1
|
Jiang W, Jiang L, Zhao X, Liu Y, Sun H, Zhou X, Liu Y, Huang S. Bioinformatics Analysis Reveals HIST1H2BH as a Novel Diagnostic Biomarker for Atrial Fibrillation-Related Cardiogenic Thromboembolic Stroke. Mol Biotechnol 2025; 67:2111-2126. [PMID: 38825608 DOI: 10.1007/s12033-024-01187-6] [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: 10/27/2023] [Accepted: 04/29/2024] [Indexed: 06/04/2024]
Abstract
Atrial fibrillation (AF) is a significant precursor to cerebral embolism. Our study sought to unearth new diagnostic biomarkers for atrial fibrillation-related cerebral embolism (AF-CE) by meticulously examining multiple GEO datasets and meta-analysis. The gene expression omnibus (GEO) database provided RNA sequencing data associated with AF and stroke. We began by pinpointing genes with varied expressions in AF-CE patient blood samples. A meta-analysis was subsequently undertaken using several RNA sequencing datasets to verify these genes. LASSO regression discerned key genes for AF-CE, with their diagnostic prowess verified through ROC curve examination. Active signaling pathways within stroke patients were discerned via GO and KEGG enrichment, with PPI interactions detailing gene interplay. Differential gene analysis revealed an upregulation of sixteen genes and a downregulation of four in stroke patient blood samples. Eight genes showcased varied expression in the meta-analysis. LASSO regression zeroed in on five of these, culminating in HIST1H2BH's identification as a characteristic gene. HIST1H2BH's prowess in predicting AF-CE was confirmed through ROC. Integrin signaling, platelet activation, ECM interactions, and the PI3K-Akt pathway were found active in stroke victims. HIST1H2BH's interaction with the notably upregulated ITGA2B was spotlighted by PPI. Additionally, HIST1H2BH exhibited links with NK cells and eosinophils. HIST1H2BH emerges as an insightful diagnostic beacon for AF-CE. Its presence, post AF, potentially modulates pathways, accentuating platelet activation and consequent thrombus generation, leading to cerebral embolism.
Collapse
Affiliation(s)
- Wenbing Jiang
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China.
| | - Lelin Jiang
- Second Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Xiaoli Zhao
- Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Yiying Liu
- Postgraduate Training Base Allianceof Wenzhou Medical University (Wenzhou Central Hosptial), Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Huanghui Sun
- The Dingli Clinical College of Wenzhou Medical University, Heart Function Examination Room, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Xinlang Zhou
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China
| | - Yin Liu
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China
| | - Shu'se Huang
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China
| |
Collapse
|
2
|
Li Y, Zeng R, Huang Y, Zhuo Y, Huang J. Integrating machine learning and single-cell sequencing to identify shared biomarkers in type 1 diabetes mellitus and clear cell renal cell carcinoma. Front Oncol 2025; 15:1543806. [PMID: 40098701 PMCID: PMC11911197 DOI: 10.3389/fonc.2025.1543806] [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: 12/11/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose Type 1 diabetes mellitus (T1DM), as an autoimmune disease, can increase susceptibility to clear cell renal cell carcinoma (ccRCC) due to its proinflammatory effects. ccRCC is characterized by its subtle onset and unfavorable prognosis. Thus, the aim of this study was to highlight prevention and early detection opportunities in high-risk populations by identifying common biomarkers for T1DM and ccRCC. Methods Based on multiple publicly available datasets, WGCNA was applied to identify gene modules closely associated with T1DM, which were then integrated with prognostic DEGs in ccRCC. Subsequently, the LASSO and SVM algorithms were employed to identify shared hub genes between the two diseases. Additionally, clinical samples were used to validate the expression patterns of these hub genes, and scRNA-seq data were utilized to analyze the cell types expressing these genes and to explore potential mechanisms of cell communication. Results Overall, three hub genes (KIF21A, PIGH, and RPS6KA2) were identified as shared biomarkers for TIDM and ccRCC. Analysis of clinical samples and multiple datasets revealed that KIF21A and PIGH were significantly downregulated and that PIG was upregulated in the disease group. KIF21A and PIGH are mainly expressed in NK and T cells, PRS6KA2 is mainly expressed in endothelial and epithelial cells, and the MIF signaling pathway may be related to hub genes. Conclusion Our results demonstrated the pivotal roles of hub genes in T1DM and ccRCC. These genes hold promise as novel biomarkers, offering potential avenues for preventive strategies and the development of new precision treatment modalities.
Collapse
Affiliation(s)
- Yi Li
- Department of Ultrasound, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Rui Zeng
- Department of Pathology, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Yuhua Huang
- Department of Ultrasound, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yumin Zhuo
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jun Huang
- Department of Ultrasound, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Hou C, Xu J, Zhou M, Huo J, Wang X, Jiang W, Su T, Wang H, Jia F. Screening of biomarkers for diagnosing chronic kidney disease and heart failure with preserved ejection fraction through bioinformatics analysis. Biochem Biophys Rep 2025; 41:101911. [PMID: 39877037 PMCID: PMC11773089 DOI: 10.1016/j.bbrep.2024.101911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Background Previous research has established that chronic kidney disease (CKD) and heart failure with preserved ejection fraction (HFpEF) often coexist. Although we have a preliminary understanding of the potential correlation between HFpEF and CKD, the underlying pathophysiological mechanisms remain unclear. This study aimed to elucidate the molecular mechanisms associated with CKD and HFpEF through bioinformatics analysis. Methods Datasets for HFpEF and CKD were obtained from the Gene Expression Omnibus (GEO) database. The R software package "limma" was employed to conduct differential expression analysis. Functional annotation was performed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO). We conducted weighted gene co-expression network analysis (WGCNA), correlation analysis with autophagy, ferroptosis, and immune-related processes, as well as transcriptional regulation analysis, immune infiltration analysis, and diagnostic performance evaluation. Finally, the diagnostic potential of the identified hub genes for CKD and HFpEF was assessed using ROC curve analysis (GSE37171). Results Differential expression analysis revealed 58 overlapping genes, comprised of 40 up-regulated and 18 down-regulated genes. Both GO and KEGG analyses indicated enriched pathways relevant to both disorders. WGCNA identified 4086 genes associated with CKD. Further comparison with differentially expressed genes (DEGs) identified three hub genes (KLF4, SCD, and SEL1L3) that were linked to autophagy, ferroptosis, and immune processes in both conditions. Additionally, a miRNA-mRNA regulatory network involving 376 miRNAs and 12 transcription factors (TFs) was constructed. ROC curve analysis was performed to evaluate the diagnostic utility of the hub genes for CKD and HFpEF. Conclusion This study elucidated shared pathogenic mechanisms and identified diagnostic markers common to both HFpEF and CKD. The identified hub genes show promise as potential tools for early diagnosis and treatment strategies for these conditions.
