1
|
Chai Y, Zheng W, Hu S, Yang T, Chen G, Huang A. Novel molecular mechanisms of gamma-aminobutyric acid production mediated by LuxS/AI-2 quorum sensing system in Limosilactobacillus fermentum B41. Int J Food Microbiol 2025; 439:111269. [PMID: 40393098 DOI: 10.1016/j.ijfoodmicro.2025.111269] [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: 02/10/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
LuxS/AI-2 quorum sensing (QS) systems promotes the production of various functional factors in lactic acid bacteria (LAB) including the production of gamma-aminobutyric acid (GABA). However, the molecular mechanisms underlying GABA biosynthesis mediated by the LuxS/AI-2 QS system in Limosilactobacillus fermentum (L. fermentum) B41 remain unclear. This study investigated the molecular mechanisms of GABA production in L. fermentum B41 by integrating LuxS/AI-2 QS system, proteomic, western blot technology, and fermented milk preparation. The results showed that autoinducer-2 (AI-2) positively correlated with cell density and GABA content of L. fermentum B41, suggesting LuxS/AI-2-QS system regulation of GABA production. Proteomic analysis revealed 657 differentially expressed proteins (DEPs) between the LuxS/AI-2-QS system peaks (10h) and lows (2 h), of which 335 were up-regulated, including the critically important s-ribosylhomocysteinase (LuxS) and GABA transaminase (ArgD). The KEGG pathway enrichment analysis revealed that the DEPs were mainly involved in cysteine and methionine metabolism and GABA biosynthesis. Additionally, western blot confirmed that LuxS and ArgD are key proteins for GABA synthesis. The GABA-enriched fermented milk produced using L. fermentum B41 demonstrated a significantly high GABA content of 201.53 ± 7.83 μg/mL (p < 0.05), indicated it promotes GABA production via LuxS/AI-2-QS system. In summary, this study providing new insights into efficient GABA production and the development of GABA-enriched fermented milk.
Collapse
Affiliation(s)
- Yunmei Chai
- Department of Food Science, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Wentao Zheng
- Department of Food Science, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Shaomei Hu
- Department of Food Science, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Tingting Yang
- Department of Food Science, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Guangwei Chen
- Department of Food Science, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Aixiang Huang
- Department of Food Science, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China.
| |
Collapse
|
2
|
Guo S, Sidhu R, Ramar V, Guo AA, Wang G, Liu M. RNA Sequencing Identifies Novel Signaling Pathways and Potential Drug Target Genes Induced by FOSL1 in Glioma Progression and Stemness. Biologics 2025; 19:157-176. [PMID: 40206361 PMCID: PMC11980931 DOI: 10.2147/btt.s509774] [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: 12/21/2024] [Accepted: 03/01/2025] [Indexed: 04/11/2025]
Abstract
Background Glioblastoma is a highly aggressive brain tumor, and the transition from the proneural to mesenchymal subtype is associated with more aggressive and therapy-resistant features. However, the signaling pathways and genes involved in this transition remain largely undefined. Methods We utilized patient-derived xenograft (PDX) samples of glioblastoma, specifically PDX-L14, which exhibit both negative and overexpressed FOSL1 expression. mRNA expression profiles were assessed by RNA sequencing in these samples, followed by gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Gene Set Enrichment Analysis (GSEA). Validation of the hub genes was performed using qPCR and immunohistochemistry assays. Results Differentially expressed genes (DEGs) between FOSL1 overexpression groups were predominantly involved in ferroptosis, immune response, angiogenesis, vascular mimicry, autophagy, epithelial-mesenchymal transition (EMT), cancer cell stemness, temozolomide (TMZ) resistance, and NF-κB signaling. Downregulated DEGs were associated with TMZ resistance, glioma proliferation, RNA processing, and Wnt/β-catenin signaling. Key enrichment pathways, including NF-κB, Want, and BMP, are all critical for maintaining glioma stemness. FOSL1 was found to regulate RNA processing and ubiquitination. Notably, 8 upregulated (ITGA5, SDC1, PHLDB2, TNFRSF8, ADAM8, TLR7, STEAP3, and POU3F2) and 4 downregulated (IFIT1, FBXO16, ARL3, and BEX1) genes were identified, with implications for glioblastoma prognosis. Conclusion This transcriptome investigation emphasizes the diverse functions of FOSL1 in different biological processes and signaling networks during the shift from proneural to mesenchymal state in glioblastoma.
Collapse
Affiliation(s)
- Shanchun Guo
- RCMI Cancer Research Center and Department of Chemistry, Xavier University, New Orleans, LA, USA
| | - Rajveer Sidhu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Vanajothi Ramar
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Alyssa A Guo
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Guangdi Wang
- RCMI Cancer Research Center and Department of Chemistry, Xavier University, New Orleans, LA, USA
| | - Mingli Liu
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
3
|
Xue J, Liu H, Jiang L, Yin Q, Chen L, Wang M. Limitations of nomogram models in predicting survival outcomes for glioma patients. Front Immunol 2025; 16:1547506. [PMID: 40170838 PMCID: PMC11959071 DOI: 10.3389/fimmu.2025.1547506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Purpose Glioma represents a prevalent and malignant tumor of the central nervous system (CNS), and it is essential to accurately predict the survival of glioma patients to optimize their subsequent treatment plans. This review outlines the most recent advancements and viewpoints regarding the application of nomograms in glioma prognosis research. Design With an emphasis on the precision and external applicability of predictive models, we carried out a comprehensive review of the literature on the application of nomograms in glioma and provided a step-by-step guide for developing and evaluating nomograms. Results A summary of thirty-nine articles was produced. The majority of nomogram-building research has used limited patient samples, disregarded the proportional hazards (PH) assumption in Cox regression models, and some of them have failed to incorporate external validation. Furthermore, the predictive capability of nomograms is influenced by the selection of incorporated risk factors. Overall, the current predictive accuracy of nomograms is moderately credible. Conclusion The development and validation of nomogram models ought to adhere to a standardized set of criteria, thereby augmenting their worth in clinical decision-making and clinician-patient communication. Prior to the clinical application of a nomogram, it is imperative to thoroughly scrutinize its statistical foundation, rigorously evaluate its accuracy, and, whenever feasible, assess its external applicability utilizing multicenter databases.
