1
|
Jiang Y, Li H, Wu S, Jiang B, Zeng L, Tang Y, Luo L, Ouyang L, Du W, Li Y. Deciphering MOSPD1's impact on breast cancer progression and therapeutic response. Biol Direct 2024; 19:88. [PMID: 39369222 PMCID: PMC11453086 DOI: 10.1186/s13062-024-00531-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/05/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Motile Sperm Domain-Containing Protein 1 (MOSPD1) has been implicated in breast cancer (BC) pathophysiology, but its exact role remains unclear. This study aimed to assess MOSPD1 expression levels in BC versus normal tissues and investigate its diagnostic potential. METHODS MOSPD1 expression was analyzed in BC and normal tissues, with Receiver Operating Characteristic analysis for diagnostic evaluation. Validation was performed using immunohistochemistry. Functional studies included tumor growth assays, MOSPD1 suppression and overexpression experiments, and testing BC cell responses to anti-PD-L1 therapy. RESULTS MOSPD1 expression was significantly higher in BC samples than normal tissues, correlating with poor clinical outcomes in BC patients. MOSPD1 suppression inhibited tumor growth, while overexpression accelerated it. Silencing MOSPD1 enhanced BC cell sensitivity to anti-PD-L1 therapy and decreased Th2 cell activity. In vivo experiments supported these findings, showing the impact of MOSPD1 on tumor growth and response to therapy. CONCLUSIONS Elevated MOSPD1 levels in BC suggest its potential as a biomarker for adverse outcomes. Targeting MOSPD1, particularly with anti-PD-L1 therapy, may effectively inhibit BC tumor growth and modulate immune responses. This study emphasizes the significance of MOSPD1 in BC pathophysiology and highlights its promise as a therapeutic target.
Collapse
Affiliation(s)
- Yiling Jiang
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, 421001, People's Republic of China
| | - Hailong Li
- Department of pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, 415000, Hunan, People's Republic of China
| | - Sixuan Wu
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, 421001, People's Republic of China
| | - Baohong Jiang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, 421001, Hunan, People's Republic of China
| | - Lijun Zeng
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, 421001, People's Republic of China
| | - Yuanbin Tang
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, 421001, People's Republic of China
| | - Lunqi Luo
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, 421001, People's Republic of China
| | - Lianjie Ouyang
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, 421001, People's Republic of China
| | - Wei Du
- Department of pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, 415000, Hunan, People's Republic of China
| | - Yuehua Li
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, 421001, People's Republic of China.
| |
Collapse
|
2
|
Gaspary JFP, Edgar L, Lopes LFD, Rosa CB, Siluk JCM. Translational insights into the hormetic potential of carbon dioxide: from physiological mechanisms to innovative adjunct therapeutic potential for cancer. Front Physiol 2024; 15:1415037. [PMID: 39086932 PMCID: PMC11288912 DOI: 10.3389/fphys.2024.1415037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
Background Carbon dioxide (CO2), traditionally viewed as a mere byproduct of cellular respiration, plays a multifaceted role in human physiology beyond simple elimination through respiration. CO2 may regulate the tumor microenvironment by significantly affecting the release of oxygen (O2) to tissues through the Bohr effect and by modulating blood pH and vasodilation. Previous studies suggest hypercapnia (elevated CO2 levels) might trigger optimized cellular mechanisms with potential therapeutic benefits. The role of CO2 in cellular stress conditions within tumor environments and its impact on O2 utilization offers a new investigative area in oncology. Objectives This study aims to explore CO2's role in the tumor environment, particularly how its physiological properties and adaptive responses can influence therapeutic strategies. Methods By applying a structured translational approach using the Work Breakdown Structure method, the study divided the analysis into six interconnected work packages to comprehensively analyze the interactions between carbon dioxide and the tumor microenvironment. Methods included systematic literature reviews, data analyses, data integration for identifying critical success factors and exploring extracellular environment modulation. The research used SMART criteria for assessing innovation and the applicability of results. Results The research revealed that the human body's adaptability to hypercapnic conditions could potentially inform innovative strategies for manipulating the tumor microenvironment. This could enhance O2 utilization efficiency and manage adaptive responses to cellular stress. The study proposed that carbon dioxide's hormetic potential could induce beneficial responses in the tumor microenvironment, prompting clinical protocols for experimental validation. The research underscored the importance of pH regulation, emphasizing CO2 and carbonic acid's role in modulating metabolic and signaling pathways related to cancer. Conclusion The study underscores CO2 as vital to our physiology and suggests potential therapeutic uses within the tumor microenvironment. pH modulation and cellular oxygenation optimization via CO2 manipulation could offer innovative strategies to enhance existing cancer therapies. These findings encourage further exploration of CO2's therapeutic potential. Future research should focus on experimental validation and exploration of clinical applications, emphasizing the need for interdisciplinary and collaborative approaches to tackle current challenges in cancer treatment.
