation-sensitive/resistant and temozolomide-sensitive/resistant GBM samples. TCGA data were utilized for subsequent analyses, including Lasso-Cox regression, risk score model construction, Kaplan-Meier survival analysis, and gene set enrichment analysis (GSEA). Hub genes were identified through survival analysis, and a gene prognostic nomogram was developed. Additionally, validation of the three-gene risk signature through multiple external cohorts and validation of protein expression levels were performed. RESULTS DEG analysis identified 111 genes associated with chemoradiotherapy resistance, providing insights into the complex landscape of GBM treatment response. The risk score model effectively stratified patients, showing significant differences in overall survival and progression-free survival. GSEA offered a deeper understanding of pathway activities, emphasizing the intricate molecular mechanisms involved. NNAT, IGFBP6, and CYGB were identified as hub genes, and a gene prognostic nomogram demonstrated predictive accuracy. CONCLUSION This study sheds light on the molecular intricacies governing GBM treatment response. The identified hub genes and the gene prognostic nomogram offer valuable tools for predicting patient outcomes and guiding personalized treatment strategies. These findings contribute to advancing our understanding of GBM biology and may pave the way for improved clinical management.
Collapse
Affiliation(s)
- Jinmin Xue
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
- Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jie Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
- Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jing Zhu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China.
- Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
2
|
Han M, Kakar M, Li W, Iqbal I, Hu X, Liu Y, Tang Q, Sun L, Shakir Y, Liu T. Targeting MDM2-p53 interaction in Glioblastoma: Transcriptomic analysis and Peptide-Based inhibition strategy. Bioorg Chem 2024; 150:107620. [PMID: 38991490 DOI: 10.1016/j.bioorg.2024.107620] [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: 05/13/2024] [Revised: 06/26/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
MDM2 is a gene that encodes a protein involved in cell survival, growth, and DNA repair. It has been implicated in the development and progression of glioblastoma (GBM). Inhibition of the MDM2-p53 interaction has emerged as a promising strategy for treating GBM. In this study, we performed comprehensive transcriptomic expression analysis from diverse datasets and observed MDM2 overexpression in a subset of GBM cases. MDM2 negatively regulates the major onco-suppressor p53. The interaction between MDM2 and p53 is a promising target for cancer therapy, as it can trigger p53-mediated cell death in response to different stress conditions, such as oncogene activation or DNA damage. In this study, we have identified a peptide-based inhibition of MDM2 as a therapeutic strategy for GBM. We have further validated the stability of the MDM2-peptide interaction using a molecular structural dynamics approach. The major trajectories, including root mean square of deviation (RMSD), root mean square of fluctuation (RMSF), and radius of gyration (RoG), indicate that the candidate peptides have a more stable binding compared to the native ligand and control drug. The stability of the binding interaction was further estimated by MMGBSA analysis, which also suggests that MDM2 has a stable binding with both peptide molecules. Based on these results, peptides P-1843 and P-3837 could be tested further for experimental validation to confirm their targeted inhibition of MDM-2. This approach could provide a highly selective and efficient inhibitor with potentially fewer side effects and less toxicity compared to small drug-based molecules.
Collapse
Affiliation(s)
- Manman Han
- Department of General Surgery, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Zhejiang Province, China
| | - Mohibullah Kakar
- Faculty of Marine Sciences, Lasbela University of Agriculture, Water and Marine Sciences (LUAWMS), Uthal, Balochistan, Pakistan
| | - Wei Li
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Imran Iqbal
- Department of PLR, Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany
| | - Xiaolin Hu
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Yiting Liu
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Qing Tang
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Lizhu Sun
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Yasmeen Shakir
- Department of Biochemistry, Hazara University, Mansehra, KPK, Pakistan.
| | - Tiantian Liu
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China.
| |
Collapse
|
3
|
Alom MW, Jibon MDK, Faruqe MO, Rahman MS, Akter F, Ali A, Rahman MM. Integrated Gene Expression Data-Driven Identification of Molecular Signatures, Prognostic Biomarkers, and Drug Targets for Glioblastoma. BIOMED RESEARCH INTERNATIONAL 2024; 2024:6810200. [PMID: 39184354 PMCID: PMC11343637 DOI: 10.1155/2024/6810200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024]
Abstract
Glioblastoma (GBM) is a highly prevalent and deadly brain tumor with high mortality rates, especially among adults. Despite extensive research, the underlying mechanisms driving its progression remain poorly understood. Computational analysis offers a powerful approach to explore potential prognostic biomarkers, drug targets, and therapeutic agents for GBM. In this study, we utilized three gene expression datasets from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) associated with GBM progression. Our goal was to uncover key molecular players implicated in GBM pathogenesis and potential avenues for targeted therapy. Analysis of the gene expression datasets revealed a total of 78 common DEGs that are potentially involved in GBM progression. Through further investigation, we identified nine hub DEGs that are highly interconnected in protein-protein interaction (PPI) networks, indicating their central role in GBM biology. Gene Ontology (GO) and pathway enrichment analyses provided insights into the biological processes and immunological pathways influenced by these DEGs. Among the nine identified DEGs, survival analysis demonstrated that increased expression of GMFG correlated with decreased patient survival rates in GBM, suggesting its potential as a prognostic biomarker and preventive target for GBM. Furthermore, molecular docking and ADMET analysis identified two compounds from the NIH clinical collection that showed promising interactions with the GMFG protein. Besides, a 100 nanosecond molecular dynamics (MD) simulation evaluated the conformational changes and the binding strength. Our study highlights the potential of GMFG as both a prognostic biomarker and a therapeutic target for GBM. The identification of GMFG and its associated pathways provides valuable insights into the molecular mechanisms driving GBM progression. Moreover, the identification of candidate compounds with potential interactions with GMFG offers exciting possibilities for targeted therapy development. However, further laboratory experiments are required to validate the role of GMFG in GBM pathogenesis and to assess the efficacy of potential therapeutic agents targeting this molecule.
Collapse
Affiliation(s)
- Md. Wasim Alom
- Department of Genetic Engineering and BiotechnologyUniversity of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Delowar Kobir Jibon
- Department of Genetic Engineering and BiotechnologyUniversity of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Omar Faruqe
- Department of Computer Science and EngineeringUniversity of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Siddikur Rahman
- Department of Genetic Engineering and BiotechnologyUniversity of Rajshahi, Rajshahi 6205, Bangladesh
| | - Farzana Akter
- Department of Genetic Engineering and BiotechnologyUniversity of Rajshahi, Rajshahi 6205, Bangladesh
| | - Aslam Ali
- Department of Genetic Engineering and BiotechnologyUniversity of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Motiur Rahman
- Department of Genetic Engineering and BiotechnologyUniversity of Rajshahi, Rajshahi 6205, Bangladesh
| |
Collapse
|
4
|
Trivic A, Milovanovic J, Kablar D, Tomic A, Folic M, Jotic A, Tomanovic N, Tomic AM, Djoric I, Jankovic M. Friend or Foe? Exploring the Role of Cytomegalovirus (HCMV) Infection in Head and Neck Tumors. Biomedicines 2024; 12:872. [PMID: 38672226 PMCID: PMC11048144 DOI: 10.3390/biomedicines12040872] [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: 03/05/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Although not regarded as an oncogenic pathogen, the human cytomegalovirus (HCMV) has been associated with a wide array of malignancies. Conversely, a number of studies report on possible anti-tumor properties of the virus, apparently mediated via HCMV-galvanized T-cell tumor killing; these were recently being investigated in clinical trials for the purposes of anti-cancer treatment by means of dendritic cell vaccines and HCMV-specific cytotoxic T cells. In the present study, we have analyzed the relation between a complement of head-and-neck tumors and HCMV infection across 73 countries worldwide using Spearman correlation, univariate and multivariate regression analysis. Intriguingly, HCMV was found to be pro-oncogenic in patients with nasopharyngeal carcinoma; contrarywise, the virus manifested an inverse (i.e., anti-tumor) association with the tumors of the lip/oral region and the salivary glands. Although this putative protective effect was noted initially for thyroid neoplasia and hypopharyngeal tumors as well, after multivariate regression analysis the connection did not hold. There was no association between laryngeal cancer and HCMV infection. It would appear that, depending on the tissue, HCMV may exert both protective and oncogenic effects. The globally observed protective feature of the virus could potentially be utilized in future therapeutic approaches for salivary tumors and neoplasia in the lip/oral region. As correlation does not necessarily imply causation, more in-depth molecular analyses from comprehensive clinical studies are warranted to substantiate our findings.
