1
|
Li F, Liu C, Nong W, Lin L, Ge Y, Luo B, Xiao S, Zhang Q, Xie X. Identification of potential biomarkers in cancer testis antigens for glioblastoma. Am J Transl Res 2023; 15:799-816. [PMID: 36915736 PMCID: PMC10006807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/16/2022] [Indexed: 03/16/2023]
Abstract
OBJECTIVE To screen and validate cancer testis antigens (CTAs) as potential biomarkers and explore their molecular mechanisms in glioblastoma (GBM). METHODS Ribonucleic acid sequencing (RNA-seq) and bioinformatics analyses were utilized to screen the highly expressed CTAs in GBM. Correlation analysis was used to identify potential biomarkers associated with tumor purity and prognosis. Immunohistochemistry was applied for detection of protein expression. Protein-protein interaction (PPI) network construction, functional enrichment analysis, and binding domain prediction were performed to investigate the underlying molecular mechanisms of GBM. RESULTS A total of 8 highly expressed CTAs were identified in GBM. One of them was PDZ-binding kinase (PBK). PBK messenger RNA (mRNA) was most highly expressed in GBM and associated with tumor purity and prognosis, PBK protein expression was also significantly increased in GBM tissues and correlated with p53 expression. Functional enrichment analysis revealed that the PBK related genes were predominantly enriched in cell cycle pathway with 38 genes enriched. The proteins encoding by these 38 genes were performed by binding domain prediction analysis, which demonstrated 15 proteins interacting with PBK. Most of these proteins were up regulated in GBM. CONCLUSION PBK is highly expressed in GBM. It may serve as a potential biomarker for GBM targeting therapy and the cell cycle modulator by interacting with certain key molecules of cell cycle in GBM.
Collapse
Affiliation(s)
- Feng Li
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China
| | - Chang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University Nanning, Guangxi, China.,Postdoctoral Research Station, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China
| | - Weixia Nong
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Lina Lin
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China
| | - Yingying Ge
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Shaowen Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University Nanning, Guangxi, China
| | - Qingmei Zhang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| | - Xiaoxun Xie
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University Nanning, Guangxi, China.,Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region Nanning, Guangxi, China
| |
Collapse
|
2
|
Correlation of Diffusion Tensor Imaging Parameters with the Pathological Grade of Brain Glioma and Expression of Vascular Endothelial Growth Factor and Ki-67. IRANIAN JOURNAL OF RADIOLOGY 2022. [DOI: 10.5812/iranjradiol-118135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background: Most brain gliomas are high-grade and likely to spread locally. Consequently, these patients commonly have a poor prognosis. Accurate identification of the malignancy grade of brain glioma before treatment is of great clinical significance. Objectives: This study aimed to explore the correlation of diffusion tensor imaging (DTI) parameters, fractional anisotropy (FA), and apparent diffusion coefficient (ADC) with the pathological grade of brain glioma and expression of vascular endothelial growth factor (VEGF) and Ki-67. Patients and Methods: A total of 116 patients were selected for this study from January 2018 to December 2019. All the participants underwent magnetic resonance imaging (MRI) and DTI before surgery, and the FA and ADC values were measured for the regions of interest. Surgically resected tumor specimens were collected for immunohistochemical assay. Finally, the FA and ADC values and positive expression rates of VEGF and Ki-67 were compared. Results: A significantly higher FA, besides the positive expression of VEGF and Ki-67, was reported in the high-grade group, whereas a lower ADC was found in this group compared to the low-grade group (P < 0.05). Areas of normal white matter and peritumoral edema had higher FA values, whereas lower ADCs were measured in these areas compared to the cerebrospinal fluid (P < 0.05). The FA of tumor parenchymal area was positively correlated with the World Health Organization (WHO) WHO class of tumors (r = 0.588, P = 0.028), and the expression of VEGF and Ki-67 was positively correlated with the WHO grade (r = 0.843, P = 0.002 and r = 0.743, P = 0.006, respectively). The FA of tumor parenchymal area was positively correlated with the expression of VEGF and Ki-67 (r = 0.654, P = 0.008 and r = 0.567, P = 0.012, respectively). However, the ADC of tumor parenchymal area was not significantly correlated with the WHO grade, VEGF expression, or Ki-67 expression (r = 0.143, P = 0.156, r = 0.232, P = 0.116, and r = 0.054, P = 0.179, respectively). Conclusion: The FA value, as a DTI parameter, is valuable for assessing the malignancy grade of tumor cells and can provide a proper reference for formulating treatment regimens for brain gliomas.