Collapse
Affiliation(s)
| | - Lee Edgar
- Elastro Crete, LLC. Research and Development Department, Veyo, UT, United States
| | - Luis Felipe Dias Lopes
- Department of Administrative Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| | - Carmen Brum Rosa
- Production Engineering Department, Federal University of Santa Maria, Santa Maria, Brazil
| | | |
Collapse
|
3
|
Zhao H, Zhou Y, Wang Z, Zhang X, Chen L, Hong Z. Plasma proteins and psoriatic arthritis: a proteome-wide Mendelian randomization study. Front Immunol 2024; 15:1417564. [PMID: 39026678 PMCID: PMC11254630 DOI: 10.3389/fimmu.2024.1417564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Background Previous epidemiological studies have identified a correlation between serum protein levels and Psoriatic Arthritis (PsA). However, the precise nature of this relationship remains uncertain. Therefore, our objective was to assess whether circulating levels of 2,923 plasma proteins are associated with the risk of PsA, utilizing the Mendelian randomization (MR) approach. Methods Two-sample MR analysis was performed to assess the causal impact of proteins on PsA risk. Exposure data for plasma proteins were sourced from a genome-wide association study (GWAS) conducted within the UK Biobank Pharma Proteomics Project, which encompassed 2,923 unique plasma proteins. The outcome data for PsA were sourced from the FinnGen study, a large-scale genomics initiative, comprising 3,537 cases and 262,844 controls. Additionally, colocalization analysis, Phenome-wide MR analysis, and candidate drug prediction were employed to identify potential causal circulating proteins and novel drug targets. Results We thoroughly assessed the association between 1,837 plasma proteins and PsA risk, identifying seven proteins associated with PsA risk. An inverse association of Interleukin-10 (IL-10) with PsA risk was observed [odds ratio (OR)=0.45, 95% confidence interval (CI), 0.28 to 0.70, P FDR=0.072]. Additionally, Apolipoprotein F (APOF) has a positive effect on PsA risk (OR=2.08, 95% CI, 1.51 to 2.86, P FDR=0.005). Subsequently, we found strong evidence indicating that IL-10 and APOF were colocalized with PsA associations (PP.H4 = 0.834 for IL-10 and PP.H4 = 0.900 for APOF). Phenome-wide association analysis suggested that these two proteins may have dual effects on other clinical traits (P FDR<0.1). Conclusion This study identified 7 plasma proteins associated with PsA risk, particularly IL-10 and APOF, which offer new insights into its etiology. Further studies are needed to assess the utility and effectiveness of these candidate proteins.