Collapse
Affiliation(s)
- Aleksandar Trivic
- Clinic for Otorhinolaryngology and Maxillofacial Surgery, University Clinical Center of Serbia, 2 Pasterova Street, 11000 Belgrade, Serbia; (A.T.); (J.M.); (M.F.); (A.J.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
| | - Jovica Milovanovic
- Clinic for Otorhinolaryngology and Maxillofacial Surgery, University Clinical Center of Serbia, 2 Pasterova Street, 11000 Belgrade, Serbia; (A.T.); (J.M.); (M.F.); (A.J.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
| | - Djurdjina Kablar
- Department for Pathology, Pathohistology and Medical Cytology, University Clinical Centre of Serbia, 11000 Belgrade, Serbia;
| | - Ana Tomic
- Center for Radiology Imaging, University Clinical Center of Serbia, 2 Pasterova Street, 11000 Belgrade, Serbia;
| | - Miljan Folic
- Clinic for Otorhinolaryngology and Maxillofacial Surgery, University Clinical Center of Serbia, 2 Pasterova Street, 11000 Belgrade, Serbia; (A.T.); (J.M.); (M.F.); (A.J.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
| | - Ana Jotic
- Clinic for Otorhinolaryngology and Maxillofacial Surgery, University Clinical Center of Serbia, 2 Pasterova Street, 11000 Belgrade, Serbia; (A.T.); (J.M.); (M.F.); (A.J.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
| | - Nada Tomanovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
- Institute of Pathology, 1 Dr. Subotica Street, 11000 Belgrade, Serbia
| | - Ana Marija Tomic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
- Institute of Pathology, 1 Dr. Subotica Street, 11000 Belgrade, Serbia
| | - Igor Djoric
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
- Clinic of Neurosurgery, University Clinical Center of Serbia, Institute of Radiology, 4 Dr. Koste Todorovića Street, 11000 Belgrade, Serbia
| | - Marko Jankovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.T.); (A.M.T.); (I.D.)
- Department of Virology, Institute of Microbiology and Immunology, 1 Dr. Subotica Street, 11000 Belgrade, Serbia
| |
Collapse
|
5
|
Jankovic M, Knezevic T, Tomic A, Milicevic O, Jovanovic T, Djunic I, Mihaljevic B, Knezevic A, Todorovic-Balint M. Human Cytomegalovirus Oncoprotection across Diverse Populations, Tumor Histologies, and Age Groups: The Relevance for Prospective Vaccinal Therapy. Int J Mol Sci 2024; 25:3741. [PMID: 38612552 PMCID: PMC11012084 DOI: 10.3390/ijms25073741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
The oncogenicity of the human cytomegalovirus (CMV) is currently being widely debated. Most recently, mounting clinical evidence suggests an anti-cancer effect via CMV-induced T cell-mediated tumor destruction. However, the data were mostly obtained from single-center studies and in vitro experiments. Broad geographic coverage is required to offer a global perspective. Our study examined the correlation between country-specific CMV seroprevalence (across 73 countries) and the age-standardized incidence rate (of 34 invasive tumors). The populations studied were stratified according to decadal age periods as the immunologic effects of CMV seropositivity may depend upon age at initial infection. The International Agency for Research on Cancer of the World Health Organization (IARC WHO) database was used. The multivariate linear regression analysis revealed a worldwide inverse correlation between CMV seroprevalence and the incidences of 62.8% tumors. Notably, this inverse link persists for all cancers combined (Spearman's ρ = -0.732, p < 0.001; β = -0.482, p < 0.001, adjusted R2 = 0.737). An antithetical and significant correlation was also observed in particular age groups for the vast majority of tumors. Our results corroborate the conclusions of previous studies and indicate that this oncopreventive phenomenon holds true on a global scale. It applies to a wide spectrum of cancer histologies, additionally supporting the idea of a common underlying mechanism-CMV-stimulated T cell tumor targeting. Although these results further advance the notion of CMV-based therapies, in-depth investigation of host-virus interactions is still warranted.
Collapse
Affiliation(s)
- Marko Jankovic
- Department of Virology, Institute of Microbiology and Immunology, 1 Dr Subotica Street, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
| | - Tara Knezevic
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
| | - Ana Tomic
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
| | - Ognjen Milicevic
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
- Institute of Medical Statistics and Informatics, 15 Dr Subotica Street, 11000 Belgrade, Serbia
| | - Tanja Jovanovic
- Institute for Biocides and Medical Ecology, 16 Trebevicka Street, 11000 Belgrade, Serbia;
| | - Irena Djunic
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovica Street, 11000 Belgrade, Serbia
| | - Biljana Mihaljevic
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovica Street, 11000 Belgrade, Serbia
| | - Aleksandra Knezevic
- Department of Virology, Institute of Microbiology and Immunology, 1 Dr Subotica Street, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
| | - Milena Todorovic-Balint
- Faculty of Medicine, University of Belgrade, 8 Dr Subotica Street, 11000 Belgrade, Serbia; (T.K.); (A.T.); (O.M.); (I.D.); (B.M.); (M.T.-B.)
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovica Street, 11000 Belgrade, Serbia
| |
Collapse
|
6
|
Zhang W, Dang R, Liu H, Dai L, Liu H, Adegboro AA, Zhang Y, Li W, Peng K, Hong J, Li X. Machine learning-based investigation of regulated cell death for predicting prognosis and immunotherapy response in glioma patients. Sci Rep 2024; 14:4173. [PMID: 38378721 PMCID: PMC10879095 DOI: 10.1038/s41598-024-54643-3] [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: 12/06/2023] [Accepted: 02/14/2024] [Indexed: 02/22/2024] Open
Abstract
Glioblastoma is a highly aggressive and malignant type of brain cancer that originates from glial cells in the brain, with a median survival time of 15 months and a 5-year survival rate of less than 5%. Regulated cell death (RCD) is the autonomous and orderly cell death under genetic control, controlled by precise signaling pathways and molecularly defined effector mechanisms, modulated by pharmacological or genetic interventions, and plays a key role in maintaining homeostasis of the internal environment. The comprehensive and systemic landscape of the RCD in glioma is not fully investigated and explored. After collecting 18 RCD-related signatures from the opening literature, we comprehensively explored the RCD landscape, integrating the multi-omics data, including large-scale bulk data, single-cell level data, glioma cell lines, and proteome level data. We also provided a machine learning framework for screening the potentially therapeutic candidates. Here, based on bulk and single-cell sequencing samples, we explored RCD-related phenotypes, investigated the profile of the RCD, and developed an RCD gene pair scoring system, named RCD.GP signature, showing a reliable and robust performance in predicting the prognosis of glioblastoma. Using the machine learning framework consisting of Lasso, RSF, XgBoost, Enet, CoxBoost and Boruta, we identified seven RCD genes as potential therapeutic targets in glioma and verified that the SLC43A3 highly expressed in glioma grades and glioma cell lines through qRT-PCR. Our study provided comprehensive insights into the RCD roles in glioma, developed a robust RCD gene pair signature for predicting the prognosis of glioma patients, constructed a machine learning framework for screening the core candidates and identified the SLC43A3 as an oncogenic role and a prediction biomarker in glioblastoma.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Ruiyue Dang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongyi Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Luohuan Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Hongwei Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Abraham Ayodeji Adegboro
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Yihao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Wang Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Kang Peng
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jidong Hong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China.