Collapse
Affiliation(s)
- Can Hou
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| | - Jiayi Xu
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| | - Min Zhou
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| | - Junyu Huo
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| | - Xiaofei Wang
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| | - Wanying Jiang
- Department of Cardiology, Changzhou Hospital of Traditional Chinese Medicine, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, 213000, Changzhou, Jiangsu Province, China
| | - Tong Su
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| | - Hui Wang
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| | - Fang Jia
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000, Changzhou, Jiangsu Province, China
| |
Collapse
|
4
|
Li J, He Q, Zheng Z, Liu C, Zhang B, Mou S, Zeng C, Sun W, Liu W, Ge P, Zhang D, Zhao J. Comprehensive Analysis and In Vitro Verification of Endothelial-Mesenchymal Transition-Related Genes in Moyamoya Disease. Mol Neurobiol 2025; 62:2515-2529. [PMID: 39134827 DOI: 10.1007/s12035-024-04423-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/06/2024] [Indexed: 01/28/2025]
Abstract
Moyamoya disease (MMD) is a rare, chronic, and progressive cerebrovascular disorder with unclear underlying causes and mechanisms. Previous studies suggest a potential involvement of endothelial-mesenchymal transition (EndMT) in the pathogenesis of MMD. This study aimed to explore the contribution of EndMT-related genes (ERGs) in MMD. Two datasets, GSE141022 and GSE157628, were integrated as the training set after batch effects removal. Differentially expressed ERGs were identified between MMD and control groups. Functional enrichment analysis and immune infiltration analysis were further performed. LASSO regression was used for hub MMD-related ERG selection. Consensus clustering was used for MMD subtype classification based on these hub MMD-related ERGs. Molecular characteristics between MMD subtypes were analyzed using WGCNA. PPI network was used to illuminate the genetic relationship. The hub MMD-related ERGs were validated in an independent testing set, GSE189993. The nomogram model was constructed and evaluated using ROC curves and calibration plots. Additionally, CCK-8, EdU, wound healing, and western blot were performed to confirm the function of the hub MMD-related ERGs. A total of 107 DE-ERGs were identified. Functional enrichment analysis showed these genes were associated with EndMT and immune response. The infiltrating levels of immune cells were commonly higher in the MMD group. LASSO regression identified 12 hub MMD-related ERGs, leading to the identification of two MMD subtypes. Four ERGs emerged as the final hub MMD-related ERGs after validation in the testing set, including CCL21, CEBPA, KRT18, and TNFRSF11A. The nomogram model exhibited excellent discrimination ability. In vitro experiments showed that CCL21, CEBPA, KRT18, and TNFRSF11A could promote proliferation, migration, and EndMT. This study investigated the potential role of EndMT in MMD and identified four hub MMD-related ERGs, providing potential therapeutic targets for MMD treatment.
Collapse
Affiliation(s)
- Junsheng Li
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhiyao Zheng
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Bojian Zhang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Siqi Mou
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chaofan Zeng
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Sun
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Liu
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peicong Ge
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Dong Zhang
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
- Department of Neurosurgery, Dongcheng District, Beijing Hospital, National Center of Gerontology, No. 1 Da Hua Road, Dong Dan, Beijing, 100730, China.
| | - Jizong Zhao
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 Nan Si Huan Xi Road, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
5
|
Ying H, Kong W, Xu X. Integrated Network Pharmacology, Machine Learning and Experimental Validation to Identify the Key Targets and Compounds of TiaoShenGongJian for the Treatment of Breast Cancer. Onco Targets Ther 2025; 18:49-71. [PMID: 39835272 PMCID: PMC11745062 DOI: 10.2147/ott.s486300] [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: 08/29/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025] Open
Abstract
Background TiaoShenGongJian (TSGJ) decoction, a traditional Chinese medicine for breast cancer, has unknown active compounds, targets, and mechanisms. This study identifies TSGJ's key targets and compounds for breast cancer treatment through network pharmacology, machine learning, and experimental validation. Methods Bioactive components and targets of TSGJ were identified from the TCMSP database, and breast cancer-related targets from GeneCards, PharmGkb, and RNA-seq datasets. Intersection of these targets revealed therapeutic targets of TSGJ. PPI analysis was performed via STRING, and machine learning methods (SVM, RF, GLM, XGBoost) identified key targets, validated by GSE70905, GSE70947, GSE22820, and TCGA-BRCA datasets. Pathway analyses and molecular docking were performed. TSGJ and core compounds' effectiveness was confirmed by MTT and RT-qPCR assays. Results 160 common targets of TSGJ were identified, with 30 hub targets from PPI analysis. Five predictive targets (HIF1A, CASP8, FOS, EGFR, PPARG) were screened via SVM. Their diagnostic, biomarker, immune, and clinical values were validated. Quercetin, luteolin, and baicalein were identified as core components. Molecular docking confirmed their strong affinities with predicted targets. These compounds modulated key targets and induced cytotoxicity in breast cancer cell lines in a similar way as TSGJ. Conclusion This study reveals the main active components and targets of TSGJ against breast cancer, supporting its potential for breast cancer prevention and treatment.
Collapse
Affiliation(s)
- Huiyan Ying
- Institute for Molecular Medicine Finland (FIMM), Hilife, University of Helsinki, Helsinki, Finland
| | - Weikaixin Kong
- Institute for Molecular Medicine Finland (FIMM), Hilife, University of Helsinki, Helsinki, Finland
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, People’s Republic of China
| | - Xiangwei Xu
- Affiliated Yongkang First People’s Hospital and School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
6
|
Yang QQ, Guo JA, Zhang K, Li SH, Xia WY, Wang DX, Xie LS, Wang JM, Wu QF. Disulfidptosis and Its Hub Gene Slc3a2 Involved in Ulcerative Colitis Pathogenesis, Disease Progression, and Patient Responses to Biologic Therapies. Int J Mol Sci 2024; 25:13506. [PMID: 39769269 PMCID: PMC11728241 DOI: 10.3390/ijms252413506] [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: 11/05/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
To analyze the role of disulfidptosis in ulcerative colitis (UC), large-scale datasets combined with weighted gene co-expression network analysis (WGCNA) and machine learning were utilized and analyzed. When the hub genes that are associated with UC disease phenotypes and have predictive performance were identified, immune cell infiltration and the CeRNA network were constructed, the role of hub genes in UC pathogenies and biotherapy were investigated, and molecular docking studies and mice-verified tests were carried out to further explore the potential core genes and potential target. Finally, we found 21 DRGs involved in UC pathogenesis, including SLC3A2, FLNA, CAPZB, TLN1, RPN1, etc. Moreover, SLC3A2, TLN1, and RPN1 show a notable correlation with UC inflammatory state, and the expression of DRGs is closely related to the response to UC biotherapy. Our study suggests that disulfidptosis plays a crucial role in the pathogenesis and disease progression of UC. Higher expression of DRGs is commonly observed in moderate to severe UC patients, which may also affect their response to biologic therapies. Among the identified genes, SLC3A2 stands out, providing new insights into the underlying mechanisms of UC and potentially serving as a novel therapeutic target for the treatment of UC.
Collapse
Affiliation(s)
- Qing-Qing Yang
- Acupuncture and Moxibustion School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Jun-An Guo
- Acupuncture and Moxibustion School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Ke Zhang
- Acupuncture and Moxibustion School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Si-Hui Li
- Acupuncture and Moxibustion School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Wan-Yu Xia
- Acupuncture and Moxibustion School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - De-Xian Wang
- College of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Lu-Shuang Xie
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Jun-Meng Wang
- Acupuncture and Moxibustion School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of Traditional Chinese Medicine), Ministry of Education, Chengdu 610075, China
- Institute of Acupuncture and Homeostasis Regulation, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Qiao-Feng Wu
- Acupuncture and Moxibustion School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of Traditional Chinese Medicine), Ministry of Education, Chengdu 610075, China
- Institute of Acupuncture and Homeostasis Regulation, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
7
|
Liu W, Pan Y. Unraveling the mechanisms underlying diabetic cataracts: insights from Mendelian randomization analysis. Redox Rep 2024; 29:2420563. [PMID: 39639475 PMCID: PMC11626871 DOI: 10.1080/13510002.2024.2420563] [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: 12/07/2024] Open
Abstract
BACKGROUND Diabetic cataract (DC) is a major cause of blindness, with its pathogenesis involving oxidative stress and ferroptosis, according to recent studies. METHODS We performed a Mendelian Randomization (MR) study using GWAS data to select SNPs and assess the causal link between diabetes and cataracts. DC datasets were analyzed for differential gene expression, WGCNA, and protein-protein interactions to identify key oxidative stress and ferroptosis genes. An SVM-RFE algorithm developed a diagnostic model, and ImmuCellAI analyzed immune infiltration patterns. RESULTS MR analysis confirmed diabetes as a cataract risk factor and identified core genes related to oxidative stress and ferroptosis in DC. Four key genes (Hspa5/Nfe2l2/Atf3/Stat3) linked to both processes were discovered. Immune infiltration analysis revealed an imbalance associated with these genes. CONCLUSIONS A functional interaction between oxidative stress and ferroptosis genes in DC is suggested, with a 4-gene model, indicating their potential as a 'bridge' in DC pathogenesis.
