1
|
Guo J, Yu H, Guo Y, Liu J, Chen Y, Li Z. Identification of endocrine disrupting chemicals targeting NTD-related hub genes during pregnancy via in silico analysis. Reprod Toxicol 2025; 134:108904. [PMID: 40187376 DOI: 10.1016/j.reprotox.2025.108904] [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: 11/25/2024] [Revised: 02/24/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Neural tube defects (NTDs) represent severe congenital malformations of the central nervous system with multifactorial etiology, involving intricate gene-environment interactions that remain incompletely characterized. Endocrine disrupting chemicals (EDCs) are exogenous substances with hormone-disrupting properties that are ubiquitous in our surroundings. These chemicals pose a significant threat to human health, contributing to a range of diseases. Pregnant women are particularly vulnerable to the effects of EDCs, as these substances can traverse the placental barrier and impact the development of both the placenta and fetus. This study utilized placental and fetal transcriptome data to identify hub genes associated with NTDs during pregnancy. By leveraging the Comparative Toxicogenomics Database (CTD), we predicted the EDCs targeting these hub genes and performed molecular docking to assess their interactions. Our findings revealed four hub genes (CTSC, FCER1G, ITGB2, and LYVE1) in NTDs, with 72 EDCs identified as their targets. Molecular docking demonstrated that atrazine, bisphenol A (BPA) and diuron exhibited stable affinity with the proteins encoded by hub genes. These findings provide new insights into the environmental endocrine disruptors that affect the development of NTDs during pregnancy.
Collapse
Affiliation(s)
- Junjie Guo
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 998 Aiying Street, Harbin, Heilongjiang 150023, China
| | - Hao Yu
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 998 Aiying Street, Harbin, Heilongjiang 150023, China
| | - Yujun Guo
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 998 Aiying Street, Harbin, Heilongjiang 150023, China
| | - Jinming Liu
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 998 Aiying Street, Harbin, Heilongjiang 150023, China
| | - Yuzhu Chen
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 998 Aiying Street, Harbin, Heilongjiang 150023, China
| | - Zhaozhu Li
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 998 Aiying Street, Harbin, Heilongjiang 150023, China.
| |
Collapse
|
2
|
Lu F, Mai Z, Zhang L, Luo H, Wang L, Li S, Zhong M. Differential Expression of Disulfidptosis-Related Genes in Spinal Cord Injury and Their Role in the Immune Microenvironment. Mol Neurobiol 2025:10.1007/s12035-025-04931-4. [PMID: 40237950 DOI: 10.1007/s12035-025-04931-4] [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: 11/25/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Spinal cord injury (SCI) often results in severe sensory, motor, and autonomic dysfunction, with limited treatment options due to complex underlying mechanisms. Disulfidptosis, a recently discovered form of cell death driven by disulfide bond accumulation, has been linked to various diseases, but its role in SCI remains unexplored. This study investigates the involvement of disulfidptosis-related genes (DRGs) in SCI to identify potential diagnostic markers and therapeutic targets. Using SCI datasets from the Gene Expression Omnibus (GEO), we conducted differential gene expression analysis, identifying key disulfidptosis-related differentially expressed genes (DRDEGs). Further analysis through gene set enrichment (GSEA) and Bayesian pathway enrichment highlighted significant involvement in pathways such as NF-κB, PI3K/Akt, and MAPK, with an emphasis on nephrin family interactions. Three core DRDEGs-HK2, Map3k8, and S100a6-were identified, and a diagnostic model built on these genes demonstrated strong predictive performance (AUC: 0.896 in training, 0.850 in validation). Additionally, real-time PCR (qRT-PCR) in an animal model validated the elevated expression of these DRDEGs in SCI samples. This research provides novel insights into disulfidptosis in SCI, suggesting these genes as promising targets for improved diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Feng Lu
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No. 128 Jinling Road, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zifeng Mai
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No. 128 Jinling Road, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Longfei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No. 128 Jinling Road, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hao Luo
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No. 128 Jinling Road, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No. 128 Jinling Road, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shihong Li
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No. 128 Jinling Road, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Maolin Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No. 128 Jinling Road, Ganzhou, Jiangxi, China.
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
3
|
Wang J, Zhong Z, Luo H, Han Q, Wu K, Jiang A, Chen L, Gao Y, Jiang Y. Modulation of brain immune microenvironment and cellular dynamics in systemic inflammation. Theranostics 2025; 15:5153-5171. [PMID: 40303348 PMCID: PMC12036864 DOI: 10.7150/thno.107061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/13/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Sepsis-associated encephalopathy (SAE) is a severe complication of sepsis, affecting approximately 70% of patients, leading to increased mortality and long-term cognitive impairments among survivors. However, there is a lack of comprehensive studies on the development of SAE, especially related to the cellular communication networks in the brain microenvironment. Methods: We evaluated the impact of myeloid cells on the brain's immune microenvironment through glial cell alterations using bulk and single-cell transcriptomics data from human and mouse models and validated this with correlative experiments. We also developed the DeconvCellLink R package to study neuroinflammation-associated cellular interaction networks. A dynamic brain immune microenvironment map showing temporal alterations in brain cellular network during systemic inflammatory reactions was constructed using time-series data. Results: While brain cellular alterations differed between human and animal models, a highly conserved set of sepsis-associated genes regulating immune microenvironment signalling was identified. The dynamic alterations in cellular interaction networks and cytokines revealed brain immune cells' temporal response to systemic inflammation. We also found that valproic acid could mitigate sepsis-induced neuroinflammation by regulating glial cell balance and modulating the neuroimmune microenvironment. Conclusion: Through dynamic cellular communication networks, the study revealed that, immune dysregulation in the inflamed brain in SAE involves overactivation of innate immunity, with neutrophils playing a crucial role, providing a scientific framework for developing novel therapeutic strategies and offering new insights into the mechanisms underlying sepsis-induced brain dysfunction.
Collapse
Affiliation(s)
- Junhao Wang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Current address: Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Zhaoqian Zhong
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qizheng Han
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Kan Wu
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Aolin Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Li Chen
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanxia Gao
- Henan Key Laboratory of Critical Care Medicine, Henan International Joint Laboratory of Infection and Immunity, Department of Critical Care Medicine and Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science Zhengzhou 451163, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Henan Key Laboratory of Critical Care Medicine, Henan International Joint Laboratory of Infection and Immunity, Department of Critical Care Medicine and Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science Zhengzhou 451163, China
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| |
Collapse
|
4
|
Ding X, Xi Y, Sheng Y, Fan Y, Yu Y. ACSL5 mediates macrophage infiltration and lipid metabolism in erythrotelangiectasia rosacea via potential pathogenic mechanisms and therapeutic targets. Sci Rep 2025; 15:11929. [PMID: 40195491 PMCID: PMC11976930 DOI: 10.1038/s41598-025-96756-3] [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: 02/10/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Abstract
Rosacea, an inflammatory skin disorder with complex pathogenesis, remains poorly understood. Through integrative bioinformatics and experimental approaches, we identified 304 differentially expressed genes in erythrotelangiectasia rosacea (ETR), primarily enriched in lipid metabolism pathways. Support vector machine (SVM), linear regression analyses and network analysis revealed ACADVL and ACSL5 as potential therapeutic targets. Immunological profiling demonstrated distinctive immune cell infiltration, with elevated M0 and M1 macrophages in patients with ETR. Immunofluorescence validation confirmed significant ACSL5 upregulation and increased M1 macrophage infiltration in the rosacea mouse model. The co-localization of ACSL5 with M1 macrophage markers suggests a mechanistic link between lipid metabolism and inflammatory responses. These findings provide new insights into ETR pathogenesis and highlight ACSL5 as a promising therapeutic target for inflammatory skin disorders.
Collapse
Affiliation(s)
- Xiaoxia Ding
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Affiliated People'S Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Youxia Xi
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yeyu Sheng
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Affiliated People'S Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yibin Fan
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Affiliated People'S Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Department of Dermatology, Zhejiang Provincial People'S Hospital, Affiliated People'S Hospital, Zhejiang, China.
| | - Yong Yu
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Affiliated People'S Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Department of Dermatology, Zhejiang Provincial People'S Hospital, Affiliated People'S Hospital, Zhejiang, China.
| |
Collapse
|
5
|
Gogoshin G, Rodin AS. Minimum uncertainty as Bayesian network model selection principle. BMC Bioinformatics 2025; 26:100. [PMID: 40200184 PMCID: PMC11980298 DOI: 10.1186/s12859-025-06104-5] [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: 09/21/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Bayesian Network (BN) modeling is a prominent methodology in computational systems biology. However, the incommensurability of datasets frequently encountered in life science domains gives rise to contextual dependence and numerical irregularities in the behavior of model selection criteria (such as MDL, Minimum Description Length) used in BN reconstruction. This renders model features, first and foremost dependency strengths, incomparable and difficult to interpret. In this study, we derive and evaluate a model selection principle that addresses these problems. RESULTS The objective of the study is attained by (i) approaching model evaluation as a misspecification problem, (ii) estimating the effect that sampling error has on the satisfiability of conditional independence criterion, as reflected by Mutual Information, and (iii) utilizing this error estimate to penalize uncertainty with the novel Minimum Uncertainty (MU) model selection principle. We validate our findings numerically and demonstrate the performance advantages of the MU criterion. Finally, we illustrate the advantages of the new model evaluation framework on real data examples. CONCLUSIONS The new BN model selection principle successfully overcomes performance irregularities observed with MDL, offers a superior average convergence rate in BN reconstruction, and improves the interpretability and universality of resulting BNs, thus enabling direct inter-BN comparisons and evaluations.