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
7
|
Shah S. Novel Therapies in Glioblastoma Treatment: Review of Glioblastoma; Current Treatment Options; and Novel Oncolytic Viral Therapies. Med Sci (Basel) 2023; 12:1. [PMID: 38249077 PMCID: PMC10801585 DOI: 10.3390/medsci12010001] [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: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
One of the most prevalent primary malignant brain tumors is glioblastoma (GB). About 6 incidents per 100,000 people are reported annually. Most frequently, these tumors are linked to a poor prognosis and poor quality of life. There has been little advancement in the treatment of GB. In recent years, some innovative medicines have been tested for the treatment of newly diagnosed cases of GB and recurrent cases of GB. Surgery, radiotherapy, and alkylating chemotherapy are all common treatments for GB. A few of the potential alternatives include immunotherapy, tumor-treating fields (TTFs), and medications that target specific cellular receptors. To provide new multimodal therapies that focus on the molecular pathways implicated in tumor initiation and progression in GB, novel medications, delivery technologies, and immunotherapy approaches are being researched. Of these, oncolytic viruses (OVs) are among the most recent. Coupling OVs with certain modern treatment approaches may have significant benefits for GB patients. Here, we discuss several OVs and how they work in conjunction with other therapies, as well as virotherapy for GB. The study was based on the PRISMA guidelines. Systematic retrieval of information was performed on PubMed. A total of 307 articles were found in a search on oncolytic viral therapies for glioblastoma. Out of these 83 articles were meta-analyses, randomized controlled trials, reviews, and systematic reviews. A total of 42 articles were from the years 2018 to 2023. Appropriate studies were isolated, and important information from each of them was understood and entered into a database from which the information was used in this article. One of the most prevalent malignant brain tumors is still GB. Significant promise and opportunity exist for oncolytic viruses in the treatment of GB and in boosting immune response. Making the most of OVs in the treatment of GB requires careful consideration and evaluation of a number of its application factors.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
8
|
Kciuk M, Yahya EB, Mohamed MMI, Abdulsamad MA, Allaq AA, Gielecińska A, Kontek R. Insights into the Role of LncRNAs and miRNAs in Glioma Progression and Their Potential as Novel Therapeutic Targets. Cancers (Basel) 2023; 15:3298. [PMID: 37444408 DOI: 10.3390/cancers15133298] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Accumulating evidence supports that both long non-coding and micro RNAs (lncRNAs and miRNAs) are implicated in glioma tumorigenesis and progression. Poor outcome of gliomas has been linked to late-stage diagnosis and mostly ineffectiveness of conventional treatment due to low knowledge about the early stage of gliomas, which are not possible to observe with conventional diagnostic approaches. The past few years witnessed a revolutionary advance in biotechnology and neuroscience with the understanding of tumor-related molecules, including non-coding RNAs that are involved in the angiogenesis and progression of glioma cells and thus are used as prognostic biomarkers as well as novel therapeutic targets. The emerging research on lncRNAs and miRNAs highlights their crucial role in glioma progression, offering new insights into the disease. These non-coding RNAs hold significant potential as novel therapeutic targets, paving the way for innovative treatment approaches against glioma. This review encompasses a comprehensive discussion about the role of lncRNAs and miRNAs in gene regulation that is responsible for the promotion or the inhibition of glioma progression and collects the existing links between these key cancer-related molecules.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Muhanad A Abdulsamad
- Department of Molecular Biology, Faculty of Science, Sabratha University, Sabratha 00218, Libya
| | - Abdulmutalib A Allaq
- Faculty of Applied Science, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
9
|
Fazlalipour M, Ghoreshi ZAS, Molaei HR, Arefinia N. The Role of DNA Viruses in Human Cancer. Cancer Inform 2023; 22:11769351231154186. [PMID: 37363356 PMCID: PMC10286548 DOI: 10.1177/11769351231154186] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/03/2023] [Indexed: 06/28/2023] Open
Abstract
This review discusses the possible involvement of infections-associated cancers in humans, with virus infections contributing 15% to 20% of total cancer cases in humans. DNA virus encoded proteins interact with host cellular signaling pathways and control proliferation, cell death and genomic integrity viral oncoproteins are known to bind cellular Deubiquitinates (DUBs) such as cyclindromatosis tumor suppressor, ubiquitin-specific proteases 7, 11, 15 and 20, and A-20 to improve their intracellular stability and cellular signaling pathways and finally transformation. Human papillomaviruses (cervical carcinoma, oral cancer and laryngeal cancer); human polyomaviruses (mesotheliomas, brain tumors); Epstein-Barr virus (B-cell lymphoproliferative diseases and nasopharyngeal carcinoma); Kaposi's Sarcoma Herpesvirus (Kaposi's Sarcoma and primary effusion lymphomas); hepatitis B (hepatocellular carcinoma (HCC)) cause up to 20% of malignancies around the world.
Collapse
Affiliation(s)
- Mehdi Fazlalipour
- WHO Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran (IPI), Tehran, Iran
- Research Center for Emerging and Reemerging Infectious diseases, Pasteur Institute of Iran (IPI), Tehran, Iran
| | | | - Hamid Reza Molaei
- Department of Medical Bacteriology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nasir Arefinia
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| |
Collapse
|
10
|
Hamad A, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recent Developments in Glioblastoma Therapy: Oncolytic Viruses and Emerging Future Strategies. Viruses 2023; 15:547. [PMID: 36851761 PMCID: PMC9958853 DOI: 10.3390/v15020547] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma is the most aggressive form of malignant brain tumor. Standard treatment protocols and traditional immunotherapy are poorly effective as they do not significantly increase the long-term survival of glioblastoma patients. Oncolytic viruses (OVs) may be an effective alternative approach. Combining OVs with some modern treatment options may also provide significant benefits for glioblastoma patients. Here we review virotherapy for glioblastomas and describe several OVs and their combination with other therapies. The personalized use of OVs and their combination with other treatment options would become a significant area of research aiming to develop the most effective treatment regimens for glioblastomas.
Collapse
Affiliation(s)
- Azzam Hamad
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
11
|
Pandeya A, Khalko RK, Singh S, Kumar M, Gosipatala SB. Hcmv-miR-UL148D regulates the staurosporine-induced apoptosis by targeting the Endoplasmic Reticulum to Nucleus signaling 1(ERN1). PLoS One 2022; 17:e0275072. [PMID: 36156601 PMCID: PMC9512192 DOI: 10.1371/journal.pone.0275072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022] Open
Abstract
The propensity of viruses to co-opt host cellular machinery by reprogramming the host's RNA-interference machinery has been a major focus of research, however, regulation of host defense mechanisms by virus-encoded miRNA, is an additional regulatory realm gaining momentum in the arena of host-viral interactions. The Human Cytomegalovirus (HCMV) miRNAs, regulate many cellular pathways alone or in concordance with HCMV proteins, thereby paving a conducive environment for successful infection in the human host. We show that HCMV miRNA, hcmv-miR-UL148D inhibits staurosporine-induced apoptosis in HEK293T cells. We establish that ERN1 mRNA is a bonafide target of hcmv-miR-UL148D and its encoded protein IRE1α is translationally repressed by the overexpression of hcmv-miR-UL148D resulting in the attenuation of apoptosis. Unlike the host microRNA seed sequence (6-8 nucleotides), hcmv-miR-UL148D has long complementarity to 3' UTR of ERN1 mRNA resulting in mRNA degradation. The repression of IRE1α by the hcmv-miR-UL148D further downregulates Xbp1 splicing and c-Jun N-terminal kinase phosphorylation thus regulating ER-stress and ER-stress induced apoptotic pathways. Strikingly, depletion of ERN1 attenuates staurosporine-induced apoptosis which further suggests that hcmv-miR-UL148D functions through regulation of its target ERN1. These results uncover a role for hcmv-miR-UL148D and its target ERN1 in regulating ER stress-induced apoptosis.
Collapse
Affiliation(s)
- Abhishek Pandeya
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Raj Kumar Khalko
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Sukhveer Singh
- Developmental Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, Uttar Pradesh, India
| | - Manish Kumar
- National Heart Lung and Blood Institute, National Institute of Health, Bethesda, Maryland, United States of America
| | - Sunil Babu Gosipatala
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
12
|
Yang T, Liu D, Fang S, Ma W, Wang Y. Cytomegalovirus and Glioblastoma: A Review of the Biological Associations and Therapeutic Strategies. J Clin Med 2022; 11:jcm11175221. [PMID: 36079151 PMCID: PMC9457369 DOI: 10.3390/jcm11175221] [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] [Received: 07/04/2022] [Revised: 08/07/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma is the most common and aggressive malignancy in the adult central nervous system. Cytomegalovirus (CMV) plays a crucial role in the pathogenesis and treatment of glioblastoma. We reviewed the epidemiology of CMV in gliomas, the mechanism of CMV-related carcinogenesis, and its therapeutic strategies, offering further clinical practice insights. To date, the CMV infection rate in glioblastoma is controversial, while mounting studies have suggested a high infection rate. The carcinogenesis mechanism of CMV has been investigated in relation to various aspects, including oncomodulation, oncogenic features, tumor microenvironment regulation, epithelial–mesenchymal transition, and overall immune system regulation. In clinical practice, the incidence of CMV-associated encephalopathy is high, and CMV-targeting treatment bears both anti-CMV and anti-tumor effects. As the major anti-CMV treatment, valganciclovir has demonstrated a promising survival benefit in both newly diagnosed and recurrent glioblastoma as an adjuvant therapy, regardless of surgery and the MGMT promoter methylation state. Immunotherapy, including DC vaccines and adoptive CMV-specific T cells, is also under investigation, and preliminary results have been promising. There are still questions regarding the significance of CMV infection and the carcinogenic mechanism of CMV. Meanwhile, studies have demonstrated the clinical benefits of anti-CMV therapy in glioblastoma. Therefore, anti-CMV therapies are worthy of further recognition and investigation.