Collapse
|
3
|
Bi SQ, Zhang QM, Zeng X, Liu C, Nong WX, Xie H, Li F, Lin LN, Luo B, Ge YY, Xie XX. Combined treatment with epigenetic agents enhances anti-tumor activity of MAGE-D4 peptide-specific T cells by upregulating the MAGE-D4 expression in glioma. Front Oncol 2022; 12:873639. [PMID: 35992806 PMCID: PMC9382192 DOI: 10.3389/fonc.2022.873639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The study evaluated the efficacy of combined epigenetic drugs of decitabine (DAC), valproic acid (VPA), and trichostatin A (TSA) on immunotherapy against glioma. METHODS The expression and prognosis of MAGE-D4 in glioma were analyzed online, and the expression of MAGE-D4 and HLA-A2 in glioma induced by epigenetic drugs was detected by qRT-PCR, Western blot, and flow cytometry. The methylation status of the MAGE-D4 promoter was determined by pyrosequencing. An HLA-A2 restricted MAGE-D4 peptide was predicted and synthesized. An affinity assay and a peptide/HLA complex stability assay were performed to determine the affinity between peptide and HLA. CCK8 assay, CFSE assay, ELISA and ELISPOT were performed to detect the function of MAGE-D4 peptide-specific T cells. Flow cytometry, ELISA, and cytotoxicity assays were used to detect the cytotoxicity effect of MAGE-D4 peptide-specific T cells combined with epigenetic drugs against glioma in vitro. Finally, the glioma-loaded mouse model was applied to test the inhibitory effect of specific T cells on gliomas in vivo. RESULTS MAGE-D4 was highly expressed in glioma and correlated with poor prognosis. Glioma cells could be induced to express MAGE-D4 and HLA-A2 by epigenetic drugs. MAGE-D4-associated peptides were found that induce DCs to stimulate the highest T-cell activities of proliferation, IL-2 excretion, and IFN-γ secretion. MAGE-D4 peptide-specific T cells treated with TSA only or combining TSA and DAC had the most cytotoxicity effect, and its cytotoxicity effect on glioma cells decreased significantly after HLA blocking. In vivo experiments also confirmed that MAGE-D4-specific T cells inhibit TSA-treated glioma. CONCLUSION MAGE-D4 is highly expressed in glioma and correlated with the prognosis of glioma. The novel MAGE-D4 peptide identified was capable of inducing MAGE-D4-specific T cells that can effectively inhibit glioma growth, and the epigenetic drug application can enhance this inhibition.