Collapse
Affiliation(s)
- Wenlan Liu
- College of Medical Technology, Xi'an Medical University, Xi'an, People’s Republic of China
| | - Yiming Pan
- College of Medical Technology, Xi'an Medical University, Xi'an, People’s Republic of China
| |
Collapse
|
8
|
Lan Y, Peng Q, Shen J, Liu H. Elucidating common biomarkers and pathways of osteoporosis and aortic valve calcification: insights into new therapeutic targets. Sci Rep 2024; 14:27827. [PMID: 39537712 PMCID: PMC11560947 DOI: 10.1038/s41598-024-78707-6] [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: 06/30/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Osteoporosis and aortic valve calcification, prevalent in the elderly, have unclear common mechanisms. This study aims to uncover them through bioinformatics analysis. METHODS Microarray data from GEO was analyzed for osteoporosis and aortic valve calcification. Differential expression analysis identified co-expressed genes. SVM-RFE and random forest selected key genes. GO and KEGG enrichment analyses were performed. Immunoinfiltration and GSEA analyses were subsequently performed. NetworkAnalyst analyzed microRNAs/TFs. HERB predicted drugs, and molecular docking assessed targeting potential. RESULTS Thirteen genes linked to osteoporosis and aortic valve calcification were identified. TNFSF11, KYNU, and HLA-DMB emerged as key genes. miRNAs, TFs, and drug predictions offered therapeutic insights. Molecular docking suggested 17-beta-estradiol and vitamin D3 as potential treatments. CONCLUSION The study clarifies shared mechanisms of osteoporosis and aortic valve calcification, identifies biomarkers, and highlights TNFSF11, KYNU, and HLA-DMB. It also suggests 17-beta-estradiol and vitamin D3 as potential effective treatments.
Collapse
Affiliation(s)
- Yujian Lan
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qingping Peng
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jianlin Shen
- Department of Orthopaedics, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
- Central Laboratory, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
| | - Huan Liu
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
9
|
Zhang X, Liu C, Cao L, Tang H, Jiang H, Hu C, Dong X, Zhou F, Qin K, Liu Q, Shen J, Zhou Y. Exploring the mechanisms of chronic obstructive pulmonary disease and Crohn's disease: a bioinformatics-based study. Sci Rep 2024; 14:27461. [PMID: 39523420 PMCID: PMC11551177 DOI: 10.1038/s41598-024-78697-5] [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: 09/04/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
This study explored the comorbid mechanisms between Crohn's disease (CD) and chronic obstructive pulmonary disease (COPD) using bioinformatics analysis. From the Gene Expression Omnibus (GEO) microarray dataset, 349 common differentially expressed genes (coDEGs) were identified, and 8 shared hub genes were found: CCL2, CXCL1, TLR2, ICAM1, PTPRC, ITGAX, PTGS2, and MMP9, which were vital for immune function and regulation of inflammatory responses. In addition, the study also analyzed the association between coDEGs and immune cell infiltration using the single-sample gene set enrichment algorithm (ssGSEA). Potential drugs related to these genes were identified using the connectivity map (CMap). These findings provided new perspectives for understanding the interaction between CD and COPD.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Caiping Liu
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Luqian Cao
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Hongguang Tang
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Haiyun Jiang
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Changjing Hu
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Xuehong Dong
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Feiyang Zhou
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Kunming Qin
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Qiang Liu
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Jinyang Shen
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China.
| | - Yue Zhou
- Department of Pharmacy, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang, 222004, Jiangsu, China.
| |
Collapse
|
10
|
Hu C, Ge M, Liu Y, Tan W, Zhang Y, Zou M, Xiang L, Song X, Guo H. From inflammation to depression: key biomarkers for IBD-related major depressive disorder. J Transl Med 2024; 22:997. [PMID: 39501335 PMCID: PMC11536793 DOI: 10.1186/s12967-024-05758-8] [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: 07/14/2024] [Accepted: 10/08/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic, inflammatory, and autoimmune disorder, and its incidence of comorbid with major depressive disorder (MDD) is significantly higher than the general population. However, many patients lack proper recognition and necessary psychological health treatments. We aimed to identify potential biomarkers and mechanisms involved in the development of IBD comorbid with MDD (IBD-MDD). METHODS We utilized IBD and MDD-related datasets from the GEO database for differential gene expression analysis, protein-protein interaction (PPI) and pathway enrichment analysis, random forest algorithm, LASSO regression analysis, and construction of a disease prediction model. We assessed the accuracy of the model using ROC curve, explored potential mechanisms through immune infiltration analysis, and validated candidate biomarkers using peripheral blood samples from patients in our center's cohort. RESULTS We identified 484 IBD-related secreted proteins and 142 key module genes associated with MDD. PPI analysis revealed two crucial modules primarily involved in inflammation and immune regulation. We identified four diagnostic genes (HGF, SPARC, ADAM12, and MMP8) from the 21 shared genes between IBD-related secreted proteins and MDD key module genes, constructed a nomogram model and confirmed its accuracy using ROC curve from an external independent dataset. Immune infiltration analysis revealed significant associations between the four diagnostic genes, and cellular immune dysregulation in MDD. Finally, we validated the expression patterns of the four diagnostic genes in our cohort. CONCLUSIONS Our study discovered four candidate biomarkers for IBD-MDD, providing new insights for the diagnosis and therapeutic intervention of serum-based IBD comorbid with MDD.
Collapse
Affiliation(s)
- Chaoqun Hu
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Mei Ge
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yan Liu
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Wei Tan
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yingzhi Zhang
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Zou
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Lingya Xiang
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xiaomei Song
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China.
| | - Hong Guo
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China.
| |
Collapse
|
11
|
Zhang WT, Ge HW, Wei Y, Gao JL, Tian F, Zhou EC. Molecular characterization of PANoptosis-related genes in chronic kidney disease. PLoS One 2024; 19:e0312696. [PMID: 39466748 PMCID: PMC11515967 DOI: 10.1371/journal.pone.0312696] [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: 11/23/2023] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Chronic kidney disease (CKD) is characterized by fibrosis and inflammation in renal tissues. Several types of cell death have been implicated in CKD onset and progression. Unlike traditional forms of cell death, PANoptosis is characterized by the crosstalk among programmed cell death pathways. However, the interaction between PANoptosis and CKD remains unclear. Here, we used bioinformatics methods to identify differentially expressed genes and differentially expressed PANoptosis-related genes (DE-PRGs) using data from the GSE37171 dataset. Following this, we further performed gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and gene set enrichment analysis using the data. We adopted a combined approach to select hub genes, using the STRING database and CytoHubba plug-in, and we used the GSE66494 as a validation dataset. In addition, we constructed ceRNA, transcription factor (TF)-gene, and drug-gene networks using Cytoscape. Lastly, we conducted immunohistochemical analysis and western blotting to validate the hub genes. We identified 57 PANoptosis-associated genes as DE-PRGs. We screened nine hub genes from the 57 DE-PRGs. We identified two hub genes (FOS and PTGS2) using the GSE66494 database, Nephroseq, immunohistochemistry, and western blotting. A common miRNA (Hsa-miR-101-3p) and three TFs (CREB1, E2F1, and RELA) may play a crucial role in the onset and progression of PANoptosis-related CKD. In our analysis of the drug-gene network, we identified eight drugs targeting FOS and 52 drugs targeting PTGS2.