Collapse
Affiliation(s)
- Tianrui Yang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Delin Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Shiyuan Fang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Correspondence: (W.M.); (Y.W.); Tel.: +86-137-0136-4566 (W.M.); +86-153-1186-0318 (Y.W.)
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Correspondence: (W.M.); (Y.W.); Tel.: +86-137-0136-4566 (W.M.); +86-153-1186-0318 (Y.W.)
| |
Collapse
|
13
|
Zheng H, Savitz J. Effect of Cytomegalovirus Infection on the Central Nervous System: Implications for Psychiatric Disorders. Curr Top Behav Neurosci 2022; 61:215-241. [PMID: 35505056 DOI: 10.1007/7854_2022_361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytomegalovirus (CMV) is a common herpesvirus that establishes lifelong latent infections and interacts extensively with the host immune system, potentially contributing to immune activation and inflammation. Given its proclivity for infecting the brain and its reactivation by inflammatory stimuli, CMV is well known for causing central nervous system complications in the immune-naïve (e.g., in utero) and in the immunocompromised (e.g., in neonates, individuals receiving transplants or cancer chemotherapy, or people living with HIV). However, its potentially pathogenic role in diseases that are characterized by more subtle immune dysregulation and inflammation such as psychiatric disorders is still a matter of debate. In this chapter, we briefly summarize the pathogenic role of CMV in immune-naïve and immunocompromised populations and then review the evidence (i.e., epidemiological studies, serological studies, postmortem studies, and recent neuroimaging studies) for a link between CMV infection and psychiatric disorders with a focus on mood disorders and schizophrenia. Finally, we discuss the potential mechanisms through which CMV may cause CNS dysfunction in the context of mental disorders and conclude with a summary of the current state of play as well as potential future research directions in this area.
Collapse
Affiliation(s)
- Haixia Zheng
- Laureate Institute for Brain Research, Tulsa, OK, USA.
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA.,Oxley College of Health Sciences, The University of Tulsa, Tulsa, OK, USA
| |
Collapse
|
14
|
Daei Sorkhabi A, Sarkesh A, Saeedi H, Marofi F, Ghaebi M, Silvestris N, Baradaran B, Brunetti O. The Basis and Advances in Clinical Application of Cytomegalovirus-Specific Cytotoxic T Cell Immunotherapy for Glioblastoma Multiforme. Front Oncol 2022; 12:818447. [PMID: 35515137 PMCID: PMC9062077 DOI: 10.3389/fonc.2022.818447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/24/2022] [Indexed: 01/28/2023] Open
Abstract
A high percentage of malignant gliomas are infected by human cytomegalovirus (HCMV), and the endogenous expression of HCMV genes and their products are found in these tumors. HCMV antigen expression and its implications in gliomagenesis have emerged as a promising target for adoptive cellular immunotherapy (ACT) strategies in glioblastoma multiforme (GB) patients. Since antigen-specific T cells in the tumor microenvironments lack efficient anti-tumor immune response due to the immunosuppressive nature of glioblastoma, CMV-specific ACT relies on in vitro expansion of CMV-specific CD8+ T cells employing immunodominant HCMV antigens. Given the fact that several hurdles remain to be conquered, recent clinical trials have outlined the feasibility of CMV-specific ACT prior to tumor recurrence with minimal adverse effects and a substantial improvement in median overall survival and progression-free survival. This review discusses the role of HCMV in gliomagenesis, disease prognosis, and recent breakthroughs in harnessing HCMV-induced immunogenicity in the GB tumor microenvironment to develop effective CMV-specific ACT.
Collapse
Affiliation(s)
- Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Ghaebi
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
| |
Collapse
|
15
|
Rong L, Li N, Zhang Z. Emerging therapies for glioblastoma: current state and future directions. J Exp Clin Cancer Res 2022; 41:142. [PMID: 35428347 PMCID: PMC9013078 DOI: 10.1186/s13046-022-02349-7] [Citation(s) in RCA: 232] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/26/2022] [Indexed: 04/15/2023] Open
Abstract
Glioblastoma (GBM) is the most common high-grade primary malignant brain tumor with an extremely poor prognosis. Given the poor survival with currently approved treatments for GBM, new therapeutic strategies are urgently needed. Advances in decades of investment in basic science of glioblastoma are rapidly translated into innovative clinical trials, utilizing improved genetic and epigenetic profiling of glioblastoma as well as the brain microenvironment and immune system interactions. Following these encouraging findings, immunotherapy including immune checkpoint blockade, chimeric antigen receptor T (CAR T) cell therapy, oncolytic virotherapy, and vaccine therapy have offered new hope for improving GBM outcomes; ongoing studies are using combinatorial therapies with the aim of minimizing adverse side-effects and augmenting antitumor immune responses. In addition, techniques to overcome the blood-brain barrier (BBB) for targeted delivery are being tested in clinical trials in patients with recurrent GBM. Here, we set forth the rationales for these promising therapies in treating GBM, review the potential novel agents, the current status of preclinical and clinical trials, and discuss the challenges and future perspectives in glioblastoma immuno-oncology.
Collapse
Affiliation(s)
- Liang Rong
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ni Li
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Zhang
- Key Laboratory of Brain, Cognition and Education Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China.
| |
Collapse
|
16
|
Sadri Nahand J, Salmaninejad A, Mollazadeh S, Tamehri Zadeh SS, Rezaee M, Sheida AH, Sadoughi F, Dana PM, Rafiyan M, Zamani M, Taghavi SP, Dashti F, Mirazimi SMA, Bannazadeh Baghi H, Moghoofei M, Karimzadeh M, Vosough M, Mirzaei H. Virus, Exosome, and MicroRNA: New Insights into Autophagy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1401:97-162. [DOI: 10.1007/5584_2022_715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Molecular Investigation of Human Cytomegalovirus and Epstein-Barr virus in Glioblastoma Brain Tumor: A Case-Control Study in Iran. IRANIAN BIOMEDICAL JOURNAL 2021; 25:426-33. [PMID: 34696577 DOI: 10.52547/ibj.25.6.426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Glioblastoma multiforme is the most invasive and lethal form of brain cancer with unclear etiology. Our study aimed to investigate the molecular prevalence of human cytomegalovirus (HCMV) and Epstein-Barr virus (EBV) infections in patients with glioblastoma multiforme (GBM). Methods This case-control study was conducted on 42 FFPE brain tumor samples from GBM patients and 42 brain autopsies from subjects without neurological disorders. The presence of EBV and HCMV DNA was determined, using PCR and nested-PCR assays, respectively. Results HCMV DNA was detected in 3 out of 42 (7.1%) of GBM samples and was absent from the control group (p = 0.07). Importantly, EBV DNA was detected in 9 out of 42 (21.4%) brain tissue specimens of GBM subjects, but again in none of the control group (p = 0.001). Conclusion Our findings indicate that infection with EBV is associated with GBM.
Collapse
|
18
|
Ulasov IV, Borovjagin A, Laevskaya A, Kamynina M, Timashev P, Cerchia L, Rozhkova EA. The IL13α 2R paves the way for anti-glioma nanotherapy. Genes Dis 2021; 10:89-100. [PMID: 37013057 PMCID: PMC10066331 DOI: 10.1016/j.gendis.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive (grade IV) gliomas characterized by a high rate of recurrence, resistance to therapy and a grim survival prognosis. The long-awaited improvement in GBM patients' survival rates essentially depends on advances in the development of new therapeutic approaches. Recent preclinical studies show that nanoscale materials could greatly contribute to the improvement of diagnosis and management of brain cancers. In the current review, we will discuss how specific features of glioma pathobiology can be employed for designing efficient targeting approaches. Moreover, we will summarize the main evidence for the potential of the IL-13R alpha 2 receptor (IL13α2R) targeting in GBM early diagnosis and experimental therapy.