Collapse
Affiliation(s)
- Jihao Xue
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Hang Liu
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Lu Jiang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qijia Yin
- Department of Urology or Nursing, Dazhou First People’s Hospital, Dazhou, Sichuan, China
- College of Nursing, Chongqing Medical University, Chongqing, Chongqing, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Ming Wang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Wu C, Qian Y, Jiang J, Li D, Feng L. Piperine inhibits the proliferation of colorectal adenocarcinoma by regulating ARL3-mediated endoplasmic reticulum stress. BIOMOLECULES & BIOMEDICINE 2025; 25:391-405. [PMID: 38972051 PMCID: PMC11734826 DOI: 10.17305/bb.2024.10525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Colorectal adenocarcinoma (COAD) is a significant cause of cancer-related mortality worldwide, necessitating the identification of novel therapeutic targets and treatments. This research aimed to investigate the role of ARL3 in COAD progression and to explore the effects of Piperine on ARL3 expression, cell proliferation, epithelial-mesenchymal transition (EMT), and endoplasmic reticulum (ER) stress. Bioinformatics analysis of The Cancer Genome Atlas (TCGA)-COAD, GSE39582, and GSE44861 datasets assessed ARL3 expression levels. Immunohistochemical data from the Human Protein Atlas (HPA) database confirmed ARL3 overexpression in COAD. The association of ARL3 with COAD clinical parameters and prognosis was also examined. COAD cells were treated with Piperine, and in vitro assays evaluated cell proliferation, apoptosis, EMT marker expression, and ER stress (ERS) responses. ARL3 overexpression in COAD correlated with poor prognosis and varied across pathological stages. Piperine treatment inhibited COAD cell proliferation in a concentration- and time-dependent manner, as indicated by reduced Ki-67 levels and decreased colony-forming ability. Piperine induced S-phase cell cycle arrest and facilitated apoptosis in COAD cells, evidenced by changes in Bax, Bcl-2, cleaved caspase-3, and cleaved Poly (ADP-ribose) polymerase (PARP) levels. Moreover, Piperine downregulated ARL3 expression in COAD cells, thereby suppressing transforming growth factor beta (TGF-β)-induced EMT. Additionally, Piperine attenuated the ARL3-mediated ER stress response, significantly reducing binding immunoglobulin protein (BiP), inositol-requiring enzyme 1 alpha (p-IRE1α), activating transcription factor 6 (ATF6), and C/EBP homologous protein (CHOP) levels. Piperine exerts anti-cancer effects in COAD by modulating ARL3 expression, disrupting cell cycle progression, inhibiting the EMT pathway, and regulating ERS. These findings suggest that Piperine holds promise as a therapeutic agent for COAD through its targeting of ARL3.
Collapse
Affiliation(s)
- Chenqu Wu
- Endoscopic Center, Central Hospital of Minhang, Shanghai, China
| | - Yanqing Qian
- Endoscopic Center, Central Hospital of Minhang, Shanghai, China
| | - Jun Jiang
- Endoscopic Center, Central Hospital of Minhang, Shanghai, China
| | - Deming Li
- Endoscopic Center, Central Hospital of Minhang, Shanghai, China
| | - Li Feng
- Endoscopic Center, Central Hospital of Minhang, Shanghai, China
| |
Collapse
|
5
|
Jiang Q, Ling GY, Yan J, Tan JY, Nong RB, Li JW, Deng T, Mo LG, Huang QR. Identification of prognostic risk score of disulfidptosis-related genes and molecular subtypes in glioma. Biochem Biophys Rep 2024; 37:101605. [PMID: 38188362 PMCID: PMC10768521 DOI: 10.1016/j.bbrep.2023.101605] [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: 08/19/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Background Programmed cell death is closely related to glioma. As a novel kind of cell death, the mechanism of disulfidptosis in glioma remains unclear. Therefore, it is of great importance to study the role of disulfidptosis-related genes (DRGs) in glioma. Methods We first investigated the genetic and transcriptional alterations of 15 DRGs. Two consensus cluster analyses were used to evaluate the association between DRGs and glioma subtypes. In addition, we constructed prognostic DRG risk scores to predict overall survival (OS) in glioma patients. Furthermore, we developed a nomogram to enhance the clinical utility of the DRG risk score. Finally, the expression levels of DRGs were verified by immunohistochemistry (IHC) staining. Results Most DRGs (14/15) were dysregulated in gliomas. The 15 DRGs were rarely mutated in gliomas, and only 50 of 987 samples (5.07 %) showed gene mutations. However, most of them had copy number variation (CNV) deletions or amplifications. Two distinct molecular subtypes were identified by cluster analysis, and DRG alterations were found to be related to the clinical characteristics, prognosis, and tumor immune microenvironment (TIME). The DRG risk score model based on 12 genes was developed and showed good performance in predicting OS. The nomogram confirmed that the risk score had a particularly strong influence on the prognosis of glioma. Furthermore, we discovered that low DRG scores, low tumor mutation burden, and immunosuppression were features of patients with better prognoses. Conclusion The DRG risk model can be used for the evaluation of clinical characteristics, prognosis prediction, and TIME estimation of glioma patients. These DRGs may be potential therapeutic targets in glioma.
Collapse
Affiliation(s)
| | | | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ju-Yuan Tan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ren-Bao Nong
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jian-Wen Li
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Teng Deng
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Li-Gen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qian-Rong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
6
|
Yin G, Huang J, Petela J, Jiang H, Zhang Y, Gong S, Wu J, Liu B, Shi J, Gao Y. Targeting small GTPases: emerging grasps on previously untamable targets, pioneered by KRAS. Signal Transduct Target Ther 2023; 8:212. [PMID: 37221195 DOI: 10.1038/s41392-023-01441-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/28/2023] [Accepted: 04/14/2023] [Indexed: 05/25/2023] Open
Abstract
Small GTPases including Ras, Rho, Rab, Arf, and Ran are omnipresent molecular switches in regulating key cellular functions. Their dysregulation is a therapeutic target for tumors, neurodegeneration, cardiomyopathies, and infection. However, small GTPases have been historically recognized as "undruggable". Targeting KRAS, one of the most frequently mutated oncogenes, has only come into reality in the last decade due to the development of breakthrough strategies such as fragment-based screening, covalent ligands, macromolecule inhibitors, and PROTACs. Two KRASG12C covalent inhibitors have obtained accelerated approval for treating KRASG12C mutant lung cancer, and allele-specific hotspot mutations on G12D/S/R have been demonstrated as viable targets. New methods of targeting KRAS are quickly evolving, including transcription, immunogenic neoepitopes, and combinatory targeting with immunotherapy. Nevertheless, the vast majority of small GTPases and hotspot mutations remain elusive, and clinical resistance to G12C inhibitors poses new challenges. In this article, we summarize diversified biological functions, shared structural properties, and complex regulatory mechanisms of small GTPases and their relationships with human diseases. Furthermore, we review the status of drug discovery for targeting small GTPases and the most recent strategic progress focused on targeting KRAS. The discovery of new regulatory mechanisms and development of targeting approaches will together promote drug discovery for small GTPases.
