1
|
Yao M, Su Y, Xiong R, Zhang X, Zhu X, Chen YC, Ao P. Deciphering the topological landscape of glioma using a network theory framework. Sci Rep 2024; 14:26724. [PMID: 39496747 PMCID: PMC11535471 DOI: 10.1038/s41598-024-77856-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 10/25/2024] [Indexed: 11/06/2024] Open
Abstract
Glioma stem cells have been recognized as key players in glioma recurrence and therapeutic resistance, presenting a promising target for novel treatments. However, the limited understanding of the role glioma stem cells play in the glioma hierarchy has drawn controversy and hindered research translation into therapies. Despite significant advances in our understanding of gene regulatory networks, the dynamics of these networks and their implications for glioma remain elusive. This study employs a systemic theoretical perspective to integrate experimental knowledge into a core endogenous network model for glioma, thereby elucidating its energy landscape through network dynamics computation. The model identifies two stable states corresponding to astrocytic-like and oligodendrocytic-like tumor cells, connected by a transition state with the feature of high stemness, which serves as one of the energy barriers between astrocytic-like and oligodendrocytic-like states, indicating the instability of glioma stem cells in vivo. We also obtained various stable states further supporting glioma's multicellular origins and uncovered a group of transition states that could potentially induce tumor heterogeneity and therapeutic resistance. This research proposes that the transition states linking both glioma stable states are central to glioma heterogeneity and therapy resistance. Our approach may contribute to the advancement of glioma therapy by offering a novel perspective on the complex landscape of glioma biology.
Collapse
Affiliation(s)
- Mengchao Yao
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai, China
| | - Yang Su
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai, China
| | - Ruiqi Xiong
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai, China
| | - Xile Zhang
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai, China
- Shanghai Shibei High School, Shanghai, China
| | - Xiaomei Zhu
- Shanghai Key Laboratory of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yong-Cong Chen
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai, China.
| | - Ping Ao
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
2
|
Samuel VP, Moglad E, Afzal M, Kazmi I, Alzarea SI, Ali H, Almujri SS, Abida, Imran M, Gupta G, Chinni SV, Tiwari A. Exploring Ubiquitin-specific proteases as therapeutic targets in Glioblastoma. Pathol Res Pract 2024; 260:155443. [PMID: 38981348 DOI: 10.1016/j.prp.2024.155443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Glioblastoma (GB) remains a formidable challenge and requires new treatment strategies. The vital part of the Ubiquitin-proteasome system (UPS) in cellular regulation has positioned it as a potentially crucial target in GB treatment, given its dysregulation oncolines. The Ubiquitin-specific proteases (USPs) in the UPS system were considered due to the garden role in the cellular processes associated with oncolines and their vital function in the apoptotic process, cell cycle regulation, and autophagy. The article provides a comprehensive summary of the evidence base for targeting USPs as potential factors for neoplasm treatment. The review considers the participation of the UPS system in the development, resulting in the importance of p53, Rb, and NF-κB, and evaluates specific goals for therapeutic administration using midnight proteasomal inhibitors and small molecule antagonists of E1 and E2 enzymes. Despite the slowed rate of drug creation, recent therapeutic discoveries based on USP system dynamics hold promise for specialized therapies. The review concludes with an analysis of future wanderers and the feasible effects of targeting USPs on personalized GB therapies, which can improve patient hydration in this current and unattractive therapeutic landscape. The manuscript emphasizes the possibility of USP oncogene therapy as a promising alternative treatment line for GB. It stresses the direct creation of research on the medical effectiveness of the approach.
Collapse
Affiliation(s)
- Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, the United Arab Emirates
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Al-Jouf, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Aseer 61421, Saudi Arabia
| | - Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, Jenjarom, Selangor 42610, Malaysia
| | - Abhishek Tiwari
- Department of Pharmacy, Pharmacy Academy, IFTM University, Lodhipur-Rajpur, Moradabad 244102, India.
| |
Collapse
|
3
|
Zheng P, Zhang X, Ren D, Bai Q. Alternative Splicing in Glioblastoma and its Clinical Implication in Outcome Prediction. Neurol India 2024; 72:846-855. [PMID: 39216044 DOI: 10.4103/neurol-india.ni_1219_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 06/08/2022] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Alternative splicing (AS) offers an important mechanism to form protein polymorphism. A growing body of evidence indicates the correlation between splicing abnormality and carcinoma. Nevertheless, an overall analysis of AS signatures in glioblastoma (GBM) is absent and urgently needed. METHODS TCGA SpliceSea data was used to evaluate the AS profiles and further classified into different AS events. The survival analysis was based on these AS events, and AS-related genes were identified and performed with enrichment analysis. At last, the splicing factor-AS regulatory network was established in Cytoscape. RESULTS Eight hundred forty-two splicing events were confirmed as prognostic molecular events in GBM. Furthermore, the final prognostic signature constructed by seven AS events gave good result with an area under the curve (AUC) of receiver operating characteristic (ROC) curve up to 0.935 for five years, showing high potency in predicting patients' outcome. We built the splicing regulatory network to show the internal relationship of splicing events in GBM. PC4 and SFRS1 interacting protein 1 (PSIP1) and histone H4 acetylation may play a significant part in the prognosis induced by splicing events. CONCLUSION In our study, a high-efficiency prognostic prediction model was built for GBM patients based on AS events, which could become potential prognostic biomarkers for GBM. Meanwhile, PSIP1 may be a critical target for pharmaceutical treatment.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, China
- Department of Key Laboratory, Shanghai Pudong New Area People's Hospital, China
| | - Xiaoxue Zhang
- Department of Key Laboratory, Shanghai Pudong New Area People's Hospital, China
| | - Dabin Ren
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, China
| | - Qingke Bai
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, China
| |
Collapse
|
4
|
Zheng P, Zhang X, Ren D, Bai Q. Identification and Prognostic Value of m6A-Related Genes in Glioblastoma. Neurol India 2024; 72:830-836. [PMID: 39216042 DOI: 10.4103/neurol-india.ni_1166_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/23/2021] [Indexed: 09/04/2024]
Abstract
BACKGROUND N6-methyladenosine (m6A) is one of the most common forms of mRNA modification, which is dynamically regulated by the m6A-related genes; however, its effect in glioblastoma (GBM) is still unknown. OBJECTIVE We sought to investigate the association between m6A-related genes (m6A-RGs) and GBM. METHODS Transcriptome data and the relevant clinical data were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases. The m6A-RGs were identified from differently expressed genes, and COX and lasso regression models were applied to locate the prognosis-related genes. RESULTS We identified 15 out of 19 m6A-RGs differentially expressed between GBM and nontumor tissues. We identified two subgroups of GBM (clusters 1 and 2) by applying consensus clustering. Compared with the cluster 1 subgroup, the cluster 1 subgroup correlates with a poorer prognosis, and most of the 19 m6A-RGs are higher expressed in cluster 1. Through univariate Cox and lasso regression model, we identified three m6A-RGs, namely HNRNPC, ALKBH5, and FTO, which were used to construct a Cox regression risk model to predict the prognosis of GBM patients. CONCLUSION We identified a valuable m6A model for predicting the prognosis of GBM patients, which can provide useful epigenetic biomarkers.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Neurosurgery, Shanghai Pudong New area People's Hospital, Shanghai, China
- Key Molecular Lab, Shanghai Pudong New area People's Hospital, Shanghai, China
| | - Xiaoxue Zhang
- Key Molecular Lab, Shanghai Pudong New area People's Hospital, Shanghai, China
| | - Dabin Ren
- Department of Neurosurgery, Shanghai Pudong New area People's Hospital, Shanghai, China
| | - Qingke Bai
- Neurology, Shanghai Pudong New area People's Hospital, Shanghai, China
| |
Collapse
|
5
|
Esteves L, Caramelo F, Roda D, Carreira IM, Melo JB, Ribeiro IP. Identification of Novel Molecular and Clinical Biomarkers of Survival in Glioblastoma Multiforme Patients: A Study Based on The Cancer Genome Atlas Data. BIOMED RESEARCH INTERNATIONAL 2024; 2024:5582424. [PMID: 38606198 PMCID: PMC11008977 DOI: 10.1155/2024/5582424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/14/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent type of brain tumour; although advancements in treatment have been made, the median survival time for GBM patients has persisted at 15 months. This study is aimed at investigating the genetic alterations and clinical features of GBM patients to find predictors of survival. GBM patients' methylation and gene expression data along with clinical information from TCGA were retrieved. The most overrepresented pathways were identified independently for each omics dataset. From the genes found in at least 30% of these pathways, one gene that was identified in both sets was further examined using the Kaplan-Meier method for survival analysis. Additionally, three groups of patients who started radio and chemotherapy at different times were identified, and the influence of these variations in treatment modality on patient survival was evaluated. Four pathways that seemed to negatively impact survival and two with the opposite effect were identified. The methylation status of PRKCB was highlighted as a potential novel biomarker for patient survival. The study also found that treatment with chemotherapy prior to radiotherapy can have a significant impact on patient survival, which could lead to improvements in clinical management and therapeutic approaches for GBM patients.
Collapse
Affiliation(s)
- Luísa Esteves
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Francisco Caramelo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Laboratory of Biostatistics and Medical Informatics, iCBR-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Domingos Roda
- Algarve Radiation Oncology Unit-Joaquim Chaves Saúde (JCS), Faro, Portugal
| | - Isabel Marques Carreira
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Joana Barbosa Melo
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ilda Patrícia Ribeiro
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| |
Collapse
|
6
|
Soleiman M, Fathi-Roudsari M, Khajeh K, Maghsoudi A. Optimization of Epigenetic Modifier Drug Combination for Synergistic Effect against Glioblastoma Multiform Cancer Cell Lines. Cancer Invest 2024; 42:319-332. [PMID: 38695671 DOI: 10.1080/07357907.2024.2345183] [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: 10/18/2023] [Accepted: 04/16/2024] [Indexed: 05/28/2024]
Abstract
Glioblastoma multiforme (GBM), is a frequent class of malignant brain tumors. Epigenetic therapy, especially with synergistic combinations is highly paid attention for aggressive solid tumors like GBM. Here, RSM optimization has been used to increase the efficient arrest of U87 and U251 cell lines due to synergistic effects. Cell lines were treated with SAHA, 5-Azacytidine, GSK-126, and PTC-209 individually and then RSM was used to find most effective combinations. Results showed that optimized combinations significantly reduce cell survival and induce cell cycle arrest and apoptosis in both cell lines. Expression of cyclin B1 and cyclin D1 were decreased while caspase3 increased expression.
Collapse
Affiliation(s)
- Morvarid Soleiman
- Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehrnoosh Fathi-Roudsari
- Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Khosro Khajeh
- Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
7
|
Yuan X, Sun X, Zhou B, Zhao S, Li Y, Ming H. HSPA4 regulated glioma progression via activation of AKT signaling pathway. Biochem Cell Biol 2024; 102:159-168. [PMID: 37339521 DOI: 10.1139/bcb-2022-0321] [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] [Indexed: 06/22/2023] Open
Abstract
Glioma is still an incurable disease with high invasiveness. Heat shock 70 kDa protein 4 (HSPA4) is a member of the HSP110 family, and is associated with the development and progression of various cancers. In the current study, we assessed the expression of HSPA4 in clinical samples, and found that HSPA4 was up-regulated in glioma tissues and correlated with tumor recurrence and grade. Survival analyses demonstrated that glioma patients with high HSPA4 expression had lower overall survival and disease-free survival times. In vitro knockdown of HSPA4 inhibited glioma cell proliferation, mediated cell cycle arrest at G2 phase and apoptosis, and reduced the migration ability. In vivo, the growth of HSPA4-knockdown xenografts was markedly suppressed compared to the tumors formed by HSPA4-positive control cells. Additionally, Gene set enrichment analyses disclosed that HSPA4 was associated with the PI3K/Akt signaling pathway. The regulatory effect of the AKT activator SC79 on cell proliferation and apoptosis was suppressed by HSPA4 knockdown, indicating that HSPA4 is capable of promoting glioma development. In summary, these data showed that HSPA4 is likely to play a pivotal role in the progression of glioma, and consequently may be a promising therapeutic target for glioma therapy.