Collapse
Affiliation(s)
- Ilya V. Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Corresponding author.
| | - Anton Borovjagin
- Department of BioMedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anastasia Laevskaya
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Margarita Kamynina
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, 4 Kosygin St, Moscow 119991, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, Moscow 119991, Russia
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), Naples 80131, Italy
| | - Elena A. Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, Argonne, IL 60439, USA
| |
Collapse
|
19
|
Krstanović F, Britt WJ, Jonjić S, Brizić I. Cytomegalovirus Infection and Inflammation in Developing Brain. Viruses 2021; 13:1078. [PMID: 34200083 PMCID: PMC8227981 DOI: 10.3390/v13061078] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a highly prevalent herpesvirus that can cause severe disease in immunocompromised individuals and immunologically immature fetuses and newborns. Most infected newborns are able to resolve the infection without developing sequelae. However, in severe cases, congenital HCMV infection can result in life-threatening pathologies and permanent damage of organ systems that possess a low regenerative capacity. Despite the severity of the problem, HCMV infection of the central nervous system (CNS) remains inadequately characterized to date. Cytomegaloviruses (CMVs) show strict species specificity, limiting the use of HCMV in experimental animals. Infection following intraperitoneal administration of mouse cytomegalovirus (MCMV) into newborn mice efficiently recapitulates many aspects of congenital HCMV infection in CNS. Upon entering the CNS, CMV targets all resident brain cells, consequently leading to the development of widespread histopathology and inflammation. Effector functions from both resident cells and infiltrating immune cells efficiently resolve acute MCMV infection in the CNS. However, host-mediated inflammatory factors can also mediate the development of immunopathologies during CMV infection of the brain. Here, we provide an overview of the cytomegalovirus infection in the brain, local immune response to infection, and mechanisms leading to CNS sequelae.
Collapse
Affiliation(s)
- Fran Krstanović
- Center for Proteomics and Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (F.K.); (S.J.)
| | - William J. Britt
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Stipan Jonjić
- Center for Proteomics and Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (F.K.); (S.J.)
| | - Ilija Brizić
- Center for Proteomics and Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (F.K.); (S.J.)
| |
Collapse
|
20
|
Chen C, Chen S, Han Z, Xie W, Zhang T, Mao C, Zhang L, Sun X, Kwok T, Shen X, Xue X. Patients with Helicobacter pylori-positive gastric cancer with human cytomegalovirus infection have a low tendency of advanced lymphatic metastasis in a Chinese population. Oncol Lett 2021; 21:402. [PMID: 33777225 PMCID: PMC7988662 DOI: 10.3892/ol.2021.12663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 07/22/2020] [Indexed: 11/18/2022] Open
Abstract
Recognized as a group I carcinogen for gastric cancer (GC) and a factor involved in the development of GC, Helicobacter pylori serves a major part in GC research. However, most studies have focused on H. pylori itself, ignoring the complicated pathogenic microbiological environment of GC and neglecting the synergistic or antagonistic effects of H. pylori with other pathogenic microorganisms. Increasing evidence has revealed that the human cytomegalovirus (HCMV) is present in several types of tumors and serves an important role in the neoplastic process of certain human malignant tumors, including GC. The aim of the present study was to explore the role of HCMV and H. pylori co-infection in GC. HCMV and H. pylori infection was analyzed in paired gastric tumor and peri-tumoral tissues from 134 (98 male and 36 female) patients using PCR. The results revealed that a total of 74 (55.2%) patients had H. pylori infection, 58 patients (43.3%) had HCMV infection, and 34 (25.4%) patients had both HCMV and H. pylori infection. Univariate and multivariate analyses demonstrated that H. pylori infection was independently associated with advanced lymphatic metastasis [P=0.007; odds ratio (OR)=3.51]. Furthermore, compared with HCMV-/H. pylori -, neither HCMV+/H. pylori - nor HCMV+/H. pylori + were associated with metastasis, but HCMV-/H. pylori + co-infection status was an independent risk factor for advanced lymphatic metastasis (P=0.005; OR=6.00). In conclusion, GC co-infected with HCMV and H. pylori exhibited a low tendency of lymph node metastasis. HCMV may interact with H. pylori to inhibit the process of lymphatic metastasis, and the mechanism requires further investigation.
Collapse
Affiliation(s)
- Chao Chen
- Department of Oncology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Zhejiang Chinese Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Sian Chen
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zheng Han
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Wangkai Xie
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Teming Zhang
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Chenchen Mao
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Liang Zhang
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiangwei Sun
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Terry Kwok
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria 3800, Australia
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria 3800, Australia
- Department of Microbiology, Monash University, Melbourne, Victoria 3800, Australia
| | - Xian Shen
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
21
|
Yu Z, Wang Y, Liu L, Zhang X, Jiang S, Wang B. Apoptosis Disorder, a Key Pathogenesis of HCMV-Related Diseases. Int J Mol Sci 2021; 22:ijms22084106. [PMID: 33921122 PMCID: PMC8071541 DOI: 10.3390/ijms22084106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) belongs to the β-herpesvirus family, which is transmitted in almost every part of the world and is carried by more than 90% of the general population. Increasing evidence indicates that HCMV infection triggers numerous diseases by disrupting the normal physiological activity of host cells, particularly apoptosis. Apoptosis disorder plays a key role in the initiation and development of multiple diseases. However, the relationship and molecular mechanism of HCMV-related diseases and apoptosis have not yet been systematically summarized. This review aims to summarize the role of apoptosis in HCMV-related diseases and provide an insight into the molecular mechanism of apoptosis induced by HCMV infection. We summarize the literature on HCMV-related diseases and suggest novel strategies for HCMV treatment by regulating apoptosis.
Collapse
Affiliation(s)
- Zhongjie Yu
- Department of Special Medicine, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266000, China;
| | - Yashuo Wang
- College of Life Sciences, Qingdao University, Qingdao 266000, China;
| | - Lili Liu
- Department of Basic Medicine, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266000, China;
| | - Xianjuan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266000, China; (X.Z.); (S.J.)
| | - Shasha Jiang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266000, China; (X.Z.); (S.J.)
| | - Bin Wang
- Department of Special Medicine, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266000, China;
- Correspondence: ; Tel.: +86-136-8532-6203
| |
Collapse
|
22
|
Suares A, Medina MV, Coso O. Autophagy in Viral Development and Progression of Cancer. Front Oncol 2021; 11:603224. [PMID: 33763351 PMCID: PMC7982729 DOI: 10.3389/fonc.2021.603224] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a complex degradative process by which eukaryotic cells capture cytoplasmic components for subsequent degradation through lysosomal hydrolases. Although this catabolic process can be triggered by a great variety of stimuli, action in cells varies according to cellular context. Autophagy has been previously linked to disease development modulation, including cancer. Autophagy helps suppress cancer cell advancement in tumor transformation early stages, while promoting proliferation and metastasis in advanced settings. Oncoviruses are a particular type of virus that directly contribute to cell transformation and tumor development. Extensive molecular studies have revealed complex ways in which autophagy can suppress or improve oncovirus fitness while still regulating viral replication and determining host cell fate. This review includes recent advances in autophagic cellular function and emphasizes its antagonistic role in cancer cells.
Collapse
Affiliation(s)
- Alejandra Suares
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Victoria Medina
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Omar Coso
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
23
|
Suares A, Medina MV, Coso O. Autophagy in Viral Development and Progression of Cancer. Front Oncol 2021. [DOI: 10.3389/fonc.2021.603224
expr 816899697 + 824303767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Autophagy is a complex degradative process by which eukaryotic cells capture cytoplasmic components for subsequent degradation through lysosomal hydrolases. Although this catabolic process can be triggered by a great variety of stimuli, action in cells varies according to cellular context. Autophagy has been previously linked to disease development modulation, including cancer. Autophagy helps suppress cancer cell advancement in tumor transformation early stages, while promoting proliferation and metastasis in advanced settings. Oncoviruses are a particular type of virus that directly contribute to cell transformation and tumor development. Extensive molecular studies have revealed complex ways in which autophagy can suppress or improve oncovirus fitness while still regulating viral replication and determining host cell fate. This review includes recent advances in autophagic cellular function and emphasizes its antagonistic role in cancer cells.