Collapse
Affiliation(s)
- Wen-tao Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong-wei Ge
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-lin Gao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Tian
- Research Center of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - En-chao Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
12
|
Wen D, Li B, Guo S, Chen L, Chen B. Exploring Pathogenic Genes in Frozen Shoulder through weighted gene co-expression network analysis and Mendelian Randomization. Int J Med Sci 2024; 21:2745-2758. [PMID: 39512681 PMCID: PMC11539380 DOI: 10.7150/ijms.98505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Background: Frozen shoulder (FS) is characterized by the thickening and fibrosis of the joint capsule, leading to joint contracture and a reduction in joint volume. The precise etiology responsible for these pathological changes remains elusive. Therefore, the primary aim of this study was to explore the potential involvement of pathogenic genes in FS and analyze their underlying roles in the disease progression. Methods: Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were employed to investigate co-expressed genes potentially associated with FS. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were conducted to elucidate the potential roles of these co-expressed genes. Subsequently, Mendelian randomization (MR) analysis was performed using expression quantitative trait loci datasets for the co-expressed genes, combined with summary statistics from the genome-wide association study of FS, aiming to identify key genes causally associated with FS. The identified key genes were further validated through reverse transcription-quantitative PCR (RT-qPCR). Additionally, a nomogram model and receiver operating characteristic (ROC) curves were established to assess the diagnostic value of the hub genes. Furthermore, the infiltration of immune cells was evaluated using the CIBERSORT algorithm and the relationship between key genes and immune-infiltrating cells was analyzed. Results: 295 overlapping co-expressed genes were identified by intersecting the differentially expressed genes with the hub genes obtained from associated modules identified through WGCNA. Utilizing MR analysis, four key genes, namely ADAMTS1, NR4A2, PARD6G and SMKR1, were found to exhibit positive causal relationships with FS, which were subsequently validated through RT-qPCR analysis. Moreover, the diagnostic value of these four key genes was demonstrated through the development of a nomogram model and the construction of ROC curves. Notably, a causal relationship between ADAMTS1 and immune cell infiltration in FS was observed. Conclusion: Our study suggested genetic predisposition to higher expression levels of ADAMTS1, NR4A2, PARD6G and SMKR1, was associated with an increased risk of FS. Further investigations elucidating the functional roles of these genes will enhance our understanding of the pathogenesis of FS and may facilitate the development of targeted treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Biao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
13
|
Zhang C, Wu Z, Song Y, Jin X, Hu J, Huang C, Zhou J, Lian J. Exploring diagnostic biomarkers of type 2 cardio-renal syndrome based on secreted proteins and bioinformatics analysis. Sci Rep 2024; 14:24612. [PMID: 39427047 PMCID: PMC11490509 DOI: 10.1038/s41598-024-75580-1] [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: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Chronic heart failure (CHF) can induce chronic kidney disease (CKD), called type 2 cardio-renal syndrome (CRS2). The mechanism is not completely clear, and there is a lack of early warning biomarkers of CKD in the context of CHF. Two CKD, one CHF-PBMC and four CHF-cardiac tissue expression profile datasets were obtained from GEO database. Differential expression analysis and WGCNA were used to detect CKD key genes and CHF-related secreted proteins. Protein-protein interaction (PPI), functional enrichment, and cMAP analysis reveal potential mechanisms and drugs of CHF-related CKD. Five machine learning algorithms were used to screen candidate biomarkers, construct a diagnostic nomogram for CKD and validate it in two external cohorts. Clinical serum samples were collected in our hospital to evaluate the correlation and diagnostic value of biomarkers and CKD. 225 CKD key genes and 316 CHF-related secreted proteins were identified. Four key subgroups, including 204 genes, were identified as CRS2-related pathogenic genes by PPI analysis. Enrichment analysis revealed that the identified subgroups exhibited significant enrichment in cytokine action, immune responses, and inflammatory processes. The cMAP analysis highlighted metiradone as a drug with greater potential for therapeutic intervention for CRS2. Utilizing five machine learning algorithms, three hub genes (CD48, COL3A1, LOXL1) were pinpointed as potential biomarkers for CKD, and a nomogram model was constructed. Receiver operating characteristic analysis demonstrated that the nomogram's area under the curve (AUC) exceeded 0.80 in both the CKD combined dataset and two external cohorts. In addition, the three biomarkers were significantly correlated with the glomerular filtration rate, and the AUC of the model predicting disease progression was 0.944. Furthermore, analysis of immune cell infiltration indicated a correlation between the three biomarkers and the infiltration fraction of macrophages, neutrophils, and other immune cells in CKD. Our clinical cohort validated the expression patterns of three biomarkers in serum, and the diagnostic model achieved an AUC of 0.876. CHF has the potential to facilitate the progression of CKD via the release of cardiac and PBMC secreted proteins. Furthermore, CD48, COL3A1, and LOXL1 have been identified as potential biomarkers for the detection of CKD.
Collapse
Affiliation(s)
- Chuanjing Zhang
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Zhuonan Wu
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Yongfei Song
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Xiaojun Jin
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Jiale Hu
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jianqing Zhou
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Jiangfang Lian
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China.
| |
Collapse
|
14
|
Chen LZ, Zheng PF, Shi XJ. Multiomics identification of ALDH9A1 as a crucial immunoregulatory molecule involved in calcific aortic valve disease. Sci Rep 2024; 14:23577. [PMID: 39384885 PMCID: PMC11464510 DOI: 10.1038/s41598-024-75115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
Mitochondrial dysfunction and immune cell infiltration play crucial yet incompletely understood roles in the pathogenesis of calcific aortic valve disease (CAVD). This study aimed to identify immune-related mitochondrial genes critical to the pathological process of CAVD using multiomics approaches. The CIBERSORT algorithm was employed to evaluate immune cell infiltration characteristics in CAVD patients. An integrative analysis combining weighted gene coexpression network analysis (WGCNA), machine learning, and summary data-based Mendelian randomization (SMR) was performed to identify key mitochondrial genes implicated in CAVD. Spearman's rank correlation analysis was also performed to assess the relationships between key mitochondrial genes and infiltrating immune cells. Compared with those in normal aortic valve tissue, an increased proportion of M0 macrophages and resting memory CD4 T cells, along with a decreased proportion of plasma cells and activated dendritic cells, were observed in CAVD patients. Additionally, eight key mitochondrial genes associated with CAVD, including PDK4, LDHB, SLC25A36, ALDH9A1, ECHDC2, AUH, ALDH2, and BNIP3, were identified through the integration of WGCNA and machine learning methods. Subsequent SMR analysis, incorporating multiomics data, such as expression quantitative trait loci (eQTLs) and methylation quantitative trait loci (mQTLs), revealed a significant causal relationship between ALDH9A1 expression and a reduced risk of CAVD. Moreover, ALDH9A1 expression was inversely correlated with M0 macrophages and positively correlated with M2 macrophages. These findings suggest that increased ALDH9A1 expression is significantly associated with a reduced risk of CAVD and that it may exert its protective effects by modulating mitochondrial function and immune cell infiltration. Specifically, ALDH9A1 may contribute to the shift from M0 macrophages to anti-inflammatory M2 macrophages, potentially mitigating the pathological progression of CAVD. In conclusion, ALDH9A1 represents a promising molecular target for the diagnosis and treatment of CAVD. However, further validation through in vivo and n vitro studies is necessary to confirm its role in CAVD pathogenesis and therapeutic potential.