Collapse
Affiliation(s)
- Xi Yuan
- Department of Radiation Oncology, Jinling Hospital of Nanjing University, No.305, Zhongshan East Road, Nanjing, Jiangsu Province 210002, China
| | - Xiangdong Sun
- Department of Radiation Oncology, Jinling Hospital of Nanjing University, No.305, Zhongshan East Road, Nanjing, Jiangsu Province 210002, China
| | - Bin Zhou
- Department of Radiation Oncology, Jinling Hospital of Nanjing University, No.305, Zhongshan East Road, Nanjing, Jiangsu Province 210002, China
| | - Shuang Zhao
- Department of Radiation Oncology, Jinling Hospital of Nanjing University, No.305, Zhongshan East Road, Nanjing, Jiangsu Province 210002, China
| | - Yikun Li
- Department of Radiation Oncology, Jinling Hospital of Nanjing University, No.305, Zhongshan East Road, Nanjing, Jiangsu Province 210002, China
| | - Haolang Ming
- Department of Neurosurgery, Tianjin Medical University General Hospital, No.154 Anshan Road, Heping District, Tianjin 300052, China
| |
Collapse
|
8
|
Zheng P, Zhang X, Ren D, Bai Q. Classification of Glioblastoma Associated with Immune Checkpoints and Tumor Microenvironment based on Immunogenomic Profiling. Neurol India 2024; 72:297-303. [PMID: 38691473 DOI: 10.4103/ni.ni_1070_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 08/01/2022] [Indexed: 05/03/2024]
Abstract
BACKGROUND Immune microenvironment is involved in tumor initiation and progression, and its effect on glioblastoma (GBM) is still unknown. OBJECT We sought to investigate the association between immune status and GBM. METHODS Transcriptome data and the relevant clinical data were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus (GEO) databases, and we identified two immune subtypes based on 29 immune-associated gene sets. RESULTS Through single-sample gene set enrichment analysis (ssGSEA), we found that the high-immunity subtype had the most tumor-infiltrating immune cells and immune checkpoint molecules in GBM patients. Furthermore, we could more effectively identify immune signature pathways in GBM. CONCLUSION After validation with the GEO dataset, we conclude that the identified GBM high-immune subtypes may be amenable to the application of novel immune therapy for GBM.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
- Department of Key Molecular Lab, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Xiaoxue Zhang
- Department of Key Molecular Lab, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Dabin Ren
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Qingke Bai
- Department of Neurology, Shanghai Pudong New Area People's Hospital, Shanghai, China
| |
Collapse
|
9
|
Li G, Ma L, Feng C, Yin H, Bao J, Wu D, Zhang Z, Li X, Li Z, Yang C, Wang H, Fang F, Hu X, Li M, Xu L, Xu Y, Liang H, Yang T, Wang J, Pan J. MZ1, a BRD4 inhibitor, exerted its anti-cancer effects by suppressing SDC1 in glioblastoma. BMC Cancer 2024; 24:220. [PMID: 38365636 PMCID: PMC10870565 DOI: 10.1186/s12885-024-11966-8] [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: 12/10/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a relatively prevalent primary tumor of the central nervous system in children, characterized by its high malignancy and mortality rates, along with the intricate challenges of achieving complete surgical resection. Recently, an increasing number of studies have focused on the crucial role of super-enhancers (SEs) in the occurrence and development of GBM. This study embarks on the task of evaluating the effectiveness of MZ1, an inhibitor of BRD4 meticulously designed to specifically target SEs, within the intricate framework of GBM. METHODS The clinical data of GBM patients was sourced from the Chinese Glioma Genome Atlas (CGGA) and the Gene Expression Profiling Interactive Analysis 2 (GEPIA2), and the gene expression data of tumor cell lines was derived from the Cancer Cell Line Encyclopedia (CCLE). The impact of MZ1 on GBM was assessed through CCK-8, colony formation assays, EdU incorporation analysis, flow cytometry, and xenograft mouse models. The underlying mechanism was investigated through RNA-seq and ChIP-seq analyses. RESULTS In this investigation, we made a noteworthy observation that MZ1 exhibited a substantial reduction in the proliferation of GBM cells by effectively degrading BRD4. Additionally, MZ1 displayed a notable capability in inducing significant cell cycle arrest and apoptosis in GBM cells. These findings were in line with our in vitro outcomes. Notably, MZ1 administration resulted in a remarkable decrease in tumor size within the xenograft model with diminished toxicity. Furthermore, on a mechanistic level, the administration of MZ1 resulted in a significant suppression of pivotal genes closely associated with cell cycle regulation and epithelial-mesenchymal transition (EMT). Interestingly, our analysis of RNA-seq and ChIP-seq data unveiled the discovery of a novel prospective oncogene, SDC1, which assumed a pivotal role in the tumorigenesis and progression of GBM. CONCLUSION In summary, our findings revealed that MZ1 effectively disrupted the aberrant transcriptional regulation of oncogenes in GBM by degradation of BRD4. This positions MZ1 as a promising candidate in the realm of therapeutic options for GBM treatment.
Collapse
Affiliation(s)
- Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Liya Ma
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, P.R. China
| | - Chenxi Feng
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Jianping Bao
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Di Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Chun Yang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Hairong Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Xiaohan Hu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Mei Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Lixiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Yunyun Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Hansi Liang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Tianquan Yang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China.
| | - Jianwei Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China.
| |
Collapse
|
10
|
Liu Y, Jin H, Liu H. Identification of T-cell exhaustion-related gene signature for predicting prognosis in glioblastoma multiforme. J Cell Mol Med 2023; 27:3503-3513. [PMID: 37635346 PMCID: PMC10660619 DOI: 10.1111/jcmm.17927] [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: 03/29/2023] [Revised: 05/16/2023] [Accepted: 05/26/2023] [Indexed: 08/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly malignant primary brain tumour with a poor prognosis in adults. Identifying biomarkers that can aid in the molecular classification and risk stratification of GBM is critical. Here, we conducted a transcriptional profiling analysis of T-cell immunity in the tumour microenvironment of GBM patients and identified two novel T cell exhaustion (TEX)-related GBM subtypes (termed TEX-C1 and TEX-C2) using the consensus clustering. Our multi-omics analysis revealed distinct immunological, molecular and clinical characteristics for these two subtypes. Specifically, the TEX-C1 subtype had higher infiltration levels of immune cells and expressed higher levels of immune checkpoint molecules than the TEX-C2 subtype. Functional analysis revealed that upregulated genes in the TEX-C1 subtype were significantly enriched in immune response and signal transduction pathways, and upregulated genes in the TEX-C2 subtype were predominantly associated with cell fate and nervous system development pathways. Notably, patients with activated T-cell activity status in the TEX-C1 subgroup demonstrated a significantly worse prognosis than those with severe T cell exhaustion status in the TEX-C2 subgroup. Finally, we proposed a machine-learning-derived novel gene signature comprising 12 TEX-related genes (12TexSig) to indicate tumour subtyping. In the TCGA cohort, the 12TexSig demonstrated the ability to accurately predict the prognosis of GBM patients, and this prognostic value was further confirmed in two independent external cohorts. Taken together, our results suggest that the TEX-derived subtyping and gene signature has the potential to serve as a clinically helpful biomarker for guiding the management of GBM patients, pending further prospective validation.
Collapse
Affiliation(s)
- Yue‐hui Liu
- Department of NeurologyAffiliated Hospital of Inner Mongolia Minzu UniversityTongliaoChina
| | - Hong‐quan Jin
- Department of NeurologyAffiliated Hospital of Inner Mongolia Minzu UniversityTongliaoChina
| | - Hai‐ping Liu
- College of Life Science and Food EngineeringInner Mongolia Minzu UniversityTongliaoChina
| |
Collapse
|
11
|
Zheng P, Zhang X, Ren D, Bai Q, Jiang P. Novel Immune-Related LncRNA Pairs are Associated with Immunol Infiltration and Survival Status in Glioblastoma. Neurol India 2023; 71:1226-1234. [PMID: 38174463 DOI: 10.4103/0028-3886.391381] [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] [Indexed: 01/05/2024]
Abstract
Background Immune-related lncRNA is involved in tumor initiation and progression, while its effect in glioblastoma (GBM) is still unknown. Objective We sought to investigate the association between immune-related lncRNA (ir-lncRNA) and GBM. Methods Transcriptomic and clinical data were obtained from the TCGA dataset, and we found 2008 ir-lncRNA differentially expressed between GBM and adjacent brain tissues. Results Appling the univariate Cox and Lasso regression model, we found 30 prognosis-related ir-lncRNA pairs to construct a Cox regression risk model to associate the outcome of GBM patients. Furthermore, with this risk model, we can identify the tumor immune infiltration status, the expression of immunosuppressive biomarkers, and chemical sensitivity in GBM patients. Conclusions We constructed an immunologic risk model with lncRNA to associate the survival outcome of GBM patients, which can provide useful biomarkers.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital; Key Molecular Lab, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Xiaoxue Zhang
- Key Molecular Lab, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Dabin Ren
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Qingke Bai
- Department of Neurology, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Ping Jiang
- Department of Nursing, Shanghai Pudong New Area People's Hospital, Shanghai, China
| |
Collapse
|
12
|
de Luna FCF, Ferreira WAS, Casseb SMM, de Oliveira EHC. Anticancer Potential of Flavonoids: An Overview with an Emphasis on Tangeretin. Pharmaceuticals (Basel) 2023; 16:1229. [PMID: 37765037 PMCID: PMC10537037 DOI: 10.3390/ph16091229] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Natural compounds with pharmacological activity, flavonoids have been the subject of an exponential increase in studies in the field of scientific research focused on therapeutic purposes due to their bioactive properties, such as antioxidant, anti-inflammatory, anti-aging, antibacterial, antiviral, neuroprotective, radioprotective, and antitumor activities. The biological potential of flavonoids, added to their bioavailability, cost-effectiveness, and minimal side effects, direct them as promising cytotoxic anticancer compounds in the optimization of therapies and the search for new drugs in the treatment of cancer, since some extensively antineoplastic therapeutic approaches have become less effective due to tumor resistance to drugs commonly used in chemotherapy. In this review, we emphasize the antitumor properties of tangeretin, a flavonoid found in citrus fruits that has shown activity against some hallmarks of cancer in several types of cancerous cell lines, such as antiproliferative, apoptotic, anti-inflammatory, anti-metastatic, anti-angiogenic, antioxidant, regulatory expression of tumor-suppressor genes, and epigenetic modulation.
Collapse
Affiliation(s)
- Francisco Canindé Ferreira de Luna
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SEAMB), Evandro Chagas Institute (IEC), BR 316, KM 7, s/n, Levilândia, Ananindeua 67030-000, Brazil; (W.A.S.F.); (E.H.C.d.O.)
| | - Wallax Augusto Silva Ferreira
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SEAMB), Evandro Chagas Institute (IEC), BR 316, KM 7, s/n, Levilândia, Ananindeua 67030-000, Brazil; (W.A.S.F.); (E.H.C.d.O.)
| | | | - Edivaldo Herculano Correa de Oliveira
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SEAMB), Evandro Chagas Institute (IEC), BR 316, KM 7, s/n, Levilândia, Ananindeua 67030-000, Brazil; (W.A.S.F.); (E.H.C.d.O.)
- Faculty of Natural Sciences, Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), Rua Augusto Correa, 01, Belém 66075-990, Brazil
| |
Collapse
|
13
|
Ferrasi AC, Puttini R, Galvani AF, Hamamoto Filho PT, Delafiori J, Argente VD, de Oliveira AN, Dias-Audibert FL, Catharino RR, Silva OC, Zanini MA, Kurokawa GA, Lima EO. Metabolomics Approach Reveals Important Glioblastoma Plasma Biomarkers for Tumor Biology. Int J Mol Sci 2023; 24:8813. [PMID: 37240159 PMCID: PMC10218163 DOI: 10.3390/ijms24108813] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/26/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Glioblastoma (GB) is the most aggressive and frequent primary malignant tumor of the central nervous system and is associated with poor overall survival even after treatment. To better understand tumor biochemical alterations and broaden the potential targets of GB, this study aimed to evaluate differential plasma biomarkers between GB patients and healthy individuals using metabolomics analysis. Plasma samples from both groups were analyzed via untargeted metabolomics using direct injection with an electrospray ionization source and an LTQ mass spectrometer. GB biomarkers were selected via Partial Least Squares Discriminant and Fold-Change analyses and were identified using tandem mass spectrometry with in silico fragmentation, consultation of metabolomics databases, and a literature search. Seven GB biomarkers were identified, some of which were unprecedented biomarkers for GB, including arginylproline (m/z 294), 5-hydroxymethyluracil (m/z 143), and N-acylphosphatidylethanolamine (m/z 982). Notably, four other metabolites were identified. The roles of all seven metabolites in epigenetic modulation, energy metabolism, protein catabolism or folding processes, and signaling pathways that activate cell proliferation and invasion were elucidated. Overall, the findings of this study highlight new molecular targets to guide future investigations on GB. These molecular targets can also be further evaluated to derive their potential as biomedical analytical tools for peripheral blood samples.
