1
|
Zhang Y, Liu J, Basang Z, Yang Q, Chen H, Chen S, Li S, Lei C, Fang M, Liu H, Jin X, Wang Y. Shared genetic features inference among hypoxia-ischemia diseases in the presence of heterogenous omics data based on a novel risk assessment method. Front Genet 2025; 16:1587854. [PMID: 40357364 PMCID: PMC12066567 DOI: 10.3389/fgene.2025.1587854] [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: 03/05/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
The hypoxia-ischemia (H-I) diseases share some common mechanisms which may help to delay the diseases' processing. However, the shared features are still unclear due to the lack of large scale high-quality multi - omics data that specifically target the same disease, population, and tissues/cells. In this study, we developed a novel risk assessment method to analyze four H-I diseases including eclampsia/preeclampsia (PE), pulmonary arterial hypertension (PAH), high-altitude polycythemia (HAPC), and ischemic stroke (IS). A combined new evaluation score was designed to integrate evaluation information from genomics, transcriptomics, proteomics, and metabolomics in previous researches. Genes were then divided into different groups according to their risk assessment score. The most significant group (direct biomarkers) contained genes with direct evidence of association to H-I disease: PIEZO2 and HPGD (shared), TSIX and SAA1 (PAH - specific), GSTM1, DNTT, and IGKC (HAPC - specific), LEP, SERPINA3, and ARHGEF4 (PE - specific), CD3D, ITK, and RPL18A (IS - specific). The groups 'Intermediate crucial biomarkers' contained genes played important roles in H-I disease related biological processes: CXCL8 (shared), HBG2, GRIN2A, and FGFBP1 (PAH - specific), FAM111B (HAPC - specific), C12orf39 and SLAMF1 (PE - specific). The genes lacking disease-association evidence but with similar characteristics with the above two groups were considered as 'potential minor-effect biomarkers': are SRRM2 - AS1 (shared), ATP8A1 (PAH - specific), RXFP1 and HJURP (HAPC - specific), HIST1H1T (PE - specific). With the development of biological experiments, these intermediate crucial and potential minor-effect biomarkers may be proved to be direct biomarkers in the future. Therefore, these biomarkers may serve as an entry point for subsequent research and are of great significance.
Collapse
Affiliation(s)
- Yifan Zhang
- BGI Research, Chongqing, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen, China
| | - Jianfeng Liu
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhuoma Basang
- High Altitude Health Science Research Center, Tibet University, Lhasa, Tibet, China
| | - Qianxun Yang
- BGI Research, Shenzhen, China
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Hongce Chen
- BGI Research, Shenzhen, China
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Shuo Chen
- BGI Research, Shenzhen, China
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, China
| | - Shaogang Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | | | - Mingyan Fang
- BGI Research, Wuhan, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, China
| | | | - Xin Jin
- BGI Research, Chongqing, China
- BGI Research, Shenzhen, China
- Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, China
| | - Yingying Wang
- BGI Research, Shenzhen, China
- Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen, China
| |
Collapse
|
2
|
Wang Y, Li K, Mo S, Yao P, Zeng J, Lu S, Qin S. Identification of common genes and pathways between type 2 diabetes and COVID-19. Front Genet 2024; 15:1249501. [PMID: 38699234 PMCID: PMC11063347 DOI: 10.3389/fgene.2024.1249501] [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: 06/28/2023] [Accepted: 03/21/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Numerous studies have reported a high incidence and risk of severe illness due to coronavirus disease 2019 (COVID-19) in patients with type 2 diabetes (T2DM). COVID-19 patients may experience elevated or decreased blood sugar levels and may even develop diabetes. However, the molecular mechanisms linking these two diseases remain unclear. This study aimed to identify the common genes and pathways between T2DM and COVID-19. METHODS Two public datasets from the Gene Expression Omnibus (GEO) database (GSE95849 and GSE164805) were analyzed to identify differentially expressed genes (DEGs) in blood between people with and without T2DM and COVID-19. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed on the common DEGs. A protein-protein interaction (PPI) network was constructed to identify common genes, and their diagnostic performance was evaluated by receiver operating characteristic (ROC) curve analysis. Validation was performed on the GSE213313 and GSE15932 datasets. A gene co-expression network was constructed using the GeneMANIA database to explore interactions among core DEGs and their co-expressed genes. Finally, a microRNA (miRNA)-transcription factor (TF)-messenger RNA (mRNA) regulatory network was constructed based on the common feature genes. RESULTS In the GSE95849 and GSE164805 datasets, 81 upregulated genes and 140 downregulated genes were identified. GO and KEGG enrichment analyses revealed that these DEGs were closely related to the negative regulation of phosphate metabolic processes, the positive regulation of mitotic nuclear division, T-cell co-stimulation, and lymphocyte co-stimulation. Four upregulated common genes (DHX15, USP14, COPS3, TYK2) and one downregulated common feature gene (RIOK2) were identified and showed good diagnostic accuracy for T2DM and COVID-19. The AUC values of DHX15, USP14, COPS3, TYK2, and RIOK2 in T2DM diagnosis were 0.931, 0.917, 0.986, 0.903, and 0.917, respectively. In COVID-19 diagnosis, the AUC values were 0.960, 0.860, 1.0, 0.9, and 0.90, respectively. Validation in the GSE213313 and GSE15932 datasets confirmed these results. The miRNA-TF-mRNA regulatory network showed that TYH2 was targeted by PITX1, PITX2, CRX, NFYA, SREBF1, RELB, NR1L2, and CEBP, whereas miR-124-3p regulates THK2, RIOK2, and USP14. CONCLUSION We identified five common feature genes (DHX15, USP14, COPS3, TYK2, and RIOK2) and their co-regulatory pathways between T2DM and COVID-19, which may provide new insights for further molecular mechanism studies.
