1
|
Wen Y, Chen J, Long T, Chen F, Wang Z, Chen S, Zhang G, Li M, Zhang S, Kang H, Feng W, Wang G. miR-6760-5p suppresses neoangiogenesis by targeting Yes-associated protein 1 in patients with moyamoya disease undergoing indirect revascularization. Gene 2025; 937:149152. [PMID: 39662645 DOI: 10.1016/j.gene.2024.149152] [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: 07/10/2024] [Revised: 11/22/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVE The aim of this research was to investigate the specific regulatory role of miR-6760-5p in angiogenesis in moyamoya disease. METHODS HUVECs were transfected with miR-6760-5p inhibitor and mimics fragments, then subjected to assays for cell proliferation, migration, and tube formation. Subsequently, downstream target genes of miR-6760-5p were predicted and the protein expression levels of these genes were evaluated. The presence of miR-6760-5p and YAP1 was verified by a dual luciferase reporter gene test, followed by an assessment of the effects of YAP1 and miR-6760-5p on the HUVECs. RESULTS Comparatively to the control group, increased expression of miR-6760-5p decreased cell growth, movement, and tube formation. YAP1 gene was discovered as a target controlled by miR-6760-5p, with subsequent investigation confirming YAP1 as a gene regulated by miR-6760-5p. Additionally, miR-6760-5p was found to counteract the angiogenesis-promoting effect of YAP1. CONCLUSION The results of this research suggest a possible link between the miR-6760-5p gene found in the cerebrospinal fluid of individuals with moyamoya disease and the process of vascularization in this particular condition. The findings indicate that miR-6760-5p may be a new molecular indicator and potential target for the diagnosis of moyamoya disease.
Collapse
Affiliation(s)
- Yunyu Wen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Junda Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Tinghan Long
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Fangzhou Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Zhibin Wang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Siyuan Chen
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Guozhong Zhang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Mingzhou Li
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Shichao Zhang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Huibin Kang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China
| | - Wenfeng Feng
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China.
| | - Gang Wang
- Neurosurgery Department, Nanfang Hospital, Southern Medical University, Located in Guangzhou, Guangdong, China; The Laboratory for Precision Neurosurgery is affiliated with Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China; The Institute of Brain Disease is part of Nanfang Hospital at Southern Medical University, Located in Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Dai J, Jiang Y, Hu H, Zhang S, Chen Y. Extracellular vesicles as modulators of glioblastoma progression and tumor microenvironment. Pathol Oncol Res 2024; 30:1611549. [PMID: 38379858 PMCID: PMC10876843 DOI: 10.3389/pore.2024.1611549] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Glioblastoma is the most aggressive brain tumor with extremely poor prognosis in adults. Routine treatments include surgery, chemotherapy, and radiotherapy; however, these may lead to rapid relapse and development of therapy-resistant tumor. Glioblastoma cells are known to communicate with macrophages, microglia, endothelial cells, astrocytes, and immune cells in the tumor microenvironment (TME) to promote tumor preservation. It was recently demonstrated that Glioblastoma-derived extracellular vesicles (EVs) participate in bidirectional intercellular communication in the TME. Apart from promoting glioblastoma cell proliferation, migration, and angiogenesis, EVs and their cargos (primarily proteins and miRNAs) can act as biomarkers for tumor diagnosis and prognosis. Furthermore, they can be used as therapeutic tools. In this review, the mechanisms of Glioblastoma-EVs biogenesis and intercellular communication with TME have been summarized. Moreover, there is discussion surrounding EVs as novel diagnostic structures and therapeutic tools for glioblastoma. Finally, unclear questions that require future investigation have been reviewed.
Collapse
Affiliation(s)
- Jie Dai
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Jiang
- Department of Neurosurgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyue Hu
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shuang Zhang
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yue Chen
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Abstract
Primary brain cancer or brain cancer is the overgrowth of abnormal or malignant cells in the brain or its nearby tissues that form unwanted masses called brain tumors. People with malignant brain tumors suffer a lot, and the expected life span of the patients after diagnosis is often only around 14 months, even with the most vigorous therapies. The blood-brain barrier (BBB) is the main barrier in the body that restricts the entry of potential chemotherapeutic agents into the brain. The chances of treatment failure or low therapeutic effects are some significant drawbacks of conventional treatment methods. However, recent advancements in nanotechnology have generated hope in cancer treatment. Nanotechnology has shown a vital role starting from the early detection, diagnosis, and treatment of cancer. These tiny nanomaterials have great potential to deliver drugs across the BBB. Beyond just drug delivery, nanomaterials can be simulated to generate fluorescence to detect tumors. The current Review discusses in detail the challenges of brain cancer treatment and the application of nanotechnology to overcome those challenges. The success of chemotherapeutic treatment or the surgical removal of tumors requires proper imaging. Nanomaterials can provide imaging and therapeutic benefits for cancer. The application of nanomaterials in the diagnosis and treatment of brain cancer is discussed in detail by reviewing past studies.