Collapse
Affiliation(s)
- Adriana C. Ferrasi
- Laboratory of Molecular Analysis and Neuro-Oncology, Department of Internal Medicine, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (A.C.F.); (R.P.); (A.F.G.); (V.D.A.); (O.C.S.); (G.A.K.)
| | - Ricardo Puttini
- Laboratory of Molecular Analysis and Neuro-Oncology, Department of Internal Medicine, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (A.C.F.); (R.P.); (A.F.G.); (V.D.A.); (O.C.S.); (G.A.K.)
| | - Aline F. Galvani
- Laboratory of Molecular Analysis and Neuro-Oncology, Department of Internal Medicine, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (A.C.F.); (R.P.); (A.F.G.); (V.D.A.); (O.C.S.); (G.A.K.)
| | - Pedro T. Hamamoto Filho
- Department of Neurology, Psychology and Psychiatry, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (P.T.H.F.); (M.A.Z.)
| | - Jeany Delafiori
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas 13.083-877, Brazil; (J.D.); (A.N.d.O.); (F.L.D.-A.); (R.R.C.)
| | - Victoria D. Argente
- Laboratory of Molecular Analysis and Neuro-Oncology, Department of Internal Medicine, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (A.C.F.); (R.P.); (A.F.G.); (V.D.A.); (O.C.S.); (G.A.K.)
| | - Arthur N. de Oliveira
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas 13.083-877, Brazil; (J.D.); (A.N.d.O.); (F.L.D.-A.); (R.R.C.)
| | - Flávia L. Dias-Audibert
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas 13.083-877, Brazil; (J.D.); (A.N.d.O.); (F.L.D.-A.); (R.R.C.)
| | - Rodrigo R. Catharino
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, University of Campinas, Campinas 13.083-877, Brazil; (J.D.); (A.N.d.O.); (F.L.D.-A.); (R.R.C.)
| | - Octavio C. Silva
- Laboratory of Molecular Analysis and Neuro-Oncology, Department of Internal Medicine, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (A.C.F.); (R.P.); (A.F.G.); (V.D.A.); (O.C.S.); (G.A.K.)
| | - Marco A. Zanini
- Department of Neurology, Psychology and Psychiatry, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (P.T.H.F.); (M.A.Z.)
| | - Gabriel A. Kurokawa
- Laboratory of Molecular Analysis and Neuro-Oncology, Department of Internal Medicine, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (A.C.F.); (R.P.); (A.F.G.); (V.D.A.); (O.C.S.); (G.A.K.)
| | - Estela O. Lima
- Laboratory of Molecular Analysis and Neuro-Oncology, Department of Internal Medicine, Botucatu Medical School, São Paulo State University, Botucatu 18.618-687, Brazil; (A.C.F.); (R.P.); (A.F.G.); (V.D.A.); (O.C.S.); (G.A.K.)
| |
Collapse
|
14
|
Zhang Y, Zhang Y, He P, Ge F, Huo Z, Qiao G. The genetic liability to rheumatoid arthritis may decrease hepatocellular carcinoma risk in East Asian population: a Mendelian randomization study. Arthritis Res Ther 2023; 25:49. [PMID: 36973792 PMCID: PMC10041783 DOI: 10.1186/s13075-023-03029-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) have a rising possibility of acquiring certain kinds of cancers than the general public. The causal risk association between RA and hepatocellular carcinoma (HCC) remains unknown. METHODS Genetic summary data from genome-wide association study (GWAS), including RA (n = 19,190) and HCC (n = 197,611), was analyzed. The inverse-variance weighted (IVW) approach was used as the principal analysis, complemented with weighted median, weighted mode, simple median method, and MR-Egger analyses. The genetic data of RA (n = 212,453) was used to verify the results in eastern Asia populations. RESULTS The results from the IVW methods indicated that genetically predicted RA was significantly linked with a declined possibility of HCC for East Asians (OR = 0.86; 95% CI: 0.78, 0.95; p = 0.003). The weighted median and the weighted mode also supported similar results (all p < 0.05). Additionally, neither the funnel plots nor the MR-Egger intercepts revealed any directional pleiotropic effects between RA and HCC. Moreover, the other set of RA data validated the results. CONCLUSION The RA may decrease the risk of being susceptible to the HCC in eastern Asia populations, which was beyond expectation. In the future, additional investigations should be made into potential biomedical mechanisms.
Collapse
Affiliation(s)
- Yuzhuo Zhang
- Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Yudong Zhang
- Department of Thoracic Surgery & Department of Cardiothoracic Surgery of East Division, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510062, Guangdong, China
| | - Peng He
- Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Fan Ge
- Guangzhou Medical University, Guangzhou, 511436, Guangdong, China.
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhenyu Huo
- Guangzhou Medical University, Guangzhou, 511436, Guangdong, China.
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
- Shantou University Medical College, Shantou, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Kundu M, Das S, Das CK, Kulkarni G, Das S, Dhara D, Mandal M. Magnolol induces cytotoxic autophagy in glioma by inhibiting PI3K/AKT/mTOR signaling. Exp Cell Res 2023; 424:113488. [PMID: 36736226 DOI: 10.1016/j.yexcr.2023.113488] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/18/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023]
Abstract
Glioma is difficult-to-treat because of its infiltrative nature and the presence of the blood-brain barrier. Temozolomide is the only FDA-approved drug for its management. Therefore, finding a novel chemotherapeutic agent for glioma is of utmost importance. Magnolol, a neolignan, has been known for its apoptotic role in glioma. In this work, we have explored a novel anti-glioma mechanism of Magnolol associated with its role in autophagy modulation. We found increased expression levels of Beclin-1, Atg5-Atg12, and LC3-II and lower p62 expression in Magnolol-treated glioma cells. PI3K/AKT/mTOR pathway proteins were also downregulated in Magnolol-treated glioma cells. Next, we treated the glioma cells with Insulin, a stimulator of PI3K/AKT/mTOR signaling, to confirm that Magnolol induced autophagy by inhibiting this pathway. Insulin reversed the effect on Magnolol-mediated autophagy induction. We also established the same in in vivo glioma model where Magnolol showed an anti-glioma effect by inducing autophagy. To confirm the cytotoxic effect of Magnolol-induced autophagy, we used Chloroquine, a late-stage autophagy inhibitor. Chloroquine efficiently reversed the anti-glioma effects of Magnolol both in vitro and in vivo. Our study revealed the cytotoxic effect of Magnolol-induced autophagy in glioma, which was not previously reported. Additionally, Magnolol showed no toxicity in non-cancerous cell lines as well as rat organs. Thus, we concluded that Magnolol is an excellent candidate for developing new therapeutic strategies for glioma management.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Chandan Kanta Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Gaurav Kulkarni
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Soumen Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Dibakar Dhara
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
16
|
Single-Cell Sequencing Reveals Necroptosis-Related Prognostic Genes of Glioblastoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2926655. [PMID: 36860730 PMCID: PMC9970716 DOI: 10.1155/2023/2926655] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/18/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023]
Abstract
Background Glioblastoma (GBM) is one of the most malignant forms of brain cancer, with the extremely lower survival rate. Necroptosis (NCPS) is also one of the most wide types of cell death, and its clinical importance in GBM is not clear. Methods We first identified necroptotic genes in GBM by single-cell RNA sequencing analysis of our surgical samples and weighted coexpression network analysis (WGNCA) from TCGA GBM data. The cox regression model with least absolute shrinkage and selection operator (LASSO) was used to construct the risk model. Then, KM plot and reactive operation curve (ROC) analysis were used to assess the prediction ability of the model. At last, the infiltrated immune cells and gene mutation profiling were investigated between the high- and low-NCPS groups as well. Result The risk model including ten necroptosis-related genes was identified as an independent risk factor for the outcome. In addition, we found that the risk model is correlated with the infiltrated immune cells and tumor mutation burden in GBM. NDUFB2 is identified to be a risk gene in GBM with bioinformatical analysis and in vitro experiment validation. Conclusion This risk model of necroptosis-related genes might provide clinical evidence for GBM interventions.
Collapse
|
17
|
Chen D, Xu Y, Gao X, Zhu X, Liu X, Yan D. A novel signature of cuproptosis-related lncRNAs predicts prognosis in glioma: Evidence from bioinformatic analysis and experiments. Front Pharmacol 2023; 14:1158723. [PMID: 37101543 PMCID: PMC10123286 DOI: 10.3389/fphar.2023.1158723] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/29/2023] [Indexed: 04/28/2023] Open
Abstract
Background: Glioma patients often experience unfavorable outcomes and elevated mortality rates. Our study established a prognostic signature utilizing cuproptosis-associated long non-coding RNAs (CRLs) and identified novel prognostic biomarkers and therapeutic targets for glioma. Methods: The expression profiles and related data of glioma patients were obtained from The Cancer Genome Atlas, an accessible online database. We then constructed a prognostic signature using CRLs and evaluated the prognosis of glioma patients by means of Kaplan-Meier survival curves and receiver operating characteristic curves. A nomogram based on clinical features was employed to predict the individual survival probability of glioma patients. Functional enrichment analysis was conducted to identify crucial CRL-related enriched biological pathways. The role of LEF1-AS1 in glioma was validated in two glioma cell lines (T98 and U251). Results: We developed and validated a prognostic model for glioma with 9 CRLs. Patients with low-risk had a considerably longer overall survival (OS). The prognostic CRL signature may serve independently as an indicator of prognosis for glioma patients. In addition, functional enrichment analysis revealed significant enrichment of multiple immunological pathways. Notable differences were observed between the two risk groups in terms of immune cell infiltration, function, and immune checkpoints. We further identified four drugs based on their different IC50 values from the two risk groups. Subsequently, we discovered two molecular subtypes of glioma (cluster one and cluster two), with the cluster one subtype exhibiting a remarkably longer OS compared to the cluster two subtype. Finally, we observed that inhibition of LEF1-AS1 curbed the proliferation, migration, and invasion of glioma cells. Conclusion: The CRL signatures were confirmed as a reliable prognostic and therapy response indicator for glioma patients. Inhibition of LEF1-AS1 effectively suppressed the growth, migration, and invasion of gliomas; therefore, LEF1-AS1 presents itself as a promising prognostic biomarker and potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuan Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xueping Gao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuqiang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Dongming Yan,
| |
Collapse
|
18
|
Zhang M, Huang J, Wang Y, Nie Q, Zhang X, Yang Y, Mao G. Cathepsin a upregulation in glioma: A potential therapeutic target associated with immune infiltration. J Med Biochem 2022; 41:459-465. [PMID: 36381072 PMCID: PMC9618335 DOI: 10.5937/jomb0-35677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/03/2022] [Indexed: 12/10/2023] Open
Abstract
BACKGROUND Glioma is the result of malignant transformation of glial cells in the white matter of the brain or spinal cord and accounts for approximately 80% of all intracranial malignancies. Cathepsin A (CTSA) is highly expressed in a variety of tumor tissues, but its role in glioma is poorly studied. This study analyses the relationship between CTSA, and glioma based on The Cancer Genome Atlas (TCGA). METHODS Data for glioma patients were collected from TCGA. The expression level of CTSA was compared between paired glioma tissues and normal tissues with Wilcoxon rank-sum test. In addition, the Wilcoxon ranksum test was also applied to analyze the relationship between clinicopathologic features and CTSA expression. Kaplan-Meier Plotter was applied to analyze OS, DSS and PFI. Immuno-infiltration analysis of BLCA was performed by single sample gene set enrichment analysis (ssGSEA) in the "GSVA" R package. RESULTS The CTSA was overexpressed in glioma tissues compared to normal tissues (P<0.001). The high expression of CTSA was significantly related to 1p/19q codeletion, IDH, WHO grade and histological type. Kaplan-Meier survival analysis showed that patients with glioma characterized with high expressed CTSA had a poorer OS (HR=2.16 P<0.001), DSS (HR=2.17 P<0.001) and PFI (HR=1.48 P<0.001) than patients with low CTSA expression. Moreover, High expressed CTSA was associated with immune cell infiltration. CONCLUSIONS CTSA may serve as a candidate prognostic biomarker for determining prognosis associated with immune infiltration in glioma.
Collapse
Affiliation(s)
- Ming Zhang
- The Third Medical Centre Chinese PLA (People's Liberation Army) General Hospital, Department of Neurosurgery, Beijing, China
| | - Jun Huang
- The Third Medical Centre Chinese PLA (People's Liberation Army) General Hospital, Department of Neurosurgery, Beijing, China
| | - Yunfei Wang
- The Third Medical Centre Chinese PLA (People's Liberation Army) General Hospital, Department of Neurosurgery, Beijing, China
| | - Qingbin Nie
- The Third Medical Centre Chinese PLA (People's Liberation Army) General Hospital, Department of Neurosurgery, Beijing, China
| | - Xinye Zhang
- The Third Medical Centre Chinese PLA (People's Liberation Army) General Hospital, Department of Neurosurgery, Beijing, China
| | - Yufeng Yang
- The Third Medical Centre Chinese PLA (People's Liberation Army) General Hospital, Department of Neurosurgery, Beijing, China
| | - Gengsheng Mao
- The Third Medical Centre Chinese PLA (People's Liberation Army) General Hospital, Department of Neurosurgery, Beijing, China
| |
Collapse
|
19
|
Wang P, Wang T, Dong L, Xu Z, Guo S, Chang C. Circular RNA circ_0079593 facilitates glioma development via modulating miR-324-5p/XBP1 axis. Metab Brain Dis 2022; 37:2389-2403. [PMID: 35793013 DOI: 10.1007/s11011-022-01040-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
Glioma is a common brain tumor with high mortality. Circular RNAs (circRNAs) play crucial roles in tumor occurrence and development. However, the function and molecular basis of circ_0079593 in glioma remain unknown. Quantitative real-time PCR (qPCR) and Western blot were used for expression determination of circ_0079593, microRNA-324-5p (miR-324-5p) and X-box binding protein 1 (XBP1). Cell Counting Kit-8 (CCK-8), colony formation, flow cytometry, transwell assays, and tube formation assay were employed to evaluate cell functions. Glycolysis was determined via detecting glucose consumption, lactate production and ATP level. The binding relationship between miR-324-5p and circ_0079593 or XBP1 was validated by dual-luciferase reporter assay and RNA Immunoprecipitation (RIP) assay. Besides, xenograft assay was applied to test tumor growth in vivo. Circ_0079593 and XBP1 levels were elevated, while miR-324-5p level was declined in glioma. Silencing of circ_0079593 restrained proliferation, mobility, angiogenesis and glycolysis and induced apoptosis in glioma cells. Circ_0079593 accelerated glioma progression via sequestering miR-324-5p, one of the targets of circ_0079593. XBP1 was a target gene of miR-324-5p, and miR-324-5p alleviated the malignant growth of glioma by repressing XBP1. Furthermore, silence of circ_0079593 hindered tumor growth in vivo. Circ_0079593 contributed to the malignant evolution of glioma via modulating miR-324-5p and downstream XBP1 gene, suggesting that circ_0079593 might be a promising therapeutic target for glioma. Circ_0079593 was boosted in glioma. Circ_0079593 depletion restrained glioma progression. Circ_0079593 triggered glioma development via miR-324-5p/XBP1 axis. Circ_0079593 silence suppressed glioma tumorigenesis in vivo.