Collapse
Affiliation(s)
- Guowei Yin
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Jing Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Johnny Petela
- Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Hongmei Jiang
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuetong Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Siqi Gong
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiaxin Wu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Bei Liu
- National Biomedical Imaging Center, School of Future Technology, Peking University, Beijing, 100871, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology, Chengdu, 610072, China.
| | - Yijun Gao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
7
|
Abulaiti A, Maimaiti A, Yiming N, Fu Q, Li S, Li Y, Wang Y, Zhou Q. Molecular subtypes based on PANoptosis-related genes and tumor microenvironment infiltration characteristics in lower-grade glioma. Funct Integr Genomics 2023; 23:84. [PMID: 36930242 DOI: 10.1007/s10142-023-01003-5] [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: 02/02/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
The growth of cancer, the effectiveness of treatment, and prognosis are all closely related to PANoptosis (include pyroptosis, apoptosis, and necroptosis). It remains unclear whether PANoptosis genes (PANGs) may contribute to lower-grade glioma (LGG) tumor microenvironment (TME). In this study, we collected 1203 LGG samples from three public databases and reported that PANoptosis involves TME interaction and prognosis. Firstly, we provided a comprehensive review of the pan-cancer landscape of PANGs in terms of expression characteristics, prognostic value, mutational profile, and pathway regulation. Then, we identified two distinct PANclusters, each with its own molecular, clinical, and immunological profile. We then developed a scoring system for LGG patients called PANscore. As well as investigating immune characteristics, tumor mutational characteristics, and drug sensitivity, we examined the differences between groups with high PANscores and those with low PANscores. Based on this PANscore and clinicopathological variables, an instant nomogram for predicting clinical survival in LGG patients was developed. Our thorough examination of PANGs in LGG revealed their probable function in TME, as well as their clinicopathological characteristics and prognosis. These discoveries could deepen our comprehension of PANGs in LGG and provide doctors fresh perspectives on how to forecast prognosis and create more efficient, individualized treatment plans.
Collapse
Affiliation(s)
- Aimitaji Abulaiti
- Department of Neurosurgery, Neurosurgery Centre, the First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, China
| | - Aierpati Maimaiti
- Department of Neurosurgery, Neurosurgery Centre, the First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, China
| | - Nadire Yiming
- Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Qiang Fu
- Department of Neurosurgery, Neurosurgery Centre, the First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, China
| | - Shaoshan Li
- Department of Neurosurgery, Neurosurgery Centre, the First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, China
| | - Yabin Li
- Department of Neurosurgery, Neurosurgery Centre, the First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, China
| | - Yongxin Wang
- Department of Neurosurgery, Neurosurgery Centre, the First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, China.
| | - Qingjiu Zhou
- Department of Neurosurgery, Neurosurgery Centre, the First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, China.
| |
Collapse
|
8
|
Ozair A, Bhat V, Alisch RS, Khosla AA, Kotecha RR, Odia Y, McDermott MW, Ahluwalia MS. DNA Methylation and Histone Modification in Low-Grade Gliomas: Current Understanding and Potential Clinical Targets. Cancers (Basel) 2023; 15:1342. [PMID: 36831683 PMCID: PMC9954183 DOI: 10.3390/cancers15041342] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Gliomas, the most common type of malignant primary brain tumor, were conventionally classified through WHO Grades I-IV (now 1-4), with low-grade gliomas being entities belonging to Grades 1 or 2. While the focus of the WHO Classification for Central Nervous System (CNS) tumors had historically been on histopathological attributes, the recently released fifth edition of the classification (WHO CNS5) characterizes brain tumors, including gliomas, using an integration of histological and molecular features, including their epigenetic changes such as histone methylation, DNA methylation, and histone acetylation, which are increasingly being used for the classification of low-grade gliomas. This review describes the current understanding of the role of DNA methylation, demethylation, and histone modification in pathogenesis, clinical behavior, and outcomes of brain tumors, in particular of low-grade gliomas. The review also highlights potential diagnostic and/or therapeutic targets in associated cellular biomolecules, structures, and processes. Targeting of MGMT promoter methylation, TET-hTDG-BER pathway, association of G-CIMP with key gene mutations, PARP inhibition, IDH and 2-HG-associated processes, TERT mutation and ARL9-associated pathways, DNA Methyltransferase (DNMT) inhibition, Histone Deacetylase (HDAC) inhibition, BET inhibition, CpG site DNA methylation signatures, along with others, present exciting avenues for translational research. This review also summarizes the current clinical trial landscape associated with the therapeutic utility of epigenetics in low-grade gliomas. Much of the evidence currently remains restricted to preclinical studies, warranting further investigation to demonstrate true clinical utility.
Collapse
Affiliation(s)
- Ahmad Ozair
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
- Faculty of Medicine, King George’s Medical University, Lucknow 226003, India
| | - Vivek Bhat
- St. John’s Medical College, Bangalore 560034, India
| | - Reid S. Alisch
- Department of Neurosurgery, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Atulya A. Khosla
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
| | - Rupesh R. Kotecha
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Yazmin Odia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Michael W. McDermott
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Miami Neuroscience Institute, Baptist Health South Florida, Miami, FL 33176, USA
| | - Manmeet S. Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
- Miami Neuroscience Institute, Baptist Health South Florida, Miami, FL 33176, USA
| |
Collapse
|
9
|
Zhu X, Chen D, Sun Y, Yang S, Wang W, Liu B, Gao P, Li X, Wu L, Ma S, Lin W, Ma J, Yan D. LncRNA WEE2-AS1 is a diagnostic biomarker that predicts poor prognoses in patients with glioma. BMC Cancer 2023; 23:120. [PMID: 36747161 PMCID: PMC9901081 DOI: 10.1186/s12885-023-10594-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Glioma is characterized by high morbidity, high mortality, and poor prognosis. Despite tremendous advances in the treatment of glioma, the prognosis of patients with glioma is still unsatisfactory. There is an urgent need to discover novel molecular markers that effectively predict prognosis in patients with glioma. The investigation of the role of WEE2-AS1 in various tumors is an emerging research field, but the biological function and prognostic value of WEE2-AS1 in glioma have rarely been reported. This study aimed to assess the value of WEE2-AS1 as a potential prognostic marker of glioma. METHODS Gene expression (RNA-Seq) data of patients with glioma were extracted from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. The Wilcoxon rank sum test was used to analyze the expression of WEE2-AS1 in the cells and tissues of glioma. The Kruskal-Wallis rank sum test, Wilcoxon rank sum test, and logistic regression were used to evaluate the relationship between clinical variables and expression of WEE2-AS1. Cox regression analysis and the Kaplan-Meier method were used to evaluate the prognostic factors in glioma. A nomogram based on Cox multivariate analysis was used to predict the impact of WEE2-AS1 on glioma prognosis. Gene Set Enrichment Analysis (GSEA) was used to identify key WEE2-AS1-associated signaling pathways. Spearman's rank correlation was used to elucidate the association between WEE2-AS1 expression and immune cell infiltration levels. RESULTS We found that WEE2-AS1 was overexpressed in a variety of cancers, including glioma. High expression of WEE2-AS1 was associated with glioma progression. We determined that the expression of WEE2-AS1 might be an independent risk factor for the survival and prognosis of patients with glioma. We further observed that the mechanism of WEE2-AS1-mediated tumorigenesis involved neuroactive ligand-receptor interaction, cell cycle, and the infiltration of immune cells into the glioma microenvironment. CONCLUSION These findings demonstrate that WEE2-AS1 is a promising biomarker for the diagnosis and prognosis of patients with glioma. An increased understanding of its effects on the regulation of cell growth may lead to the development of clinical applications that improve the prognostic status of patients with glioma.