Collapse
|
24
|
Zhang Q, Song X, Ma P, Lv L, Zhang Y, Deng J, Zhang Y. Human Cytomegalovirus miR-US33as-5p Targets IFNAR1 to Achieve Immune Evasion During Both Lytic and Latent Infection. Front Immunol 2021; 12:628364. [PMID: 33746965 PMCID: PMC7973039 DOI: 10.3389/fimmu.2021.628364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/04/2021] [Indexed: 11/21/2022] Open
Abstract
As the first line of antiviral defense, type I interferon (IFN) binds IFN receptor 1 (IFNAR1) and IFNAR2 to activate the Jak-STAT signal transduction pathway, producing IFN-stimulated genes (ISGs) to control viral infection. The mechanisms by which human cytomegalovirus (HCMV) counteracts the IFN pathway are only partially defined. We show that miR-US33as-5p encoded by HCMV is expressed in both lytic and latent infection. By analysis with RNA hybrid and screening with luciferase reporter assays, we identified IFNAR1 as a target of hcmv-miR-US33as-5p, which was further verified by examining the expression of two IFNAR1 mutants and the binding of IFNAR1 to miR-US33as-5p/miR-US33as-5p-M1/miR-US33as-5p-M2. We found that after the transfection of miR-US33as-5p mimics into different cell lines, the phosphorylation of downstream proteins and ISG expression were downregulated. Immunofluorescence showed that the miR-US33as-5p mimics also inhibited STAT1 translocation into the nucleus. Furthermore, we constructed HCMV with mutant miR-US33as-5p and determined that the mutation did not affect HCMV replication. We found that MRC-5/human foreskin fibroblast (HFF) cells infected with ΔmiRNA HCMV exhibited higher IFNAR1 and ISG expression and a reduced viral load in the presence of exogenous IFN than cells infected with WT HCMV did, confirming that the knockout of miR-US33as-5p impaired viral resistance to IFN. Finally, we tested the effect of ΔmiRNA HCMV on THP-1 and d-THP-1 cells, common in vitro models of latent infection and reactivation, respectively. Again, we found that cells infected with ΔmiRNA HCMV showed a reduced viral load in the presence of IFN than the control cells did, confirming that miR-US33as-5p also affects IFN resistance during both latency and reactivation. These results indicate a new microRNA (miRNA)-based immune evasion mechanism employed by HCMV to achieve lifelong infection.
Collapse
Affiliation(s)
- Qian Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Xin Song
- Department of Otolaryngology Head and Neck Surgery, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ping Ma
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Liping Lv
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Yangyang Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Jiang Deng
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| | - Yanyu Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China.,Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, China
| |
Collapse
|
25
|
The Association of Human Herpesviruses with Malignant Brain Tumor Pathology and Therapy: Two Sides of a Coin. Int J Mol Sci 2021; 22:ijms22052250. [PMID: 33668202 PMCID: PMC7956256 DOI: 10.3390/ijms22052250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
The role of certain viruses in malignant brain tumor development remains controversial. Experimental data demonstrate that human herpesviruses (HHVs), particularly cytomegalovirus (CMV), Epstein–Barr virus (EBV) and human herpes virus 6 (HHV-6), are implicated in brain tumor pathology, although their direct role has not yet been proven. CMV is present in most gliomas and medulloblastomas and is known to facilitate oncomodulation and/or immunomodulation, thus promoting cancer cell proliferation, invasion, apoptosis, angiogenesis, and immunosuppression. EBV and HHV-6 have also been detected in brain tumors and high-grade gliomas, showing high rates of expression and an inflammatory potential. On the other hand, due to the neurotropic nature of HHVs, novel studies have highlighted the engagement of such viruses in the development of new immunotherapeutic approaches in the context of oncolytic viral treatment and vaccine-based strategies against brain tumors. This review provides a comprehensive evaluation of recent scientific data concerning the emerging dual role of HHVs in malignant brain pathology, either as potential causative agents or as immunotherapeutic tools in the fight against these devastating diseases.
Collapse
|
26
|
Semmes EC, Hurst JH, Walsh KM, Permar SR. Cytomegalovirus as an immunomodulator across the lifespan. Curr Opin Virol 2020; 44:112-120. [PMID: 32818717 DOI: 10.1016/j.coviro.2020.07.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022]
Abstract
Human cytomegalovirus (HCMV) is a nearly ubiquitous β-herpesvirus that establishes latent infection in the majority of the world's population. HCMV infection profoundly influences the host immune system and, perhaps more than any other human pathogen, has been shown to create a lasting imprint on human T and NK cell compartments. HCMV-seropositivity has been associated with both beneficial effects, such as increased vaccine responsiveness or heterologous protection against infections, and deleterious effects, such as pathological neurodevelopmental sequelae from congenital infection in utero and cumulative damage from chronic lifelong latency into old age. The significance of many of these associations is unclear, as studies into the causal mechanisms linking HCMV and these disease outcomes are lacking; however, HCMV-mediated changes to the immune system may play a key role. This review examines how HCMV impacts the host immune system in an age-dependent manner with important implications for human immunophenotypes and long-term disease risk.
Collapse
Affiliation(s)
- Eleanor C Semmes
- Medical Scientist Training Program, Duke University, Durham, NC, USA; Children's Health and Discovery Institute, Department of Pediatrics, Duke University, Durham, NC, USA
| | - Jillian H Hurst
- Children's Health and Discovery Institute, Department of Pediatrics, Duke University, Durham, NC, USA; Department of Pediatrics, Division of Infectious Diseases, Duke University, Durham NC, USA
| | - Kyle M Walsh
- Children's Health and Discovery Institute, Department of Pediatrics, Duke University, Durham, NC, USA; Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Sallie R Permar
- Children's Health and Discovery Institute, Department of Pediatrics, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, Duke University, Durham, NC, USA; Department of Pediatrics, Division of Infectious Diseases, Duke University, Durham NC, USA.
| |
Collapse
|
27
|
Britt WJ. Human Cytomegalovirus Infection in Women With Preexisting Immunity: Sources of Infection and Mechanisms of Infection in the Presence of Antiviral Immunity. J Infect Dis 2020; 221:S1-S8. [PMID: 32134479 PMCID: PMC7057782 DOI: 10.1093/infdis/jiz464] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) infection remains an important cause of neurodevelopmental sequelae in infants infected in utero. Unique to the natural history of perinatal HCMV infections is the occurrence of congenital HCMV infections (cCMV) in women with existing immunity to HCMV, infections that have been designated as nonprimary maternal infection. In maternal populations with a high HCMV seroprevalence, cCMV that follows nonprimary maternal infections accounts for 75%-90% of all cases of cCMV infections as well as a large proportion of infected infants with neurodevelopmental sequelae. Although considerable effort has been directed toward understanding immune correlates that can modify maternal infections and intrauterine transmission, the source of virus leading to nonprimary maternal infections and intrauterine transmission is not well defined. Previous paradigms that included reactivation of latent virus as the source of infection in immune women have been challenged by studies demonstrating acquisition and transmission of antigenically distinct viruses, a finding suggesting that reinfection through exposure to an exogenous virus is responsible for some cases of nonprimary maternal infection. Additional understanding of the source(s) of virus that leads to nonprimary maternal infection will be of considerable value in the development and testing of interventions such as vaccines designed to limit the incidence of cCMV in populations with high HCMV seroprevalence.
Collapse
Affiliation(s)
- William J Britt
- Departments of Pediatrics, Microbiology, Neurobiology, University of Alabama School of Medicine, Birmingham, Alabama
| |
Collapse
|
28
|
Human cytomegalovirus ie2 affects the migration of glioblastoma by mediating the different splicing patterns of RON through hnRNP A2B1. Neuroreport 2019; 30:805-811. [PMID: 31283707 DOI: 10.1097/wnr.0000000000001277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glioblastoma is the most aggressive intracranial tumor and diffuse migration is the leading cause of death. Recent evidence has indicated that heterogeneous nuclear ribonucleoprotein A2B1 (hnRNP A2B1) is overexpressed in human glioblastoma tissue and enhances glioblastoma invasion in vitro. We found by mass spectrometry that hnRNP A2B1 interacts with human cytomegalovirus (HCMV) immediate early 86 protein (IE86, ie2 gene-encoded) in malignant glioma cells (U87MG) infected with HCMV. However, the role of hnRNP A2 B1 in glioblastoma development remains poorly understood. Here, we report that hnRNP A2B1 is highly expressed in the HCMV·ie2 transgenic mice model. This phenomenon was confirmed in U87MG cell lines transfected with pEGFP-N3-ie2 plasmid. In addition, hnRNP A2B1 knockdown in U87MG cells inhibited tumor migration, and this effect might be mediated by hnRNP A2B1 through effects on splicing patterns of RON. Our data suggested that HCMV· ie2 promotes glioblastoma migration by regulating hnRNP A2B1 expression.