Collapse
Affiliation(s)
- Pengcheng Wang
- Department of Neurosurgery, Weifang People's Hospital, No 151 Guangwen Street, Kuiwen district, Weifang, 261000, China
| | - Tong Wang
- Department of Neurosurgery, Weifang People's Hospital, No 151 Guangwen Street, Kuiwen district, Weifang, 261000, China
| | - Lei Dong
- Department of Neurosurgery, Weifang People's Hospital, No 151 Guangwen Street, Kuiwen district, Weifang, 261000, China
| | - Zhenkuan Xu
- Department of Neurosurgery, Second Hospital of Shandong University, Jinan, China
| | - Shouzhong Guo
- Department of Neurosurgery, Linyi People's Hospital, Linyi, China
| | - Chengyue Chang
- Department of Neurosurgery, Weifang People's Hospital, No 151 Guangwen Street, Kuiwen district, Weifang, 261000, China.
| |
Collapse
|
20
|
Lan C, Zhang H, Wang K, Liu X, Zhao Y, Guo Z, Zhang N, Zhou Y, Gao M, Gu F, Ma Y. The alternative splicing of intersectin 1 regulated by PTBP1 promotes human glioma progression. Cell Death Dis 2022; 13:835. [PMID: 36171198 PMCID: PMC9519902 DOI: 10.1038/s41419-022-05238-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 01/23/2023]
Abstract
Intersectin 1 (ITSN1) contains two isoforms: ITSN1-S and ITSN1-L, which are highly regulated by alternative splicing. Our previous results showed that the two isoforms of ITSN1 displayed opposite functions: ITSN1-S promoted glioma development, while ITSN1-L exerted an inhibitory role in glioma progression. In this study, our transcriptome analysis using a large glioma cohort indicated that the ratio of ITSN1-S/ITSN1-L was positively correlated with glioma grading and poor prognosis. We identified the RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1) as an ITSN1 pre-mRNA interaction protein through RNA pull-down assay and RNA immunoprecipitation assay. Knockdown of PTBP1 decreased the ratio of ITSN1-S/ITSN1-L. Minigene reporter assay and mutation analyses further confirmed PTBP1 targeted polypyrimidine sequences on ITSN1 exon 30 (TTGCACTTCAGTATTTT) and promoted the inclusion of ITSN1 exon 30. Subsequently, silencing PTBP1 inhibited glioma cell proliferation, migration, and invasion by down-regulating the ratio of ITSN1-S/ITSN1-L. Taken together, our study provides a novel mechanism that PTBP1 modulates the alternative splicing of ITSN1 and promotes glioma proliferation and motility by up-regulating the ratio of ITSN1-S/ITSN1-L, thereby highlighting that PTBP1 may be an attractive therapeutic target for gliomas.
Collapse
Affiliation(s)
- Chungen Lan
- grid.411918.40000 0004 1798 6427Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Huikun Zhang
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Kezhen Wang
- grid.411918.40000 0004 1798 6427Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xiaoli Liu
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Yawen Zhao
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Zhifang Guo
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Ning Zhang
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Yongxia Zhou
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Manzhi Gao
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Feng Gu
- grid.411918.40000 0004 1798 6427Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Yongjie Ma
- grid.411918.40000 0004 1798 6427Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China ,grid.411918.40000 0004 1798 6427Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China ,grid.411918.40000 0004 1798 6427Key Laboratory of Cancer Prevention and Therapy, Tianjin, China ,grid.265021.20000 0000 9792 1228Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|
21
|
Zheng P, Zhang X, Ren D, Zhang Y. RP11-552D4.1: a novel m6a-related LncRNA associated with immune status in glioblastoma. Aging (Albany NY) 2022; 14:7348-7363. [PMID: 35852867 PMCID: PMC9550243 DOI: 10.18632/aging.204177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 07/05/2022] [Indexed: 11/25/2022]
Abstract
Glioblastoma (GBM) is the most malignant form of brain cancer in the world. Nevertheless, the survival rate of patients with GBM is extremely low. N6-methyladenosine (m6A) and long noncoding RNAs (lncRNAs) conduct important biological functions in patients’ survival status and the immunotherapeutic response. Here, m6A-related lncRNAs were identified by a co-expression method. Univariate and multivariate Cox regression together with LASSO were applied to establish the risk model. Kaplan-Meier and ROC analysis were applied to evaluate the prediction power of this risk model. Finally, the related immune profiling and chemical sensitivity targets were also investigated. The risk model holding three m6A-related lncRNAs was confirmed as an independent predictor for the prognosis. Furthermore, we found the risk model based on m6A-related lncRNAs is associated with the immune status, immunosuppressive biomarkers, and chemo-sensitivity in GBM patients. The RP11-552D4.1 is found to facilitate neuronal proliferation. This risk model consisted of m6A-related lncRNAs may be available for the clinical interventions in GBM patients.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Neurosurgery, Shanghai Pudong New Area People’s Hospital, Shanghai, China
- Key Molecular Lab, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Xiaoxue Zhang
- Key Molecular Lab, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Dabin Ren
- Department of Neurosurgery, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Yisong Zhang
- Department of Neurosurgery, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| |
Collapse
|
22
|
Khabibov M, Garifullin A, Boumber Y, Khaddour K, Fernandez M, Khamitov F, Khalikova L, Kuznetsova N, Kit O, Kharin L. Signaling pathways and therapeutic approaches in glioblastoma multiforme (Review). Int J Oncol 2022; 60:69. [PMID: 35445737 PMCID: PMC9084550 DOI: 10.3892/ijo.2022.5359] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/30/2022] [Indexed: 12/04/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of primary brain tumor and is associated with a poor clinical prognosis. Despite the progress in the understanding of the molecular and genetic changes that promote tumorigenesis, effective treatment options are limited. The present review intended to identify and summarize major signaling pathways and genetic abnormalities involved in the pathogenesis of GBM, as well as therapies that target these pathways. Glioblastoma remains a difficult to treat tumor; however, in the last two decades, significant improvements in the understanding of GBM biology have enabled advances in available therapeutics. Significant genomic events and signaling pathway disruptions (NF‑κB, Wnt, PI3K/AKT/mTOR) involved in the formation of GBM were discussed. Current therapeutic options may only marginally prolong survival and the current standard of therapy cures only a small fraction of patients. As a result, there is an unmet requirement for further study into the processes of glioblastoma pathogenesis and the discovery of novel therapeutic targets in novel signaling pathways implicated in the evolution of glioblastoma.
Collapse
Affiliation(s)
- Marsel Khabibov
- Department of Oncology, I. M. Sechenov First Moscow State Medical University, 119992 Moscow, Russia
| | - Airat Garifullin
- Department of Histology, Bashkir State Medical University, 450000 Ufa, Russia
| | - Yanis Boumber
- Division of Hematology/Oncology at The Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Karam Khaddour
- Department of Hematology and Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Manuel Fernandez
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Firat Khamitov
- Department of Histology, Bashkir State Medical University, 450000 Ufa, Russia
| | - Larisa Khalikova
- Department of Histology, Bashkir State Medical University, 450000 Ufa, Russia
| | - Natalia Kuznetsova
- Department of Neuro-Oncology, National Medical Research Center for Oncology, 344037 Rostov-on-Don, Russia
| | - Oleg Kit
- Abdominal Oncology Department, National Medical Research Center for Oncology, 344037 Rostov-on-Don, Russia
| | - Leonid Kharin
- Abdominal Oncology Department, National Medical Research Center for Oncology, 344037 Rostov-on-Don, Russia
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
23
|
Kannan S, Kannan Murugan A, Balasubramaniam S, Kannan Munirajan A, Alzahrani AS. Gliomas: Genetic alterations, mechanisms of metastasis, recurrence, drug resistance, and recent trends in molecular therapeutic options. Biochem Pharmacol 2022; 201:115090. [PMID: 35577014 DOI: 10.1016/j.bcp.2022.115090] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022]
Abstract
Glioma is the most common intracranial tumor with poor treatment outcomes and has high morbidity and mortality. Various studies on genomic analyses of glioma found a variety of deregulated genes with somatic mutations including TERT, TP53, IDH1, ATRX, TTN, etc. The genetic alterations in the key genes have been demonstrated to play a crucial role in gliomagenesis by modulating important signaling pathways that alter the fundamental intracellular functions such as DNA damage and repair, cell proliferation, metabolism, growth, wound healing, motility, etc. The SPRK1, MMP2, MMP9, AKT, mTOR, etc., genes, and noncoding RNAs (miRNAs, lncRNAs, circRNAs, etc) were shown mostly to be implicated in the metastases of glioma. Despite advances in the current treatment strategies, a low-grade glioma is a uniformly fatal disease with overall median survival of ∼5-7 years while the patients bearing high-grade tumors display poorer median survival of ∼9-10 months mainly due to aggressive metastasis and therapeutic resistance. This review discusses the spectrum of deregulated genes, molecular and cellular mechanisms of metastasis, recurrence, and its management, the plausible causes for the development of therapy resistance, current treatment options, and the recent trends in malignant gliomas. Understanding the pathogenic mechanisms and advances in molecular genetics would aid in the novel diagnosis, prognosis, and translation of pathogenesis-based treatment opportunities which could pave the way for precision medicine in glioma.
Collapse
Affiliation(s)
- Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE UK
| | - Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh - 11211 Saudi Arabia.
| | | | - Arasambattu Kannan Munirajan
- Department of Genetics, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai, 600113 India
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh - 11211 Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh - 11211 Saudi Arabia
| |
Collapse
|
24
|
Roy PK, Biswas A, Deepak K, Mandal M. An insight into the ubiquitin-proteasomal axis and related therapeutic approaches towards central nervous system malignancies. Biochim Biophys Acta Rev Cancer 2022; 1877:188734. [PMID: 35489645 DOI: 10.1016/j.bbcan.2022.188734] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/12/2022] [Accepted: 04/22/2022] [Indexed: 10/18/2022]
Abstract
The Ubiquitin-Protease system (UPS) is a major destruction system that is responsible for the elimination of dysfunctional/misfolded proteins, thus acting as a pivotal regulator of protein homeostasis in eukaryotic cells. In this review, the UPS system and its various functions in the cell and their detailed impact such as cell cycle control, DNA damage response, apoptosis, and cellular stress regulations have been elucidated with a focus on the central nervous system. Since the Ubiquitin-Protease pathway(UPP) plays a prominent role in the sculpting of the CNS cells and their maintenance, it is naturally deeply involved in many malignancies that develop due to dysregulation of the UPS. Understanding the major disruptive players of the UPS in the development of these malignancies, for example, insoluble protein aggregates or inclusion bodies deposits due to malfunctioning of the UPS has paved the pathway for the development of new therapeutics. Here, the de-regulation of the UPS at various checkpoints in CNS malignancies has been detailed, thus facilitating an easy comprehension of the different targets that remain to be explored yet. The present therapeutic advancements in the field of CNS malignancies management through UPS targeting have also been included thus broadening the scope of drug development. Thus, this review while shedding sufficient light on the details of the UPS system and its connection to CNS malignancies, also opens new avenues for therapeutic advancements in the form of novel targetable UPP proteins and their interactions.
Collapse
Affiliation(s)
- Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Angana Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India..