Collapse
Affiliation(s)
- Lu-Zhu Chen
- Department of Cardiology, The Central Hospital of ShaoYang, No. 36 QianYuan Lane, Daxiang District, Shaoyang, 422000, Hunan, China
| | - Peng-Fei Zheng
- Cardiology Department, Hunan Provincial People's Hospital, No.61 West Jiefang Road, Furong District, Changsha, 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, No.61 West Jiefang Road, Furong District, Changsha, 410000, Hunan, China
- Institute of cardiovascular epidemiology, Hunan Provincial People's Hospital, No.61 West Jiefang Road, Furong District, Changsha, 410000, Hunan, China
| | - Xiang-Jiang Shi
- Department of Cardiology, The Central Hospital of ShaoYang, No. 36 QianYuan Lane, Daxiang District, Shaoyang, 422000, Hunan, China.
| |
Collapse
|
15
|
Li G, Lu Z, Chen Z. Identification of common signature genes and pathways underlying the pathogenesis association between nonalcoholic fatty liver disease and heart failure. Front Immunol 2024; 15:1424308. [PMID: 39351239 PMCID: PMC11439677 DOI: 10.3389/fimmu.2024.1424308] [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: 04/28/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) and heart failure (HF) are related conditions with an increasing incidence. However, the mechanism underlying their association remains unclear. This study aimed to explore the shared pathogenic mechanisms and common biomarkers of NAFLD and HF through bioinformatics analyses and experimental validation. METHODS NAFLD and HF-related transcriptome data were extracted from the Gene Expression Omnibus (GEO) database (GSE126848 and GSE26887). Differential analysis was performed to identify common differentially expressed genes (co-DEGs) between NAFLD and HF. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were conducted to explore the functions and regulatory pathways of co-DEGs. Protein-protein interaction (PPI) network and support vector machine-recursive feature elimination (SVM-RFE) methods were used to screen common key DEGs. The diagnostic value of common key DEGs was assessed by receiver operating characteristic (ROC) curve and validated with external datasets (GSE89632 and GSE57345). Finally, the expression of biomarkers was validated in mouse models. RESULTS A total of 161 co-DEGs were screened out in NAFLD and HF patients. GO, KEGG, and GSEA analyses indicated that these co-DEGs were mainly enriched in immune-related pathways. PPI network revealed 14 key DEGs, and SVM-RFE model eventually identified two genes (CD163 and CCR1) as common key DEGs for NAFLD and HF. Expression analysis revealed that the expression levels of CD163 and CCR1 were significantly down-regulated in HF and NAFLD patients. ROC curve analysis showed that CD163 and CCR1 had good diagnostic values for HF and NAFLD. Single-gene GSEA suggested that CD163 and CCR1 were mainly engaged in immune responses and inflammation. Experimental validation indicated unbalanced macrophage polarization in HF and NAFLD mouse models, and the expression of CD163 and CCR1 were significantly down-regulated. CONCLUSION This study identified M2 polarization impairment characterized by decreased expression of CD163 and CCR1 as a common pathogenic pathway in NAFLD and HF. The downregulation of CD163 and CCR1 may reflect key pathological changes in the development and progression of NAFLD and HF, suggesting their potential as diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Gerui Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| | - Zhengjie Lu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ze Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Kachanova OS, Boyarskaya NV, Docshin PM, Scherbinin TS, Zubkova VG, Saprankov VL, Uspensky VE, Mitrofanova LB, Malashicheva AB. Ex vivo model of pathological calcification of human aortic valve. Front Cardiovasc Med 2024; 11:1411398. [PMID: 39280032 PMCID: PMC11394195 DOI: 10.3389/fcvm.2024.1411398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
The development of drug therapy for the pathological calcification of the aortic valve is still an open issue due to the lack of effective treatment strategies. Currently, the only option for treating this condition is surgical correction and symptom management. The search for models to study the safety and efficacy of anti-calcifying drugs requires them to not only be as close as possible to in vivo conditions, but also to be flexible with regard to the molecular studies that can be applied to them. The ex vivo model has several advantages, including the ability to study the effect of a drug on human cells while preserving the original structure of the valve. This allows for a better understanding of how different cell types interact within the valve, including non-dividing cells. The aim of this study was to develop a reproducible ex vivo calcification model based on valves from patients with calcific aortic stenosis. We aimed to induce spontaneous calcification in valve tissue fragments under osteogenic conditions, and to demonstrate the possibility of significantly suppressing it using a calcification inhibitor. To validate the model, we tested a Notch inhibitor Crenigacestat (LY3039478), which has been previously shown to have an anti-calcifying effect on interstitial cell of the aortic valve. We demonstrate here an approach to testing calcification inhibitors using an ex vivo model of cultured human aortic valve tissue fragments. Thus, we propose that ex vivo models may warrant further investigation for their utility in studying aortic valve disease and performing pre-clinical assessment of drug efficacy.
Collapse
Affiliation(s)
- O S Kachanova
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - N V Boyarskaya
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - P M Docshin
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - T S Scherbinin
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - V G Zubkova
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - V L Saprankov
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - V E Uspensky
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - L B Mitrofanova
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - A B Malashicheva
- Research Laboratory of Diseases with Excessive Calcification, Almazov National Medical Research Centre, Saint Petersburg, Russia
| |
Collapse
|
17
|
Pei J, Zhang J, Yu C, Luo J, Wen S, Hua Y, Wei G. Transcriptomics-based exploration of shared M1-type macrophage-related biomarker in acute kidney injury after kidney transplantation and acute rejection after kidney transplantation. Transpl Immunol 2024; 85:102066. [PMID: 38815767 DOI: 10.1016/j.trim.2024.102066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Macrophage type 1 (M1) cells are associated with both acute kidney injury (AKI) during kidney transplantation and acute rejection (AR) after kidney transplantation. Our study explored M1-related biomarkers involved in both AKI and AR and their potential biological functions. METHODS Based on the Gene Expression Omnibus (GEO) database, the immune cell infiltration levels and differentially expressed genes were examined in AKI and AR in the kidney transplantation; M1-related genes shared in AKI and AR were identified using weighted gene co-expression analysis (WGCNA) system. Subsequently, protein-protein interaction (PPI) networks and machine learning methods to identify Hub genes and construct diagnostic models. Both AKI model and AR rat models were built to validate the expressions of Hub genes and test the injury phenotype, oxidative stress markers, and inflammatory factors. Finally, the transcription factor (TF)-Hub gene and micro-RNA (miRNA)-Hub gene regulatory networks were constructed based on identified Hub genes. RESULTS Out of 2167 differential expression genes (DEGs) in AKI and 2100 DEGs in AR, four M1-related Hub genes were obtained by PPI networks and machine learning methods, namely GBP2, TYROBP, CCR5, and TLR8. The calibration curves in the nomogram diagnostic model for these four Hub genes suggested the same predictive probability as an ideal model for AKI and AR after kidney transplantation (AUC values of the area under the ROC curve were all >0.7). The same observations were confirmed in ischemia reperfusion injury (IRI) and AR rat models by identifying common four Hub genes (GBP2, TYROBP, TLR8, and CCR5). Western blots showed that these four Hub genes were significantly different in rat models of IRI and AR (all p<0.05). Compared with the control group, IRI and AR groups showed aggravated histopathological damage and increased secretion of oxidative stress markers and inflammatory factors in rat kidneys (all p<0.05). Finally, TF-Hub and miRNA-Hub gene regulatory networks were constructed to provide a theoretical basis for the regulation of Hub genes. CONCLUSION We identified four macrophage M1-related Hub genes shared among AKI and AR after kidney transplantation. These genes may be considered for diagnosis of AKI and AR after kidney transplantation.