Collapse
Affiliation(s)
- Heran Zhao
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Zhou
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziyan Wang
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Zhang
- College of Orthopedics and Traumatology, Guangxi University of Chinese Medicine, Nanning, China
| | - Leilei Chen
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhinan Hong
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
4
|
Zhang J, Zhang Q, Hu W, Liang Y, Jiang D, Chen H. A transcriptome-wide association study identified susceptibility genes for hepatocellular carcinoma in East Asia. Gastroenterol Rep (Oxf) 2024; 12:goae057. [PMID: 38846986 PMCID: PMC11153834 DOI: 10.1093/gastro/goae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/07/2024] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and is prevalent in East Asia. Although genome-wide association studies (GWASs) of HCC have identified 23 risk regions, the susceptibility genes underlying these associations largely remain unclear. To identify novel candidate genes for HCC, we conducted liver single-tissue and cross-tissue transcriptome-wide association studies (TWASs) in two populations of East Asia. Methods GWAS summary statistics of 2,514 subjects (1,161 HCC cases and 1,353 controls) from the Chinese Qidong cohort and 161,323 subjects (2,122 HCC cases and 159,201 controls) from the BioBank Japan project were used to conduct TWAS analysis. The single-tissue and cross-tissue TWAS approaches were both used to detect the association between susceptible genes and the risk of HCC. TWAS identified genes were further annotated by Metascape, UALCAN, GEPIA2, and DepMap. Results We identified 22 novel genes at 16 independent loci significantly associated with HCC risk after Bonferroni correction. Of these, 13 genes were located in novel regions. Besides, we found 83 genes overlapped in the Chinese and Japanese cohorts with P < 0.05, of which, three genes (NUAK2, HLA-DQA1, and ATP6V1G2) were discerned by both single-tissue and cross-tissue TWAS approaches. Among the genes identified through TWAS, a significant proportion of them exhibit a credible role in HCC biology, such as FAM96B, HSPA5, POLRMT, MPHOSPH10, and RABL2A. HLA-DQA1, NUAK2, and HSPA5 associated with the process of carcinogenesis in HCC as previously reported. Conclusions Our findings highlight the value of leveraging the gene expression data to identify new candidate genes beyond the GWAS associations and could further provide a genetic insight for the biology of HCC.
Collapse
Affiliation(s)
- Jingjing Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- School of Public Health (Shenzhen), Shenzhen campus of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| | - Qingrong Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- School of Public Health (Shenzhen), Shenzhen campus of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| | - Wenyan Hu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- School of Public Health (Shenzhen), Shenzhen campus of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| | - Yuxuan Liang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- School of Public Health (Shenzhen), Shenzhen campus of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| | - Deke Jiang
- Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Haitao Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- School of Public Health (Shenzhen), Shenzhen campus of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| |
Collapse
|
5
|
Zhan Z, Lin K, Wang T. Construction of oxidative phosphorylation-related prognostic risk score model in uveal melanoma. BMC Ophthalmol 2024; 24:204. [PMID: 38698303 PMCID: PMC11067154 DOI: 10.1186/s12886-024-03441-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Uveal melanoma (UVM) is a malignant intraocular tumor in adults. Targeting genes related to oxidative phosphorylation (OXPHOS) may play a role in anti-tumor therapy. However, the clinical significance of oxidative phosphorylation in UVM is unclear. METHOD The 134 OXPHOS-related genes were obtained from the KEGG pathway, the TCGA UVM dataset contained 80 samples, served as the training set, while GSE22138 and GSE39717 was used as the validation set. LASSO regression was carried out to identify OXPHOS-related prognostic genes. The coefficients obtained from Cox multivariate regression analysis were used to calculate a risk score, which facilitated the construction of a prognostic model. Kaplan-Meier survival analysis, logrank test and ROC curve using the time "timeROC" package were conducted. The immune cell frequency in low- and high-risk group was analyzed through Cibersort tool. The specific genomic alterations were analyzed by "maftools" R package. The differential expressed genes between low- or high-risk group were analyzed and performed Gene Ontology (GO) and GSEA. Finally, we verified the function of CYC1 in UVM by gene silencing in vitro. RESULTS A total of 9 OXPHOS-related prognostic genes were identified, including NDUFB1, NDUFB8, ATP12A, NDUFA3, CYC1, COX6B1, ATP6V1G2, ATP4B and NDUFB4. The UVM prognostic risk model was constructed based on the 9 OXPHOS-related prognostic genes. The prognosis of patients in the high-risk group was poorer than low-risk group. Besides, the ROC curve demonstrated that the area under the curve of the model for predicting the 1 to 5-year survival rate of UVM patients were all more than 0.88. External validation in GSE22138 and GSE39717 dataset revealed that these 9 genes could also be utilized to evaluate and predict the overall survival of patients with UVM. The risk score levels related to immune cell frequency and specific genomic alterations. The DEGs between the low- and high- risk group were enriched in tumor OXPHOS and immune related pathway. In vitro experiments, CYC1 silencing significantly inhibited UVM cell proliferation and invasion, induced cell apoptosis. CONCLUSION In sum, a prognostic risk score model based on oxidative phosphorylation-related genes in UVM was developed to enhance understanding of the disease. This prognostic risk score model may help to find potential therapeutic targets for UVM patients. CYC1 acts as an oncogene role in UVM.