Collapse
Affiliation(s)
- Xuqiang Zhu
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Di Chen
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Yiyu Sun
- grid.16821.3c0000 0004 0368 8293Department of Neurosurgery, School of Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, 200030 Shanghai, China
| | - Shuo Yang
- grid.16821.3c0000 0004 0368 8293Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Weiguang Wang
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Bing Liu
- grid.16821.3c0000 0004 0368 8293Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Peng Gao
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Xueyuan Li
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Lixin Wu
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Siqi Ma
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Wenyang Lin
- grid.412633.10000 0004 1799 0733Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, Henan China
| | - Jiwei Ma
- grid.493088.e0000 0004 1757 7279Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100 Henan Shanghai, China
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
10
|
Bader JM, Deigendesch N, Misch M, Mann M, Koch A, Meissner F. Proteomics separates adult-type diffuse high-grade gliomas in metabolic subgroups independent of 1p/19q codeletion and across IDH mutational status. Cell Rep Med 2023; 4:100877. [PMID: 36584682 PMCID: PMC9873829 DOI: 10.1016/j.xcrm.2022.100877] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 07/15/2022] [Accepted: 12/07/2022] [Indexed: 12/30/2022]
Abstract
High-grade adult-type diffuse gliomas are malignant neuroepithelial tumors with poor survival rates in combined chemoradiotherapy. The current WHO classification is based on IDH1/2 mutational and 1p/19q codeletion status. Glioma proteome alterations remain undercharacterized despite their promise for a better molecular patient stratification and therapeutic target identification. Here, we use mass spectrometry to characterize 42 formalin-fixed, paraffin-embedded (FFPE) samples from IDH-wild-type (IDHwt) gliomas, IDH-mutant (IDHmut) gliomas with and without 1p/19q codeletion, and non-neoplastic controls. Based on more than 5,500 quantified proteins and 5,000 phosphosites, gliomas separate by IDH1/2 mutational status but not by 1p/19q status. Instead, IDHmut gliomas split into two proteomic subtypes with widespread perturbations, including aerobic/anaerobic energy metabolism. Validations with three independent glioma proteome datasets confirm these subgroups and link the IDHmut subtypes to the established proneural and classic/mesenchymal subtypes in IDHwt glioma. This demonstrates common phenotypic subtypes across the IDH status with potential therapeutic implications for patients with IDHmut gliomas.
Collapse
Affiliation(s)
- Jakob Maximilian Bader
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nikolaus Deigendesch
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Martin Misch
- Department of Neurosurgery, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Arend Koch
- Department of Neuropathology, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany.
| | - Felix Meissner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Department of Systems Immunology and Proteomics, Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany.
| |
Collapse
|
11
|
Gui Z, Ying X, Liu C. NXPH4 Used as a New Prognostic and Immunotherapeutic Marker for Muscle-Invasive Bladder Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4271409. [PMID: 36245981 PMCID: PMC9553512 DOI: 10.1155/2022/4271409] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022]
Abstract
Background One of the most common malignant tumors of the urinary system is muscle-invasive bladder cancer (MIBC). With the increased use of immunotherapy, its importance in the field of cancer is becoming abundantly evident. This study classifies MIBC according to GSVA score from the perspective of the GSEA immune gene set. Methods This study integrated the sequencing and clinical data of MIBC patients in TCGA and GEO databases, then scored the data using the GSVA algorithm, the CNMF algorithm was implemented to divide the subtypes of GEO and TCGA datasets, respectively, and finally screened and determined the key pathways in combination with clinical data. Simultaneously, LASSO Cox regression model was constructed based on key pathway genes to assess the model's predictive ability (ROC) and describe the immune landscape differences between high- and low-risk groups; key genes were further analyzed and verified in patient tissues. Results 404 TCGA and 297 GEO datasets were divided into C1-3 groups (TCGA-C1:120/C2:152/C3:132; GEO- C1:112/C2:101/C3:84), of which TCGA-C2 (n = 152) subtype and GEO-C1 (n = 112) subtype had the worst prognosis. LASSO Cox regression model with ROC (train set = 0.718, test set = 0.667) could be constructed. When combined with the Cancer Immunome Atlas database, it was found that patients with high-risk scores were more sensitive to PD-1 inhibitor and PD-1 inhibitor combined with CTLA-4. NXPH4, as a key gene, plays a role in MIBC with tissue validation results show that nxph4 is highly expressed in tumor. Conclusion The immune gene score of MIBC data in TCGA and GEO databases was successfully evaluated using GSVA in this research. The lasso Cox expression model was successfully constructed by screening immune genes, the high-risk group had a worse prognosis and higher sensitivity to immunotherapy, PD-1 inhibitors or PD-1 combined with CTLA-4 inhibitors can be preferentially used in high-risk patients who are sensitive to immunotherapy, and NXPH4 may be a molecular target to adjust the effect of immunotherapy.