Collapse
Affiliation(s)
- Jun Pei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jie Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Chengjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jin Luo
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Sheng Wen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Yi Hua
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| |
Collapse
|
18
|
Huang Y, Yue S, Qiao J, Dong Y, Liu Y, Zhang M, Zhang C, Chen C, Tang Y, Zheng J. Identification of diagnostic genes and drug prediction in metabolic syndrome-associated rheumatoid arthritis by integrated bioinformatics analysis, machine learning, and molecular docking. Front Immunol 2024; 15:1431452. [PMID: 39139563 PMCID: PMC11320606 DOI: 10.3389/fimmu.2024.1431452] [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: 05/12/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Background Interactions between the immune and metabolic systems may play a crucial role in the pathogenesis of metabolic syndrome-associated rheumatoid arthritis (MetS-RA). The purpose of this study was to discover candidate biomarkers for the diagnosis of RA patients who also had MetS. Methods Three RA datasets and one MetS dataset were obtained from the Gene Expression Omnibus (GEO) database. Differential expression analysis, weighted gene co-expression network analysis (WGCNA), and machine learning algorithms including Least Absolute Shrinkage and Selection Operator (LASSO) regression and Random Forest (RF) were employed to identify hub genes in MetS-RA. Enrichment analysis was used to explore underlying common pathways between MetS and RA. Receiver operating characteristic curves were applied to assess the diagnostic performance of nomogram constructed based on hub genes. Protein-protein interaction, Connectivity Map (CMap) analyses, and molecular docking were utilized to predict the potential small molecule compounds for MetS-RA treatment. qRT-PCR was used to verify the expression of hub genes in fibroblast-like synoviocytes (FLS) of MetS-RA. The effects of small molecule compounds on the function of RA-FLS were evaluated by wound-healing assays and angiogenesis experiments. The CIBERSORT algorithm was used to explore immune cell infiltration in MetS and RA. Results MetS-RA key genes were mainly enriched in immune cell-related signaling pathways and immune-related processes. Two hub genes (TYK2 and TRAF2) were selected as candidate biomarkers for developing nomogram with ideal diagnostic performance through machine learning and proved to have a high diagnostic value (area under the curve, TYK2, 0.92; TRAF2, 0.90). qRT-PCR results showed that the expression of TYK2 and TRAF2 in MetS-RA-FLS was significantly higher than that in non-MetS-RA-FLS (nMetS-RA-FLS). The combination of CMap analysis and molecular docking predicted camptothecin (CPT) as a potential drug for MetS-RA treatment. In vitro validation, CPT was observed to suppress the cell migration capacity and angiogenesis capacity of MetS-RA-FLS. Immune cell infiltration results revealed immune dysregulation in MetS and RA. Conclusion Two hub genes were identified in MetS-RA, a nomogram for the diagnosis of RA and MetS was established based on them, and a potential therapeutic small molecule compound for MetS-RA was predicted, which offered a novel research perspective for future serum-based diagnosis and therapeutic intervention of MetS-RA.
Collapse
Affiliation(s)
- Yifan Huang
- Department of Orthopedics, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Songkai Yue
- Department of Orthopedics, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Jinhan Qiao
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yonghui Dong
- Department of Orthopedics, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Yunke Liu
- Department of Orthopedics, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Meng Zhang
- Department of Orthopedics, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Cheng Zhang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Chuanliang Chen
- Clinical Bioinformatics Experimental Center, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Yuqin Tang
- Clinical Bioinformatics Experimental Center, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Jia Zheng
- Department of Orthopedics, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Ran Z, Mu BR, Zhu T, Zhang Y, Luo JX, Yang X, Li B, Wang DM, Lu MH. Predicting biomarkers related to idiopathic pulmonary fibrosis: Robust ranking aggregation analysis and animal experiment verification. Int Immunopharmacol 2024; 139:112766. [PMID: 39067403 DOI: 10.1016/j.intimp.2024.112766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/22/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and incurable lung disease characterized by unknown etiology. This study employs robust ranking aggregation to identify consistent differential genes across multiple datasets, aiming to enhance prognostic evaluation and facilitate the development of more effective immunotherapy strategies for IPF. Using the GSE10667, GSE110147, and GSE24206 datasets, the analysis identifies 92 robust differentially expressed genes (DEGs), including SPP1, IGF1, ASPN, and KLHL13, highlighted as potential biomarkers through machine learning and experimental validation. Additionally, significant differences in immune cell types between IPF samples and controls, such as Plasma cells, Macrophages M0, Mast cells resting, T cells CD8, and NK cells resting, inform the construction of diagnostic and survival prediction models, demonstrating good applicability. These findings provide insights into IPF pathophysiology and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Zhao Ran
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ben-Rong Mu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Zhu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia-Xin Luo
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiong Yang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Li
- Department of Respiratory Medicine, Guangyuan Hospital of Traditional Chinese Medicine, No.133 Jianshe Road, Lizhou District, Guangyuan 628099, Sichuan, China
| | - Dong-Mei Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Mei-Hong Lu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
20
|
De Paolis E, Tilocca B, Inchingolo R, Lombardi C, Perrucci A, Maneri G, Roncada P, Varone F, Luca R, Urbani A, Minucci A, Santonocito C. The novel CFTR haplotype E583G/F508del in CFTR-related disorder. Mol Biol Rep 2024; 51:849. [PMID: 39052151 PMCID: PMC11272816 DOI: 10.1007/s11033-024-09732-x] [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: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND CFTR-related disorder (CFTR-RD) is a clinical entity associated to complex diagnostic paths and newly upgraded standard of care. In CFTR-RD, CFTR genotyping represents a diagnostic surrogate marker. In case of novel haplotype, the diagnosis could represents an area of concern. We described the molecular evaluation of the rare CFTR variant E583G identified in trans with the F508del in a novel haplotype. METHODS AND RESULTS An adult woman was referred to our pulmonary unit for persistent respiratory symptoms. CFTR Next Generation Sequencing was performed to evaluate full-gene mutational status. The variant identified was evaluated for its pathogenicity integrating clinical evidences with dedicated bioinformatics analyses. Clinical evaluation of patient matched with a mono-organ CFTR-RD diagnosis. Genotyping revealed the novel CFTR haplotype F508del/E583G. Multiple evidences of a deleterious effect of the CFTR E583G rare variant emerged from the bioinformatics analyses performed. CONCLUSIONS Guidelines for CFTR-RD are available with the purpose of harmonizing clinical and molecular investigations. In such context, the identification of novel CFTR haplotype need to a deeper evaluation with a combination of skills. The novel E583G variant could be considered of clinical interest and overall a CFTR-RD Variants of Varying Clinical Consequences.
Collapse
Affiliation(s)
- Elisa De Paolis
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, 00168, Italy
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Bruno Tilocca
- Department of Health Science, University "Magna Graecia" of Catanzaro, Catanzaro, 88100, Italy
| | - Riccardo Inchingolo
- Pulmonary Medicine Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carla Lombardi
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessia Perrucci
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, 00168, Italy
| | - Giulia Maneri
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, 00168, Italy
| | - Paola Roncada
- Department of Health Science, University "Magna Graecia" of Catanzaro, Catanzaro, 88100, Italy
| | - Francesco Varone
- Pulmonary Medicine Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Richeldi Luca
- Pulmonary Medicine Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of Sacread Heart, Rome, 1-00168, Italy
| | - Andrea Urbani
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, Largo Agostino Gemelli,, Rome, 00168, Italy
| | - Angelo Minucci
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, 00168, Italy
| | - Concetta Santonocito
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, 00168, Italy.