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India..
| |
Collapse
|
25
|
Ghaffarian Zirak R, Tajik H, Asadi J, Hashemian P, Javid H. The Role of Micro RNAs in Regulating PI3K/AKT Signaling Pathways in Glioblastoma. IRANIAN JOURNAL OF PATHOLOGY 2022; 17:122-136. [PMID: 35463721 PMCID: PMC9013863 DOI: 10.30699/ijp.2022.539029.2726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/02/2022] [Indexed: 12/21/2022]
Abstract
Glioblastoma is a type of brain cancer with aggressive and invasive nature. Such features result from increased proliferation and migration and also poor apoptosis of glioma cells leading to resistance to current treatments such as chemotherapy and radiotherapy. In recent studies, micro RNAs have been introduced as a novel target for treating glioblastoma via regulation of apoptotic signaling pathway, remarkably PI3K/AKT, which affect cellular functions and blockage or progression of the tumor. In this review, we focus on PI3K/AKT signaling pathway and other related apoptotic processes contributing to glioblastoma and investigate the role of micro RNAs interfering in apoptosis, invasion and proliferation of glioma through such apoptotic processes pathways. Databases NCBI, PubMed, and Web of Science were searched for published English articles using keywords such as 'miRNA OR microRNA', 'Glioblastoma', 'apoptotic pathways', 'PI3K and AKT', 'Caspase signaling Pathway' and 'Notch pathway'. Most articles were published from 7 May 2015 to 16 June 2020. This study focused on PI3K/AKT signaling pathway affecting glioma cells in separated subparts. Also, other related apoptotic pathways as the Caspase cycle and Notch have been also investigated. Nearly 40 miRNAs were found as tumor suppressors or onco-miRNA, and their targets, which regulated subcomponents participating in proliferation, invasion, and apoptosis of the tumoral cells. Our review reveals that miRNAs affect key molecules in signaling apoptotic pathways, partly PI3K/AKT, making them potential therapeutic targets to overcome the tumor. However, their utility as a novel treatment for glioblastoma requires further examination and investigation.
Collapse
Affiliation(s)
- Roshanak Ghaffarian Zirak
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hurie Tajik
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Science, Shahrekord, Iran.,Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Jahanbakhsh Asadi
- Department of Clinical Biochemistry, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Pedram Hashemian
- Jahad Daneshgahi Research Committee, Jahad Daneshgahi Institute, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Li L, Yu J, Cheng S, Peng Z, Ben-David Y, Luo H. Transcription factor Fli-1 as a new target for antitumor drug development. Int J Biol Macromol 2022; 209:1155-1168. [PMID: 35447268 DOI: 10.1016/j.ijbiomac.2022.04.076] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023]
Abstract
The transcription factor Friend leukemia virus integration 1 (Fli-1) belonging to the E26 Transformation-Specific (ETS) transcription factor family is not only expressed in normal cells such as hematopoietic stem cells and vascular endothelial cells, but also abnormally expressed in various malignant tumors including Ewing sarcoma, Merkel cell sarcoma, small cell lung carcinoma, benign or malignant hemangioma, squamous cell carcinoma, adenocarcinoma, bladder cancer, leukemia, and lymphoma. Fli-1 binds to the promoter or enhancer of the target genes and participates in a variety of physiological and pathological processes of tumor cells, including cell growth, proliferation, differentiation, and apoptosis. The expression of Fli-1 gene is related to the specific biological functions and characteristics of the tissue in which it is located. In tumor research, Fli-1 gene is used as a specific marker for the occurrence, metastasis, efficacy, and prognosis of tumors, thus, a potential new target for tumor diagnosis and treatment. These studies indicated that Fli-1 may be a specific candidate for antitumor drug development. Recent studies identified small molecules regulating Fli-1 thanks to our screened strategy of natural products and their derivatives. Therefore, in this review, the advanced research on Fli-1 as a target for antitumor drug development is analyzed in different cancers. The inhibitors and agonists of Fli-1 that regulate its expression are introduced and their clinical applications in the treatment of cancer, thus providing new therapeutic strategies.
Collapse
Affiliation(s)
- Lanlan Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; College of Pharmacy, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Jia Yu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Sha Cheng
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Zhilin Peng
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Heng Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China.
| |
Collapse
|
27
|
Liu SH, Wang QZ, Liu T, Bai R, Ma MM, Liu QL, Zhou HG, Liu J, Wang M. Enhanced Glioblastoma Selectivity of Harmine via the Albumin Carrier. J Biomed Nanotechnol 2022; 18:1052-1063. [PMID: 35854453 DOI: 10.1166/jbn.2022.3321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Glioblastoma, the most common tumor in the brain, has witnessed very little clinical progress over the last decades. Exploring and discovering new therapeutic strategies for glioblastoma has become a critical problem. Harmine (HM), belonging to the beta-carboline alkaloid, is a natural product and isolated from the seeds of Peganum harmala L., which own notable antitumor activity in vitro. However, the poor water solubility and less selectivity of HM severely limit its clinical use. For enhancing its selective ability to tumor cells, we fabricated a kind of protein nanoparticles (BSA-HM NPs), composed of the modified bovine serum albumin (BSA) and HM. It was substantiated through in vitro and in vivo experiment that BSA-HM NPs could predominantly accumulate in tumor tissues and exhibited remarkably enhanced antitumor efficacy. This study provides a promising strategy to improve the bioavailability and avoid side effects of HM as antitumor agents by choosing BSA as carriers.
Collapse
Affiliation(s)
- Shi-Hui Liu
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, P. R. China
| | - Qing-Zhen Wang
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, P. R. China
| | - Tao Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Ru Bai
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Man-Man Ma
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Qiao-Lin Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Hui-Ge Zhou
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Jing Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Mei Wang
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, P. R. China
| |
Collapse
|
28
|
Overexpression of prothymosin-alpha in glioma is associated with tumor aggressiveness and poor prognosis. Biosci Rep 2022; 42:231053. [PMID: 35297481 PMCID: PMC9069441 DOI: 10.1042/bsr20212685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 12/05/2022] Open
Abstract
Prothymosin-α (PTMA), a nuclear protein, is strikingly associated with unfavorable clinical outcomes in many cancers. However, no information about its clinical relevance in glioma was available. Therefore in the present study, we evaluated the prognostic utility of this protein in a cohort of 81 glioma patients. The PTMA expression was assessed by immunohistochemical analysis, quantitative PCR, and Western blotting. Furthermore, the association of PTMA with clinicopathological features and molecular alterations were assessed in the patient cohort and validated in multiomics datasets, The Cancer Genome Atlas (TCGA; n=667) and Chinese Glioma Genome Atlas (CGGA; n=1013). We observed an increase in PTMA expression with increasing histological grades of this malignancy. PTMA immunostaining also displayed a strong positive association with the MIB-1 index. Univariate analysis revealed a superior prognostic value of PTMA to predict overall survival (OS) as compared with the routinely used markers (p53, isocitrate dehydrogenase (IDH) 1 (IDH1), α-thalassemia/intellectual disability syndrome X-linked (ATRX), and Ki-67). Interestingly, in Cox regression analysis it emerged as an independent predictor of OS (hazard ratio (HR) = 13.71, 95% CI = 5.96–31.52, P<0.0001). Thus, our results demonstrate the potential prognostic utility of PTMA in glioma which may prove useful in the management of this deadly malignancy.
Collapse
|
29
|
NOX2-Induced High Glycolytic Activity Contributes to the Gain of COL5A1-Mediated Mesenchymal Phenotype in GBM. Cancers (Basel) 2022; 14:cancers14030516. [PMID: 35158782 PMCID: PMC8833670 DOI: 10.3390/cancers14030516] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) is the most aggressive type of glioma and exhibits extensive heterogeneity and poor prognosis with a high recurrence rate. Among the genetic alterations in GBM with different phenotypic states, a mesenchymal subtype has been associated with a worse outcome in patients with GBM. The mechanisms for the gain of the mesenchymal subtype in GBM remain unclear. Our aim was to investigate whether NOX2-induced high glycolytic activity could contribute to the gain of the mesenchymal phenotype in GBM. We revealed that NOX2-induced high glycolytic activity can induce the gain of the COL5A1-mediated mesenchymal phenotype in GBM. Our findings will provide the molecular mechanism by which NOX2 contributes to the gain of mesenchymal phenotype in GBM. Abstract The alteration of the cellular metabolism is a hallmark of glioma. The high glycolytic phenotype is a critical factor in the pathogenesis of high-grade glioma, including glioblastoma multiforme (GBM). GBM has been stratified into three subtypes as the proneural, mesenchymal, and classical subtypes. High glycolytic activity was found in mesenchymal GBM relative to proneural GBM. NADPH oxidase 2 (NOX2) has been linked to cellular metabolism and epithelial-mesenchymal transition (EMT) in tumors. The role of NOX2 in the regulation of the high glycolytic phenotype and the gain of the mesenchymal subtype in glioma remain unclear. Here, our results show that the levels of NOX2 were elevated in patients with GBM. NOX2 induces hexokinase 2 (HK2)-dependent high glycolytic activity in U87MG glioma cells. High levels of NOX2 are correlated with high levels of HK2 and glucose uptake in patients with GBM relative to benign glioma. Moreover, NOX2 increases the expression of mesenchymal-subtype-related genes, including COL5A1 and FN1 in U87MG glioma cells. High levels of NOX2 are correlated with high levels of COL5A1 and the accumulation of extracellular matrix (ECM) in patients with GBM relative to benign glioma. Furthermore, high levels of HK2 are correlated with high levels of COL5A1 in patients with GBM relative to benign glioma. Our results suggest that NOX2-induced high glycolytic activity contributes to the gain of the COL5A1-mediated mesenchymal phenotype in GBM.
Collapse
|
30
|
Dhar D, Ghosh S, Das S, Chatterjee J. A review of recent advances in magnetic nanoparticle-based theranostics of glioblastoma. Nanomedicine (Lond) 2022; 17:107-132. [PMID: 35000429 DOI: 10.2217/nnm-2021-0348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Rapid vascular growth, infiltrative cells and high tumor heterogenicity are some glioblastoma multiforme (GBM) characteristics, making it the most lethal form of brain cancer. Low efficacy of the conventional treatment modalities leads to rampant disease progression and a median survival of 15 months. Magnetic nanoparticles (MNPs), due to their unique physical features/inherent abilities, have emerged as a suitable theranostic platform for targeted GBM treatment. Thus, new strategies are being designed to enhance the efficiency of existing therapeutic techniques such as chemotherapy, radiotherapy, and so on, using MNPs. Herein, the limitations of the current therapeutic strategies, the role of MNPs in mitigating those inadequacies, recent advances in the MNP-based theranostics of GBM and possible future directions are discussed.
Collapse
Affiliation(s)
- Dhruba Dhar
- School of Medical Sciences & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India
| | - Swachhatoa Ghosh
- School of Medical Sciences & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India
| | - Soumen Das
- School of Medical Sciences & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India
| | - Jyotirmoy Chatterjee
- School of Medical Sciences & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India
| |
Collapse
|
31
|
Ullah MA, Alam S, Farzana M, Tayab Moin A, Binte Sayed Prapty CN, Zohora US, Rahman MS. Prognostic and therapeutic value of LSM5 gene in human brain cancer Glioma: An omics database exploration approach. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
32
|
Marker DF, Agnihotri S, Amankulor N, Murdoch GH, Pearce TM. The dominant TP53 hotspot mutation in IDH -mutant astrocytoma, R273C, has distinctive pathologic features and sex-specific prognostic implications. Neurooncol Adv 2022; 4:vdab182. [PMID: 35047821 PMCID: PMC8760900 DOI: 10.1093/noajnl/vdab182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Infiltrative astrocytic tumors with and without isocitrate dehydrogenase (IDH) mutation frequently contain mutations in the TP53 tumor suppressor gene. Disruption of normal p53 protein activity confers neoplastic cells with a number of oncogenic properties and is a common feature of aggressive malignancies. However, the high prevalence of TP53 mutation and its pathogenic role in IDH-mutant (IDHmut) astrocytoma is not well understood. METHODS We performed a retrospective analysis of molecular and clinical data from patients with IDHmut astrocytoma at the University of Pittsburgh Medical Center between 2015 and 2019 as our initial cohort. We validated and expanded our findings using molecular and clinical data from The Cancer Genome Atlas. RESULTS We show that the TP53 mutational spectrum in IDHmut astrocytomas is dominated by a single hotspot mutation that codes for the R273C amino acid change. This mutation is not enriched in IDH-wildtype astrocytomas. The high prevalence of TP53 R273C mutation is not readily explained by known mutagenic mechanisms, and TP53 R273C mutant tumors have lower transcriptional levels of proliferation-related genes compared to IDHmut astrocytomas harboring other forms of mutant p53. Despite lower proliferation, TP53 R273C mutant tumors tend to progress more quickly and have a shorter overall survival than those with other TP53 mutations, particularly in male patients. CONCLUSIONS Our findings suggest that compared to other TP53 mutations, IDHmut astrocytomas may select for TP53 R273C mutations during tumorigenesis. The genotype, sex, and mutation-specific findings are clinically relevant and should prompt further investigation of TP53 R273C.