Collapse
Affiliation(s)
- Ya Wang
- Gastroenterology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Endocrinology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Kai Li
- Orthopedics Department, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Shuangyang Mo
- Gastroenterology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Peishan Yao
- Gastroenterology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiaxing Zeng
- Department of Traumatic Surgery, Microsurgery, and Hand Surgery, Guangxi Zhuang Autonomous Region People’s Hospital, Nanning, Guangxi, China
| | - Shunyu Lu
- Department of Pharmacy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shanyu Qin
- Gastroenterology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
3
|
Li Y, Zhang C, Zhao Z. KNOCKDOWN OF CIRC_0114428 ALLEVIATES LPS-INDUCED HK2 CELL APOPTOSIS AND INFLAMMATION INJURY VIA TARGETING MIR-215-5P/TRAF6/NF-ΚB AXIS IN SEPTIC ACUTE KIDNEY INJURY. Shock 2024; 61:620-629. [PMID: 38010029 DOI: 10.1097/shk.0000000000002245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
ABSTRACT Background: Sepsis is a systemic inflammatory disease that can cause multiple organ damage. Circular RNAs (circRNAs) have been reported to play a regulatory role in sepsis-induced acute kidney injury (AKI); however, the role of circ_0114428 has not been studied. Methods: In this study, HK2 cells were treated with different concentrations of LPS to induce cell damage, and then the expressions of circ_0114428, microRNA-215-5p (miR-215-5p), and tumor necrosis factor receptor-associated factor 6 (TRAF6) were detected by quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot examined the Bax and cleaved-Caspase-3 proteins. Cell proliferation was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and thymidine analog 5-ethynyl-2'-deoxyuridine (EdU) assay. In addition, cell apoptosis was detected by flow cytometry, and the levels of inflammatory factors were detected by enzyme-linked immunosorbent assay. Results: After LPS treatment with different concentrations, we found that LPS at 10 μg/mL had the best effect on HK2 cells. Circ_0114428 was highly expressed in sepsis-AKI patients and LPS-treated HK2 cells. Knockdown of circ_0114428 restored the effects of LPS treatment on proliferation, apoptosis, and inflammatory response of HK2 cells. MiR-215-5p was a target of circ_0114428, and TRAF6 was a downstream target of miR-215-5p. Circ_0114428 regulated TRAF6 expression by sponging miR-215-5p in LPS-treated HK2 cells. Circ_0114428 regulated LPS-induced NF-κB signaling in HK2 cells by targeting miR-215-5p/TRAF6 axis. Conclusion: Circ_0114428 knockdown abolished the cell proliferation, apoptosis, and inflammatory damage in LPS-induced HK2 cells by targeting miR-215-5p/TRAF6/NF-κB.