Collapse
Affiliation(s)
- Grigoriy Gogoshin
- Department of Computational and Quantitative Medicine, Beckman Research Institute, and Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| | - Andrei S Rodin
- Department of Computational and Quantitative Medicine, Beckman Research Institute, and Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| |
Collapse
|
6
|
Liu JY, Weng KQ, Gao QD, Xue XY, Xu N. Molecular subtyping of renal clear cell carcinoma based on prognostic RASSF family genes and validation of C1QL1 as a key prognostic marker. Sci Rep 2025; 15:9786. [PMID: 40118977 PMCID: PMC11928449 DOI: 10.1038/s41598-025-94978-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/18/2025] [Indexed: 03/24/2025] Open
Abstract
RASSF family proteins play crucial roles in mitosis, apoptosis, cell migration, adhesion, and immune functions, primarily acting as tumor suppressors. Their roles in renal clear cell carcinoma (ccRCC) are not fully understood. We analyzed the expression and prognostic significance of RASSF genes in 573 ccRCC samples from TCGA and GEO, identifying two molecular subtypes with distinct characteristics. We developed a RAS score to assess prognosis and molecular status and investigated the key gene C1QL1 through in vitro assays. Four RASSF genes were identified as associated with prognosis and progression in ccRCC. Based on their co-expression, we defined two patient subtypes, one with poorer prognosis. A higher RAS score correlated with advanced disease and worse outcomes but indicated a favorable response to specific inhibitors, supporting personalized treatment strategies. Additionally, VHL mutations may cause abnormal SFMBT1 expression, leading to high C1QL1 levels, which promote tumor progression via the YAP-EMT pathway. Our findings highlight the potential of RASSF-based molecular subtypes and scoring systems in personalizing ccRCC treatment. Understanding C1QL1's role may facilitate the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Jin-Yu Liu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, The Affiliated Hospital of Putian University, Putian, 351100, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
| | - Kang-Qiang Weng
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Qin-Dong Gao
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
7
|
Yu Y, Liu X, Liu W, Yuan H, Han Q, Shi J, Xue Y, Li Y. Decoding the cytokine code for heart failure based on bioinformatics, machine learning and Bayesian networks. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167701. [PMID: 39909085 DOI: 10.1016/j.bbadis.2025.167701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/29/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Despite maximal pharmacological treatment guided by clinical guidelines, the prognosis of heart failure (HF) remains poor, posing a significant public health burden. This necessitates uncovering novel pathological and cardioprotective pathways. Targeting cytokines presents a promising therapeutic strategy for HF, yet their intricate mechanisms in HF progression remain obscure. METHODS HF datasets were obtained from the GEO database. Cytokine-related genes were identified through WGCNA and the CytReg database. GO and KEGG enrichment analyses were conducted using the clusterProfiler package. Reactome pathway enrichment analysis and Bayesian regulatory network construction were performed using the CBNplot package. Key genes were identified via LASSO regression and RF algorithms, with diagnostic accuracy evaluated by ROC curves. Potential therapeutic drugs were predicted using the DSigDB database, and immune cell infiltration was assessed with the CIBERSORT package. RESULTS We identified 13 cytokine-related genes associated with HF. Enrichment analyses indicated these genes mediate inflammatory responses and immune cell recruitment. Bayesian network analysis revealed two cytokine regulatory chains: IL34-CCL5-CCL4 and IL34-CCL5-CXCL12. Machine learning algorithms identified five key cytokine genes: CCL4, CCL5, CXCL12, CXCL14, and IL34. The DSigDB database predicted 47 potential therapeutic drugs, including Proscillaridin. Immune infiltration analysis showed significant differences in seven immune cell types between HF and healthy samples. CONCLUSION Our study provides insights into cytokines' molecular mechanisms in HF pathophysiology and highlights potential immunomodulatory strategies, gene therapies, and candidate drugs. Future research should validate these findings in clinical settings to develop effective HF therapies.
Collapse
Affiliation(s)
- Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiujuan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Wenwen Liu
- Traditional Chinese Medicine Hospital of Jimo District, Qingdao 266200, China
| | - Huajing Yuan
- Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Quancheng Han
- Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
8
|
Yang XY, Chen N, Wen Q, Zhou Y, Zhang T, Zhou J, Liang CH, Han LP, Wang XY, Kang QM, Zheng XX, Zhai XJ, Jiang HY, Shen TH, Xiao JW, Zou YX, Deng Y, Lin S, Duan JJ, Wang J, Yu SC. The microenvironment cell index is a novel indicator for the prognosis and therapeutic regimen selection of cancers. J Transl Med 2025; 23:61. [PMID: 39806464 PMCID: PMC11727790 DOI: 10.1186/s12967-024-05950-w] [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: 07/29/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND It is worthwhile to establish a prognostic prediction model based on microenvironment cells (MCs) infiltration and explore new treatment strategies for triple-negative breast cancer (TNBC). METHODS The xCell algorithm was used to quantify the cellular components of the TNBC microenvironment based on bulk RNA sequencing (bulk RNA-seq) data. The MCs index (MCI) was constructed using the least absolute shrinkage and selection operator Cox (LASSO-Cox) regression analysis. Single-cell RNA sequencing (scRNA-seq), spatially resolved transcriptomics (SRT), and multiplex immunofluorescence (mIF) staining analyses verified MCI. The mechanism of action of the MCI was investigated in tumor-bearing mice. RESULTS MCI consists of the six types of MCs, which can precisely predict the prognosis of the TNBC patients. scRNA-seq, SRT, and mIF analyses verified the existence and proportions of these cells. Furthermore, combined with the spatial distribution characteristics of the six types of MCs, an MCI-enhanced (MCI-e) model was constructed, which could predict the prognosis of the TNBC patients more accurately. More importantly, inhibition of the insulin signaling pathway activated in the cancer cells of the MCIhigh the TNBC patients significantly prolonged the survival time of tumor-bearing mice. CONCLUSIONS Overall, our results demonstrate that MCs infiltration can be exploited as a novel indicator for the prognosis and therapeutic regimen selection of the TNBC patients.
Collapse
Affiliation(s)
- Xian-Yan Yang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-Feng Laboratory, Chongqing, 401329, China
| | - Nian Chen
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Qian Wen
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Yu Zhou
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Tao Zhang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Ji Zhou
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Cheng-Hui Liang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Li-Ping Han
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Xiao-Ya Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Qing-Mei Kang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Xiao-Xia Zheng
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Xue-Jia Zhai
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Hong-Ying Jiang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Tian-Hua Shen
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Jin-Wei Xiao
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Yu-Xin Zou
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Yun Deng
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
| | - Shuang Lin
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-Feng Laboratory, Chongqing, 401329, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China.
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China.
- Jin-Feng Laboratory, Chongqing, 401329, China.
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China.
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China.
- Jin-Feng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
9
|
Huang S, Tan C, Chen W, Zhang T, Xu L, Li Z, Chen M, Yuan X, Chen C, Yan Q. Multiomics identification of programmed cell death-related characteristics for nonobstructive azoospermia based on a 675-combination machine learning computational framework. Genomics 2025; 117:110977. [PMID: 39662639 DOI: 10.1016/j.ygeno.2024.110977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/29/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Abnormal programmed cell death (PCD) plays a central role in spermatogenic dysfunction. However, the molecular mechanisms and biomarkers of PCD in patients with nonobstructive azoospermia (NOA) remain unclear. METHODS The genetic conditions of NOA patients were analysed using bulk transcriptomic, single-cell transcriptomic, single nucleotide polymorphism (SNP), and clinical data from multiple centres. A total of 675 machine learning methods were applied to construct models from 12 different PCDs and to screen for distinctive genes. A new PCDscore system was created to measure the degree of PCD in patients. Using the NOA mouse model, TUNEL, qRT-PCR, Western blotting, and immunohistochemistry (IHC) were utilized to validate the PCD status in NOA testes and the expression levels of hub PCD-related genes (PCDRGs). Mouse testicular samples were used for sequencing of the whole transcriptome. The sequencing results were used to evaluate the correlation between PCD scores and expression of hub genes. RESULTS A PCDscore system was built using 12 characteristic PCDRGs chosen by machine learning. PCD scores correlated with gene interaction and immune activity changes. Leydig, Sertoli, and T cells were prominent in cell interactions with PCDscore changes. PCDscore in the NOA mouse testis was increased. Among the 12 PCDRGs, BCL2L14, GGA1, GPX4, PHKG2, and SLC39A8 were strongly linked to spermatogenesis. BCL2L14, GGA1, GPX4, and PHKG2 strongly correlated with PCD statuses. The changes in the expression of these genes may be due to the effects of SNPs, which may lead to the male reproductive system disorders. CONCLUSIONS Our study provides new insights into PCD-related mechanisms in NOA patients via multiomics and proposes reliable models for the diagnosis of NOA via the use of PCD biomarkers. A deeper understanding of these mechanisms may aid in the clinical diagnosis and treatment of NOA.
Collapse
Affiliation(s)
- Shuqiang Huang
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Cuiyu Tan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Wanru Chen
- The Third School of Clinical Medicine, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Tongtong Zhang
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Liying Xu
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Zhihong Li
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Miaoqi Chen
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Xiaojun Yuan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Cairong Chen
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China; Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China.
| | - Qiuxia Yan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China; Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China.
| |
Collapse
|
10
|
Qi J, Li L, Gao B, Dai K, Shen K, Wu X, Li H, Yu Z, Wang Z, Wang Z. Prognostic prediction and immune checkpoint profiling in glioma patients through neddylation-associated features. Gene 2024; 930:148835. [PMID: 39127414 DOI: 10.1016/j.gene.2024.148835] [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: 02/02/2024] [Revised: 07/11/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Gliomas are the most common primary malignant tumours of the central nervous system, and neddylation may be a potential target for the treatment of gliomas. Our study analysed neddylation's potential role in gliomas of different pathological types and its correlation with immunotherapy. METHODS Genes required for model construction were sourced from existing literature, and their expression data were extracted from the TCGA and CGGA databases. LASSO regression was employed to identify genes associated with the prognosis of glioma patients in TCGA and to establish a clinical prognostic model. Biological changes in glioma cell lines following intervention with hub genes were evaluated using the CCK-8 assay and transwell assay. The genes implicated in the model construction were validated across various cell lines using Western blot. We conducted analyses to examine correlations between model scores and clinical data, tumor microenvironments, and immune checkpoints. Furthermore, we investigated potential differences in molecular functions and mechanisms among different groups. RESULTS We identified 249 genes from the Reactome database and analysed their expression profiles in the TCGA and CGGA databases. After using LASSO-Cox, four genes (BRCA1, BIRC5, FBXL16 and KLHL25, p < 0.05) with significant correlations were identified. We selected FBXL16 for validation in in vitro experiments. Following FBXL16 overexpression, the proliferation, migration, and invasion abilities of glioma cell lines all showed a decrease. Then, we constructed the NEDD Index for gliomas. The nomogram indicated that this model could serve as an independent prognostic marker. Analysis of the tumour microenvironment and immune checkpoints revealed that the NEDD index was also correlated with immune cell infiltration and the expression levels of various immune checkpoints. CONCLUSION The NEDD index can serve as a practical tool for predicting the prognosis of glioma patients, and it is correlated with immune cell infiltration and the expression levels of immune checkpoints.