Collapse
Affiliation(s)
- Daniel F Marker
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nduka Amankulor
- Department of Neurosurgery and Brain Tumor Center, Abramson Cancer Center, The University of Pennsylvania, Philadelphia, PA, USA
| | - Geoffrey H Murdoch
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas M Pearce
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
33
|
Li Z, Li M, Xia P, Wang L, Lu Z. LncRNA FOXD3-AS1 Promotes Tumorigenesis of Glioma via Targeting miR-128-3p/ SZRD1 Axis. Cancer Manag Res 2021; 13:9037-9048. [PMID: 34916848 PMCID: PMC8666723 DOI: 10.2147/cmar.s324920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Background The aim of the current study was to investigate the roles of LncRNA FOXD3-AS1 (FOXD3-AS1) in the glioma progression, and its underlying mechanism of competing endogenous RNA (ceRNA) network of FOXD3-AS1/miR-128-3p/SZRD1. Materials and Methods The FOXD3-AS1 expression and its prognostic relation were detected by bioinformatics tool. Next, glioma cell lines (HS683, U251, T98G, and SNB-19) were used to verify the FOXD3-AS1 expression. Furthermore, the roles of the FOXD3-AS1/miR-128-3p/SZRD1 axis on the glioma development in vitro and in vivo were examined. Results Bioinformatics analysis showed that FOXD3-AS1 was upregulated in the glioma and linked with poor prognosis. Consistently, FOXD3-AS1 level was overexpressed in the glioma cell lines (HS683 and U251). Subsequently, we verified that silencing of FOXD3-AS1 (si-FOXD3-AS1) restrained the cell proliferation, invasion, and tumor growth in vivo, and induced G0/G1 arrest, and promoted apoptosis. Further study also stated that FOXD3-AS1 interacted with miR-128-3p and SZRD1 was the target gene of miR-128-3p. Moreover, overexpression of miR-128-3p restrained the cell proliferation and metastasis of glioma, and reduced the SZRD1 level. Rescue assay illustrated that miR-128-3p inhibitor could reverse the suppressive impact of si-FOXD3-AS1 on the glioma progression. Similarly, SZRD1 overexpression could neutralize the influences of miR-128-3p mimic on glioma progression. Conclusion FOXD3-AS1 promoted the tumorigenesis of glioma, and exerted its function to modulate SZRD1 by targeting miR-128-3p.
Collapse
Affiliation(s)
- Zhang Li
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China
| | - Ming Li
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China
| | - Pengcheng Xia
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China
| | - Lili Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China
| |
Collapse
|
34
|
Spellerberg R, Benli-Hoppe T, Kitzberger C, Berger S, Schmohl KA, Schwenk N, Yen HY, Zach C, Schilling F, Weber WA, Kälin RE, Glass R, Nelson PJ, Wagner E, Spitzweg C. Selective sodium iodide symporter ( NIS) genetherapy of glioblastoma mediatedby EGFR-targeted lipopolyplexes. Mol Ther Oncolytics 2021; 23:432-446. [PMID: 34853814 PMCID: PMC8604759 DOI: 10.1016/j.omto.2021.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 12/18/2022] Open
Abstract
Lipo-oligomers, post-functionalized with ligands to enhance targeting, represent promising new vehicles for the tumor-specific delivery of therapeutic genes such as the sodium iodide symporter (NIS). Due to its iodide trapping activity, NIS is a powerful theranostic tool for diagnostic imaging and the application of therapeutic radionuclides. 124I PET imaging allows non-invasive monitoring of the in vivo biodistribution of functional NIS expression, and application of 131I enables cytoreduction. In our experimental design, we used epidermal growth factor receptor (EGFR)-targeted polyplexes (GE11) initially characterized in vitro using 125I uptake assays. Mice bearing an orthotopic glioblastoma were treated subsequently with mono-dibenzocyclooctyne (DBCO)-PEG24-GE11/NIS or bisDBCO-PEG24-GE11/NIS, and 24-48 h later, 124I uptake was assessed by positron emission tomography (PET) imaging. The best-performing polyplex in the imaging studies was then selected for 131I therapy studies. The in vitro studies showed EGFR-dependent and NIS-specific transfection efficiency of the polyplexes. The injection of monoDBCO-PEG24-GE11/NIS polyplexes 48 h before 124I application was characterized to be the optimal regime in the imaging studies and was therefore used for an 131I therapy study, showing a significant decrease in tumor growth and a significant extension of survival in the therapy group. These studies demonstrate the potential of EGFR-targeted polyplex-mediated NIS gene therapy as a new strategy for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Rebekka Spellerberg
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Teoman Benli-Hoppe
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, 81377 Munich, Germany
| | - Carolin Kitzberger
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, 81377 Munich, Germany
| | - Kathrin A Schmohl
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Nathalie Schwenk
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Hsi-Yu Yen
- Institute of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Christian Zach
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Roland E Kälin
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany.,Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany.,Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany.,German Cancer Consortium (DKTK), partner site 80336 Munich and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter J Nelson
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, 81377 Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IV, University Hospital, LMU Munich, 81377 Munich, Germany.,Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
35
|
Sun C, Zheng X, Sun Y, Yu J, Sheng M, Yan S, Zhu Q, Lan Q. Identification of IGF2BP3 as an Adverse Prognostic Biomarker of Gliomas. Front Genet 2021; 12:743738. [PMID: 34721530 PMCID: PMC8551830 DOI: 10.3389/fgene.2021.743738] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/29/2021] [Indexed: 11/29/2022] Open
Abstract
N6-methyladenosine (m6A) RNA modification can alter gene expression and function by regulating RNA splicing, stability, translocation, and translation. It is involved in various types of cancer. However, its role in gliomas is not well known. This study aimed to determine the prognostic value of the m6A RNA methylation regulator in gliomas and investigate the underlying mechanisms of the aberrant expression of m6A-related genes.mRNA expression profiles and clinical information of 448 glioma samples were obtained from The Cancer Genome Atlas and cBioportal. The expression of m6A-related genes in normal controls and low-grade glioma and glioblastoma was obtained from Gene Expression Profiling Interactive Analysis. Further, m6A-related gene expression and its relationship with prognosis were obtained through The Chinese Glioma Genome Atlas (CGGA). Multivariate Cox regression analyses were performed, and a nomogram was built with potential risk factors based on a multivariate Cox analysis to predict survival probability. Online tools such as Gene Set Enrichment Analysis, STRING, Cytoscape, and Molecular Complex Detection were applied for bioinformatics analysis and to investigate the underlying mechanisms of the aberrant expression of m6A-related genes. The multivariate Cox regression analysis found that higher expression levels of YTHDC2 and insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3, also called IMP3) were independent negative and positive prognostic factors for overall survival (OS), respectively. Data from the CGGA database showed that IGF2BP3 expression increased when the tumor grade increased. Receiver operating characteristic (ROC) curve was used to evaluate the predictive specificity and sensitivity. The area under the ROC curve indicated that the OS prediction was 0.92 (1-year) and 0.917 (3-years), indicating that m6A-related genes could predict patient survival. In addition, IGF2BP3 was closely related to the shorter survival period of patients. Copy number variation and DNA methylation, but not somatic mutations, might contribute to the abnormal upregulation of IGF2BP3 in gliomas. Significantly altered genes were identified, and the protein–protein interaction network was constructed. Based on the data presented, our study identified several m6A-related genes, especially IGF2BP3, that could be potential prognostic biomarkers of gliomas. The study unveiled the potential regulatory mechanism of IGF2BP3 in gliomas.
Collapse
Affiliation(s)
- Chao Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yingxin Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ju Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Minfeng Sheng
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Suji Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qing Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
36
|
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: 0.7] [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
|
37
|
Regulatory interplay between microRNAs and WNT pathway in glioma. Biomed Pharmacother 2021; 143:112187. [PMID: 34560532 DOI: 10.1016/j.biopha.2021.112187] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/10/2021] [Indexed: 12/17/2022] Open
Abstract
Glioma is one of the most common neoplasms of the central nervous system with a poor survival. Due to the obstacles in treating this disease, a part of recent studies mainly focuses on identifying the underlying molecular mechanisms that contribute to its malignancy. Altering microRNAs (miRNAs) expression pattern has been identified obviously in many cancers. Through regulating various targets and signaling pathways, miRNAs play a pivotal role in cancer progression. As one of the essential signaling pathways, WNT pathway is dysregulated in many cancers, and a growing body of evidence emphasis its dysregulation in glioma. Herein, we provide a comprehensive review of miRNAs involved in WNT pathway in glioma. Moreover, we show the interplay between miRNAs and WNT pathway in regulating different processes such as proliferation, invasion, migration, radio/chemotherapy resistance, and epithelial-mesenchymal-transition. Then, we introduce several drugs and treatments against glioma, which their effects are mediated through the interplay of WNT pathway and miRNAs.
Collapse
|
38
|
Nsairat H, Khater D, Odeh F, Al-Adaileh F, Al-Taher S, Jaber AM, Alshaer W, Al Bawab A, Mubarak MS. Lipid nanostructures for targeting brain cancer. Heliyon 2021; 7:e07994. [PMID: 34632135 PMCID: PMC8488847 DOI: 10.1016/j.heliyon.2021.e07994] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/28/2021] [Accepted: 09/09/2021] [Indexed: 01/02/2023] Open
Abstract
Advancements in both material science and bionanotechnology are transforming the health care sector. To this end, nanoparticles are increasingly used to improve diagnosis, monitoring, and therapy. Huge research is being carried out to improve the design, efficiency, and performance of these nanoparticles. Nanoparticles are also considered as a major area of research and development to meet the essential requirements for use in nanomedicine where safety, compatibility, biodegradability, biodistribution, stability, and effectiveness are requirements towards the desired application. In this regard, lipids have been used in pharmaceuticals and medical formulations for a long time. The present work focuses on the use of lipid nanostructures to combat brain tumors. In addition, this review summarizes the literature pertaining to solid lipid nanoparticles (SLN) and nanostructured lipid carriers (LNC), methods of preparation and characterization, developments achieved to overcome blood brain barrier (BBB), and modifications used to increase their effectiveness.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman 11931, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
| | - Fedaa Al-Adaileh
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
| | - Suma Al-Taher
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
| | - Areej M. Jaber
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman 11942, Jordan
| | | |
Collapse
|
39
|
Zhao J, Jiang Y, Chen L, Ma Y, Zhang H, Zhou J, Li H, Jing Z. The EIF4A3/CASC2/RORA Feedback Loop Regulates the Aggressive Phenotype in Glioblastomas. Front Oncol 2021; 11:699933. [PMID: 34408982 PMCID: PMC8366401 DOI: 10.3389/fonc.2021.699933] [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: 04/24/2021] [Accepted: 07/16/2021] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is a common and refractory subtype of high-grade glioma with a poor prognosis. The epithelial-mesenchymal transition (EMT) is an important cause of enhanced glioblastoma invasiveness and tumor recurrence. Our previous study found that retinoic acid receptor-related orphan receptor A (RORA) is a nuclear receptor and plays an important role in inhibiting proliferation and tumorigenesis of glioma. We further confirmed RORA was downregulated in GBM. Thus, we determined whether RORA was involved in the migration, invasion, and EMT of GBM. Human GBM cell lines, U87 and T98G, and patient-derived glioma stem cells (GSCs), GSC2C and GSC4D, were used for in vitro and in vivo experiments. The expressions of RORA, CASC2, and EIF4A3 in GBM cells and GSCs were detected by RT-qPCR and western blotting. The biological effects of RORA, CASC2, and EIF4A3 on GBM migration, invasion, and EMT were evaluated using the migration assay, transwell assay, immunofluorescence staining, and xenograft experiments. We found that RORA inhibited the migration, invasion, and EMT of GBM. CASC2 could bind to, maintain the stability, and promote the nuclear translocation of RORA protein. EIF4A3 could downregulate CASC2 expression via inducing its cleavage, while RORA transcriptionally inhibited EIF4A3 expression, which formed a feedback loop among EIF4A3/CASC2/RORA. Moreover, gene set enrichment analysis (GSEA) and in vitro and in vivo experiments showed RORA inhibited the aggressiveness of GBM by negatively regulating the TGF-β1/Smad signaling pathway. Therefore, The EIF4A3/CASC2/RORA feedback loop regulated TGF-β1/Smad signaling pathway might become a promising therapeutic strategy for GBM treatment.