Collapse
|
29
|
Piper K, Foster H, Gabel B, Nabors B, Cobbs C. Glioblastoma Mimicking Viral Encephalitis Responds to Acyclovir: A Case Series and Literature Review. Front Oncol 2019; 9:8. [PMID: 30723703 PMCID: PMC6350341 DOI: 10.3389/fonc.2019.00008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/03/2019] [Indexed: 12/14/2022] Open
Abstract
Viral encephalitis and glioblastoma are both relatively rare conditions with poor prognoses. While the clinical and radiographic presentations of these diseases are often distinctly different, viral encephalitis can sometimes masquerade as glioblastoma. Rarely, glioblastoma can also be misdiagnosed as viral encephalitis. In some cases where a high-grade glioma was initially diagnosed as viral encephalitis, antiviral administration has proven effective for relieving early symptoms. We present three cases in which patients presented with symptoms and radiographic findings suggestive of viral encephalitis and experienced dramatic clinical improvement following treatment with acyclovir, only to later be diagnosed with glioblastoma in the region of suspected encephalitis and ultimately succumb to tumor progression.
Collapse
Affiliation(s)
- Keenan Piper
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States
| | - Haidn Foster
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States.,University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Brandon Gabel
- Department of Neurological Surgery, University of California San Diego Medical Center, San Diego, CA, United States
| | - Burt Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Charles Cobbs
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States
| |
Collapse
|
30
|
Modulation of radiation sensitivity and antitumor immunity by viral pathogenic factors: Implications for radio-immunotherapy. Biochim Biophys Acta Rev Cancer 2018; 1871:126-137. [PMID: 30605716 DOI: 10.1016/j.bbcan.2018.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/17/2018] [Accepted: 12/03/2018] [Indexed: 02/07/2023]
Abstract
Several DNA viruses including Human Papillomavirus (HPV), Epstein-Barr virus (EBV), and Human cytomegalovirus (HCMV) are mechanistically associated with the development of human cancers (HPV, EBV) and/or modulation of the immune system (HCMV). Moreover, a number of distinct mechanisms have been described regarding the modulation of tumor cell response to ionizing radiation and evasion from the host immune system by viral factors. There is further accumulating interest in the treatment with immune-modulatory therapies such as immune checkpoint inhibitors for malignancies with a viral etiology. Also, patients with HPV-positive tumors have a significantly improved prognosis that is attributable to increased intrinsic radiation sensitivity and may also arise from modulation of a cytotoxic T cell response in the tumor microenvironment (TME). In this review, we will highlight recent advances in the understanding of the biological basis of radiation response mediated by viral pathogenic factors and evasion from and modulation of the immune system by viruses.
Collapse
|
31
|
Kim JW, Kane JR, Panek WK, Young JS, Rashidi A, Yu D, Kanojia D, Hasan T, Miska J, Gómez-Lim MA, Ulasov IV, Balyasnikova IV, Ahmed AU, Wainwright DA, Lesniak MS. A Dendritic Cell-Targeted Adenoviral Vector Facilitates Adaptive Immune Response Against Human Glioma Antigen (CMV-IE) and Prolongs Survival in a Human Glioma Tumor Model. Neurotherapeutics 2018; 15:1127-1138. [PMID: 30027430 PMCID: PMC6277295 DOI: 10.1007/s13311-018-0650-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Antitumor immunotherapeutic strategies represent an especially promising set of approaches with rapid translational potential considering the dismal clinical context of high-grade gliomas. Dendritic cells (DCs) are the body's most professional antigen-presenting cells, able to recruit and activate T cells to stimulate an adaptive immune response. In this regard, specific loading of tumor-specific antigen onto dendritic cells potentially represents one of the most advanced strategies to achieve effective antitumor immunization. In this study, we developed a DC-specific adenoviral (Ad) vector, named Ad5scFvDEC205FF, targeting the DC surface receptor, DEC205. In vitro analysis shows that 60% of DCs was infected by this vector while the infectivity of other control adenoviral vectors was less than 10%, demonstrating superior infectivity on DCs. Moreover, an average of 14% of DCs were infected by Ad5scFvDEC205FF-GFP, while less than 3% of non-DCs were infected following in vivo administration, demonstrating highly selective in vivo DC infection. Importantly, vaccination with this vehicle expressing human glioma-specific antigen, Ad5scFvDEC205FF-CMV-IE, shows a prolonged survival benefit in GL261CMV-IE-implanted murine glioma models (p < 0.0007). Furthermore, when rechallenged, cancerous cells were completely rejected. In conclusion, our novel, viral-mediated, DC-based immunization approach has the significant therapeutic potential for patients with high-grade gliomas.
Collapse
Affiliation(s)
- Julius W Kim
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - J Robert Kane
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Wojciech K Panek
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Jacob S Young
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Dou Yu
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Tanwir Hasan
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Miguel A Gómez-Lim
- Departamento de Ingeniería Genética, CINVESTAV Irapuato, Km 9.6 Libramiento Norte Carretera Irapuato-León, 36821, Irapuato, Guanajuato, Mexico
| | - Ilya V Ulasov
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Atique U Ahmed
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, Illinois, 60611, USA.
| |
Collapse
|
32
|
More than just oncogenes: mechanisms of tumorigenesis by human viruses. Curr Opin Virol 2018; 32:48-59. [PMID: 30268926 DOI: 10.1016/j.coviro.2018.09.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/05/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022]
Abstract
Most humans are infected with at least one of the known human cancer viruses during their lifetimes. While the initial infection with these viruses does not cause major disease, infected cells can acquire cancer hallmarks, particularly upon immunosuppression or exposure to co-carcinogenic stimuli. Even though cancer formation represents a rare outcome of a viral infection, approximately one out of eight human cancers has a viral etiology. Viral cancers present unique opportunities for prophylaxis, diagnosis, and therapy, as demonstrated by the success of HBV and HPV vaccines and HCV antivirals in decreasing the incidence of tumors that are caused by these viruses. Here we review common characteristics and mechanisms of action of the human oncogenic viruses.
Collapse
|
33
|
Dos Santos CJ, Ferreira Castro FL, de Aguiar RB, Menezes IG, Santos AC, Paulus C, Nevels M, Carlan da Silva MC. Impact of human cytomegalovirus on glioblastoma cell viability and chemotherapy treatment. J Gen Virol 2018; 99:1274-1285. [PMID: 30045780 DOI: 10.1099/jgv.0.001118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The relationship between human cytomegalovirus (HCMV) and tumours has been extensively investigated, mainly in glioblastoma multiforme (GBM), a malignant tumour of the central nervous system with low overall survival rates. Several reports have demonstrated the presence of HCMV in GBM, although typically restricted to a low number of cells, and studies have indicated that viral proteins have the ability to dysregulate cellular processes and increase tumour malignancy. Treatment of GBM involves the use of the chemotherapeutic agents temozolomide (TMZ) and carmustine (bis-chloroethylnitrosourea, BCNU), which lead to the attachment of adducts to the DNA backbone, causing errors during replication and consequent cell death. It is known that HCMV infection can modulate DNA repair pathways, but what effects the virus may exhibit during chemotherapy are unknown. Here we approach this question by analysing HCMV infection and viral protein accumulation in GBM cell lines with different genotypes and their response to TMZ and BCNU in the presence of the virus. We demonstrate that A172, TP365MG and U251MG GBM cells are efficiently infected by both low-passage (TB40E) and high-passage (AD169) HCMV strains. However, the GBM cell lines vary widely in their permissiveness to viral gene expression and exhibit very different patterns of immediate early, early and late protein accumulation. HCMV reduces the viability of permissive GBM cells in a multiplicity-dependent manner in both the absence and presence of TMZ or BNCU. In sum, we demonstrate that GBM cell lines are equally susceptible but differentially permissive to infection by both low- and high-passage strains of HCMV. This observation not only indicates that viral replication is largely controlled by cellular factors in this system, but also provides a possible explanation for why viral gene products are only found in a subset of cells in GBM tumours. Furthermore, we conclude that the virus does not confer increased resistance to chemotherapeutic drugs in various GBM cell lines, but instead reduces tumour cell viability. These results highlight that the oncomodulatory potential of HCMV is not limited to cancer-promoting activities, but also includes adverse effects on tumour cell proliferation or survival.