Collapse
Affiliation(s)
- Juxing Qi
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Longyuan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Bixi Gao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Kun Dai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Kecheng Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Xin Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
11
|
Lei X, Xiao R, Chen Z, Ren J, Zhao W, Tang W, Wen K, Zhu Y, Li X, Ouyang S, Xu A, Hu Y, Bi E. Augmenting antitumor efficacy of Th17-derived Th1 cells through IFN-γ-induced type I interferon response network via IRF7. Proc Natl Acad Sci U S A 2024; 121:e2412120121. [PMID: 39541355 PMCID: PMC11588128 DOI: 10.1073/pnas.2412120121] [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: 06/19/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
The importance of CD4+ T cells in cancer immunotherapy has gained increasing recognition. Particularly, a specific subset of CD4+ T cells coexpressing the T helper type 1 (Th1) and Th17 markers has demonstrated remarkable antitumor potential. However, the underlying mechanisms governing the differentiation of these cells and their subsequent antitumor responses remain incompletely understood. Single-cell RNA sequencing (scRNA-seq) data reanalysis demonstrated the presence of Th171 cells within tumors. Subsequent trajectory analysis found that these Th171 cells are initially primed under Th17 conditions and then converted into IFN-γ-producing cells. Following the in vivo differentiation trajectory of Th171 cells, we successfully established in vitro Th171 cell culture. Transcriptomic profiling has unveiled a substantial resemblance between in vitro-generated Th171 cells and their tumor-infiltrating counterparts. Th171 cells exhibit more potent antitumor responses than Th1 or Th17 cells. Additionally, Th171chimeric antigen receptor T (CAR-T) cells eradicate solid tumors more efficiently. Importantly, Th171 cells display an early exhaustion phenotype while retaining stemness. Mechanistically, Th171 cells migrate faster and accumulate more in tumors in an extracellular matrix protein 1 (ECM1)-dependent manner. Furthermore, we show that IFN-γ up-regulated IRF7 to promote the type I interferon response network and ECM1 expression but decreased the exhaustion status in Th171 cells. Taken together, our findings position Th171 cells as a great candidate for improving targeted immunotherapies in solid malignancies.
Collapse
Affiliation(s)
- Xiaoyi Lei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Ruipei Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Zhe Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Jie Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Wenli Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Wenting Tang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Kang Wen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| | - Yihan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Xinru Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan523808, China
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| | - Yu Hu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Enguang Bi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| |
Collapse
|
12
|
Wei R, Xie H, Zhou Y, Chen X, Zhang L, Bui B, Liu X. VCAN in the extracellular matrix drives glioma recurrence by enhancing cell proliferation and migration. Front Neurosci 2024; 18:1501906. [PMID: 39554845 PMCID: PMC11565936 DOI: 10.3389/fnins.2024.1501906] [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: 09/25/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Gliomas are the most prevalent primary malignant intracranial tumors, characterized by high rates of therapy resistance, recurrence, and mortality. A major factor contributing to the poor prognosis of gliomas is their ability to diffusely infiltrate surrounding and even distant brain tissues, rendering complete total resection almost impossible and leading to frequent recurrences. The extracellular matrix (ECM) plays a key role in the tumor microenvironment and may significantly influence glioma progression, recurrence, and therapeutic response. Methods In this study, we first identified the ECM and the Versican (VCAN), a key ECM protein, as critical contributors to glioma recurrence through a comprehensive analysis of transcriptomic data comparing recurrent and primary gliomas. Using single-cell sequencing, we revealed heterogeneous distribution patterns and extensive intercellular communication among ECM components. External sequencing and immunohistochemical (IHC) staining further validated that VCAN is significantly upregulated in recurrent gliomas and is associated with poor patient outcomes. Results Functional assays conducted in glioma cell lines overexpressing VCAN demonstrated that VCAN promotes cell proliferation and migration via the PI3K/Akt/AP-1 signaling pathway. Furthermore, inhibiting the PI3K/Akt pathway effectively blocked VCAN-mediated glioma progression. Conclusion These findings provide valuable insights into the mechanisms underlying glioma recurrence and suggest that targeting both VCAN and the PI3K/Akt pathway could represent a promising therapeutic strategy for managing recurrent gliomas.
Collapse
Affiliation(s)
- Ruolun Wei
- Department of Neurosurgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Haoyun Xie
- Biochemistry and Molecular Biology, Georgetown University, Washington, DC, United States
| | - Yukun Zhou
- Department of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xuhao Chen
- Department of Pathophysiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Liwei Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Brandon Bui
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
- Department of Human Biology, Stanford University, Stanford, CA, United States
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Han R, Sun X, Wu Y, Yang YH, Wang QC, Zhang XT, Ding T, Yang JT. Proteomic and Phosphoproteomic Profiling of Matrix Stiffness-Induced Stemness-Dormancy State Transition in Breast Cancer Cells. J Proteome Res 2024; 23:4658-4673. [PMID: 39298182 DOI: 10.1021/acs.jproteome.4c00563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The dormancy of cancer stem cells is a major factor leading to drug resistance and a high rate of late recurrence and mortality in estrogen receptor-positive (ER+) breast cancer. Previously, we demonstrated that a stiffer matrix induces tumor cell dormancy and drug resistance, whereas a softened matrix promotes tumor cells to exhibit a stem cell state with high proliferation and migration. In this study, we present a comprehensive analysis of the proteome and phosphoproteome in response to gradient changes in matrix stiffness, elucidating the mechanisms behind cell dormancy-induced drug resistance. Overall, we found that antiapoptotic and membrane transport processes may be involved in the mechanical force-induced dormancy resistance of ER+ breast cancer cells. Our research provides new insights from a holistic proteomic and phosphoproteomic perspective, underscoring the significant role of mechanical forces stemming from the stiffness of the surrounding extracellular matrix as a critical regulatory factor in the tumor microenvironment.
Collapse
Affiliation(s)
- Rong Han
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Xu Sun
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Yue Wu
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Ye-Hong Yang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Qiao-Chu Wang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Xu-Tong Zhang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Tao Ding
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Jun-Tao Yang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| |
Collapse
|
14
|
Deng J, Li K, Luo W. Singular Value Decomposition-Driven Non-negative Matrix Factorization with Application to Identify the Association Patterns of Sarcoma Recurrence. Interdiscip Sci 2024; 16:554-567. [PMID: 38424397 DOI: 10.1007/s12539-024-00606-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 03/02/2024]
Abstract
Sarcomas are malignant tumors from mesenchymal tissue and are characterized by their complexity and diversity. The high recurrence rate making it important to understand the mechanisms behind their recurrence and to develop personalized treatments and drugs. However, previous studies on the association patterns of multi-modal data on sarcoma recurrence have overlooked the fact that genes do not act independently, but rather function within signaling pathways. Therefore, this study collected 290 whole solid images, 869 gene and 1387 pathway data of over 260 sarcoma samples from UCSC and TCGA to identify the association patterns of gene-pathway-cell related to sarcoma recurrences. Meanwhile, considering that most multi-modal data fusion methods based on the joint non-negative matrix factorization (NMF) model led to poor experimental repeatability due to random initialization of factorization parameters, the study proposed the singular value decomposition (SVD)-driven joint NMF model by applying the SVD method to calculate initialized weight and coefficient matrices to achieve the reproducibility of the results. The results of the experimental comparison indicated that the SVD algorithm enhances the performance of the joint NMF algorithm. Furthermore, the representative module indicated a significant relationship between genes in pathways and image features. Multi-level analysis provided valuable insights into the connections between biological processes, cellular features, and sarcoma recurrence. In addition, potential biomarkers were uncovered, while various mechanisms of sarcoma recurrence were identified from an imaging genetic perspective. Overall, the SVD-NMF model affords a novel perspective on combining multi-omics data to explore the association related to sarcoma recurrence.
Collapse
Affiliation(s)
- Jin Deng
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, 510642, China
- Pazhou Lab, Guangzhou, 510335, China
| | - Kaijun Li
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, 510642, China
| | - Wei Luo
- College of Mathematics and Informatics, South China Agricultural University, Guangzhou, 510642, China.
- Pazhou Lab, Guangzhou, 510335, China.
| |
Collapse
|
15
|
Huang S, Tan C, Zheng J, Huang Z, Li Z, Lv Z, Chen W, Chen M, Yuan X, Chen C, Yan Q. Identification of RNMT as an immunotherapeutic and prognostic biomarker: From pan-cancer analysis to lung squamous cell carcinoma validation. Immunobiology 2024; 229:152836. [PMID: 39018675 DOI: 10.1016/j.imbio.2024.152836] [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: 04/05/2024] [Revised: 06/23/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Dysregulation of RNA guanine-7 methyltransferase (RNMT) plays a crucial role in the tumor progression and immune responses. However, the detailed role of RNMT in pan-cancer is still unknown. METHODS Bulk transcriptomic data of pan-cancer were obtained from the Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Cancer Cell Line Encyclopedia (CCLE) databases. Single-cell transcriptomic and proteomics data of lung squamous cell carcinoma (LUSC) were analyzed in the Tumor Immune Single-cell Hub 2 (TISCH2) and Clinical Proteomic Tumor Analysis Consortium (CPTAC) databases, respectively. The correlation between RNMT expression and cancer prognosis was analyzed by Cox proportional hazards regression and Kaplan-Meier analyses. The correlation of RNMT expression with common immunoregulators, tumor mutation burden (TMB), microsatellite instability (MSI), mismatch repair (MMR), and DNA methyltransferase (DNMT) was analyzed. Additionally, the correlation between RNMT expression and immune infiltration level was evaluated. A total of 1287 machine learning combinations were used to construct prognostic models for LUSC. qRT-PCR and Western blot were used to validate the bioinformatics findings of RNMT upregulation in LUSC. RESULTS RNMT was widely expressed across different cancers, with significant correlation to prognosis in cancers such as kidney chromophobe (KICH) (p = 0.0033, HR = 7.12), liver hepatocellular carcinoma (LIHC) (p = 0.01, HR = 1.41), and others. Notably, RNMT participates in the regulation of the tumor microenvironment. RNMT expression positively correlated with immune cell expression (Spearman's rank correlation, p < 0.05). Moreover, RNMT expression was strongly associated with immunoregulators, TMB, MSI, MMR, and DNMT in most cancer types. Notably, RNMT expression displayed excellent prognostic and immunological performance in LUSC. The expression of RNMT was mainly enriched in B cells of LUSC tissues. qRT-PCR and Western blot verified the high expression of RNMT in LUSC. CONCLUSION RNMT expression widely correlated with prognosis and immune infiltration in various tumors, especially LUSC. The RNMT detection may provide a new idea for future tumor immune studies and treatment strategies.