Collapse
Affiliation(s)
- Yogita Ale
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand 248007, India
| | - Nidhi Nainwal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand 248007, India
| |
Collapse
|
4
|
Musatova OE, Rubtsov YP. Effects of glioblastoma-derived extracellular vesicles on the functions of immune cells. Front Cell Dev Biol 2023; 11:1060000. [PMID: 36960410 PMCID: PMC10028257 DOI: 10.3389/fcell.2023.1060000] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 02/22/2023] [Indexed: 03/09/2023] Open
Abstract
Glioblastoma is the most aggressive variant of glioma, the tumor of glial origin which accounts for 80% of brain tumors. Glioblastoma is characterized by astoundingly poor prognosis for patients; a combination of surgery, chemo- and radiotherapy used for clinical treatment of glioblastoma almost inevitably results in rapid relapse and development of more aggressive and therapy resistant tumor. Recently, it was demonstrated that extracellular vesicles produced by glioblastoma (GBM-EVs) during apoptotic cell death can bind to surrounding cells and change their phenotype to more aggressive. GBM-EVs participate also in establishment of immune suppressive microenvironment that protects glioblastoma from antigen-specific recognition and killing by T cells. In this review, we collected present data concerning characterization of GBM-EVs and study of their effects on different populations of the immune cells (T cells, macrophages, dendritic cells, myeloid-derived suppressor cells). We aimed at critical analysis of experimental evidence in order to conclude whether glioblastoma-derived extracellular vesicles are a major factor in immune evasion of this deadly tumor. We summarized data concerning potential use of GBM-EVs for non-invasive diagnostics of glioblastoma. Finally, the applicability of approaches aimed at blocking of GBM-EVs production or their fusion with target cells for treatment of glioblastoma was analyzed.
Collapse
Affiliation(s)
- Oxana E. Musatova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
| | - Yury P. Rubtsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
- N.N.Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Moscow, Russia
- *Correspondence: Yury P. Rubtsov,
| |
Collapse
|
5
|
Shao W, Azam Z, Guo J, To SST. Oncogenic potential of PIK3CD in glioblastoma is exerted through cytoskeletal proteins PAK3 and PLEK2. J Transl Med 2022; 102:1314-1322. [PMID: 35851857 DOI: 10.1038/s41374-022-00821-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
The Class IA phosphoinositide-3-kinase catalytic isoforms p110α, p110β, and p110δ have been implicated to play vital but overlapping roles in various cancers, including glioblastoma (GBM). We have previously shown that PIK3CD, encoding p110δ, is highly expressed in multiple glioma cell lines and involved in glioma cell migration and invasion. Based on the RNA sequencing data from The Cancer Genome Atlas (TCGA) database, we found the level of PIK3CD expression is significantly higher in GBM than WHO grade II and III gliomas and is closely related to poor survival. To further dissect the oncogenic roles of PIK3CD in glioma progression, we employed CRISPR/Cas9 to completely abrogate its expression in the GBM cell line U87-MG and have successfully isolated two knockout clones with different gene modifications. As expected, the knockout clones exhibited significantly lower migration and invasion capabilities when compared with their parental cells. Interestingly, knockout of PIK3CD also dramatically reduced the colony formation ability of the knockout cells. Further study revealed that PIK3CD deficiency could negate tumorigenesis in nude mice. To determine the downstream effect of PIK3CD depletion, we performed RT2 profiler PCR array of selected gene sets and found that knockout of PIK3CD impaired the activity of p-21 activated kinase 3 (PAK3) and pleckstrin 2 (PLEK2), molecules involved in cancer cell migration and proliferation. This explains why the glioma cells without the PIK3CD expression exhibited weaker oncogenic features. Further, RNAseq analysis of parent and knockout clones revealed that this interaction might happen through axonogenesis signaling pathway. Taken together, we demonstrated that PIK3CD could be a potential prognostic factor and therapeutic target for GBM patients.