Collapse
Affiliation(s)
- Zhiyun Zhan
- Ophthalmology Department, First Affiliated Hospital of Fujian Medical University, No. 20, Chazhong Road, Taijiang District, 350004, Fuzhou, Fujian, China
| | - Kun Lin
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, 516 Jinrong South Road, 350001, Fuzhou, China
| | - Tingting Wang
- Ophthalmology Department, First Affiliated Hospital of Fujian Medical University, No. 20, Chazhong Road, Taijiang District, 350004, Fuzhou, Fujian, China.
| |
Collapse
|
6
|
Luo S, Liu Z, Chang H, Cheng X, Qian R, Gao Y, Hou C. Potential value of expression of receptor accessory protein 4 for evaluating the prognosis of lower-grade glioma patients. Aging (Albany NY) 2024; 16:6188-6211. [PMID: 38552216 PMCID: PMC11042925 DOI: 10.18632/aging.205695] [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: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 04/23/2024]
Abstract
BACKGROUND REEP4 is involved in the regulation of the biological process of mitosis. Lower grade glioma (LGG), as a malignant tumor, is accompanied by abnormalities in mitosis, but there have been no reports of REEP4 so far. METHODS We collected transcriptome data, DNA methylation data and the clinical characteristics of thousands of patients with LGG. Various big data analysis methods and molecular biology experiments were employed to reveal the impact of REEP4 on the pathological process of LGG. RESULTS It was found that the expression of REEP4 was significantly elevated and negatively regulated by its methylation site. Therefore, both the high expression of REEP4 and low methylation state of cg16311504 showed that the patients are correlated with lower patient survival rate. In addition, high REEP4 expression participates in the regulation of various cancer-related cellular signaling pathways, such as the cell cycle, MAPK signaling pathway, NOD-like receptor signaling pathway, etc. More importantly, the level of immune cell infiltration significantly increased in the high expression group of REEP4 in the LGG tumor microenvironment and REEP4 has a high positive correlation with PD-L1 and other immune checkpoints. CONCLUSIONS In brief, this study is the first to introduce REEP4 in malignant tumors, which can be used as an independent risk factor that participates in the malignant process of LGG. More importantly, REEP4 has the potential to become a new star in the field of anti-tumor treatment.