Collapse
Affiliation(s)
- Junshuang Zhao
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yang Jiang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lian Chen
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yue Ma
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haiying Zhang
- International Education College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jinpeng Zhou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Hao Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Zhitao Jing
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
40
|
Henao-Restrepo J, Caro-Urrego YA, Barrera-Arenas LM, Arango-Viana JC, Bermudez-Munoz M. Expression of activator proteins of SHH/GLI and PI3K/Akt/mTORC1 signaling pathways in human gliomas is associated with high grade tumors. Exp Mol Pathol 2021; 122:104673. [PMID: 34371011 DOI: 10.1016/j.yexmp.2021.104673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/25/2021] [Accepted: 08/03/2021] [Indexed: 01/15/2023]
Abstract
Recent findings have demonstrated a synergic crosstalk between SHH/GLI and PI3K/Akt/mTORC1 signaling in glioblastoma progression cells in vitro and in tumors in mice, but it is not known if this also occurs in human gliomas. We then aimed to investigate the expression of key proteins of these pathways in different human gliomas. The expression of PTEN, phospho-Akt (Ser473), phospho-S6K1 (Thr389), SHH, GLI1, GLI2 and GLI3 was assessed by immunohistochemistry in gliomas and in control brain tissues. The pattern of expression of each protein was established according to glioma type, glioma grade and to cell type; the relative expression of each protein was used to perform statistical analyses. We found that the expression of proteins of both signaling pathways differs between normal brain and glioma tissues. For instance, normal astrocytes had a different protein expression pattern compared with reactive and tumoral astrocytes. Interestingly, we detected a recurrent pattern of expression of GLI3 in oligodendrocytes and of phospho-S6K1 in mitotic neoplastic cells. We also identified differences of cell signaling according to glioma type: oligodendrogliomas and ependymomas are related with the expression of SHH/GLI proteins. Finally, we detected that high grade gliomas statistically correlate with the expression of GLI1 and GLI2, and that GLI1, GLI2, phospho-Akt and phospho-S6K1 are more expressed in patients with less survival, suggesting that activation of these cell signaling influences glioma outcome and patient survival. In summary, our results show that proteins of PI3K/Akt/mTORC1 and SHH/GLI pathways are differentially expressed in human gliomas according to tumor type and grade, and suggest that the activation of these signaling networks is associated with glioma progression.
Collapse
Affiliation(s)
- Julián Henao-Restrepo
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Calle 67 #53-108, 050010 Medellín, Colombia.
| | - Yudys Anggelly Caro-Urrego
- Department of Pathology, Faculty of Medicine, University of Antioquia, Cra. 51d #62-29, 050010 Medellín, Colombia
| | - Lina Marcela Barrera-Arenas
- Grupo de Investigaciones Biomédicas, Health Sciences Faculty, University Corporation Remington, Calle 51 #51-27, Medellín, Colombia.
| | - Juan Carlos Arango-Viana
- Department of Pathology, Faculty of Medicine, University of Antioquia, Cra. 51d #62-29, 050010 Medellín, Colombia.
| | - Maria Bermudez-Munoz
- Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Calle 67 #53-108, 050010 Medellín, Colombia.
| |
Collapse
|
41
|
Roy PK, Rajesh Y, Mandal M. Therapeutic targeting of membrane-associated proteins in central nervous system tumors. Exp Cell Res 2021; 406:112760. [PMID: 34339674 DOI: 10.1016/j.yexcr.2021.112760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/28/2021] [Accepted: 07/28/2021] [Indexed: 12/09/2022]
Abstract
The activity of the most complex system, the central nervous system (CNS) is profoundly regulated by a huge number of membrane-associated proteins (MAP). A minor change stimulates immense chemical changes and the elicited response is organized by MAP, which acts as a receptor of that chemical or channel enabling the flow of ions. Slight changes in the activity or expression of these MAPs lead to severe consequences such as cognitive disorders, memory loss, or cancer. CNS tumors are heterogeneous in nature and hard-to-treat due to random mutations in MAPs; like as overexpression of EGFRvIII/TGFβR/VEGFR, change in adhesion molecules α5β3 integrin/SEMA3A, imbalance in ion channel proteins, etc. Extensive research is under process for developing new therapeutic approaches using these proteins such as targeted cytotoxic radiotherapy, drug-delivery, and prodrug activation, blocking of receptors like GluA1, developing viral vector against cell surface receptor. The combinatorial approach of these strategies along with the conventional one might be more potential. Henceforth, our review focuses on in-depth analysis regarding MAPs aiming for a better understanding for developing an efficient therapeutic approach for targeting CNS tumors.
Collapse
Affiliation(s)
- Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, India
| | - Yetirajam Rajesh
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, India.
| |
Collapse
|
42
|
Zorzan M, Del Vecchio C, Vogiatzis S, Saccon E, Parolin C, Palù G, Calistri A, Mucignat-Caretta C. Targeting the Regulatory Subunit R2Alpha of Protein Kinase A in Human Glioblastoma through shRNA-Expressing Lentiviral Vectors. Viruses 2021; 13:v13071361. [PMID: 34372567 PMCID: PMC8310305 DOI: 10.3390/v13071361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/25/2021] [Accepted: 07/10/2021] [Indexed: 01/17/2023] Open
Abstract
Glioblastoma is the most malignant and most common form of brain tumor, still today associated with a poor 14-months median survival from diagnosis. Protein kinase A, particularly its regulatory subunit R2Alpha, presents a typical intracellular distribution in glioblastoma cells compared to the healthy brain parenchyma and this peculiarity might be exploited in a therapeutic setting. In the present study, a third-generation lentiviral system for delivery of shRNA targeting the regulatory subunit R2Alpha of protein kinase A was developed. Generated lentiviral vectors are able to induce an efficient and stable downregulation of R2Alpha in different cellular models, including non-stem and stem-like glioblastoma cells. In addition, our data suggest a potential correlation between silencing of the regulatory subunit of protein kinase A and reduced viability of tumor cells, apparently due to a reduction in replication rate. Thus, our findings support the role of protein kinase A as a promising target for novel anti-glioma therapies.
Collapse
|
43
|
Chen L, Zhou J, Li L, Zhao J, Li H, Zheng W, Xu J, Jing Z. SLC39A7 promotes malignant behaviors in glioma via the TNF-α-mediated NF-κB signaling pathway. J Cancer 2021; 12:4530-4541. [PMID: 34149917 PMCID: PMC8210565 DOI: 10.7150/jca.54158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 05/03/2021] [Indexed: 11/05/2022] Open
Abstract
Purpose: Several studies have indicated that SLC39A7 plays an important role in tumor progression; however, little is known about the function and mechanism of SLC39A7 in glioma. In this study, we aimed to explore the role of SLC39A7 in glioma development. Patients and methods: Bioinformatic analysis was used to predict the role of SLC39A7 in glioma. Cell viability and Edu assays were used to detect the proliferation of glioma cells. A transwell assay was used to measure the invasion and migration of glioma cells. Western blotting, qPCR and ELISA were used to detect the expression of all molecules. Results: SLC39A7 was found to be highly expressed in high-grade glioma patients with a poor prognosis. Our results indicated that SLC39A7 significantly promoted the proliferation, invasion and migration of glioma cells. Furthermore, SLC39A7 promoted tumorigenesis in orthotopic models. We determined that SLC39A7 promotes the malignant behaviors of glioma by activating the TNF-α-mediated NF-κB signaling pathway. Conclusion: Our study revealed that SLC39A7 promotes the proliferation, invasion and migration of glioma cells via the TNF-α-mediated NF-κB signaling pathway, which provides potential targets for glioma therapy.
Collapse
Affiliation(s)
- Lian Chen
- Department of Neurosurgery, the First Hospital of China Medical University, NO. 155 North Nanjing Street, Shenyang, 110001 China
| | - Jinpeng Zhou
- Department of Neurosurgery, the First Hospital of China Medical University, NO. 155 North Nanjing Street, Shenyang, 110001 China
| | - Long Li
- Department of Neurosurgery, the First Hospital of China Medical University, NO. 155 North Nanjing Street, Shenyang, 110001 China
| | - Junshuang Zhao
- Department of Neurosurgery, the First Hospital of China Medical University, NO. 155 North Nanjing Street, Shenyang, 110001 China
| | - Hao Li
- Department of Neurosurgery, the First Hospital of China Medical University, NO. 155 North Nanjing Street, Shenyang, 110001 China
| | - Wei Zheng
- Department of Histology and Embryology, College of Basic Medical Science, China Medical University, NO. 77 Puhe Road, Shenyang, 110122 China
| | - Jinkun Xu
- Department of Neurosurgery, the First Hospital of China Medical University, NO. 155 North Nanjing Street, Shenyang, 110001 China
| | - Zhitao Jing
- Department of Neurosurgery, the First Hospital of China Medical University, NO. 155 North Nanjing Street, Shenyang, 110001 China
| |
Collapse
|
44
|
Carvalho IC, Mansur AAP, Carvalho SM, Mansur HS. Nanotheranostics through Mitochondria-targeted Delivery with Fluorescent Peptidomimetic Nanohybrids for Apoptosis Induction of Brain Cancer Cells. Nanotheranostics 2021; 5:213-239. [PMID: 33614399 PMCID: PMC7893535 DOI: 10.7150/ntno.54491] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/26/2021] [Indexed: 01/23/2023] Open
Abstract
Overview: Malignant brain tumors remain one of the greatest challenges faced by health professionals and scientists among the utmost lethal forms of cancer. Nanotheranostics can play a pivotal role in developing revolutionary nanoarchitectures with multifunctional and multimodal capabilities to fight cancer. Mitochondria are vital organelles to eukaryotic cells, which have been recognized as a significant target in cancer therapy where, by damaging the mitochondria, it will cause irreparable cell death or apoptosis. Methods: We designed and produced novel hybrid nanostructures comprising a fluorescent semiconductor core (AgInS2, AIS) and cysteine-modified carboxymethylcellulose (termed thiomer, CMC_Cys) conjugated with mitochondria-targeting peptides (KLA) forming a macromolecular shell for combining bioimaging and for inducing brain cancer cell (U-87 MG) death. Results: The optical and physicochemical properties of the nanoconjugates demonstrated suitability as photoluminescent nanostructures for cell bioimaging and intracellular tracking. Additionally, the results proved a remarkable killing activity towards glioblastoma cells of cysteine-bearing CMC conjugates coupled with KLA peptides through the half-maximal effective concentration values, approximately 70-fold higher compared to the conjugate analogs without Cys residues. Moreover, these thiomer-based pro-apoptotic drug nanoconjugates displayed higher lethality against U-87 MG cancer cells than doxorubicin, a model drug in chemotherapy, although extremely toxic. Remarkably, these peptidomimetic nanohybrids demonstrated a relative "protective effect" regarding healthy cells while maintaining high killing activity towards malignant brain cells. Conclusion: These findings pave the way for developing hybrid nanoarchitectures applied as targeted multifunctional platforms for simultaneous imaging and therapy against cancer while minimizing the high systemic toxicity and side-effects of conventional drugs in anticancer chemotherapy.
Collapse
Affiliation(s)
| | | | | | - Herman S. Mansur
- Center of Nanoscience, Nanotechnology, and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Belo Horizonte/MG, Brazil
| |
Collapse
|
45
|
Tan JY, Wijesinghe IVS, Alfarizal Kamarudin MN, Parhar I. Paediatric Gliomas: BRAF and Histone H3 as Biomarkers, Therapy and Perspective of Liquid Biopsies. Cancers (Basel) 2021; 13:cancers13040607. [PMID: 33557011 PMCID: PMC7913734 DOI: 10.3390/cancers13040607] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Gliomas are major causes of worldwide cancer-associated deaths in children. Generally, paediatric gliomas can be classified into low-grade and high-grade gliomas. They differ significantly from adult gliomas in terms of prevalence, molecular alterations, molecular mechanisms and predominant histological types. The aims of this review article are: (i) to discuss the current updates of biomarkers in paediatric low-grade and high-grade gliomas including their diagnostic and prognostic values, and (ii) to discuss potential targeted therapies in treating paediatric low-grade and high-grade gliomas. Our findings revealed that liquid biopsy is less invasive than tissue biopsy in obtaining the samples for biomarker detections in children. In addition, future clinical trials should consider blood-brain barrier (BBB) penetration of therapeutic drugs in paediatric population. Abstract Paediatric gliomas categorised as low- or high-grade vary markedly from their adult counterparts, and denoted as the second most prevalent childhood cancers after leukaemia. As compared to adult gliomas, the studies of diagnostic and prognostic biomarkers, as well as the development of therapy in paediatric gliomas, are still in their infancy. A body of evidence demonstrates that B-Raf Proto-Oncogene or V-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF) and histone H3 mutations are valuable biomarkers for paediatric low-grade gliomas (pLGGs) and high-grade gliomas (pHGGs). Various diagnostic methods involving fluorescence in situ hybridisation, whole-genomic sequencing, PCR, next-generation sequencing and NanoString are currently used for detecting BRAF and histone H3 mutations. Additionally, liquid biopsies are gaining popularity as an alternative to tumour materials in detecting these biomarkers, but still, they cannot fully replace solid biopsies due to several limitations. Although histone H3 mutations are reliable prognosis biomarkers in pHGGs, children with these mutations have a dismal prognosis. Conversely, the role of BRAF alterations as prognostic biomarkers in pLGGs is still in doubt due to contradictory findings. The BRAF V600E mutation is seen in the majority of pLGGs (as seen in pleomorphic xanthoastrocytoma and gangliomas). By contrast, the H3K27M mutation is found in the majority of paediatric diffuse intrinsic pontine glioma and other midline gliomas in pHGGs. pLGG patients with a BRAF V600E mutation often have a lower progression-free survival rate in comparison to wild-type pLGGs when treated with conventional therapies. BRAF inhibitors (Dabrafenib and Vemurafenib), however, show higher overall survival and tumour response in BRAF V600E mutated pLGGs than conventional therapies in some studies. To date, targeted therapy and precision medicine are promising avenues for paediatric gliomas with BRAF V600E and diffuse intrinsic pontine glioma with the H3K27M mutations. Given these shortcomings in the current treatments of paediatric gliomas, there is a dire need for novel therapies that yield a better therapeutic response. The present review discusses the diagnostic tools and the perspective of liquid biopsies in the detection of BRAF V600E and H3K27M mutations. An in-depth understanding of these biomarkers and the therapeutics associated with the respective challenges will bridge the gap between paediatric glioma patients and the development of effective therapies.