Collapse
Affiliation(s)
- Wei Shao
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
| | - Zulfikar Azam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
| | - Jintao Guo
- Department of Translational Medicine, Medical College of Xiamen University, Xiamen, 361102, China
| | - Shing Shun Tony To
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China.
| |
Collapse
|
6
|
Zhang S, Zhang W, Wu B, Xia L, Li L, Jin K, Zou Y, Sun C. Hub gene target of glioblastoma: LOX, SERPINH1 and TGFBI. Medicine (Baltimore) 2022; 101:e31418. [PMID: 36397358 PMCID: PMC9666166 DOI: 10.1097/md.0000000000031418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma (GBM) is a malignant tumor. The long-term prognosis of the patients is poor. Therefore, it is of important clinical value to further explore the pathogenesis and look for molecular markers for early diagnosis and targeted treatment. Two expression profiling datasets [GSE50161 (GPL570 platform), GSE116520 (GPL10558 platform)] were respectively downloaded from the gene expression omnibus database. Volcano diagrams show the Differently expressed genes (DEGs) of GSE50161 and GSE116520. A Venn diagram revealed 467 common DEGs between the 2 datasets. Lysyl oxidase (LOX), serpin family H member 1 (SERPINH1) and transforming growth factor beta induced (TGFBI) were negatively correlated with the overall survival rate in patients with GBM. The hub genes are high in GBM tumor tissues. The relative expression levels of LOX, SERPINH1 and TGFBI were significantly higher in GBM samples, compared with the normal brain tissues groups. Bioinformatics technology could be a useful tool to predict progression of GBM and to explore the mechanism of GBM.LOX, SERPINH1 and TGFBI may be involved in the mechanism of the occurrence and development of GBM, and may be used as molecular targets for early diagnosis and specific treatment. DEGs identified using GEO2R. Functional annotation of DEGs using Kyoto Encyclopedia of Genes and Genomes and gene body pathway enrichment analysis. Construction of a protein-protein interaction network. The pathway and process enrichment analysis of the hub genes were performed by Metascape. Survival analysis was performed in gene expression profiling interactive analysis. Real-time fluorescent quantitative polymerase chain reaction assay was performed to verify. The animal model was established for western blot test analysis.
Collapse
Affiliation(s)
- Shuyuan Zhang
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Weiwei Zhang
- Department of Operating Theater, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Bin Wu
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Liang Xia
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Liwen Li
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Kai Jin
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Yangfan Zou
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Caixing Sun
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
- * Correspondence: Caixing Sun, Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China (e-mail: )
| |
Collapse
|
7
|
The Potential Application of Extracellular Vesicles from Liquid Biopsies for Determination of Pharmacogene Expression. Pharmaceuticals (Basel) 2022; 15:ph15020252. [PMID: 35215364 PMCID: PMC8879428 DOI: 10.3390/ph15020252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Pharmacogenomics (PGx) entails the study of heritability of drug response. This may include both variability in genes related to pharmacokinetics (drug absorption, distribution, metabolism and excretion) and pharmacodynamics (e.g., drug receptors or signaling pathways). Individualizing drug therapy taking into account the genetic profile of the patient has the potential to make drug therapy safer and more effective. Currently, this approach relies on the determination of genetic variants in pharmacogenes by genotyping. However, it is widely acknowledged that large variability in gene expression is attributed to non-structural genetic variants. Therefore, at least from a theoretical viewpoint individualizing drug therapy based upon expression of pharmacogenes rather than on genotype may be advantageous but has been difficult to implement in the clinical setting. Extracellular vesicles (EVs) are lipid encapsulated structures that contain cargo such as lipids, nucleic acids and proteins. Since their cargo is tissue- and cell-specific they can be used to determine the expression of pharmacogenes in the liver. In this review, we describe methods of EV isolation and the potential of EVs isolated from liquid biopsies as a tool to determine the expression of pharmacogenes for use in personalized medicine.