Collapse
Affiliation(s)
- Shuping Luo
- Department of Colorectal Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou 450003, Henan, China
| | - Haigang Chang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Xingbo Cheng
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou 450003, Henan, China
| | - Rongjun Qian
- Department of Neurosurgery, Henan Provincial People’s Hospital, People’s Hospital of Henan University, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou 450003, Henan, China
| | - Chaofeng Hou
- Department of Colorectal Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Giambra M, Di Cristofori A, Raimondo F, Rigolio R, Conconi D, Chiarello G, Tabano SM, Antolini L, Nicolini G, Bua M, Ferlito D, Carrabba G, Giussani CG, Lavitrano M, Bentivegna A. Vacuolar Proton-Translocating ATPase May Take Part in the Drug Resistance Phenotype of Glioma Stem Cells. Int J Mol Sci 2024; 25:2743. [PMID: 38473989 DOI: 10.3390/ijms25052743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The vacuolar proton-translocating ATPase (V-ATPase) is a transmembrane multi-protein complex fundamental in maintaining a normal intracellular pH. In the tumoral contest, its role is crucial since the metabolism underlying carcinogenesis is mainly based on anaerobic glycolytic reactions. Moreover, neoplastic cells use the V-ATPase to extrude chemotherapy drugs into the extra-cellular compartment as a drug resistance mechanism. In glioblastoma (GBM), the most malignant and incurable primary brain tumor, the expression of this pump is upregulated, making it a new possible therapeutic target. In this work, the bafilomycin A1-induced inhibition of V-ATPase in patient-derived glioma stem cell (GSC) lines was evaluated together with temozolomide, the first-line therapy against GBM. In contrast with previous published data, the proposed treatment did not overcome resistance to the standard therapy. In addition, our data showed that nanomolar dosages of bafilomycin A1 led to the blockage of the autophagy process and cellular necrosis, making the drug unusable in models which are more complex. Nevertheless, the increased expression of V-ATPase following bafilomycin A1 suggests a critical role of the proton pump in GBM stem components, encouraging the search for novel strategies to limit its activity in order to circumvent resistance to conventional therapy.
Collapse
Affiliation(s)
- Martina Giambra
- PhD Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Andrea Di Cristofori
- PhD Program in Neuroscience, University of Milano-Bicocca, 20900 Monza, Italy
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Francesca Raimondo
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberta Rigolio
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Donatella Conconi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Gaia Chiarello
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- Pathology, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Silvia Maria Tabano
- Laboratory of Medical Genetics, Ospedale Maggiore Policlinico, IRCCS Ca' Granda, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Laura Antolini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Gabriella Nicolini
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Miriam Bua
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Davide Ferlito
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Giorgio Carrabba
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Carlo Giorgio Giussani
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Neurosurgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Marialuisa Lavitrano
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Angela Bentivegna
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
8
|
Qi C, Lei L, Hu J, Ou S. Establishment and validation of a novel integrin-based prognostic gene signature that sub-classifies gliomas and effectively predicts immunosuppressive microenvironment. Cell Cycle 2023; 22:1259-1283. [PMID: 37096960 PMCID: PMC10193886 DOI: 10.1080/15384101.2023.2205204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/20/2022] [Accepted: 04/17/2023] [Indexed: 04/26/2023] Open
Abstract
The integrin family members play a key role in cancer immunomodulation and prognosis. We comprehensively analyzed the expression patterns and clinical significance of integrin family-related genes in gliomas. A total of 2293 gliomas from the Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA) and Gliovis platform were enrolled for analyses. Twenty-six integrin coding genes showed different expression patterns between glioma and normal brain tissues. We screened an integrin family-related gene signature (ITGA5, ITGA9, ITGAE, ITGB7 and ITGB8) that showed independent prognostic value and sub-classified gliomas into different prognostic and molecular clusters, further composed an integrin-based risk score model associated with glioma malignant clinical features, overall survival (OS), and immune microenvironment alterations. Besides, glioma patients with high-risk scores showed chemotherapeutic resistance and more immune cells infiltration as well as high immune checkpoints expression. Concurrently, we also revealed that high-risk score group presented resistance to T cell-mediated cancer killing process and lower rates of response to immune checkpoint blockade (ICB) treatment. In conclusion, our study identified a valuable integrin gene signature that predicted gliomas OS effectively, and sub-classified them into different phenotypes and accompanied with immunological changes, possibly acted as a biomarker for ICB treatment.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