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, Largo Agostino Gemelli,, Rome, 00168, Italy.
| |
Collapse
|
21
|
Gong Z, Huang X, Cao Q, Wu Y, Zhang Q. A CLRN3-Based CD8 + T-Related Gene Signature Predicts Prognosis and Immunotherapy Response in Colorectal Cancer. Biomolecules 2024; 14:891. [PMID: 39199281 PMCID: PMC11352867 DOI: 10.3390/biom14080891] [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: 04/24/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) ranks among the most prevalent malignancies affecting the gastrointestinal tract. The infiltration of CD8+ T cells significantly influences the prognosis and progression of tumor patients. METHODS This study establishes a CRC immune risk model based on CD8+ T cell-related genes. CD8+ T cell-related genes were identified through Weighted Gene Co-expression Network Analysis (WGCNA), and the enriched gene sets were annotated via Gene Ontology (GO) and Reactome pathway analysis. Employing machine learning methods, including the Least Absolute Shrinkage and Selection Operator (LASSO) algorithm and Random Forest (RF), we identified nine genes associated with CD8+ T-cell infiltration. The infiltration levels of immune cells in CRC tissues were assessed using the ssGSEA algorithm. RESULTS These genes provide a foundation for constructing a prognostic model. The TCGA-CRC sample model's prediction scores were categorized, and the prediction models were validated through Cox regression analysis and Kaplan-Meier curve analysis. Notably, although CRC tissues with higher risk scores exhibited elevated levels of CD8+ T-cell infiltration, they also demonstrated heightened expression of immune checkpoint genes. Furthermore, comparison of microsatellite instability (MSI) and gene mutations across the immune subgroups revealed notable gene variations, particularly with APC, TP53, and TNNT1 showing higher mutation frequencies. Finally, the predictive model's efficacy was corroborated through the use of Tumor Immune Dysfunction and Exclusion (TIDE), Immune Profiling Score (IPS), and immune escape-related molecular markers. The predictive model was validated through an external cohort of CRC and the Bladder Cancer Immunotherapy Cohort. CLRN3 expression levels in tumor and adjacent normal tissues were assessed using quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Subsequent in vitro and in vivo experiments demonstrated that CLRN3 knockdown significantly attenuated the malignant biological behavior of CRC cells, while overexpression had the opposite effect. CONCLUSIONS This study presents a novel prognostic model for CRC, providing a framework for enhancing the survival rates of CRC patients by targeting CD8+ T-cell infiltration.
Collapse
Affiliation(s)
- Zhiwen Gong
- Department of Thoracic Surgery, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China; (Z.G.); (Q.C.)
| | - Xiuting Huang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China;
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Qingdong Cao
- Department of Thoracic Surgery, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China; (Z.G.); (Q.C.)
| | - Yuanquan Wu
- Department of Gastrointestinal Surgery, The Affiliated Kashi Hospital, Sun Yat-Sen University, Kashi 844000, China
| | - Qunying Zhang
- Department of Geriatrics, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| |
Collapse
|
22
|
Zhang C, Song Y, Cen L, Huang C, Zhou J, Lian J. Screening of Secretory Proteins Linking Major Depressive Disorder with Heart Failure Based on Comprehensive Bioinformatics Analysis and Machine Learning. Biomolecules 2024; 14:793. [PMID: 39062507 PMCID: PMC11275063 DOI: 10.3390/biom14070793] [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: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) plays a crucial role in the occurrence of heart failure (HF). This investigation was undertaken to explore the possible mechanism of MDD's involvement in HF pathogenesis and identify candidate biomarkers for the diagnosis of MDD with HF. METHODS GWAS data for MDD and HF were collected, and Mendelian randomization (MR) analysis was performed to investigate the causal relationship between MDD and HF. Differential expression analysis (DEA) and WGCNA were used to detect HF key genes and MDD-associated secretory proteins. Protein-protein interaction (PPI), functional enrichment, and cMAP analysis were used to reveal potential mechanisms and drugs for MDD-related HF. Then, four machine learning (ML) algorithms (including GLM, RF, SVM, and XGB) were used to screen candidate biomarkers, construct diagnostic nomograms, and predict MDD-related HF. Furthermore, the MCPcounter algorithm was used to explore immune cell infiltration in HF, and MR analysis was performed to explore the causal effect of immunophenotypes on HF. Finally, the validation of the association of MDD with reduced left ventricular ejection fraction (LVEF) and the performance assessment of diagnostic biomarkers was accomplished based on animal models mimicking MDD. RESULTS The MR analysis showed that the MDD was linked to an increased risk of HF (OR = 1.129, p < 0.001). DEA combined with WGCNA and secretory protein gene set identified 315 HF key genes and 332 MDD-associated secretory proteins, respectively. Through PPI and MCODE analysis, 78 genes were pinpointed as MDD-related pathogenic genes for HF. The enrichment analysis revealed that these genes were predominantly enriched in immune and inflammatory regulation. Through four ML algorithms, two hub genes (ISLR/SFRP4) were identified as candidate HF biomarkers, and a nomogram was developed. ROC analysis showed that the AUC of the nomogram was higher than 0.90 in both the HF combined dataset and two external cohorts. In addition, an immune cell infiltration analysis revealed the immune dysregulation in HF, with ISLR/SFRP4 displaying notable associations with the infiltration of B cells, CD8 T cells, and fibroblasts. More importantly, animal experiments showed that the expression levels of ISLR (r = -0.653, p < 0.001) and SFRP4 (r = -0.476, p = 0.008) were significantly negatively correlated with LVEF. CONCLUSIONS The MR analysis indicated that MDD is a risk factor for HF at the genetic level. Bioinformatics analysis and the ML results suggest that ISLR and SFRP4 have the potential to serve as diagnostic biomarkers for HF. Animal experiments showed a negative correlation between the serum levels of ISLR/SFRP4 and LVEF, emphasizing the need for additional clinical studies to elucidate their diagnostic value.
Collapse
Affiliation(s)
- Chuanjing Zhang
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Yongfei Song
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Lichao Cen
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Chen Huang
- Department of Genetics, Xi’an Jiaotong University, Xi’an 710049, China;
| | - Jianqing Zhou
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| | - Jiangfang Lian
- Ningbo University Health Science Center, Ningbo 315040, China; (C.Z.); (Y.S.); (L.C.); (J.Z.)
| |
Collapse
|
23
|
Wang Y, Xie D, Ma S, Shao N, Zhang X, Wang X. Exploring the common mechanism of vascular dementia and inflammatory bowel disease: a bioinformatics-based study. Front Immunol 2024; 15:1347415. [PMID: 38736878 PMCID: PMC11084673 DOI: 10.3389/fimmu.2024.1347415] [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: 11/30/2023] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Objective Emerging evidence has shown that gut diseases can regulate the development and function of the immune, metabolic, and nervous systems through dynamic bidirectional communication on the brain-gut axis. However, the specific mechanism of intestinal diseases and vascular dementia (VD) remains unclear. We designed this study especially, to further clarify the connection between VD and inflammatory bowel disease (IBD) from bioinformatics analyses. Methods We downloaded Gene expression profiles for VD (GSE122063) and IBD (GSE47908, GSE179285) from the Gene Expression Omnibus (GEO) database. Then individual Gene Set Enrichment Analysis (GSEA) was used to confirm the connection between the two diseases respectively. The common differentially expressed genes (coDEGs) were identified, and the STRING database together with Cytoscape software were used to construct protein-protein interaction (PPI) network and core functional modules. We identified the hub genes by using the Cytohubba plugin. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were applied to identify pathways of coDEGs and hub genes. Subsequently, receiver operating characteristic (ROC) analysis was used to identify the diagnostic ability of these hub genes, and a training dataset was used to verify the expression levels of the hub genes. An alternative single-sample gene set enrichment (ssGSEA) algorithm was used to analyze immune cell infiltration between coDEGs and immune cells. Finally, the correlation between hub genes and immune cells was analyzed. Results We screened 167 coDEGs. The main articles of coDEGs enrichment analysis focused on immune function. 8 shared hub genes were identified, including PTPRC, ITGB2, CYBB, IL1B, TLR2, CASP1, IL10RA, and BTK. The functional categories of hub genes enrichment analysis were mainly involved in the regulation of immune function and neuroinflammatory response. Compared to the healthy controls, abnormal infiltration of immune cells was found in VD and IBD. We also found the correlation between 8 shared hub genes and immune cells. Conclusions This study suggests that IBD may be a new risk factor for VD. The 8 hub genes may predict the IBD complicated with VD. Immune-related coDEGS may be related to their association, which requires further research to prove.