Collapse
Affiliation(s)
- Yan Li
- Department of Emergency Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Chunmei Zhang
- Department of Critical Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhongyan Zhao
- Department of Critical Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Li L, Li C, Cao S, Zhou G, Jiang Y, Feng J. Circ-SUZ12 Protects Cardiomyocytes from Hypoxia-Induced Dysfunction Through Upregulating SUZ12 Expression to Activate Wnt/β-catenin Signaling Pathway. Int Heart J 2023; 64:1113-1124. [PMID: 37967992 DOI: 10.1536/ihj.22-452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Acute myocardial infarction (AMI) is a common coronary artery disease. This study attempted to reveal the impact of circ-SUZ12 (hsa_circ_0042961) on cardiomyocyte injury after exposure to hypoxia.Circ-SUZ12 was screened out from the GEO dataset GSE169594. RNA expression and protein level were detected by quantitative real-time PCR (qRT-PCR) and Western blot, respectively. The characteristics of circ-SUZ12 were identified by measuring its resistance to Rnase R or actinomycin D (Act D) treatment. CCK-8 and EdU assays were performed to explore the viability of AC16 cells. Cell apoptosis was assessed through TUNEL assay and flow cytometry analysis. Mechanism experiments were performed to investigate the downstream molecular mechanism of circ-SUZ12.Circ-SUZ12 was highly expressed in blood samples of AMI patients in the GEO dataset and lowly expressed in hypoxia-treated cardiomyocytes. Overexpression of circ-SUZ12 reversed hypoxia-induced cardiomyocyte injury. Circ-SUZ12 regulated SUZ12 polycomb repressive complex 2 subunit (SUZ12) expression by recruiting FUS protein. SUZ12 activated the Wnt/β-catenin signaling pathway by increasing the H3K27me3 level in microRNA (miR)-526b-5p promoter to release catenin beta 1 (CTNNB1). CTNNB1 depletion reversed the effect of circ-SUZ12 on the viability and apoptosis of hypoxia-induced cardiomyocytes.Circ-SUZ12 protects cardiomyocytes from hypoxia-induced dysfunction through upregulating SUZ12 expression to activate the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Long Li
- Department of Cardiology, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Medical University of Anhui)
| | - Chao Li
- Department of Cardiology, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Medical University of Anhui)
| | - Shuai Cao
- Department of Cardiology, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Medical University of Anhui)
| | - Gaoliang Zhou
- Department of Cardiology, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Medical University of Anhui)
| | - Yongjin Jiang
- Department of Cardiology, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Medical University of Anhui)
| | - Jun Feng
- Department of Cardiology, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Medical University of Anhui)
| |
Collapse
|
5
|
Yu CJ, Xia F, Ruan L, Hu SP, Zhu WJ, Yang K. Circ_0004771 Promotes Hypoxia/Reoxygenation Induced Cardiomyocyte Injury via Activation of Mitogen-Activated Protein Kinase Signaling Pathway. Int Heart J 2023; 64:1125-1132. [PMID: 37967979 DOI: 10.1536/ihj.23-333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
This study aimed to observe the mechanism and effect of circ_0004771 on cardiomyocyte injury in acute myocardial infarction (AMI). The differences in circ_0004771 expression in the blood of AMI patients and healthy volunteers were observed by Real-Time Quantitative Reverse Transcription-Polymerase Chain Reaction. AMI cell models were constructed by hypoxia/reoxygenation (H/R)-induced injury in human cardiomyocytes (AC16 cells). The changes of circ_0004771 expression in AMI cells were observed. After transfection with the knockdown or overexpression of circ_0004771 vector in AMI cells, Cell Counting Kit-8 (CCK-8) assay and propidium iodide/FITC-Annexin V staining were performed to detect cell proliferation and apoptosis levels, extracellular lactate dehydrogenase (LDH) activity, malondialdehyde (MDA) concentration, and superoxide dismutase (SOD) activity. Expression levels of Mitogen-activated protein kinase (MAPK) signaling pathway-related proteins (p-MEK1/2, MEK1/2, p-ERK1/2, ERK1/2), and endoplasmic reticulum (ER) stress proteins (GRP78 and CHOP-1) were observed in each group of cells by western blot method. The expression level of circ_0004771 was significantly reduced in both clinical samples and cells of AMI. When circ_0004771 was knocked down in AMI cells, it resulted in a decrease in cell proliferation level and significant increase in apoptosis level. The inhibition of circ_0004771 expression caused leakage of LDH in AMI cells, accumulation of intracellular MDA, and inhibition of SOD activity. In addition, the knockdown of circ_0004771 significantly increased the levels of p-MEK1/2, p-ERK1/2, GRP78, and CHOP-1 in H/R-induced AC16 cells. However, the overexpression of circ_0004771 resulted in the opposite result as when circ_0004771 was knocked down. A low level of circ_0004771 in AMI activates the MAPK signaling pathway in cardiomyocytes as well as encourages intracellular oxidative stress and ER stress, thereby inhibiting cell proliferation and promoting apoptosis.