Collapse
Affiliation(s)
- Zhiming Gui
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Xiaoling Ying
- Laboratory of Translational Medicine, The First Affiliated Hospital of Sun Yat sen University, 510000, China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
12
|
HOXC6 Regulates the Epithelial-Mesenchymal Transition through the TGF-β/Smad Signaling Pathway and Predicts a Poor Prognosis in Glioblastoma. JOURNAL OF ONCOLOGY 2022; 2022:8016102. [PMID: 35571491 PMCID: PMC9098331 DOI: 10.1155/2022/8016102] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 11/18/2022]
Abstract
Background The HOX gene family of transcription factors, characterized by conserved homeodomains, is positively correlated with the resistance to chemotherapy drugs and poor prognosis, as well as the initiating potential of gliomas. However, there are few studies regarding the HOXC6 gene in glioma cells. Therefore, in the present study, we explored the regulatory roles and detailed mechanisms underlying the relationship between HOXC6 and the progression of GBM. Methods The expression levels and prognostic value of HOXC6 in GBM were evaluated using the data obtained from the GCCA, GEPIA, and ONCOMINE databases. The relationship between GBM prognosis and levels of HOXC6 was identified using Kaplan-Meier curves. The protein levels of HOXC6 in GBM and adjacent normal tissues were identified via Western blot and immunohistochemistry (IHC) staining methods. Lentiviruses containing full-length HOXC6 and HOXC6 specific siRNA sequences were used to overexpress and knock down, respectively, the expression of HOXC6 in U87 and U251 cells. The role of HOXC6 in the regulation of migration and proliferation of GBM cells was accessed using Transwell, wound healing, CCK-8, and colony formation assays. The activation of the TGF-β/Smad signaling pathway was detected via Western blotting. Results Compared to normal tissues and control cells, GBM tissues and cell lines showed higher expressions of HOXC6. The expression of HOXC6 was associated with disease-free and the overall survival of GBM patients. Additionally, positive correlations between the expression of HOXC6 and the migration and proliferation of GBM cells were observed in vitro. The mechanistic analyses indicated that HOXC6 exerts its promotive effect on the progression and invasion of glioma cells by promoting the activation of the EMT and TGF-β/Smad signaling pathways. Conclusions HOXC6 enhances the migration and proliferation of GBM by activating the EMT signaling pathway.
Collapse
|
13
|
Role of Calcium Signaling Pathway-Related Gene Regulatory Networks in Ischemic Stroke Based on Multiple WGCNA and Single-Cell Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:8060477. [PMID: 34987704 PMCID: PMC8720592 DOI: 10.1155/2021/8060477] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/20/2021] [Accepted: 11/27/2021] [Indexed: 01/28/2023]
Abstract
Background This study is aimed at investigating the changes in relevant pathways and the differential expression of related gene expression after ischemic stroke (IS) at the single-cell level using multiple weighted gene coexpression network analysis (WGCNA) and single-cell analysis. Methods The transcriptome expression datasets of IS samples and single-cell RNA sequencing (scRNA-seq) profiles of cerebrovascular tissues were obtained by searching the Gene Expression Omnibus (GEO) database. First, gene pathway scoring was calculated via gene set variation analysis (GSVA) and was imported into multiple WGCNA to acquire key pathways and pathway-related hub genes. Furthermore, SCENIC was used to identify transcription factors (TFs) regulating these core genes using scRNA-seq data. Finally, the pseudotemporal trajectory analysis was used to analyse the role of these TFs on various cell types under hypoxic and normoxic conditions. Results The scores of 186 KEGG pathways were obtained via GSVA using microarray expression profiles of 40 specimens. WGCNA of the KEGG pathways revealed the two following pathways: calcium signaling pathway and neuroactive ligand-receptor interaction pathways. Subsequently, WGCNA of the gene expression matrix of the samples revealed the calcium signaling pathway-related genes (AC079305.10, BCL10, BCL2A1, BRE-AS1, DYNLL2, EREG, and PTGS2) that were identified as core genes via correlation analysis. Furthermore, SCENIC and pseudotemporal analysis revealed JUN, IRF9, ETV5, and PPARA score gene-related TFs. Jun was found to be associated with hypoxia in endothelial cells, whereas Irf9 and Etv5 were identified as astrocyte-specific TFs associated with oxygen concentration in the mouse cerebral cortex. Conclusions Calcium signaling pathway-related genes (AC079305.10, BCL10, BCL2A1, BRE-AS1, DYNLL2, EREG, and PTGS2) and TFs (JUN, IRF9, ETV5, and PPARA) were identified to play a key role in IS. This study provides a new perspective and basis for investigating the pathogenesis of IS and developing new therapeutic approaches.
Collapse
|
14
|
Brito C, Costa-Silva B, Barral DC, Pojo M. Unraveling the Relevance of ARL GTPases in Cutaneous Melanoma Prognosis through Integrated Bioinformatics Analysis. Int J Mol Sci 2021; 22:9260. [PMID: 34502169 PMCID: PMC8431576 DOI: 10.3390/ijms22179260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/23/2022] Open
Abstract
Cutaneous melanoma (CM) is the deadliest skin cancer, whose molecular pathways underlying its malignancy remain unclear. Therefore, new information to guide evidence-based clinical decisions is required. Adenosine diphosphate (ADP)-ribosylation factor-like (ARL) proteins are membrane trafficking regulators whose biological relevance in CM is undetermined. Here, we investigated ARL expression and its impact on CM prognosis and immune microenvironment through integrated bioinformatics analysis. Our study found that all 22 ARLs are differentially expressed in CM. Specifically, ARL1 and ARL11 are upregulated and ARL15 is downregulated regardless of mutational frequency or copy number variations. According to TCGA data, ARL1 and ARL15 represent independent prognostic factors in CM as well as ARL11 based on GEPIA and OncoLnc. To investigate the mechanisms by which ARL1 and ARL11 increase patient survival while ARL15 reduces it, we evaluated their correlation with the immune microenvironment. CD4+ T cells and neutrophil infiltrates are significantly increased by ARL1 expression. Furthermore, ARL11 expression was correlated with 17 out of 21 immune infiltrates, including CD8+ T cells and M2 macrophages, described as having anti-tumoral activity. Likewise, ARL11 is interconnected with ZAP70, ADAM17, and P2RX7, which are implicated in immune cell activation. Collectively, this study provides the first evidence that ARL1, ARL11, and ARL15 may influence CM progression, prognosis, and immune microenvironment remodeling.