Collapse
Affiliation(s)
- Shuqiang Huang
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Cuiyu Tan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Jinzhen Zheng
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Zhugu Huang
- School of Pediatrics, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhihong Li
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Ziyin Lv
- The First School of Clinical Medicine, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Wanru Chen
- The Third School of Clinical Medicine, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Miaoqi Chen
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Xiaojun Yuan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Cairong Chen
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China; Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China.
| | - Qiuxia Yan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China; Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China.
| |
Collapse
|
16
|
Lv W, Xie H, Wu S, Dong J, Jia Y, Ying H. Identification of key metabolism-related genes and pathways in spontaneous preterm birth: combining bioinformatic analysis and machine learning. Front Endocrinol (Lausanne) 2024; 15:1440436. [PMID: 39229380 PMCID: PMC11368757 DOI: 10.3389/fendo.2024.1440436] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Background Spontaneous preterm birth (sPTB) is a global disease that is a leading cause of death in neonates and children younger than 5 years of age. However, the etiology of sPTB remains poorly understood. Recent evidence has shown a strong association between metabolic disorders and sPTB. To determine the metabolic alterations in sPTB patients, we used various bioinformatics methods to analyze the abnormal changes in metabolic pathways in the preterm placenta via existing datasets. Methods In this study, we integrated two datasets (GSE203507 and GSE174415) from the NCBI GEO database for the following analysis. We utilized the "Deseq2" R package and WGCNA for differentially expressed genes (DEGs) analysis; the identified DEGs were subsequently compared with metabolism-related genes. To identify the altered metabolism-related pathways and hub genes in sPTB patients, we performed multiple functional enrichment analysis and applied three machine learning algorithms, LASSO, SVM-RFE, and RF, with the hub genes that were verified by immunohistochemistry. Additionally, we conducted single-sample gene set enrichment analysis to assess immune infiltration in the placenta. Results We identified 228 sPTB-related DEGs that were enriched in pathways such as arachidonic acid and glutathione metabolism. A total of 3 metabolism-related hub genes, namely, ANPEP, CKMT1B, and PLA2G4A, were identified and validated in external datasets and experiments. A nomogram model was developed and evaluated with 3 hub genes; the model could reliably distinguish sPTB patients and term labor patients with an area under the curve (AUC) > 0.75 for both the training and validation sets. Immune infiltration analysis revealed immune dysregulation in sPTB patients. Conclusion Three potential hub genes that influence the occurrence of sPTB through shadow participation in placental metabolism were identified; these results provide a new perspective for the development and targeting of treatments for sPTB.
Collapse
Affiliation(s)
- Wenqi Lv
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Han Xie
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Shengyu Wu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Jiaqi Dong
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Yuanhui Jia
- Department of Clinical Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, sChina
| | - Hao Ying
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
- Department of Clinical Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, sChina
| |
Collapse
|
17
|
Tan C, Huang S, Xu L, Zhang T, Yuan X, Li Z, Chen M, Chen C, Yan Q. Cross-talk between oxidative stress and lipid metabolism regulators reveals molecular clusters and immunological characterization in polycystic ovarian syndrome. Lipids Health Dis 2024; 23:248. [PMID: 39143634 PMCID: PMC11325768 DOI: 10.1186/s12944-024-02237-3] [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: 06/16/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Changes in the oxidative stress and lipid metabolism (OSLM) pathways play important roles in polycystic ovarian syndrome (PCOS) pathogenesis and development. Consequently, a systematic analysis of genes related to OSLM was conducted to identify molecular clusters and explore new biomarkers that are helpful for the diagnostic of PCOS. METHODS Gene expression and clinical data from 22 PCOS women and 14 normal women were obtained from the GEO database (GSE34526, GSE95728, and GSE106724). Consensus clustering identified OSLM-related molecular clusters, and WGCNA revealed co-expression patterns. The immune microenvironment was quantitatively assessed utilizing the CIBERSORT algorithm. Multiple machine learning models and connectivity map analyses were subsequently applied to explore potential biomarkers for PCOS, and nomograms were employed to develop a predictive multigene model of PCOS. Finally, the OSLM status of PCOS and the hub genes expression profiles were preliminarily verified using TUNEL, qRT‒PCR, western blot, and IHC assays in a PCOS mouse model. RESULTS 19 differential expression genes (DEGs) related to OSLM were identified. Based on 19 DEGs that were strongly influenced by OSLM, PCOS patients were stratified into two distinct clusters, designated Cluster 1 and Cluster 2. Distinct differences in the immune cell proportions existed in normal and two PCOS clusters. The random forest showed the best results, with the least cross-entropy and the utmost AUC (cross-entropy: 0.111 AUC: 0.960). Among the 19 OSLM-related genes, CXCR1, ACP5, CEACAM3, S1PR4, and TCF7 were identified by a Bayesian network and had a good fit with PCOS disease risk by the nomogram (AUC: 0.990 CI: 0.968-1.000). TUNEL assays revealed more severe DNA damage within the ovarian granule cells of PCOS mice than in those of normal mice (P < 0.001). The RNA and protein expression levels of the five hub genes were significantly elevated in PCOS mice, which was consistent with the results of the bioinformatics analyses. CONCLUSION A novel predictive model was constructed for PCOS patients and five hub genes were identified as potential biomarkers to offer novel insights into clinical diagnostic strategies for PCOS.
Collapse
Affiliation(s)
- Cuiyu Tan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Shuqiang Huang
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Liying Xu
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Tongtong Zhang
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Xiaojun Yuan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Zhihong Li
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Miaoqi Chen
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Cairong Chen
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China.
- Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China.
| | - Qiuxia Yan
- Center for Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China.
- Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, China.
| |
Collapse
|
18
|
Zhu Y, Chen Y, Zu Y. Leveraging a neutrophil-derived PCD signature to predict and stratify patients with acute myocardial infarction: from AI prediction to biological interpretation. J Transl Med 2024; 22:612. [PMID: 38956669 PMCID: PMC11221097 DOI: 10.1186/s12967-024-05415-0] [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: 12/11/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Programmed cell death (PCD) has recently been implicated in modulating the removal of neutrophils recruited in acute myocardial infarction (AMI). Nonetheless, the clinical significance and biological mechanism of neutrophil-related PCD remain unexplored. METHODS We employed an integrative machine learning-based computational framework to generate a predictive neutrophil-derived PCD signature (NPCDS) within five independent microarray cohorts from the peripheral blood of AMI patients. Non-negative matrix factorization was leveraged to develop an NPCDS-based AMI subtype. To elucidate the biological mechanism underlying NPCDS, we implemented single-cell transcriptomics on Cd45+ cells isolated from the murine heart of experimental AMI. We finally conducted a Mendelian randomization (MR) study and molecular docking to investigate the therapeutic value of NPCDS on AMI. RESULTS We reported the robust and superior performance of NPCDS in AMI prediction, which contributed to an optimal combination of random forest and stepwise regression fitted on nine neutrophil-related PCD genes (MDM2, PTK2B, MYH9, IVNS1ABP, MAPK14, GNS, MYD88, TLR2, CFLAR). Two divergent NPCDS-based subtypes of AMI were revealed, in which subtype 1 was characterized as inflammation-activated with more vibrant neutrophil activities, whereas subtype 2 demonstrated the opposite. Mechanically, we unveiled the expression dynamics of NPCDS to regulate neutrophil transformation from a pro-inflammatory phase to an anti-inflammatory phase in AMI. We uncovered a significant causal association between genetic predisposition towards MDM2 expression and the risk of AMI. We also found that lidoflazine, isotetrandrine, and cepharanthine could stably target MDM2. CONCLUSION Altogether, NPCDS offers significant implications for prediction, stratification, and therapeutic management for AMI.
Collapse
Affiliation(s)
- Yihao Zhu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Yuxi Chen
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Yao Zu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, People's Republic of China.
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, People's Republic of China.
- Marine Biomedical Science and Technology Innovation Platform of Lin-Gang Special Area, Shanghai, 201306, People's Republic of China.
| |
Collapse
|
19
|
Guo T, Wang J, Meng X, Wang Y, Lou Y, Ma J, Xu S, Ni X, Jia Z, Jin L, Wang C, Chen Q, Li P, Huang Y, Ren S. Deciphering the role of zinc homeostasis in the tumor microenvironment and prognosis of prostate cancer. Discov Oncol 2024; 15:207. [PMID: 38833013 DOI: 10.1007/s12672-024-01006-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Dysregulation of zinc homeostasis is widely recognized as a hallmark feature of prostate cancer (PCa) based on the compelling clinical and experimental evidence. Nevertheless, the implications of zinc dyshomeostasis in PCa remains largely unexplored. METHODS In this research, the zinc homeostasis pattern subtype (ZHPS) was constructed according to the profile of zinc homeostasis genes. The identified subtypes were assessed for their immune functions, mutational landscapes, biological peculiarities and drug susceptibility. Subsequently, we developed the optimal signature, known as the zinc homeostasis-related risk score (ZHRRS), using the approach won out in multifariously machine learning algorithms. Eventually, clinical specimens, Bayesian network inference and single-cell sequencing were used to excavate the underlying mechanisms of MT1A in PCa. RESULTS The zinc dyshomeostasis subgroup, ZHPS2, possessed a markedly worse prognosis than ZHPS1. Moreover, ZHPS2 demonstrated a more conspicuous genomic instability and better therapeutic responses to docetaxel and olaparib than ZHPS1. Compared with traditional clinicopathological characteristics and 35 published signatures, ZHRRS displayed a significantly improved accuracy in prognosis prediction. The diagnostic value of MT1A in PCa was substantiated through analysis of clinical samples. Additionally, we inferred and established the regulatory network of MT1A to elucidate its biological mechanisms. CONCLUSIONS The ZHPS classifier and ZHRRS model hold great potential as clinical applications for improving outcomes of PCa patients.
Collapse
Affiliation(s)
- Tao Guo
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Wang
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiangyu Meng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ye Wang
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yihaoyun Lou
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jianglei Ma
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shuang Xu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangyu Ni
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zongming Jia
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lichen Jin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chengyu Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyang Chen
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Peng Li
- Department of Urology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China.