Collapse
Affiliation(s)
- Yujiao Wang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Daojun Xie
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shijia Ma
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Nan Shao
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiaoyan Zhang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xie Wang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
24
|
Lv X, Fan Q, Li X, Li P, Wan Z, Han X, Wang H, Wang X, Wu L, Huo B, Yang L, Chen G, Zhang Y. Identification of renal ischemia reperfusion injury-characteristic genes, pathways and immunological micro-environment features through bioinformatics approaches. Aging (Albany NY) 2024; 16:2123-2140. [PMID: 38329418 PMCID: PMC10911371 DOI: 10.18632/aging.205471] [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: 09/22/2023] [Accepted: 12/15/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND Biomarkers and pathways associated with renal ischemia reperfusion injury (IRI) had not been well unveiled. This study was intended to investigate and summarize the regulatory networks for related hub genes. Besides, the immunological micro-environment features were evaluated and the correlations between immune cells and hub genes were also explored. METHODS GSE98622 containing mouse samples with multiple IRI stages and controls was collected from the GEO database. Differentially expressed genes (DEGs) were recognized by the R package limma, and the GO and KEGG analyses were conducted by DAVID. Gene set variation analysis (GSVA) and weighted gene coexpression network analysis (WGCNA) had been implemented to uncover changed pathways and gene modules related to IRI. Besides the known pathways such as apoptosis pathway, metabolic pathway, and cell cycle pathways, some novel pathways were also discovered to be critical in IRI. A series of novel genes associated with IRI was also dug out. An IRI mouse model was constructed to validate the results. RESULTS The well-known IRI marker genes (Kim1 and Lcn2) and novel hub genes (Hbegf, Serpine2, Apbb1ip, Trip13, Atf3, and Ncaph) had been proved by the quantitative real-time polymerase chain reaction (qRT-PCR). Thereafter, miRNAs targeted to the dysregulated genes were predicted and the miRNA-target network was constructed. Furthermore, the immune infiltration for these samples was predicted and the results showed that macrophages infiltrated to the injured kidney to affect the tissue repair or fibrosis. Hub genes were significantly positively or negatively correlated with the macrophage abundance indicating they played a crucial role in macrophage infiltration. CONCLUSIONS Consequently, the pathways, hub genes, miRNAs, and the immune microenvironment may explain the mechanism of IRI and might be the potential targets for IRI treatments.
Collapse
Affiliation(s)
- Xinghua Lv
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Qian Fan
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University Eye Institute, Nankai University, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Xuanjie Li
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Peng Li
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhanhai Wan
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xuena Han
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hao Wang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xiaoxia Wang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lin Wu
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Bin Huo
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Li Yang
- Lanzhou First People's Hospital, Lanzhou, Gansu, China
| | - Gen Chen
- Department of Microbiology, School of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Yan Zhang
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
25
|
Wu L, Li S, Wu C, Wu S, Lin Y, Wei D. Causal relationship between systemic lupus erythematosus and primary liver cirrhosis based on two-sample bidirectional Mendelian randomization and transcriptome overlap analysis. Arthritis Res Ther 2024; 26:10. [PMID: 38167341 PMCID: PMC10762944 DOI: 10.1186/s13075-023-03235-z] [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: 10/22/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Overlapping cases of systemic lupus erythematosus (SLE) and primary biliary cirrhosis (PBC) are rare and have not yet been fully proven to be accidental or have a common genetic basis. METHODS Two-sample bidirectional Mendelian randomization (MR) analysis was applied to explore the potential causal relationship between SLE and PBC. The heterogeneity and reliability of MR analysis were evaluated through Cochran's Q-test and sensitivity test, respectively. Next, transcriptome overlap analysis of SLE and PBC was performed using the Gene Expression Omnibus database to identify the potential mechanism of hub genes. Finally, based on MR analysis, the potential causal relationship between hub genes and SLE or PBC was validated again. RESULTS The MR analysis results indicated that SLE and PBC were both high-risk factors for the occurrence and development of the other party. On the one hand, MR analysis had heterogeneity, and on the other hand, it also had robustness. Nine hub genes were identified through transcriptome overlap analysis, and machine learning algorithms were used to verify their high recognition efficiency for SLE patients. Finally, based on MR analysis, it was verified that there was no potential causal relationship between the central gene SOCS3 and SLE, but it was a high-risk factor for the potential risk of PBC. CONCLUSION The two-sample bidirectional MR analysis revealed that SLE and PBC were high-risk factors for each other, indicating that they had similar genetic bases, which could to some extent overcome the limitation of insufficient overlap in case samples of SLE and PBC. The analysis of transcriptome overlapping hub genes provided a theoretical basis for the potential mechanisms and therapeutic targets of SLE with PBC overlapping cases.
Collapse
Affiliation(s)
- Linyong Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525000, People's Republic of China
| | - Songhua Li
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525000, People's Republic of China.
| | - Chaojun Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525000, People's Republic of China
| | - Shaofeng Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525000, People's Republic of China
| | - Yan Lin
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525000, People's Republic of China
| | - Dayou Wei
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, Guangdong, 525000, People's Republic of China.
| |
Collapse
|
26
|
Shi H, Yuan X, Liu G, Fan W. Identifying and Validating GSTM5 as an Immunogenic Gene in Diabetic Foot Ulcer Using Bioinformatics and Machine Learning. J Inflamm Res 2023; 16:6241-6256. [PMID: 38145013 PMCID: PMC10748866 DOI: 10.2147/jir.s442388] [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: 10/31/2023] [Accepted: 12/12/2023] [Indexed: 12/26/2023] Open
Abstract
Background A diabetic foot ulcer (DFU) is a serious, long-term condition associated with a significant risk of disability and mortality. However, research on its biomarkers is still limited. This study utilizes bioinformatics and machine learning methods to identify immune-related biomarkers for DFU and validates them through external datasets and animal experiments. Methods This study used bioinformatics and machine learning to analyze microarray data from the Gene Expression Omnibus (GEO) database to identify key genes associated with DFU. Animal experiments were conducted to validate these findings. This research employs the datasets GSE68183 and GSE80178 retrieved from the GEO database as the training dataset for building a gene machine learning model, and after conducting differential analysis on the data, this study used package glmnet and package e1071 to construct LASSO and SVM-RFE machine learning models, respectively. Subsequently, we validated the model using the training set and validation set (GSE134431). We conducted enrichment analysis, including GSEA and GSVA, on the model genes. We also performed immune functional analysis and immune-related analysis on the model genes. Finally, we conducted immunohistochemistry (IHC) validation on the model genes. Results This study identifies GSTM5 as a potential immune-related key target in DFU using machine learning and bioinformatics methods. Subsequent validation through external datasets and IHC experiments also confirms GSTM5 as a critical biomarker for DFU. The gene may be associated with T cells regulatory (Tregs) and T cells follicular helper, and it influences the NF-κB, GnRH, and MAPK signaling pathway. Conclusion This study identified and validated GSTM5 as a biomarker for DFU. This finding may potentially provide a target for immune therapy for DFU.
Collapse
Affiliation(s)
- Hongshuo Shi
- Department of Peripheral Vascular Surgery, Institute of Surgery of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xin Yuan
- Department of Peripheral Vascular Surgery, Institute of Surgery of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Guobin Liu
- Department of Peripheral Vascular Surgery, Institute of Surgery of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Weijing Fan
- Department of Peripheral Vascular Surgery, Institute of Surgery of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|