Collapse
Affiliation(s)
- Cheila Brito
- Unidade de Investigação em Patobiologia Molecular (UIPM) do Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal;
| | - Bruno Costa-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, Avenida de Brasília, 1400-038 Lisbon, Portugal;
| | - Duarte C. Barral
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal;
| | - Marta Pojo
- Unidade de Investigação em Patobiologia Molecular (UIPM) do Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal;
| |
Collapse
|
15
|
Zhang J, Gu J, Guo S, Huang W, Zheng Y, Wang X, Zhang T, Zhao W, Ni B, Fan Y, Wang H. Establishing and validating a pathway prognostic signature in pancreatic cancer based on miRNA and mRNA sets using GSVA. Aging (Albany NY) 2020; 12:22840-22858. [PMID: 33197892 PMCID: PMC7746356 DOI: 10.18632/aging.103965] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 07/30/2020] [Indexed: 12/25/2022]
Abstract
Pancreatic cancer (PC) is a severe disease with the highest mortality rate among various cancers. It is urgent to find an effective and accurate way to predict the survival of PC patients. Gene set variation analysis (GSVA) was used to establish and validate a miRNA set-based pathway prognostic signature for PC (miPPSPC) and a mRNA set-based pathway prognostic signature for PC (mPPSPC) in independent datasets. An optimized miPPSPC was constructed by combining clinical parameters. The miPPSPC, optimized miPPSPC and mPPSPC were established and validated to predict the survival of PC patients and showed excellent predictive ability. Four metabolic pathways and one oxidative stress pathway were identified in the miPPSPC, whereas linoleic acid metabolism and the pentose phosphate pathway were identified in the mPPSPC. Key factors of the pentose phosphate pathway and linoleic acid metabolism, G6PD and CYP2C8/9/18/19, respectively, are related to the survival of PC patients according to our tissue microarray. Thus, the miPPSPC, optimized miPPSPC and mPPSPC can predict the survival of PC patients efficiently and precisely. The metabolic and oxidative stress pathways may participate in PC progression.
Collapse
Affiliation(s)
- Junfeng Zhang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| | - Jianyou Gu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, P R China
| | - Shixiang Guo
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| | - Wenjie Huang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, P R China
| | - Yao Zheng
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| | - Xianxing Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| | - Tao Zhang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| | - Weibo Zhao
- PLA Strategic Support Force Characteristic Medical Center (The 306th Hospital of PLA), Beijing 100101, P R China
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P R China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing 400038, P R China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing 400038, P R China
| | - Yingfang Fan
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, P R China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| |
Collapse
|
16
|
Xiao K, Tan J, Yuan J, Peng G, Long W, Su J, Xiao Y, Xiao Q, Wu C, Qin C, Hu L, Liu K, Liu S, Zhou H, Ning Y, Ding X, Liu Q. Prognostic value and immune cell infiltration of hypoxic phenotype-related gene signatures in glioblastoma microenvironment. J Cell Mol Med 2020; 24:13235-13247. [PMID: 33009892 PMCID: PMC7701576 DOI: 10.1111/jcmm.15939] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/14/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a malignant intracranial tumour with the highest proportion and lethality. It is characterized by invasiveness and heterogeneity. However, the currently available therapies are not curative. As an essential environmental cue that maintains glioma stem cells, hypoxia is considered the cause of tumour resistance to chemotherapy and radiation. Growing evidence shows that immunotherapy focusing on the tumour microenvironment is an effective treatment for GBM; however, the current clinicopathological features cannot predict the response to immunotherapy and provide accurate guidance for immunotherapy. Based on the ESTIMATE algorithm, GBM cases of The Cancer Genome Atlas (TCGA) data set were classified into high- and low-immune/stromal score groups, and a four-gene tumour environment-related model was constructed. This model exhibited good efficiency at forecasting short- and long-term prognosis and could also act as an independent prognostic biomarker. Additionally, this model and four of its genes (CLECL5A, SERPING1, CHI3L1 and C1R) were found to be associated with immune cell infiltration, and further study demonstrated that these four genes might drive the hypoxic phenotype of perinecrotic GBM, which affects hypoxia-induced glioma stemness. Therefore, these might be important candidates for immunotherapy of GBM and deserve further exploration.
Collapse
Affiliation(s)
- Kai Xiao
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Jian Yuan
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Gang Peng
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Wenyong Long
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Jun Su
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Yao Xiao
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Qun Xiao
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Changwu Wu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China.,Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Chaoying Qin
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Lili Hu
- Medical College of Hunan Normal University, Changsha, China
| | - Kaili Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, China
| | - Shunlian Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, China
| | - Hao Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, China
| | - Yichong Ning
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, China
| | - Xiaofeng Ding
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| |
Collapse
|
17
|
Tan Y, Zhang S, Xiao Q, Wang J, Zhao K, Liu W, Huang K, Tian W, Niu H, Lei T, Shu K. Prognostic significance of ARL9 and its methylation in low-grade glioma. Genomics 2020; 112:4808-4816. [PMID: 32882327 PMCID: PMC7462573 DOI: 10.1016/j.ygeno.2020.08.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
This study aimed to determine the value of ARL9 expression or methylation as a biomarker for LGG survival. We investigated the expression, methylation, prognosis and immune significance of ARL9 through bioinformatics analysis. ARL9 is negatively regulated by ARL9 methylation, leading to its low expression in LGG tissues. Both low ARL9 expression and hypermethylation predicted favorable OS and PFS in LGG patients, according to the TCGA database. Cox regression demonstrated that low ARL9 expression and ARL9 hypermethylation were independent biomarkers for OS. Moreover, three other glioma databases were utilized to verify the prognostic role of ARL9 in LGG, and the similar results were reached. A meta-analysis revealed that low ARL9 expression was closely relevant to better OS. Finally, ARL9 expression exhibited a close correlation with some immune cells, especially CD8+ T cells. ARL9 could constitute a promising prognostic biomarker, and probably plays an important role in immune cell infiltration in LGG. This is the first study to report the clinical and prognostic significance of ARL9, a methylation-driven gene,in LGG. Meta-analysis could be used for bioinformatics analysis to assess the overall effect of the gene from different datasets. ARL9 probably plays a role in the infiltration of immune cells, and acts as a promising prognostic marker in LGG patients.