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Shancheng Ren
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
20
|
Han T, Xu W, Wang X, Gao J, Zhang S, Yang L, Wang M, Li C, Li X. Emodin-8-O-β-D-glucopyranoside-induced hepatotoxicity and gender differences in zebrafish as revealed by integration of metabolomics and transcriptomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155411. [PMID: 38518638 DOI: 10.1016/j.phymed.2024.155411] [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: 06/13/2023] [Revised: 10/27/2023] [Accepted: 02/01/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Emodin-8-O-β-D-glucopyranoside (Em8G) is an active ingredient of traditional Chinese medicine Rhei Radix et Rhizoma and Polygonum multiflorum Thunb.. And it caused hepatotoxicity, while the underlying mechanism was not clear yet. PURPOSE We aimed to explore the detrimental effects of Em8G on the zebrafish liver through the metabolome and transcriptome integrated analysis. STUDY DESIGN AND METHODS In this study, zebrafish larvae were used in acute toxicity tests to reveal the hepatotoxicity of Em8G. Adult zebrafish were then used to evaluate the gender differences in hepatotoxicity induced by Em8G. Integration of transcriptomic and metabolomic analysis was used further to explore the molecular mechanisms underlying gender differences in hepatotoxicity. RESULTS Our results showed that under non-lethal concentration exposure conditions, hepatotoxicity was observed in Em8G-treated zebrafish larvae, including changes in liver transmittance, liver area, hepatocyte apoptosis and hepatocyte vacuolation. Male adult zebrafish displayed a higher Em8G-induced hepatotoxicity than female zebrafish, as demonstrated by the higher mortality and histopathological alterations. The results of transcriptomics combined with metabolomics showed that Em8G mainly affected carbohydrate metabolism (such as TCA cycle) in male zebrafish and amino acid metabolism (such as arginine and proline metabolism) in females, suggesting that the difference of energy metabolism disorder may be the potential mechanism of male and female liver toxicity induced by Em8G. CONCLUSIONS This study provided the direct evidence for the hepatotoxicity of Em8G to zebrafish models in vivo, and brought a new insight into the molecular mechanisms of Em8G hepatotoxicity, which can guide the rational application of this phytotoxin. In addition, our findings revealed gender differences in the hepatotoxicity of Em8G to zebrafish, which is related to energy metabolism and provided a methodological reference for evaluating hepatotoxic drugs with gender differences.
Collapse
Affiliation(s)
- Ting Han
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenjuan Xu
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xuan Wang
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiahui Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuyan Zhang
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Linlin Yang
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Min Wang
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunshuai Li
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiangri Li
- Centre of TCM Processing Research / Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
21
|
Liang H, Zheng Y, Huang Z, Dai J, Yao L, Xie D, Chen D, Qiu H, Wang H, Li H, Leng J, Tang Z, Zhang D, Zhou H. Pan-cancer analysis for the prognostic and immunological role of CD47: interact with TNFRSF9 inducing CD8 + T cell exhaustion. Discov Oncol 2024; 15:149. [PMID: 38720108 PMCID: PMC11078914 DOI: 10.1007/s12672-024-00951-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/27/2024] [Indexed: 05/12/2024] Open
Abstract
PURPOSE The research endeavors to explore the implications of CD47 in cancer immunotherapy effectiveness. Specifically, there is a gap in comprehending the influence of CD47 on the tumor immune microenvironment, particularly in relation to CD8 + T cells. Our study aims to elucidate the prognostic and immunological relevance of CD47 to enhance insights into its prospective utilities in immunotherapeutic interventions. METHODS Differential gene expression analysis, prognosis assessment, immunological infiltration evaluation, pathway enrichment analysis, and correlation investigation were performed utilizing a combination of R packages, computational algorithms, diverse datasets, and patient cohorts. Validation of the concept was achieved through the utilization of single-cell sequencing technology. RESULTS CD47 demonstrated ubiquitous expression across various cancer types and was notably associated with unfavorable prognostic outcomes in pan-cancer assessments. Immunological investigations unveiled a robust correlation between CD47 expression and T-cell infiltration rather than T-cell exclusion across multiple cancer types. Specifically, the CD47-high group exhibited a poorer prognosis for the cytotoxic CD8 + T cell Top group compared to the CD47-low group, suggesting a potential impairment of CD8 + T cell functionality by CD47. The exploration of mechanism identified enrichment of CD47-associated differentially expressed genes in the CD8 + T cell exhausted pathway in multiple cancer contexts. Further analyses focusing on the CD8 TCR Downstream Pathway and gene correlation patterns underscored the significant involvement of TNFRSF9 in mediating these effects. CONCLUSION A robust association exists between CD47 and the exhaustion of CD8 + T cells, potentially enabling immune evasion by cancer cells and thereby contributing to adverse prognostic outcomes. Consequently, genes such as CD47 and those linked to T-cell exhaustion, notably TNFRSF9, present as promising dual antigenic targets, providing critical insights into the field of immunotherapy.
Collapse
Affiliation(s)
- Hongxin Liang
- Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China
| | - Yong Zheng
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zekai Huang
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jinchi Dai
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Lintong Yao
- Southern Medical University, Guangzhou, 510515, China
| | - Daipeng Xie
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Duo Chen
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hongrui Qiu
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Huili Wang
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Hao Li
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jinhang Leng
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ziming Tang
- Southern Medical University, Guangzhou, 510515, China
| | - Dongkun Zhang
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Haiyu Zhou
- Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China.
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
22
|
Cao Y, Fu A, Liu C. Exploring the NRF2-TP53 Signaling Network Through Machine Learning and Pan-Cancer Analysis: Identifying Potential targets for Cancer Prognosis Related to Oxidative Stress. Adv Biol (Weinh) 2024; 8:e2300659. [PMID: 38519438 DOI: 10.1002/adbi.202300659] [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: 12/03/2023] [Revised: 01/24/2024] [Indexed: 03/24/2024]
Abstract
Oxidative stress (OXS) is closely related to tumor prognosis and immune response, while TP53 integrated with NRF2 is closely associated with the regulation of cancer-related OXS. Hence, constructing a TP53-NRF2 integrated OXS signature of pan-cancer is essential in predicting survival prognosis and facilitating cancer drug treatment. The pan-cancer analysis acquired the Cancer Genome Atlas (TCGA) transcriptome sequencing data from UCSC Xena, which consisted of 33 cancer types (n = 10 440). The Random Forest, Lasso regression, and Cox regression analyses are used to construct an OXS score based on 25 OXS genes. Following this, based on the OXS signature, patients are categorized into low- and high-risk groups. The disparities between the two cohorts regarding survival prognosis, immune infiltration, and drug sensitivity are delved deeply. The expression level of genes is confirmed using immunohistochemistry. The prognosis of pan-cancer patients is adequately predicted by the OXS signature with the assistance of the machine-learning algorithm. A highly accurate nomogram is developed by combining the OXS signature and clinical features. The presence of immune cells indicated that the OXS signature can be associated with the critical pathways of immunotherapy for all types of cancer, and BCL2 showed promising results. Distinct inter-group differences are observed in the OXS signature for frequently utilized antineoplastic medications in clinical settings, including first-line drugs suggested in the guidelines. In summary, by conducting a thorough analysis of OXS genes, a new model based on OXSscore is successfully developed. This model can predict the clinical prognosis and drug sensitivity of pan-cancer with high accuracy. Potential stars in the field of cancer-related anti-OXS may include drugs that target BCL2.
Collapse
Affiliation(s)
- Yuchen Cao
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Ao Fu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Chunjun Liu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| |
Collapse
|
23
|
Bai R, Luo Y. Exploring the role of mitochondrial-associated and peripheral neuropathy genes in the pathogenesis of diabetic peripheral neuropathy. BMC Neurol 2024; 24:95. [PMID: 38481183 PMCID: PMC10936109 DOI: 10.1186/s12883-024-03589-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is a prevalent and serious complication of diabetes mellitus, impacting the nerves in the limbs and leading to symptoms like pain, numbness, and diminished function. While the exact molecular and immune mechanisms underlying DPN remain incompletely understood, recent findings indicate that mitochondrial dysfunction may play a role in the advancement of this diabetic condition. METHODS Two RNA transcriptome datasets (codes: GSE185011 and GSE95849), comprising samples from diabetic peripheral neuropathy (DPN) patients and healthy controls (HC), were retrieved from the Gene Expression Omnibus (GEO) database hosted by the National Center for Biotechnology Information (NCBI). Subsequently, differential expression analysis and gene set enrichment analysis were performed. Protein-protein interaction (PPI) networks were constructed to pinpoint key hub genes associated with DPN, with a specific emphasis on genes related to mitochondria and peripheral neuropathy disease (PND) that displayed differential expression. Additionally, the study estimated the levels of immune cell infiltration in both the HC and DPN samples. To validate the findings, quantitative polymerase chain reaction (qPCR) was employed to confirm the differential expression of selected genes in the DPN samples. RESULTS This research identifies four hub genes associated mitochondria or PN. Furthermore, the analysis revealed increased immune cell infiltration in DPN tissues, particularly notable for macrophages and T cells. Additionally, our investigation identified potential drug candidates capable of regulating the expression of the four hub genes. These findings were corroborated by qPCR results, reinforcing the credibility of our bioinformatics analysis. CONCLUSIONS This study provides a comprehensive overview of the molecular and immunological characteristics of DPN, based on both bioinformatics and experimental methods.
Collapse
Affiliation(s)
- Ruojing Bai
- Department of Geriatric Medicine, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Yuanyuan Luo
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
24
|
Xu X, Yu Y, Zhang W, Ma W, He C, Qiu G, Wang X, Liu Q, Zhao M, Xie J, Tao F, Perry JM, Liu Q, Rao S, Kang X, Zhao M, Jiang L. SHP-1 inhibition targets leukaemia stem cells to restore immunosurveillance and enhance chemosensitivity by metabolic reprogramming. Nat Cell Biol 2024; 26:464-477. [PMID: 38321204 DOI: 10.1038/s41556-024-01349-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 01/03/2024] [Indexed: 02/08/2024]
Abstract
Leukaemia stem cells (LSCs) in acute myeloid leukaemia present a considerable treatment challenge due to their resistance to chemotherapy and immunosurveillance. The connection between these properties in LSCs remains poorly understood. Here we demonstrate that inhibition of tyrosine phosphatase SHP-1 in LSCs increases their glycolysis and oxidative phosphorylation, enhancing their sensitivity to chemotherapy and vulnerability to immunosurveillance. Mechanistically, SHP-1 inhibition leads to the upregulation of phosphofructokinase platelet (PFKP) through the AKT-β-catenin pathway. The increase in PFKP elevates energy metabolic activities and, as a consequence, enhances the sensitivity of LSCs to chemotherapeutic agents. Moreover, the upregulation of PFKP promotes MYC degradation and, consequently, reduces the immune evasion abilities of LSCs. Overall, our study demonstrates that targeting SHP-1 disrupts the metabolic balance in LSCs, thereby increasing their vulnerability to chemotherapy and immunosurveillance. This approach offers a promising strategy to overcome LSC resistance in acute myeloid leukaemia.
Collapse
Affiliation(s)
- Xi Xu
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, Guangdong, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yanhui Yu
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Wenwen Zhang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, Guangdong, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Ma
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, Guangdong, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chong He
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, Guangdong, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guo Qiu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyi Wang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, Guangdong, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiong Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Minyi Zhao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiayi Xie
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, Guangdong, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fang Tao
- Children's Mercy Hospital, University of Kansas Medical Center, University of Missouri, Kansas City, MO, USA
| | - John M Perry
- Children's Mercy Hospital, University of Kansas Medical Center, University of Missouri, Kansas City, MO, USA
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Xunlei Kang
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, USA.
| | - Meng Zhao
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University Guangzhou, Guangdong, China.