- Department of Neurosurgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Lei Lei
- Department of Rheumatology and Immunology, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Jinqu Hu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shaowu Ou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
9
|
SARS-CoV-2 Pattern Provides a New Scoring System and Predicts the Prognosis and Immune Therapeutic Response in Glioma. Cells 2022; 11:cells11243997. [PMID: 36552760 PMCID: PMC9777143 DOI: 10.3390/cells11243997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Glioma is the most common primary malignancy of the adult central nervous system (CNS), with a poor prognosis and no effective prognostic signature. Since late 2019, the world has been affected by the rapid spread of SARS-CoV-2 infection. Research on SARS-CoV-2 is flourishing; however, its potential mechanistic association with glioma has rarely been reported. The aim of this study was to investigate the potential correlation of SARS-CoV-2-related genes with the occurrence, progression, prognosis, and immunotherapy of gliomas. METHODS SARS-CoV-2-related genes were obtained from the human protein atlas (HPA), while transcriptional data and clinicopathological data were obtained from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases. Glioma samples were collected from surgeries with the knowledge of patients. Differentially expressed genes were then identified and screened, and seven SARS-CoV-2 related genes were generated by LASSO regression analysis and uni/multi-variate COX analysis. A prognostic SARS-CoV-2-related gene signature (SCRGS) was then constructed based on these seven genes and validated in the TCGA validation cohort and CGGA cohort. Next, a nomogram was established by combining critical clinicopathological data. The correlation between SCRGS and glioma related biological processes was clarified by Gene set enrichment analysis (GSEA). In addition, immune infiltration and immune score, as well as immune checkpoint expression and immune escape, were further analyzed to assess the role of SCRGS in glioma-associated immune landscape and the responsiveness of immunotherapy. Finally, the reliability of SCRGS was verified by quantitative real-time polymerase chain reaction (qRT-PCR) on glioma samples. RESULTS The prognostic SCRGS contained seven genes, REEP6, CEP112, LARP4B, CWC27, GOLGA2, ATP6AP1, and ERO1B. Patients were divided into high- and low-risk groups according to the median SARS-CoV-2 Index. Overall survival was significantly worse in the high-risk group than in the low-risk group. COX analysis and receiver operating characteristic (ROC) curves demonstrated excellent predictive power for SCRGS for glioma prognosis. In addition, GSEA, immune infiltration, and immune scores indicated that SCRGS could potentially predict the tumor microenvironment, immune infiltration, and immune response in glioma patients. CONCLUSIONS The SCRGS established here can effectively predict the prognosis of glioma patients and provide a potential direction for immunotherapy.
Collapse
|
10
|
Li B, Liu Y, Sun S. Pump proton inhibitors display anti-tumour potential in glioma. Cell Prolif 2022:e13321. [PMID: 35961680 DOI: 10.1111/cpr.13321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/28/2022] [Accepted: 07/14/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVES Glioma is one of the most aggressive brain tumours with poor overall survival despite advanced technology in surgical resection, chemotherapy and radiation. Progression and recurrence are the hinge causes of low survival. Our aim is to explain the concrete mechanism in the proliferation and progression of tumours based on tumour microenvironment (TME). The main purpose is to illustrate the mechanism of proton pump inhibitors (PPIs) in affecting acidity, hypoxia, oxidative stress, inflammatory response and autophagy based on the TME to induce apoptosis and enhance the sensitivity of chemoradiotherapy. FINDINGS TME is the main medium for tumour growth and progression. Acidity, hypoxia, inflammatory response, autophagy, angiogenesis and so on are the main causes of tumour progress. PPIs, as a common clinical drug to inhibit gastric acid secretion, have the advantages of fast onset, long action time and small adverse reactions. Nowadays, several kinds of literature highlight the potential of PPIs in inhibiting tumour progression. However, long-term use of PPIs alone also has obvious side effects. Therefore, till now, how to apply PPIs to promote the effect of radio-chemotherapy and find the concrete dose and concentration of combined use are novel challenges. CONCLUSIONS PPIs display the potential in enhancing the sensitivity of chemoradiotherapy to defend against glioma based on TME. In the clinic, it is also necessary to explore specific concentrations and dosages in synthetic applications.
Collapse
Affiliation(s)
- Bihan Li
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Ying Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Shilong Sun
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|