Collapse
|
8
|
Śledzińska P, Bebyn MG, Furtak J, Kowalewski J, Lewandowska MA. Prognostic and Predictive Biomarkers in Gliomas. Int J Mol Sci 2021; 22:ijms221910373. [PMID: 34638714 PMCID: PMC8508830 DOI: 10.3390/ijms221910373] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Gliomas are the most common central nervous system tumors. New technologies, including genetic research and advanced statistical methods, revolutionize the therapeutic approach to the patient and reveal new points of treatment options. Moreover, the 2021 World Health Organization Classification of Tumors of the Central Nervous System has fundamentally changed the classification of gliomas and incorporated many molecular biomarkers. Given the rapid progress in neuro-oncology, here we compile the latest research on prognostic and predictive biomarkers in gliomas. In adult patients, IDH mutations are positive prognostic markers and have the greatest prognostic significance. However, CDKN2A deletion, in IDH-mutant astrocytomas, is a marker of the highest malignancy grade. Moreover, the presence of TERT promoter mutations, EGFR alterations, or a combination of chromosome 7 gain and 10 loss upgrade IDH-wildtype astrocytoma to glioblastoma. In pediatric patients, H3F3A alterations are the most important markers which predict the worse outcome. MGMT promoter methylation has the greatest clinical significance in predicting responses to temozolomide (TMZ). Conversely, mismatch repair defects cause hypermutation phenotype predicting poor response to TMZ. Finally, we discussed liquid biopsies, which are promising diagnostic, prognostic, and predictive techniques, but further work is needed to implement these novel technologies in clinical practice.
Collapse
Affiliation(s)
- Paulina Śledzińska
- Department of Thoracic Surgery and Tumors, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-067 Torun, Poland
- The F. Lukaszczyk Oncology Center, Molecular Oncology and Genetics Department, Innovative Medical Forum, 85-796 Bydgoszcz, Poland
| | - Marek G Bebyn
- The F. Lukaszczyk Oncology Center, Molecular Oncology and Genetics Department, Innovative Medical Forum, 85-796 Bydgoszcz, Poland
- Faculty of Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, 85-681 Bydgoszcz, Poland
- Franciszek Lukaszczyk Oncology Center, Department of Neurooncology and Radiosurgery, 85-796 Bydgoszcz, Poland
| | - Janusz Kowalewski
- Department of Thoracic Surgery and Tumors, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-067 Torun, Poland
| | - Marzena A Lewandowska
- Department of Thoracic Surgery and Tumors, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-067 Torun, Poland
- The F. Lukaszczyk Oncology Center, Molecular Oncology and Genetics Department, Innovative Medical Forum, 85-796 Bydgoszcz, Poland
| |
Collapse
|
9
|
Zhao X, Xiao Z, Li B, Li H, Yang B, Li T, Mei Z. miRNA-21 may serve as a promising noninvasive marker of glioma with a high diagnostic performance: a pooled analysis of 997 patients. Ther Adv Med Oncol 2021; 13:1758835920987650. [PMID: 33613699 PMCID: PMC7871292 DOI: 10.1177/1758835920987650] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Although various serum and tissue biomarkers have been investigated for
glioma diagnosis, no gold standard has been identified. miRNA-21 was
demonstrated to be a promising biomarker for the diagnosis of various brain
tumors, whereas there remains uncertainty concerning whether miRNA-21 could
be used as a good clinical diagnostic biomarker for glioma. The current
meta-analysis aimed to evaluate the diagnostic accuracy of miRNA-21 as a
potent biomarker in adults with suspected glioma. Methods: The Pubmed and Embase databases were searched systematically from inception
to January 2020 to identify relevant research reports. Pooled sensitivity,
specificity, positive likelihood ratio (PLR), negative likelihood ratio
(NLR), and diagnostic odds ratio (DOR) were calculated. Summary receiver
operating characteristic (SROC) curves were used to evaluate the overall
diagnostic performance. Meta-regression and subgroup analyses were conducted
to determine the source of heterogeneity and test the robustness of the
results. Results: From 5394 citations with 997 subjects that met the inclusion criteria, 11
studies were selected. Summary estimates of the diagnostic performance of
miRNA-21 were as follows: sensitivity, 0.83 [95% confidence interval (CI):
0.73–0.89]; specificity, 0.92 (95% CI: 0.85–0.96); PLR, 10.20 (95% CI:
5.10–20.30); NLR, 0.19 (95% CI: 0.12–0.31); and DOR, 54 (95% CI: 19–155).
The area under the SROC curve was 0.94 (95% CI: 0.92–0.96). Deeks’s funnel
plot revealed no evidence of publication bias (p = 0.59).
Meta-regression analysis suggested that study publication year could
attribute to the heterogeneity. Subgroup analysis found miRNA-21 had a
constant high diagnostic accuracy across different ethnicity, glioma grade,
sample source, and study region. Conclusion: This meta-analysis demonstrated that miRNA-21 has high diagnostic performance
and could serve as a promising noninvasive diagnostic marker for glioma.