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Linjia Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
25
|
Huang Y, Liu H, Liu B, Chen X, Li D, Xue J, Li N, Zhu L, Yang L, Xiao J, Liu C. Quantified pathway mutations associate epithelial-mesenchymal transition and immune escape with poor prognosis and immunotherapy resistance of head and neck squamous cell carcinoma. BMC Med Genomics 2024; 17:49. [PMID: 38331768 PMCID: PMC10854145 DOI: 10.1186/s12920-024-01818-6] [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: 06/14/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Pathway mutations have been calculated to predict the poor prognosis and immunotherapy resistance in head and neck squamous cell carcinoma (HNSCC). To uncover the unique markers predicting prognosis and immune therapy response, the accurate quantification of pathway mutations are required to evaluate epithelial-mesenchymal transition (EMT) and immune escape. Yet, there is a lack of score to accurately quantify pathway mutations. MATERIAL AND METHODS Firstly, we proposed Individualized Weighted Hallmark Gene Set Mutation Burden (IWHMB, https://github.com/YuHongHuang-lab/IWHMB ) which integrated pathway structure information and eliminated the interference of global Tumor Mutation Burden to accurately quantify pathway mutations. Subsequently, to further elucidate the association of IWHMB with EMT and immune escape, support vector machine regression model was used to identify IWHMB-related transcriptomic features (IRG), while Adversarially Regularized Graph Autoencoder (ARVGA) was used to further resolve IRG network features. Finally, Random walk with restart algorithm was used to identify biomarkers for predicting ICI response. RESULTS We quantified the HNSCC pathway mutation signatures and identified pathway mutation subtypes using IWHMB. The IWHMB-related transcriptomic features (IRG) identified by support vector machine regression were divided into 5 communities by ARVGA, among which the Community 1 enriching malignant mesenchymal components promoted EMT dynamically and regulated immune patterns associated with ICI responses. Bridge Hub Gene (BHG) identified by random walk with restart was key to IWHMB in EMT and immune escape, thus, more predictive for ICI response than other 70 public signatures. CONCLUSION In summary, the novel pathway mutation scoring-IWHMB suggested that the elevated malignancy mediated by pathway mutations is a major cause of poor prognosis and immunotherapy failure in HNSCC, and is capable of identifying novel biomarkers to predict immunotherapy response.
Collapse
Affiliation(s)
- Yuhong Huang
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Han Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Bo Liu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, China
| | - Xiaoyan Chen
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Danya Li
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Junyuan Xue
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Nan Li
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Lei Zhu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Liu Yang
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China
| | - Jing Xiao
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
| | - Chao Liu
- Department of Oral Pathology, Dalian Medical University School of Stomatology, Dalian, China.
- Academician Laboratory of Immunology and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
| |
Collapse
|
26
|
Zhu Y, Chen B, Zu Y. Identifying OGN as a Biomarker Covering Multiple Pathogenic Pathways for Diagnosing Heart Failure: From Machine Learning to Mechanism Interpretation. Biomolecules 2024; 14:179. [PMID: 38397416 PMCID: PMC10886937 DOI: 10.3390/biom14020179] [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: 11/21/2023] [Revised: 01/14/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The pathophysiologic heterogeneity of heart failure (HF) necessitates a more detailed identification of diagnostic biomarkers that can reflect its diverse pathogenic pathways. METHODS We conducted weighted gene and multiscale embedded gene co-expression network analysis on differentially expressed genes obtained from HF and non-HF specimens. We employed a machine learning integration framework and protein-protein interaction network to identify diagnostic biomarkers. Additionally, we integrated gene set variation analysis, gene set enrichment analysis (GSEA), and transcription factor (TF)-target analysis to unravel the biomarker-dominant pathways. Leveraging single-sample GSEA and molecular docking, we predicted immune cells and therapeutic drugs related to biomarkers. Quantitative polymerase chain reaction validated the expressions of biomarkers in the plasma of HF patients. A two-sample Mendelian randomization analysis was implemented to investigate the causal impact of biomarkers on HF. RESULTS We first identified COL14A1, OGN, MFAP4, and SFRP4 as candidate biomarkers with robust diagnostic performance. We revealed that regulating biomarkers in HF pathogenesis involves TFs (BNC2, MEOX2) and pathways (cell adhesion molecules, chemokine signaling pathway, cytokine-cytokine receptor interaction, oxidative phosphorylation). Moreover, we observed the elevated infiltration of effector memory CD4+ T cells in HF, which was highly related to biomarkers and could impact immune pathways. Captopril, aldosterone antagonist, cyclopenthiazide, estradiol, tolazoline, and genistein were predicted as therapeutic drugs alleviating HF via interactions with biomarkers. In vitro study confirmed the up-regulation of OGN as a plasma biomarker of HF. Mendelian randomization analysis suggested that genetic predisposition toward higher plasma OGN promoted the risk of HF. CONCLUSIONS We propose OGN as a diagnostic biomarker for HF, which may advance our understanding of the diagnosis and pathogenesis of HF.
Collapse
Affiliation(s)
- Yihao Zhu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Bin Chen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Lin-gang), Shanghai 201306, China
| | - Yao Zu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai 201306, China
| |
Collapse
|
27
|
Guo C, Yang X, Li L. Pyroptosis-Related Gene Signature Predicts Prognosis and Response to Immunotherapy and Medication in Pediatric and Young Adult Osteosarcoma Patients. J Inflamm Res 2024; 17:417-445. [PMID: 38269108 PMCID: PMC10807455 DOI: 10.2147/jir.s440425] [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] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024] Open
Abstract
Purpose Pyroptosis, a new form of inflammatory programmed cell death, has recently gained attention. However, the impact of the expression levels of pyroptosis-related genes (PRGs) on the overall survival (OS) of osteosarcoma patients remains unclear. This study aims to investigate the impact of the expression levels of PRGs on the OS of pediatric and young adult patients with osteosarcoma. Patients and Methods Transcriptome matrix datasets of normal muscle or skeletal tissues from the Genotype-Tissue Expression (GTEx) project and osteosarcoma specimen the National Cancer Institute's (NCI) Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database were used to identify pyroptosis-related genes (PRGs) associated with prognosis. The National Center for Biotechnology Information's (NCBI) GSE21257 dataset was employed to validate the predictive value of the pyroptosis-related signature (PRS). Additionally, reverse transcription polymerase chain reaction (RT-qPCR) experiment was performed in normal and osteosarcoma cell lines. Results The study identified 18 differentially expressed PRGs (DEPRGs) between normal muscle or skeletal tissues and tumor samples. Multiple machine learning techniques were used to select PRGs, resulting in the identification of four hub PRGs. A PRS-score was calculated for each sample based on the expression of these four hub PRGs, and samples were categorized into low and high PRS-score level groups. It was confirmed that metastatic status and PRS-score level are independent prognostic predictors. A nomogram model for predicting OS of osteosarcoma patients was constructed. Single-cell RNA-sequencing data display the expression patterns of the hub PRGs. RT-qPCR data results were found to be consistent with the differential expression analysis performed on TARGET and GTEx samples. Conclusion The study developed a novel pyroptosis-related gene signature that can stratify pediatric and young adult osteosarcoma patients into different risk groups, thus predicting their response to immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Chaofan Guo
- Department of Orthopedics, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi Province, People’s Republic of China
- Department of Spine Surgery, Fifth Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, People’s Republic of China
| | - Xin Yang
- Department of Neurosurgery, Chongqing Fourth People’s Hospital, Chongqing, People’s Republic of China
| | - Lijun Li
- Department of Orthopedics, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi Province, People’s Republic of China
- Department of Spine Surgery, Fifth Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, People’s Republic of China
| |
Collapse
|
28
|
Li Z, Peng L, Sun L, Si J. A link between mitochondrial damage and the immune microenvironment of delayed onset muscle soreness. BMC Med Genomics 2023; 16:196. [PMID: 37612729 PMCID: PMC10464284 DOI: 10.1186/s12920-023-01621-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Delayed onset muscle soreness (DOMS) is a self-healing muscle pain disorder. Inflammatory pain is the main feature of DOMS. More and more researchers have realized that changes in mitochondrial morphology are related to pain. However, the role of mitochondria in the pathogenesis of DOMS and the abnormal immune microenvironment is still unknown. METHODS Mitochondria-related genes and gene expression data were obtained from MitoCarta3.0 and NCBI GEO databases. The network of mitochondrial function and the immune microenvironment of DOMS was constructed by computer algorithm. Subsequently, the skeletal muscle of DOMS rats was subjected to qPCR to verify the bioinformatics results. DOMS and non-DOMS histological samples were further studied by staining and transmission electron microscopy. RESULTS Bioinformatics results showed that expression of mitochondria-related genes was changed in DOMS. The results of qPCR showed that four hub genes (AMPK, PGC1-α, SLC25A25, and ARMCX1) were differentially expressed in DOMS. These hub genes are related to the degree of skeletal muscle immune cell infiltration, mitochondrial respiratory chain complex, DAMPs, the TCA cycle, and mitochondrial metabolism. Bayesian network inference showed that IL-6 and PGC1-α may be the main regulatory genes of mitochondrial damage in DOMS. Transmission electron microscopy revealed swelling of skeletal muscle mitochondria and disorganization of myofilaments. CONCLUSIONS Our study found that skeletal muscle mitochondrial damage is one of the causes of inflammatory factor accumulation in DOMS. According to the screened-out hub genes, this study provides a reference for follow-up clinical application.