Collapse
Affiliation(s)
- Shui-Qing Bi
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Department of Neurosurgery, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qing-Mei Zhang
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Key laboratory of Preclinical Medicine, Education Department of Guangxi Zhuang Autonomous region, Nanning, China
| | - Xia Zeng
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Chang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wei-Xia Nong
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Huan Xie
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Feng Li
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Li-Na Lin
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Bin Luo
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Key laboratory of Preclinical Medicine, Education Department of Guangxi Zhuang Autonomous region, Nanning, China
| | - Ying-Ying Ge
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiao-Xun Xie
- Department of Histology and Embryology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Key laboratory of Preclinical Medicine, Education Department of Guangxi Zhuang Autonomous region, Nanning, China
| |
Collapse
|
4
|
Expression and Prognostic Value of Melanoma-Associated Antigen D2 in Gliomas. Brain Sci 2022; 12:brainsci12080986. [PMID: 35892426 PMCID: PMC9330880 DOI: 10.3390/brainsci12080986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
Abstract
Introduction: The melanoma-associated antigen D2 (MAGED2) is one of the melanoma-associated antigen family members. It is commonly overexpressed in a variety of malignancies. However, the mechanism and function of MAGED2 in glioma remain unknown. Methods: The MAGED2 expression level and the correlations between clinical characteristics were analyzed with the data from the CGGA and TCGA datasets. MAGED2 expression in 98 glioma tissues was measured using RT-qPCR, Western blot, and immunohistochemistry. CCK-8, colony formation, and EdU assays were used to assess the effect of MAGED2 on U251-MG cell proliferation. Flow cytometry was used to track changes in the cell cycle and cell apoptosis following plasmid transfection with CRISPRi. Results: MAGED2 was shown to be highly expressed in glioma tissues, and high MAGED2 expression predicted poor prognosis. Furthermore, MAGED2 knockdown significantly inhibited the proliferation of U251-MG cells by preventing cell cycle arrest at the G0/G1 phase and triggering apoptosis. In line with in vitro findings, the results of the xenograft experiment and immunohistochemistry also showed that MAGED2 suppression inhibited tumor development and decreased Ki-67 expression levels. Conclusions: MAGED2 may be a possible biomarker for glioma and an important prognostic factor for glioma patients.
Collapse
|
5
|
Arora M, Kumari S, Singh J, Chopra A, Chauhan SS. Downregulation of Brain Enriched Type 2 MAGEs Is Associated With Immune Infiltration and Poor Prognosis in Glioma. Front Oncol 2020; 10:573378. [PMID: 33425727 PMCID: PMC7787151 DOI: 10.3389/fonc.2020.573378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Melanoma associated antigen (MAGE) is an extensively studied family of tumor-associated genes that share a common MAGE homology domain (MHD). Based upon their expression pattern, MAGE genes have been broadly classified into type 1 MAGEs (T1Ms) and type 2 MAGEs (T2Ms) categories. Interestingly, several T2Ms are highly expressed in the brain and involved in the regulation of neuronal development, differentiation, and survival. Available literature suggests possible tumor suppressor functions of a few T2Ms, while information available about their expression, regulation, and clinical significance in glioma is scanty. This prompted us to perform a comprehensive analysis of T2M expression in glioma. Gene expression data from glioma datasets: Oncomine, TCGA, and REMBRANDT study, were used to assess the mRNA expression of T2M genes (MAGED1, MAGED2, MAGED3, MAGED4, MAGED4B, MAGEE1, MAGEE2, MAGEF1, MAGEH1, MAGEL2, NSMCE3, and NDN), and their association with clinical characteristics and composition of the tumor microenvironment. Further, mutation, copy number alteration, and DNA methylation data from TCGA were assessed for determining potential mechanisms of T2Ms expression in glioma. Expression analysis revealed overexpression of MAGED subfamily genes in glioma, while other genes of this family exhibited reduced expression in advanced grades of this malignancy. Further, the expression of T2Ms exhibited varying extent of positive correlations with each other. Amongst downregulated T2Ms, MAGEH1 expression exhibited negative correlations with DNA methylation. Additionally, genes associated with MAGEH1 were enriched in Myc and Hedgehog signaling. Furthermore, T2Ms downregulation was associated with immune infiltration in glioma tissues and poor overall survival of glioma patients. In multivariate Cox regression analysis, MAGEH1 emerged as an independent prognosticator in lower grade glioma. Conclusively, these results suggest that expression of T2Ms is associated with important clinical and molecular features in glioma. Mechanistic studies may further provide novel insights into their role in glioma progression.
Collapse
Affiliation(s)
- Mohit Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sarita Kumari
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (Dr. BRA-IRCH), All India Institute of Medical Sciences, New Delhi, India
| | - Jay Singh
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (Dr. BRA-IRCH), All India Institute of Medical Sciences, New Delhi, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (Dr. BRA-IRCH), All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|