Further large prospective studies are needed to validate its diagnostic
value and its prognostic significance and therapeutic effects.
Collapse
Affiliation(s)
- Xinli Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhihong Xiao
- Department of Spine Surgery, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Bin Li
- Department of Neurosurgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongwei Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, Henan Province, 450052, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Road, Xi'an 710032, China
| | - Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Anorectal Disease Institute of Shuguang Hospital, 528 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
10
|
Abstract
This review considers glioma molecular markers in brain tissues and body fluids, shows the pathways of their formation, and describes traditional methods of analysis. The most important optical properties of glioma markers in the terahertz (THz) frequency range are also presented. New metamaterial-based technologies for molecular marker detection at THz frequencies are discussed. A variety of machine learning methods, which allow the marker detection sensitivity and differentiation of healthy and tumor tissues to be improved with the aid of THz tools, are considered. The actual results on the application of THz techniques in the intraoperative diagnosis of brain gliomas are shown. THz technologies’ potential in molecular marker detection and defining the boundaries of the glioma’s tissue is discussed.
Collapse
|
11
|
Azam Z, TO ST, Tannous BA. Mesenchymal Transformation: The Rosetta Stone of Glioblastoma Pathogenesis and Therapy Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002015. [PMID: 33240762 PMCID: PMC7675056 DOI: 10.1002/advs.202002015] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/23/2020] [Indexed: 05/06/2023]
Abstract
Despite decades of research, glioblastoma (GBM) remains invariably fatal among all forms of cancers. The high level of inter- and intratumoral heterogeneity along with its biological location, the brain, are major barriers against effective treatment. Molecular and single cell analysis identifies different molecular subtypes with varying prognosis, while multiple subtypes can reside in the same tumor. Cellular plasticity among different subtypes in response to therapies or during recurrence adds another hurdle in the treatment of GBM. This phenotypic shift is induced and sustained by activation of several pathways within the tumor itself, or microenvironmental factors. In this review, the dynamic nature of cellular shifts in GBM and how the tumor (immune) microenvironment shapes this process leading to therapeutic resistance, while highlighting emerging tools and approaches to study this dynamic double-edged sword are discussed.
Collapse
Affiliation(s)
- Zulfikar Azam
- Experimental Therapeutics and Molecular Imaging UnitDepartment of NeurologyNeuro‐Oncology DivisionMassachusetts General Hospital and Harvard Medical SchoolBostonMA02129USA
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Shing‐Shun Tony TO
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Bakhos A. Tannous
- Experimental Therapeutics and Molecular Imaging UnitDepartment of NeurologyNeuro‐Oncology DivisionMassachusetts General Hospital and Harvard Medical SchoolBostonMA02129USA
| |
Collapse
|
12
|
Simon T, Jackson E, Giamas G. Breaking through the glioblastoma micro-environment via extracellular vesicles. Oncogene 2020; 39:4477-4490. [PMID: 32366909 PMCID: PMC7269906 DOI: 10.1038/s41388-020-1308-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/20/2022]
Abstract
Glioblastoma (GBM) is the most common and most aggressive brain tumour. Prognosis remains poor, despite the combined treatment of radio- and chemotherapy following surgical removal. GBM cells coexist with normal non-neoplastic cells, including endothelial cells, astrocytes and immune cells, constituting a complex and dynamic tumour micro-environment (TME). Extracellular vesicles (EVs) provide a critical means of bidirectional inter-cellular communication in the TME. Through delivery of a diverse range of genomic, lipidomic and proteomic cargo to neighbouring and distant cells, EVs can alter the phenotype and function of the recipient cell. As such, EVs have demonstrated their role in promoting angiogenesis, immune suppression, invasion, migration, drug resistance and GBM recurrence. Moreover, EVs can reflect the phenotype of the cells within the TME. Thus, in conjunction with their accessibility in biofluids, they can potentially serve as a biomarker reservoir for patient prognosis, diagnosis and predictive therapeutic response as well as treatment follow-up. Furthermore, together with the ability of EVs to cross the blood-brain barrier undeterred and through the exploitation of their cargo, EVs may provide an effective mean of drug delivery to the target site. Unveiling the mechanisms by which EVs within the GBM TME are secreted and target recipient cells may offer an indispensable understanding of GBM that holds the potential to provide a better prognosis and overall quality of life for GBM patients.
Collapse
Affiliation(s)
- Thomas Simon
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK.
| | - Ellen Jackson
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK.
| |
Collapse
|