Collapse
Affiliation(s)
- Zheng Li
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China
| | - Lina Peng
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China
| | - Lili Sun
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China.
| | - Juncheng Si
- College of Sport Human Sciences, Harbin Sport University, No. 1, Dacheng Road, Nangang District, 150008, Harbin, China
| |
Collapse
|
29
|
Tao W, Yang X, Zhang Q, Bi S, Yao Z. Optimal treatment for post-MI heart failure in rats: dapagliflozin first, adding sacubitril-valsartan 2 weeks later. Front Cardiovasc Med 2023; 10:1181473. [PMID: 37383701 PMCID: PMC10296765 DOI: 10.3389/fcvm.2023.1181473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/19/2023] [Indexed: 06/30/2023] Open
Abstract
Background Based on previous research, both dapagliflozin (DAPA) and sacubitril-valsartan (S/V) improve the prognosis of patients with heart failure (HF). Our study aims to investigate whether the early initiation of DAPA or the combination of DAPA with S/V in different orders would exert a greater protective effect on heart function than that of S/V alone in post-myocardial infarction HF (post-MI HF). Methods Rats were randomized into six groups: (A) Sham; (B) MI; (C) MI + S/V (1st d); (D) MI + DAPA (1st d); (E) MI + S/V (1st d) + DAPA (14th d); (F) MI + DAPA (1st d) + S/V (14th d). The MI model was established in rats via surgical ligation of the left anterior descending coronary artery. Histology, Western blotting, RNA-seq, and other approaches were used to explore the optimal treatment to preserve the heart function in post-MI HF. A daily dose of 1 mg/kg DAPA and 68 mg/kg S/V was administered. Results The results of our study revealed that DAPA or S/V substantially improved the cardiac structure and function. DAPA and S/V monotherapy resulted in comparable reduction in infarct size, fibrosis, myocardium hypertrophy, and apoptosis. The administration of DAPA followed by S/V results in a superior improvement in heart function in rats with post-MI HF than those in other treatment groups. The administration of DAPA following S/V did not result in any additional improvement in heart function as compared to S/V monotherapy in rats with post-MI HF. Our findings further suggest that the combination of DAPA and S/V should not be administered within 3 days after acute myocardial infarction (AMI), as it resulted in a considerable increase in mortality. Our RNA-Seq data revealed that DAPA treatment after AMI altered the expression of genes related to myocardial mitochondrial biogenesis and oxidative phosphorylation. Conclusions Our study revealed no notable difference in the cardioprotective effects of singular DAPA or S/V in rats with post-MI HF. Based on our preclinical investigation, the most effective treatment strategy for post-MI HF is the administration of DAPA during the 2 weeks, followed by the addition of S/V to DAPA later. Conversely, adopting a therapeutic scheme whereby S/V was administered first, followed by later addition of DAPA, failed to further improve the cardiac function compared to S/V monotherapy.
Collapse
Affiliation(s)
- Wenqi Tao
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Xiaoyu Yang
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Qing Zhang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuli Bi
- School of Medicine, Nankai University, Tianjin, China
| | - Zhuhua Yao
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| |
Collapse
|
30
|
Zhang H, Feng H, Yu T, Zhang M, Liu Z, Ma L, Liu H. Construction of an oxidative stress-related lncRNAs signature to predict prognosis and the immune response in gastric cancer. Sci Rep 2023; 13:8822. [PMID: 37258567 DOI: 10.1038/s41598-023-35167-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/13/2023] [Indexed: 06/02/2023] Open
Abstract
Oxidative stress, as a characteristic of cellular aerobic metabolism, plays a crucial regulatory role in the development and metastasis of gastric cancer (GC). Long noncoding RNAs (lncRNAs) are important regulators in GC development. However, research on the prognostic patterns of oxidative stress-related lncRNAs (OSRLs) and their functions in the immune microenvironment is currently insufficient. We identified the OSRLs signature (DIP2A-IT1, DUXAP8, TP53TG1, SNHG5, AC091057.1, AL355001.1, ARRDC1-AS1, and COLCA1) from 185 oxidative stress-related genes in The Cancer Genome Atlas (TCGA) cohort via random survival forest and Cox analyses, and the results were subsequently validated in the Gene Expression Omnibus (GEO) dataset. The patients were divided into high- and low-risk groups by the risk score of the OSRLs signature. Longer overall survival was detected in the low-risk group than in the high-risk group in both the TCGA cohort (P < 0. 001, HR = 0.43, 95% CI 0.31-0.62) and the GEO cohort (P = 0.014, HR = 0.67, 95% CI 0.48-0.93). Next, multivariate Cox analysis identified that the risk model was an independent prognostic characteristic (HR > 1, P = 0.005), and time-dependent receiver operating characteristic (ROC) curve analysis and nomogram analysis were utilized to evaluate the predictive ability of the risk model. Next, gene set enrichment analysis revealed that the immune-related pathway, Wnt/[Formula: see text]-catenin signature, mammalian target of rapamycin complex 1 signature, and cytokine‒cytokine receptor interaction was enriched. High-risk patients were more responsive to CD200, TNFSF4, TNFSF9, and BTNL2 immune checkpoint blockade. The results of qRT‒PCR further proved the accuracy of our bioinformatic analysis. Overall, our study identified a novel OSRLs signature that can serve as a promising biomarker and prognostic indicator, which provides a personalized predictive approach for patient prognosis evaluation and treatment.
Collapse
Affiliation(s)
- Hui Zhang
- School of Life Science, Liaoning University, Shenyang, 110036, China
| | - Huawei Feng
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
- Key Laboratory of Computational Simulation and Information Processing of Biomacromolecules of Liaoning Province, Shenyang, 110036, China
- Liaoning Provincial Engineering Laboratory of Molecular Modeling and Design for Drug, Shenyang, 110036, China
- Key Laboratory for Simulating Computation and Information Processing of Bio-Macromolecules of Shenyang, Shenyang, 110036, China
| | - Tiansong Yu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
| | - Man Zhang
- School of Life Science, Liaoning University, Shenyang, 110036, China
| | - Zhikui Liu
- Liaoning Huikang Testing and Evaluation Technology Co, Shenyang, 110036, China
| | - Lidan Ma
- Dandong Customs Integrated Technical Service Center, Dandong, 118000, China
| | - Hongsheng Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China.
- Key Laboratory of Computational Simulation and Information Processing of Biomacromolecules of Liaoning Province, Shenyang, 110036, China.
- Liaoning Provincial Engineering Laboratory of Molecular Modeling and Design for Drug, Shenyang, 110036, China.
- Key Laboratory for Simulating Computation and Information Processing of Bio-Macromolecules of Shenyang, Shenyang, 110036, China.
| |
Collapse
|
31
|
Liu Y, Zhou S, Wang L, Xu M, Huang X, Li Z, Hajdu A, Zhang L. Machine learning approach combined with causal relationship inferring unlocks the shared pathomechanism between COVID-19 and acute myocardial infarction. Front Microbiol 2023; 14:1153106. [PMID: 37065165 PMCID: PMC10090501 DOI: 10.3389/fmicb.2023.1153106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 02/27/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundIncreasing evidence suggests that people with Coronavirus Disease 2019 (COVID-19) have a much higher prevalence of Acute Myocardial Infarction (AMI) than the general population. However, the underlying mechanism is not yet comprehended. Therefore, our study aims to explore the potential secret behind this complication.Materials and methodsThe gene expression profiles of COVID-19 and AMI were acquired from the Gene Expression Omnibus (GEO) database. After identifying the differentially expressed genes (DEGs) shared by COVID-19 and AMI, we conducted a series of bioinformatics analytics to enhance our understanding of this issue.ResultsOverall, 61 common DEGs were filtered out, based on which we established a powerful diagnostic predictor through 20 mainstream machine-learning algorithms, by utilizing which we could estimate if there is any risk in a specific COVID-19 patient to develop AMI. Moreover, we explored their shared implications of immunology. Most remarkably, through the Bayesian network, we inferred the causal relationships of the essential biological processes through which the underlying mechanism of co-pathogenesis between COVID-19 and AMI was identified.ConclusionFor the first time, the approach of causal relationship inferring was applied to analyzing shared pathomechanism between two relevant diseases, COVID-19 and AMI. Our findings showcase a novel mechanistic insight into COVID-19 and AMI, which may benefit future preventive, personalized, and precision medicine.Graphical abstract
Collapse
Affiliation(s)
- Ying Liu
- Department of Cardiology, Sixth Medical Center, PLA General Hospital, Beijing, China
| | - Shujing Zhou
- Department of Data Science and Visualization, Faculty of Informatics, University of Debrecen, Debrecen, Hungary
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Longbin Wang
- Department of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Xu
- Department of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xufeng Huang
- Department of Data Science and Visualization, Faculty of Informatics, University of Debrecen, Debrecen, Hungary
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - Andras Hajdu
- Department of Data Science and Visualization, Faculty of Informatics, University of Debrecen, Debrecen, Hungary
- *Correspondence: Andras Hajdu,
| | - Ling Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
- Ling Zhang,
| |
Collapse
|
32
|
Ning J, Sun K, Wang X, Fan X, Jia K, Cui J, Ma C. Use of machine learning-based integration to develop a monocyte differentiation-related signature for improving prognosis in patients with sepsis. Mol Med 2023; 29:37. [PMID: 36941583 PMCID: PMC10029317 DOI: 10.1186/s10020-023-00634-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/13/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Although significant advances have been made in intensive care medicine and antibacterial treatment, sepsis is still a common disease with high mortality. The condition of sepsis patients changes rapidly, and each hour of delay in the administration of appropriate antibiotic treatment can lead to a 4-7% increase in fatality. Therefore, early diagnosis and intervention may help improve the prognosis of patients with sepsis. METHODS We obtained single-cell sequencing data from 12 patients. This included 14,622 cells from four patients with bacterial infectious sepsis and eight patients with sepsis admitted to the ICU for other various reasons. Monocyte differentiation trajectories were analyzed using the "monocle" software, and differentiation-related genes were identified. Based on the expression of differentiation-related genes, 99 machine-learning combinations of prognostic signatures were obtained, and risk scores were calculated for all patients. The "scissor" software was used to associate high-risk and low-risk patients with individual cells. The "cellchat" software was used to demonstrate the regulatory relationships between high-risk and low-risk cells in a cellular communication network. The diagnostic value and prognostic predictive value of Enah/Vasp-like (EVL) were determined. Clinical validation of the results was performed with 40 samples. The "CBNplot" software based on Bayesian network inference was used to construct EVL regulatory networks. RESULTS We systematically analyzed three cell states during monocyte differentiation. The differential analysis identified 166 monocyte differentiation-related genes. Among the 99 machine-learning combinations of prognostic signatures constructed, the Lasso + CoxBoost signature with 17 genes showed the best prognostic prediction performance. The highest percentage of high-risk cells was found in state one. Cell communication analysis demonstrated regulatory networks between high-risk and low-risk cell subpopulations and other immune cells. We then determined the diagnostic and prognostic value of EVL stabilization in multiple external datasets. Experiments with clinical samples demonstrated the accuracy of this analysis. Finally, Bayesian network inference revealed potential network mechanisms of EVL regulation. CONCLUSIONS Monocyte differentiation-related prognostic signatures based on the Lasso + CoxBoost combination were able to accurately predict the prognostic status of patients with sepsis. In addition, low EVL expression was associated with poor prognosis in sepsis.