Collapse
Affiliation(s)
| | | | | | - Ishwar Parhar
- Correspondence: ; Tel.: +603-5514-6304; Fax: +603-5515-6341
| |
Collapse
|
46
|
Yu G, Butler MK, Abdelmaksoud A, Pang Y, Su YT, Rae Z, Dadkhah K, Kelly MC, Song YK, Wei JS, Terabe M, Atony R, Mentges K, Theeler BJ, Penas-Prado M, Butman J, Camphausen K, Zaghloul KA, Nduom E, Quezado M, Aldape K, Armstrong TS, Gilbert MR, Gulley JL, Khan J, Wu J. Case Report: Single-Cell Transcriptomic Analysis of an Anaplastic Oligodendroglioma Post Immunotherapy. Front Oncol 2021; 10:601452. [PMID: 33520712 PMCID: PMC7841290 DOI: 10.3389/fonc.2020.601452] [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: 09/01/2020] [Accepted: 11/20/2020] [Indexed: 11/13/2022] Open
Abstract
Glioma is the most common primary malignant brain tumor with a poor prognosis. Immune checkpoint inhibitors have been of great interest in investigation of glioma treatments. Here, we report single-cell transcriptomic analyses of two tumor areas from an oligodendroglioma taken from a patient who had multiple tumor recurrences, following several chemotherapies and radiation treatments. The patient subsequently received nivolumab and was considered have disease progression based on conventional diagnostic imaging after two cycles of treatment. He underwent a debulking surgical resection and pathological diagnosis was recurrent disease. During the surgery, tumor tissues were also collected from the enhancing and non-enhancing areas for a scRNAseq analysis to investigate the tumor microenvironment of these radiographically divergent areas. The scRNAseq analysis reveals a plethora of immune cells, suggesting that the increased mass observed on MRI may be partially a result of immune cell infiltration. The patient continued to receive immunotherapy after a short course of palliative radiation and remained free of disease progression for at least 12 months after the last surgery, suggesting a sustained response to immunotherapy. The scRNAseq analysis indicated that the radiological progression was in large part due to immune cell infiltrate and continued immunotherapy led to a positive clinical outcome in a patient who would have otherwise been admitted to hospice care with halting of immunotherapy. Our study demonstrates the potential of scRNAseq analyses in understanding the tumor microenvironment, which may assist the clinical decision-making process for challenging glioma cases following immunotherapy.
Collapse
Affiliation(s)
- Guangyang Yu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Madison K. Butler
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Abdalla Abdelmaksoud
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ying Pang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yu-Ting Su
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Zachary Rae
- Single Cell Analysis Facility, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Kimia Dadkhah
- Single Cell Analysis Facility, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Michael C. Kelly
- Single Cell Analysis Facility, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Young K. Song
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jun S. Wei
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Masaki Terabe
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ramya Atony
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kelly Mentges
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Brett J. Theeler
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Marta Penas-Prado
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - John Butman
- Diagnostic Radiology Department, The Clinical Center of the National Institutes of Health, Bethesda, MD, United States
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kareem A. Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Edjah Nduom
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Terri S. Armstrong
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mark R. Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
47
|
Nunna RS, Khalid S, Behbahani M, Mehta AI. Pediatric primary high-grade spinal glioma: a National Cancer Database analysis of current patterns in treatment and outcomes. Childs Nerv Syst 2021; 37:185-193. [PMID: 32533298 DOI: 10.1007/s00381-020-04722-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 06/03/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Pediatric primary high-grade spinal glioma (p-HGSG) is an extremely rare disease process, with little data within the current literature. Akin to primary high-grade gliomas, this cancer has been exemplified by dismal prognosis and poor response to modern treatment paradigms. This study seeks to investigate the current trends affecting overall survival using the National Cancer Database (NCDB). METHODS The NCDB was queried for p-HGSG between 2004 and 2016, by utilizing the designated diagnosis codes. Kaplan-Meier curves were generated, and log-rank testing was performed to analyze factors affecting overall survival. In addition, a Cox proportional-hazards model was used to perform multivariate regression analysis of survival outcomes. RESULTS A cohort of 97 patients was identified with a histologically confirmed p-HGSG. The overall incidence of p-HGSG in all pediatric spinal cord tumors is 7.5%, with a mean survival time of 25.3 months (SD, 21.0) and 5-year overall survival of 17.0%. The majority of patients underwent surgery (n = 87, 89.7%), radiotherapy (n = 73, 75.3%), and chemotherapy (n = 60, 61.9%). Univariate, multivariate, and Kaplan-Meier log-rank testing failed to demonstrate an association between performing surgery, extent of resection, radiotherapy, or chemotherapy with improved survival outcomes. CONCLUSIONS The current study constitutes the largest retrospective analysis of p-HGSGs to date, finding that current treatment options of surgery, radiotherapy, and chemotherapy have unclear benefit. This disease process has a poor prognosis without a current modality of treatment that conclusively alters survival. The risks and side effects of these treatment modalities must be carefully considered in such a highly aggressive disease process, especially given potentially limited survival benefits.
Collapse
Affiliation(s)
- Ravi S Nunna
- Department of Neurosurgery, University of Illinois at Chicago, 912 S. Wood Street, 451-N, Chicago, IL, 60612, USA
| | - Syed Khalid
- Department of Neurosurgery, University of Illinois at Chicago, 912 S. Wood Street, 451-N, Chicago, IL, 60612, USA
| | - Mandana Behbahani
- Department of Neurosurgery, University of Illinois at Chicago, 912 S. Wood Street, 451-N, Chicago, IL, 60612, USA
| | - Ankit I Mehta
- Department of Neurosurgery, University of Illinois at Chicago, 912 S. Wood Street, 451-N, Chicago, IL, 60612, USA.
| |
Collapse
|
48
|
Gahoi N, Syed P, Choudhary S, Epari S, Moiyadi A, Varma SG, Gandhi MN, Srivastava S. A Protein Microarray-Based Investigation of Cerebrospinal Fluid Reveals Distinct Autoantibody Signature in Low and High-Grade Gliomas. Front Oncol 2020; 10:543947. [PMID: 33415070 PMCID: PMC7784397 DOI: 10.3389/fonc.2020.543947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Gliomas are one of the most aggressive primary brain tumors arising from neural progenitor cells. Delayed diagnosis, invasive biopsy, and diagnostic challenges stems the need for specific, minimally-invasive, and early diagnostic biomarkers. Tumor-associated (TA) autoantibodies are measurable in the biofluids long before the onset of the symptoms, suggesting their role in early diagnosis and clinical management of the patients. In the current study, cerebrospinal fluid (CSF) samples from patients with low-grade glioma (LGG) and the Glioblastoma multiforme (GBM) that characterizes advanced disease were compared with healthy control samples to identify putative TA autoantibodies, using protein microarrays. The CSF samples from LGGs (n = 10), GBM (n = 7) were compared with the control CSF samples (n = 6). Proteins showing significant antigenic response were cross-verified. Proteins NOL4 (a cancer-testis antigen) and KALRN showed an antigenic response in the CSF of GBM patients, whereas, UTP4 and CCDC28A showed an antigenic response in low grade gliomas when compared with the control samples. TA autoantibodies identified in this study from the CSF of the patients could supplement current screening modalities. Further validation of these TA autoantibodies on a larger clinical cohort could provide cues towards relevance of these proteins in early diagnosis of the disease.
Collapse
Affiliation(s)
- Nikita Gahoi
- Wadhwani Research Center for Biosciences and Bioengineering, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India.,Centre for Research in Nanotechnology and Sciences, Indian Institute of Technology Bombay, Mumbai, India
| | - Parvez Syed
- Wadhwani Research Center for Biosciences and Bioengineering, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India.,Inme Oy, Turku, Finland
| | - Saket Choudhary
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India.,Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Centre, Mumbai, India
| | - Aliasgar Moiyadi
- Neurosurgical Services, Department of Surgical Oncology, Tata Memorial Centre and Homi Bhabha National Institute, Mumbai, India
| | - Santosh G Varma
- Deptartment of Biochemistry, Grant Govt. Medical College and Sir JJ Group of Hospitals, Mumbai, India.,Department of Biochemistry, BJ Medical College and Sassoon Hospital, Pune, India
| | - Mayuri N Gandhi
- Centre for Research in Nanotechnology and Sciences, Indian Institute of Technology Bombay, Mumbai, India
| | - Sanjeeva Srivastava
- Wadhwani Research Center for Biosciences and Bioengineering, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
49
|
Lidocaine inhibits glioma cell proliferation, migration and invasion by modulating the circEZH2/miR-181b-5p pathway. Neuroreport 2020; 32:52-60. [PMID: 33252475 DOI: 10.1097/wnr.0000000000001560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lidocaine is well known as a local anesthetic that has been reported to play an antitumor role in numerous cancers, including glioma. Circular RNAs (circRNAs) play multiple biological roles in cancers. The aim of this study was to determine the effects of lidocaine in glioma in vitro and in vivo and explore functional mechanisms. METHODS The effects of lidocaine on glioma progression were investigated by cell proliferation, migration and invasion using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay, colony formation assay and transwell assay. The expression of CD133 and glial fibrillary acidic protein (GFAP) was quantified by western blot to assess cell differentiation. The expression of circEZH2 and miR-181b-5p was detected by a quantitative real-time PCR (qRT-PCR). The target relationship between circEZH2 and miR-181b-5p was verified by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. The effect of lidocaine on tumor growth in vivo was investigated by establishing Xenograft models. RESULTS Lidocaine inhibited proliferation, migration, invasion and induced differentiation of glioma cells in vitro. Lidocaine suppressed the expression of circEZH2, and circEZH2 was highly expressed in glioma tissues and cells. CircEZH2 overexpression partly inhibited the function of lidocaine. CircEZH2 was a sponge of miR-181b-5p, and miR-181b-5p was downregulated in glioma tissues and cells. Besides, miR-181b-5p restoration reversed the effects of circEZH2 overexpression to repress the malignant behaviors of glioma cells. In addition, lidocaine mediated the circEZH2/miR-181b-5p axis to inhibit tumor growth in vivo. CONCLUSION Lidocaine suppressed glioma progression by modulating the circEZH2/miR-181b-5p pathway.
Collapse
|
50
|
Sapio L, Salzillo A, Ragone A, Illiano M, Spina A, Naviglio S. Targeting CREB in Cancer Therapy: A Key Candidate or One of Many? An Update. Cancers (Basel) 2020; 12:cancers12113166. [PMID: 33126560 PMCID: PMC7693618 DOI: 10.3390/cancers12113166] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Only 5% of all drug-related targets currently move from preclinical to clinical in cancer, and just some of them achieve patient’s bedside. Among others, intratumor heterogeneity and preclinical cancer model limitations actually represent the main reasons for this failure. Cyclic-AMP response element-binding protein (CREB) has been defined as a proto-oncogene in different tumor types, being involved in maintenance and progression. Due to its relevance in tumor pathophysiology, many CREB inhibitor compounds have been developed and tested over the years. Herein, we examine the current state-of-the-art of both CREB and CREB inhibitors in cancer, retracing some of the most significant findings of the last years. While the scientific statement confers on CREB a proactive role in cancer, its therapeutic potential is still stuck at laboratory bench. Therefore, pursuing every concrete result to achieve CREB inhibition in clinical might give chance and future to cancer patients worldwide. Abstract Intratumor heterogeneity (ITH) is considered the major disorienting factor in cancer treatment. As a result of stochastic genetic and epigenetic alterations, the appearance of a branched evolutionary shape confers tumor plasticity, causing relapse and unfavorable clinical prognosis. The growing evidence in cancer discovery presents to us “the great paradox” consisting of countless potential targets constantly discovered and a small number of candidates being effective in human patients. Among these, cyclic-AMP response element-binding protein (CREB) has been proposed as proto-oncogene supporting tumor initiation, progression and metastasis. Overexpression and hyperactivation of CREB are frequently observed in cancer, whereas genetic and pharmacological CREB downregulation affects proliferation and apoptosis. Notably, the present review is designed to investigate the feasibility of targeting CREB in cancer therapy. In particular, starting with the latest CREB evidence in cancer pathophysiology, we evaluate the advancement state of CREB inhibitor design, including the histone lysine demethylases JMJD3/UTX inhibitor GSKJ4 that we newly identified as a promising CREB modulator in leukemia cells. Moreover, an accurate analysis of strengths and weaknesses is also conducted to figure out whether CREB can actually represent a therapeutic candidate or just one of the innumerable preclinical cancer targets.
Collapse
|