Collapse
Affiliation(s)
- Chun-Jun Yu
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Feng Xia
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Lin Ruan
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Sheng-Peng Hu
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Wen-Jie Zhu
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Kai Yang
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| |
Collapse
|
6
|
Ai L, Liu Y, Chen Y. Diagnostic and treatment value of XPNPEP3 in acute myocardial infarction. J Thorac Dis 2023; 15:4976-4986. [PMID: 37868889 PMCID: PMC10586987 DOI: 10.21037/jtd-23-1203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023]
Abstract
Background At present, acute myocardial infarction (AMI) is a serious cardiovascular disease with high morbidity and mortality. Discovering biomarkers of AMI is important for clinical diagnosis and needs. Therefore, this study aimed to elucidate the role of XPNPEP3 as a potential biomarker for AMI. Methods Expression profiling data were downloaded for AMI patients and healthy patients in the GSE24548 and GSE24519 datasets, respectively. The limma package in R was conducted to determine differentially expressed microRNA (DEmiRNA)/messenger RNA (mRNA) [differentially expressed genes (DEGs)]. TargetScan and Cytoscape were used to build regulatory network of miRNA-mRNA. The Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) and Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) were applied to determine immune cell score. The gene set variation analysis (GSVA) package was used to calculate pathway score. Key drugs were determined by protein-protein interaction (PPI) and molecular docking. Results Totals of 36 DEmiRNAs and 63 DEGs were determined in the GSE24584 dataset and GSE24519 dataset, respectively, and then we constructed a miRNA-mRNA network including 31 DEmiRNAs and 47 DEGs. The correlation analysis between immune cells and 47 DEGs identified that XPNPEP3 was most associated with AMI. Furthermore, XPNPEP3 was negatively correlated with inflammatory response score. A diagnosis model based on XPNPEP3 expression showed an area under the curve (AUC) of 93.38%, and 159 genes were highly correlated with XPNPEP3. Molecular docking analysis showed that DB06909 had the lowest docking score with XPNPEP3, revealing it to be a potential XPNPEP3 inhibitor. Conclusions This work discovered that XPNPEP3 is correlated with the development of AMI. These findings may provide theoretical basis for the diagnosis and treatment of AMI.
Collapse
Affiliation(s)
- Lunna Ai
- Medical School of Chinese PLA and Department of Cardiovascular Medicine, the First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yuqi Liu
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiovascular Medicine, the First Medical Centre of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Liu H, Qin S, Zhao Y, Gao L, Zhang C. Construction of the ceRNA network in the progression of acute myocardial infarction. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2022; 12:283-297. [PMID: 36743510 PMCID: PMC9890199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/11/2022] [Indexed: 02/07/2023]
Abstract
Acute myocardial infarction (AMI) is a common disease that induced by sudden occlusion of a coronary artery and myocardial necrosis, which causes a great medical burden worldwide. Noncoding RNAs, such as circRNA, lncRNA and miRNA, play crucial roles in the progression of cardiovascular diseases. However, the circRNA-miRNA-mRNA network in the occurrence and development of AMI needs further investigation. In this study, we downloaded three AMI datasets, including circRNA (GSE160717), miRNA (GSE24591), and mRNA (GSE66360) from GEO database. The differentially expressed candidates, and GO and KEGG functions were analyzed by RStudio, and subsequently import to PPI and Cytoscape to obtain the hub genes. By using the starbase target prediction database, we further screen the ceRNA network of circRNA-miRNA-mRNA based on the selected differentially expressed candidates. We found 46 differential expressed mRNAs, 65 miRNAs, and five circRNAs. GO functions and KEGG enrichment of the 46 mRNAs focused on immune response and functions, involving IL-17 signaling pathway, Toll-like receptor signaling pathway, cytokine-cytokine receptor interaction, TNF signaling pathway, chemokine signaling pathway, and NF-kappaB signaling pathway, which may aggravate the pathologies of AMI. PPI and Cytoscape analysis showed 10 hub genes, including TLR2, IL1B, CCL4, CCL3, CCR5, TREM1, CXCL2, NLRP3, CSF3, and CCL20. By using starbase and circinteractome databases, ceRNA network construction showed that circRNA_023461 and circRNA_400027 regulate several miRNA-mRNA axes in AMI. In summary, this study uncovered the circRNA-miRNA-mRNA network based on three AMI datasets. The differentially expressed genes, including CCL20, CCL4, CSF3, and IL1B, focus on immune functions and pathways. Furthermore, circRNA_023461 and circRNA_400027 regulate several miRNA-mRNA axes, exerting important roles in AMI progression. Our founding provides new insights into AMI and improve the therapeutic strategies for AMI.
Collapse
|