Collapse
Affiliation(s)
- Yutang Tan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Suojun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qungen Xiao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Junwen Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kai Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weihua Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kuan Huang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weidong Tian
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Neurosurgery, First Affiliated Hospital of Medical College, Shihezi University, Xinjiang 832000, China
| | - Hongquan Niu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
18
|
Wu H, Wang C, Liu J, Zhou D, Chen D, Liu Z, Wu A, Yang L, Chang J, Luo C, Cheng W, Shen S, Bai Y, Mu X, Li C, Wang Z, Chen L. Evaluation of a tumor electric field treatment system in a rat model of glioma. CNS Neurosci Ther 2020; 26:1168-1177. [PMID: 32734621 PMCID: PMC7564191 DOI: 10.1111/cns.13441] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
Objective Glioma is a devastating disease lacking effective treatment. Tumor electric field therapy is emerging as a novel non‐invasive therapy. The current study evaluates the efficacy and safety of a self‐designed tumor electric field therapy system (TEFTS ASCLU‐300) in a rat orthotopic transplantation model of glioma. Methods A model of intracranial orthotopic transplantation was established in rats using glioma C6 cells. For electric field therapy, glioma‐bearing rats were exposed to alternating electric fields generated by a self‐developed TEFTS starting on either 1st (Group 2) or 3rd (Group 3) day after transplantation, while other conditions were maintained the same as non‐treated rats (Group 1). Glioma size, body weight, and overall survival (OS) were compared between groups. Immunohistochemical staining was applied to access tumor cell death and microvessel density within the tumor. In addition, the systemic effects of TEFTS on blood cells, vital organs, and hepatorenal functions were evaluated. Results TEFTS treatment significantly elongated the OS of tumor‐bearing rats compared with non‐treated rats (non‐treated vs treated: 24.77 ± 7.08 days vs 40.31 ± 19.11 days, P = .0031). Continuous TEFTS treatment starting on 1st or 3rd day significantly reduced glioma size at 2 and 3 weeks after tumor cell inoculation (Week 2: Group 1:289.95 ± 101.69 mm3; Group 2:70.45 ± 17.79 mm3; Group 3:73.88 ± 33.21 mm3, P < .0001. Week 3: Group 1:544.096 ± 78.53 mm3; Group 2:187.58 ± 78.44 mm3; Group 3:167.14 ± 109.96 mm3, P = .0005). Continuous treatment for more than 4 weeks inhibited tumor growth. The TEFTS treatment promoted tumor cell death, as demonstrated by increased number of Caspase 3+ cells within the tumor (non‐treated vs treated: 38.06 ± 10.04 vs 68.57 ± 8.09 cells/field, P = .0007), but had minimal effect on microvessel density, as shown by CD31 expression (non‐treated vs treated: 1.63 ± 0.09 vs 1.57 ± 0.13% of positively stained areas, P > .05). No remarkable differences were observed in hepatorenal function, blood cell counts, or other vital organs between non‐treated and treated groups. Conclusion The TEFTS developed by our research team was proved to be effective and safe to inhibit tumor growth and improve general outcomes in a rat model of brain glioma.
Collapse
Affiliation(s)
- Hao Wu
- Chinese PLA Institute of Neurosurgery, Chinese PLA General Hospital and PLA Medical College, Beijing, China
| | - Chenxi Wang
- National Institutes for Food and Drug Control, Beijing, China
| | - Jialin Liu
- Chinese PLA Institute of Neurosurgery, Chinese PLA General Hospital and PLA Medical College, Beijing, China
| | - Dan Zhou
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Dikang Chen
- Hunan An Tai Kang Cheng Biotechnology Co., Ltd, Changsha, China
| | - Zhixiong Liu
- Xiangya Hospital, Central South University, Changsha, China
| | - Anhua Wu
- The First Hospital of China Medical University, Shenyang, China
| | - Lin Yang
- Chinese PLA Institute of Neurosurgery, Chinese PLA General Hospital and PLA Medical College, Beijing, China
| | | | - Chengke Luo
- Xiangya Hospital, Central South University, Changsha, China
| | - Wen Cheng
- The First Hospital of China Medical University, Shenyang, China
| | - Shuai Shen
- The First Hospital of China Medical University, Shenyang, China
| | - Yunjuan Bai
- Chinese PLA Institute of Neurosurgery, Chinese PLA General Hospital and PLA Medical College, Beijing, China
| | - Xuetao Mu
- The Third Medical Center of The General Hospital of PLA, Beijing, China
| | - Chong Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhifei Wang
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ling Chen
- Chinese PLA Institute of Neurosurgery, Chinese PLA General Hospital and PLA Medical College, Beijing, China
| |
Collapse
|
19
|
Gorenjak V, Vance DR, Dade S, Stathopoulou MG, Doherty L, Xie T, Murray H, Masson C, Lamont J, Fitzgerald P, Visvikis-Siest S. Epigenome-wide association study in healthy individuals identifies significant associations with DNA methylation and PBMC extract VEGF-A concentration. Clin Epigenetics 2020; 12:79. [PMID: 32503626 PMCID: PMC7273671 DOI: 10.1186/s13148-020-00874-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Vascular endothelial growth factor A (VEGF-A) is a chemokine that induces proliferation and migration of vascular endothelial cells and is essential for both physiological and pathological angiogenesis. It is known for its high heritability (> 60%) and involvement in most common morbidities, which makes it a potentially interesting biomarker. Large GWAS studies have already assessed polymorphisms related to VEGF-A. However, no previous research has provided epigenome-wide insight in regulation of VEGF-A. METHODS VEGF-A concentrations of healthy participants from the STANISLAS Family Study (n = 201) were comprehensively assessed for association with DNA methylation. Genome-wide DNA methylation profiles were determined in whole blood DNA using the 450K Infinium BeadChip Array (Illumina). VEGF-A concentration in PBMC extracts was detected using a high-sensitivity multiplex Cytokine Array (Randox Laboratories, UK). RESULTS Epigenome-wide association analysis identified 41 methylation sites significantly associated with VEGF-A concentrations derived from PBMC extracts. Twenty CpG sites within 13 chromosomes reached Holm-Bonferroni significance. Significant values ranged from P = 1.08 × 10-7 to P = 5.64 × 10-15. CONCLUSION This study exposed twenty significant CpG sites linking DNA methylation to VEGF-A concentration. Methylation detected in promoter regions, such as TPX2 and HAS-1, could explain previously reported associations with the VEGFA gene. Methylation may also help in the understanding of the regulatory mechanisms of other genes located in the vicinity of detected CpG sites.
Collapse
Affiliation(s)
- Vesna Gorenjak
- IGE-PCV, Inserm, Université de Lorraine, F-54000, Nancy, France
| | - Dwaine R Vance
- Randox Laboratories Limited, Crumlin, Co. Antrim, Northern Ireland, UK
| | - Sébastien Dade
- IGE-PCV, Inserm, Université de Lorraine, F-54000, Nancy, France
| | | | - Lauren Doherty
- Randox Laboratories Limited, Crumlin, Co. Antrim, Northern Ireland, UK
| | - Ting Xie
- IGE-PCV, Inserm, Université de Lorraine, F-54000, Nancy, France
| | - Helena Murray
- Randox Laboratories Limited, Crumlin, Co. Antrim, Northern Ireland, UK
| | | | - John Lamont
- Randox Laboratories Limited, Crumlin, Co. Antrim, Northern Ireland, UK
| | - Peter Fitzgerald
- Randox Laboratories Limited, Crumlin, Co. Antrim, Northern Ireland, UK
| | - Sophie Visvikis-Siest
- IGE-PCV, Inserm, Université de Lorraine, F-54000, Nancy, France.