Collapse
Affiliation(s)
- Claudia Januário Dos Santos
- 1Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | | | - Rodrigo Barbosa de Aguiar
- 2Department of Biophysics, Paulista Medical School, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Isabela Godoy Menezes
- 1Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - Ana Carolina Santos
- 1Center for Natural and Humanities Sciences, Federal University of ABC (UFABC), São Bernardo do Campo, Brazil
| | - Christina Paulus
- 3Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Michael Nevels
- 3Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | | |
Collapse
|
34
|
Advancing Our Understanding of Protective Maternal Immunity as a Guide for Development of Vaccines To Reduce Congenital Cytomegalovirus Infections. J Virol 2018; 92:JVI.00030-18. [PMID: 29343580 DOI: 10.1128/jvi.00030-18] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the most common congenitally transmitted pathogen worldwide, impacting an estimated 1 million newborns annually. Congenital HCMV (cCMV) infection is a major global contributor to long-term neurologic deficits, including deafness, microcephaly, and neurodevelopmental delay, as well as to fetal loss and occasional infant mortality. Accordingly, design of a maternal vaccine to prevent cCMV continues to be a top public health priority. Nevertheless, we remain without a licensed vaccine. Maternal immunity provides partial protection, as the risk of vertical HCMV transmission from chronically infected mothers is reduced compared to settings in which the mother is newly infected during pregnancy. Therefore, an understanding of the maternal immune correlates of protection against cCMV is critical to informing design of an efficacious maternal vaccine. Although vaccine development is being assiduously pursued by a large number of pharmaceutical manufacturers, biotechnology organizations, and academic researchers, some pessimism has been expressed regarding the issue of whether a vaccine to protect against cCMV is possible. This pessimism is based on observations that natural immunity is not completely protective against maternal reinfection and congenital transmission. However, we assert that optimism regarding vaccine development is indeed justified, on the basis of accruing evidence of immune correlates of protection-readily achievable by vaccination-that are associated with reduced transmission of HCMV to the fetus in seronegative women. In light of the substantial burden on society conferred by cCMV infection, even a modest reduction in the occurrence of this fetal disease is an important public health goal and justifies aggressive clinical evaluation of vaccines currently in the pipeline.
Collapse
|
35
|
Du Y, Zhang G, Liu Z. Human cytomegalovirus infection and coronary heart disease: a systematic review. Virol J 2018; 15:31. [PMID: 29409508 PMCID: PMC5801777 DOI: 10.1186/s12985-018-0937-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 01/18/2018] [Indexed: 12/15/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) infection is closely associated with coronary heart disease. Main body of the abstract In 1987, Adam et al. were the first to report an association between HCMV infection and atherosclerosis (AS), and later, many serum epidemiology and molecular biology studies showed that HCMV-infected endothelial cells play an important role in the development of AS. As patients with HCMV are generally susceptible to coronary heart disease, and with the increasing elderly population, a review of recent studies focusing on the relationships of HCMV infection and coronary heart disease is timely and necessary. Short conclusion The role of HCMV infection in the development of AS needs further study, since many remaining issues need to be explored and resolved. For example, whether HCMV promotes the development of coronary AS, and what the independent factors that lead to coronary artery AS by viral infection are. A comprehensive understanding of HCMV infection is needed in order to develop better strategies for preventing AS.
Collapse
Affiliation(s)
- Yu Du
- Department of Microbiology, Weifang Medical University, Weifang, 261053, China
| | - Guangxue Zhang
- Department of Clinical Laboratory, Shandong Qingzhou Rongjun Hospital, Qingzhou, 262500, China
| | - Zhijun Liu
- Department of Microbiology, Weifang Medical University, Weifang, 261053, China.
| |
Collapse
|
36
|
Abstract
Glioblastoma (GBM) is a rare tumor and one of the most challenging malignancies to treat in all of oncology. Although advances have been made in the treatment of GBM, encouraging outcomes typically are not observed; patients diagnosed with these tumors generally have a dismal prognosis and poor quality of life as the disease progresses. This review summarizes the clinical presentation of GBM, diagnostic methods, evidentiary basis for the current standards of care, and investigational approaches to treat or manage GBM. Because the track record for developing effective therapies for GBM has been dismal, we also review the challenges to successful therapeutic and biomarker development.
Collapse
Affiliation(s)
- Brian M. Alexander
- Brian M. Alexander, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, MA; and Timothy F. Cloughesy, University of California Los Angeles, Los Angeles, CA
| | - Timothy F. Cloughesy
- Brian M. Alexander, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, MA; and Timothy F. Cloughesy, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
37
|
Lack of human cytomegalovirus expression in single cells from glioblastoma tumors and cell lines. J Neurovirol 2017; 23:671-678. [PMID: 28695489 DOI: 10.1007/s13365-017-0543-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/23/2017] [Accepted: 06/08/2017] [Indexed: 12/21/2022]
Abstract
The relationship between human cytomegalovirus (HCMV) and glioblastoma (GBM) is an ongoing debate with extensive evidence supporting or refuting its existence through molecular assays, pre-clinical studies, and clinical trials. We focus primarily on the crux of the debate, detection of HCMV in GBM samples using molecular assays. We propose that these differences in detection could be affected by cellular heterogeneity. To take this into account, we align the single-cell RNA sequencing (scRNA-seq) reads from five GBM tumors and two cell lines to HCMV and analyze the alignments for evidence of (i) complete viral transcripts and (ii) low-abundance viral reads. We found that neither tumor nor cell line samples showed conclusive evidence of full HCMV viral transcripts. We also identified low-abundance reads aligned across all tumors, with two tumors having higher alignment rates than the rest of the tumor samples. This work is meant to rigorously test for HCMV RNA expression at a single cell level in GBM samples and examine the possible utility of single cell data in tumor virology.
Collapse
|
38
|
Ma B, Yuan Z, Zhang L, Lv P, Yang T, Gao J, Pan N, Wu Q, Lou J, Han C, Zhang B. Long non-coding RNA AC023115.3 suppresses chemoresistance of glioblastoma by reducing autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1393-1404. [PMID: 28499919 DOI: 10.1016/j.bbamcr.2017.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/13/2022]
Abstract
Malignant glioma is an aggressive brain cancer that responds poorly to chemotherapy. However, the molecular mechanism underlying the development of chemoresistance in glioma is not well-understood. In this study, we show that long non-coding RNA AC023115.3 is induced by cisplatin in human glioblastoma cells and that elevated AC023115.3 promotes cisplatin-induced apoptosis by inhibiting autophagy. Further mechanistic studies revealed that AC023115.3 acts as a competing endogenous RNA for miR-26a and attenuates the inhibitory effect of miR-26a on GSK3β, a proline-directed serine-threonine kinase that promotes the degradation of Mcl1, leading to an increase in GSK3β and a decrease in autophagy. Additionally, we discovered that AC023115.3 improves chemosensitivity of glioma cells to cisplatin by regulating the miR-26a-GSK3β-Mcl1 pathway. Thus, these data indicate that the AC023115.3-miR-26a-GSK3β signalling axis plays an important role in reducing the chemoresistance of glioma.
Collapse
Affiliation(s)
- Binbin Ma
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116027, China
| | - Zhongbo Yuan
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116027, China
| | - Li Zhang
- Laboratory of Pathogenic Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116027, China
| | - Peng Lv
- Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian 116027, China
| | - Ting Yang
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116027, China
| | - Jinxia Gao
- Department of Anaesthesiology, Second Affiliated Hospital, Dalian Medical University, 116027, China
| | - Ning Pan
- Department of Anaesthesiology, Second Affiliated Hospital, Dalian Medical University, 116027, China
| | - Qiong Wu
- Department of Neurology of Dalian Municipal Central Hospital Affiliated, Dalian Medical University, 116033, China
| | - Jiacheng Lou
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116027, China
| | - Chuanchun Han
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116027, China.
| | - Bo Zhang
- Department of Neurosurgery, Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116027, China.
| |
Collapse
|