Collapse
Affiliation(s)
- Jingyuan Ning
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Keran Sun
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xuan Wang
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China
- Department of Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiaoqing Fan
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Keqi Jia
- Department of Pathology, Shijiazhuang People's Hospital, Shijiazhuang, People's Republic of China
| | - Jinlei Cui
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Cuiqing Ma
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
33
|
Single-cell Sequence Analysis Combined with Multiple Machine Learning to Identify Markers in Sepsis Patients: LILRA5. Inflammation 2023:10.1007/s10753-023-01803-8. [PMID: 36920635 DOI: 10.1007/s10753-023-01803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023]
Abstract
Sepsis is a disease with a very high mortality rate, mainly involving an immune-dysregulated response due to bacterial infection. Most studies are currently limited to the whole blood transcriptome level; however, at the single cell level, there is still a great deal unknown about specific cell subsets and disease markers. We obtained 29 peripheral blood single-cell sequencing data, including 66,283 cells from 10 confirmed samples of sepsis infection and 19 healthy samples. Cells related to the sepsis phenotype were identified and characterized by the "scissor" method. The regulatory relationships of sepsis-related phenotype cells in the cellular communication network were clarified using the "cell chat" method. The least absolute shrinkage and selection operator (LASSO), support vector machine (SVM), and random forest (RF) were used to identify sepsis signature genes of diagnostic value. External validation was performed using multiple datasets from the GEO database (GSE28750, GSE185263, GSE57065) and 40 clinical samples. Bayesian algorithm was used to calculate the regulatory network of LILRA5 co-expressed genes. The stability of atenolol-targeting LILRA5 was determined by molecular docking techniques. Ultimately, action trajectory and survival analyses demonstrate the effectiveness of atenolol-targeted LILRA5 in treating patients with sepsis. We successfully identified 1215 healthy phenotypic cells and 462 sepsis phenotypic cells. We focused on 447 monocytes of the sepsis phenotype. Among the cellular communications, there were a large number of differences between these cells and other immune cells showing a significant inflammatory phenotype compared to the healthy phenotypic cells. Together, the three machine learning algorithms identified the LILRA5 marker gene in sepsis patients, and validation results from multiple external datasets as well as real-world clinical samples demonstrated the robust diagnostic performance of LILRA5. The AUC values of LILRA5 in the external datasets GSE28750, GSE185263, and GSE57065 could reach 0.875, 0.940, and 0.980, in that order. Bayesian networks identified a large number of unknown regulatory relationships for LILRA5 co-expression. Molecular docking results demonstrated the possibility of atenolol targeting LILRA5 for the treatment of sepsis. Behavioral trajectory analysis and survival analysis demonstrate that atenolol has a desirable therapeutic effect. LILRA5 is a marker gene in sepsis patients, and atenolol can stably target LILRA5.
Collapse
|
34
|
Chang W, Li H, Ou W, Wang SY. A novel zinc metabolism-related gene signature to predict prognosis and immunotherapy response in lung adenocarcinoma. Front Immunol 2023; 14:1147528. [PMID: 37033934 PMCID: PMC10079938 DOI: 10.3389/fimmu.2023.1147528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Background Zinc is a key mineral element in regulating cell growth, development, and immune system. We constructed the zinc metabolism-related gene signature to predict prognosis and immunotherapy response for lung adenocarcinoma (LUAD). Methods Zinc metabolism-associated gene sets were obtained from Molecular Signature Database. Then, the zinc metabolism-related gene signature (ZMRGS) was constructed and validated. After combining with clinical characteristics, the nomogram for practical application was constructed. The differences in biological pathways, immune molecules, and tumor microenvironment (TME) between the different groups were analyzed. Tumor Immune Dysfunction and Exclusion algorithm (TIDE) and two immunotherapy datasets were used to evaluate the immunotherapy response. Results The signature was constructed according to six key zinc metabolism-related genes, which can well predict the prognosis of LUAD patients. The nomogram also showed excellent prediction performance. Functional analysis showed that the low-risk group was in the status of immune activation. More importantly, the lower risk score of LUAD patients showed a higher response rate to immunotherapy. Conclusion The state of zinc metabolism is closely connected to prognosis, tumor microenvironment, and response to immunotherapy. The zinc metabolism-related signature can well evaluate the prognosis and immunotherapy response for LUAD patients.
Collapse
Affiliation(s)
- Wuguang Chang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hongmu Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wei Ou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- *Correspondence: Si-Yu Wang, ; Wei Ou,
| | - Si-Yu Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- *Correspondence: Si-Yu Wang, ; Wei Ou,
| |
Collapse
|
35
|
Ye Q, Guo NL. Inferencing Bulk Tumor and Single-Cell Multi-Omics Regulatory Networks for Discovery of Biomarkers and Therapeutic Targets. Cells 2022; 12:101. [PMID: 36611894 PMCID: PMC9818242 DOI: 10.3390/cells12010101] [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: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
There are insufficient accurate biomarkers and effective therapeutic targets in current cancer treatment. Multi-omics regulatory networks in patient bulk tumors and single cells can shed light on molecular disease mechanisms. Integration of multi-omics data with large-scale patient electronic medical records (EMRs) can lead to the discovery of biomarkers and therapeutic targets. In this review, multi-omics data harmonization methods were introduced, and common approaches to molecular network inference were summarized. Our Prediction Logic Boolean Implication Networks (PLBINs) have advantages over other methods in constructing genome-scale multi-omics networks in bulk tumors and single cells in terms of computational efficiency, scalability, and accuracy. Based on the constructed multi-modal regulatory networks, graph theory network centrality metrics can be used in the prioritization of candidates for discovering biomarkers and therapeutic targets. Our approach to integrating multi-omics profiles in a patient cohort with large-scale patient EMRs such as the SEER-Medicare cancer registry combined with extensive external validation can identify potential biomarkers applicable in large patient populations. These methodologies form a conceptually innovative framework to analyze various available information from research laboratories and healthcare systems, accelerating the discovery of biomarkers and therapeutic targets to ultimately improve cancer patient survival outcomes.
Collapse
Affiliation(s)
- Qing Ye
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Nancy Lan Guo
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
36
|
Zhu Y, Yang X, Zu Y. Integrated analysis of WGCNA and machine learning identified diagnostic biomarkers in dilated cardiomyopathy with heart failure. Front Cell Dev Biol 2022; 10:1089915. [PMID: 36544902 PMCID: PMC9760806 DOI: 10.3389/fcell.2022.1089915] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/23/2022] [Indexed: 12/08/2022] Open
Abstract
The etiologies and pathogenesis of dilated cardiomyopathy (DCM) with heart failure (HF) remain to be defined. Thus, exploring specific diagnosis biomarkers and mechanisms is urgently needed to improve this situation. In this study, three gene expression profiling datasets (GSE29819, GSE21610, GSE17800) and one single-cell RNA sequencing dataset (GSE95140) were obtained from the Gene Expression Omnibus (GEO) database. GSE29819 and GSE21610 were combined into the training group, while GSE17800 was the test group. We used the weighted gene co-expression network analysis (WGCNA) and identified fifteen driver genes highly associated with DCM with HF in the module. We performed the least absolute shrinkage and selection operator (LASSO) on the driver genes and then constructed five machine learning classifiers (random forest, gradient boosting machine, neural network, eXtreme gradient boosting, and support vector machine). Random forest was the best-performing classifier established on five Lasso-selected genes, which was utilized to select out NPPA, OMD, and PRELP for diagnosing DCM with HF. Moreover, we observed the up-regulation mRNA levels and robust diagnostic accuracies of NPPA, OMD, and PRELP in the training group and test group. Single-cell RNA-seq analysis further demonstrated their stable up-regulation expression patterns in various cardiomyocytes of DCM patients. Besides, through gene set enrichment analysis (GSEA), we found TGF-β signaling pathway, correlated with NPPA, OMD, and PRELP, was the underlying mechanism of DCM with HF. Overall, our study revealed NPPA, OMD, and PRELP serving as diagnostic biomarkers for DCM with HF, deepening the understanding of its pathogenesis.
Collapse
Affiliation(s)
- Yihao Zhu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Xiaojing Yang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Yao Zu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China,Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China,*Correspondence: Yao Zu,
| |
Collapse
|
37
|
Feng T, Wu T, Zhang Y, Zhou L, Liu S, Li L, Li M, Hu E, Wang Q, Fu X, Zhan L, Xie Z, Xie W, Huang X, Shang X, Yu G. Stemness Analysis Uncovers That The Peroxisome Proliferator-Activated Receptor Signaling Pathway Can Mediate Fatty Acid Homeostasis In Sorafenib-Resistant Hepatocellular Carcinoma Cells. Front Oncol 2022; 12:912694. [PMID: 35957896 PMCID: PMC9361019 DOI: 10.3389/fonc.2022.912694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) stem cells are regarded as an important part of individualized HCC treatment and sorafenib resistance. However, there is lacking systematic assessment of stem-like indices and associations with a response of sorafenib in HCC. Our study thus aimed to evaluate the status of tumor dedifferentiation for HCC and further identify the regulatory mechanisms under the condition of resistance to sorafenib. Datasets of HCC, including messenger RNAs (mRNAs) expression, somatic mutation, and clinical information were collected. The mRNA expression-based stemness index (mRNAsi), which can represent degrees of dedifferentiation of HCC samples, was calculated to predict drug response of sorafenib therapy and prognosis. Next, unsupervised cluster analysis was conducted to distinguish mRNAsi-based subgroups, and gene/geneset functional enrichment analysis was employed to identify key sorafenib resistance-related pathways. In addition, we analyzed and confirmed the regulation of key genes discovered in this study by combining other omics data. Finally, Luciferase reporter assays were performed to validate their regulation. Our study demonstrated that the stemness index obtained from transcriptomic is a promising biomarker to predict the response of sorafenib therapy and the prognosis in HCC. We revealed the peroxisome proliferator-activated receptor signaling pathway (the PPAR signaling pathway), related to fatty acid biosynthesis, that was a potential sorafenib resistance pathway that had not been reported before. By analyzing the core regulatory genes of the PPAR signaling pathway, we identified four candidate target genes, retinoid X receptor beta (RXRB), nuclear receptor subfamily 1 group H member 3 (NR1H3), cytochrome P450 family 8 subfamily B member 1 (CYP8B1) and stearoyl-CoA desaturase (SCD), as a signature to distinguish the response of sorafenib. We proposed and validated that the RXRB and NR1H3 could directly regulate NR1H3 and SCD, respectively. Our results suggest that the combined use of SCD inhibitors and sorafenib may be a promising therapeutic approach.
Collapse
Affiliation(s)
- Tingze Feng
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tianzhi Wu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanxia Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lang Zhou
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shanshan Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Country Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ming Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Erqiang Hu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qianwen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaocong Fu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Li Zhan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zijing Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenqin Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xianying Huang
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Xianying Huang, ; Xuan Shang, ; Guangchuang Yu,
| | - Xuan Shang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Xianying Huang, ; Xuan Shang, ; Guangchuang Yu,
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Xianying Huang, ; Xuan Shang, ; Guangchuang Yu,
| |
Collapse
|