- Department of Internal Medicine and Geriatrics, CHU Technopôle Nancy-Brabois, Rue du Morvan, F-54511, Vandoeuvre-lès-, Nancy, France.
- INSERM UMR U1122, IGE-PCV, Faculté de Pharmacie-Université de Lorraine, 30 rue Lionnois, 54000, Nancy, France.
| |
Collapse
|
20
|
Han W, Shi J, Cao J, Dong B, Guan W. Current advances of long non-coding RNAs mediated by wnt signaling in glioma. Pathol Res Pract 2020; 216:153008. [PMID: 32703485 DOI: 10.1016/j.prp.2020.153008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/14/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022]
Abstract
Glioma is the most common and aggressive brain tumor in the central nervous system (CNS), in which Wnt signaling pathway has been verified to play a pivotal role in regulating the initiation and progression. Currently, numerous studies have indicated that long non-coding RNAs (lncRNAs) have critical functions across biological processes including cell proliferation, colony formation, migration, invasion and apoptosis via Wnt signaling pathway in glioma. This review depicts canonical and non-canonical Wnt/β-catenin signaling pathway properties and relative processing mechanisms in gliomas, and summarizes the function and regulation of lncRNAs mediated by Wnt signaling pathway in the development and progression of glioma. Ultimately, we hope to seek out promising biomarkers and reliable therapeutic targets for glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiachao Cao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
21
|
Casalou C, Ferreira A, Barral DC. The Role of ARF Family Proteins and Their Regulators and Effectors in Cancer Progression: A Therapeutic Perspective. Front Cell Dev Biol 2020; 8:217. [PMID: 32426352 PMCID: PMC7212444 DOI: 10.3389/fcell.2020.00217] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
The Adenosine diphosphate-Ribosylation Factor (ARF) family belongs to the RAS superfamily of small GTPases and is involved in a wide variety of physiological processes, such as cell proliferation, motility and differentiation by regulating membrane traffic and associating with the cytoskeleton. Like other members of the RAS superfamily, ARF family proteins are activated by Guanine nucleotide Exchange Factors (GEFs) and inactivated by GTPase-Activating Proteins (GAPs). When active, they bind effectors, which mediate downstream functions. Several studies have reported that cancer cells are able to subvert membrane traffic regulators to enhance migration and invasion. Indeed, members of the ARF family, including ARF-Like (ARL) proteins have been implicated in tumorigenesis and progression of several types of cancer. Here, we review the role of ARF family members, their GEFs/GAPs and effectors in tumorigenesis and cancer progression, highlighting the ones that can have a pro-oncogenic behavior or function as tumor suppressors. Moreover, we propose possible mechanisms and approaches to target these proteins, toward the development of novel therapeutic strategies to impair tumor progression.
Collapse
Affiliation(s)
- Cristina Casalou
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Andreia Ferreira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Duarte C Barral
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
22
|
Wang Y, Zhao W, Xiao Z, Guan G, Liu X, Zhuang M. A risk signature with four autophagy-related genes for predicting survival of glioblastoma multiforme. J Cell Mol Med 2020; 24:3807-3821. [PMID: 32065482 PMCID: PMC7171404 DOI: 10.1111/jcmm.14938] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/05/2019] [Accepted: 12/17/2019] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a devastating brain tumour without effective treatment. Recent studies have shown that autophagy is a promising therapeutic strategy for GBM. Therefore, it is necessary to identify novel biomarkers associated with autophagy in GBM. In this study, we downloaded autophagy-related genes from Human Autophagy Database (HADb) and Gene Set Enrichment Analysis (GSEA) website. Least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression analysis were performed to identify genes for constructing a risk signature. A nomogram was developed by integrating the risk signature with clinicopathological factors. Time-dependent receiver operating characteristic (ROC) curve and calibration plot were used to evaluate the efficiency of the prognostic model. Finally, four autophagy-related genes (DIRAS3, LGALS8, MAPK8 and STAM) were identified and were used for constructing a risk signature, which proved to be an independent risk factor for GBM patients. Furthermore, a nomogram was developed based on the risk signature and clinicopathological factors (IDH1 status, age and history of radiotherapy or chemotherapy). ROC curve and calibration plot suggested the nomogram could accurately predict 1-, 3- and 5-year survival rate of GBM patients. For function analysis, the risk signature was associated with apoptosis, necrosis, immunity, inflammation response and MAPK signalling pathway. In conclusion, the risk signature with 4 autophagy-related genes could serve as an independent prognostic factor for GBM patients. Moreover, we developed a nomogram based on the risk signature and clinical traits which was validated to perform better for predicting 1-, 3- and 5-year survival rate of GBM.
Collapse
Affiliation(s)
- Yulin Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | | | - Zhe Xiao
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Gefei Guan
- Department of NeurosurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Xin Liu
- Department of StomatologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Minghua Zhuang
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
23
|
Zhang J, Zhang Q, Sun C, Huang Y, Zhang J, Wang Q. Clinical relevance of ARF/ARL family genes and oncogenic function of ARL4C in endometrial cancer. Biomed Pharmacother 2020; 125:110000. [PMID: 32070877 DOI: 10.1016/j.biopha.2020.110000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
Members of ADP-ribosylation factor (ARF)/ARF-like protein (ARL) family regulate malignant phenotype of cancer cells. The present study aims to investigate the clinical relevance of ARF/ARL family members in endometrial cancer. We report that several ARF/ARL family genes serve as prognostic biomarkers for endometrial cancer. Through a combination of TCGA database and immunohistochemistry analysis, we revealed that ARL4C, a member of ARL family, was overexpressed in endometrial cancer and might function as an oncogene in endometrial carcinogenesis. Gene set enrichment analysis (GSEA) and functional studies demonstrated that cell cycle and cell adhesion pathways were the potential mechanism of ARL4C in promoting endometrial cancer cell proliferation, migration and invasion. Moreover, we also observed the involvement of ARL4C in metformin-inhibited cellular proliferation of endometrial cancer. Collectively, knowledge of the expression and function of ARF/ARL family genes could provide a potential therapeutic strategy for endometrial cancer.
Collapse
Affiliation(s)
- Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qinyi Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Cong Sun
- Department of Obstetrics and Gynecology, First Hospital of Zibo, Shandong, China
| | - Yanjuan Huang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jing Zhang
- Department of Integrated Therapy, Shanghai Cancer Center, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Qingying Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|