1
|
Zou E, Xu X, Chen L. Potential of plasma biomarkers for heart failure prediction, management, and prognosis: A multiomics perspective. Heart Fail Rev 2025; 30:55-67. [PMID: 39377997 DOI: 10.1007/s10741-024-10443-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 12/15/2024]
Abstract
Heart failure (HF) remains a major global health challenge, and more effective and comprehensive plasma biomarkers are needed to effectively treat HF patients. Multiomics studies have shown that DNA fragments, noncoding RNAs, proteins, and metabolites may be potential plasma biomarkers for HF. However, comprehensive reviews that focus on research on plasma biomarkers for HF from an omics perspective are lacking. This review summarizes the applications of various omics approaches in the exploration of biomarkers related to the risk assessment, diagnosis, subtype classification, medical management, and prognosis prediction of HF. Moreover, as heart transplantation and left ventricular assistant device (LVAD) implantation are terminal therapies for end-stage HF patients, this review also discusses the role of cell-free DNA as a biomarker for cardiac transplant rejection and omics studies of plasma biomarkers in patients who respond to LVAD therapy. Our findings suggest that future omics research on HF biomarkers should employ integrated multiomics methods and expand the sample size to increase the robustness of the results and that the identified biomarkers should be further validated in large cohorts.
Collapse
Affiliation(s)
- Erhou Zou
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Huang N, Liu C, Liu Z, Lei H. Disulfidptosis-related gene in acute myocardial infarction and immune microenvironment analysis: A bioinformatics analysis and validation. PLoS One 2024; 19:e0314935. [PMID: 39666769 PMCID: PMC11637291 DOI: 10.1371/journal.pone.0314935] [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: 06/07/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024] Open
Abstract
Disulfidptosis is a newly discovered method of cell death. However, no studies have fully elucidated the role of disulfidptosis-related genes (DSRGs) in acute myocardial infarction (AMI). The potential role of DSRGs in AMI was analyzed through a comprehensive bioinformatics approach. Finally, hub genes were verified in vitro by qPCR. Sixteen DE-DSRGs were in the AMI. Thereafter, seven hub genes were determined by machine learning algorithms, which had potential diagnostic value in AMI. The risk model showed a robust diagnostic value (area under curve, AUC = 0.940). Prognostic analysis revealed the potential prognostic value of INF2 and CD2AP. Immune landscape analysis showed that hub genes were closely related to the immune microenvironment. By predictive analysis, we obtained four miRNAs, thirteen small molecule drugs, and five TFs closely related to hub genes. Experimental verification revealed that Slc3a2 and Inf2 were significantly up-regulated and Dstn was significantly down-regulated in the hypoxic model. Our study demonstrated that DSRGs are disorderedly expressed in AMI and identified seven hub genes through machine learning. In addition, a diagnostic model was constructed based on hub genes, providing a new perspective for the early diagnosis of AMI.
Collapse
Affiliation(s)
- Nan Huang
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, Hunan Province, PR China
| | - Chan Liu
- Clinical Pharmacy, Liuyang People’s Hospital, Liuyang, Hunan Province, China
| | - Zheng Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, Hunan Province, PR China
| | - Haibo Lei
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, Hunan Province, PR China
| |
Collapse
|
3
|
Fang C, Sun S, Chen W, Huang D, Wang F, Wei W, Wang W. Bioinformatics analysis of the role of cuproptosis gene in acute myocardial infarction. Minerva Cardiol Angiol 2024; 72:595-606. [PMID: 38842240 DOI: 10.23736/s2724-5683.23.06493-1] [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: 06/07/2024]
Abstract
BACKGROUND Immune infiltration plays a vital role in the course of acute myocardial infarction (AMI). Cuproptosis is a new type of programmed cell death discovered recently. Currently, there is no study on the mechanism of cuproptosis gene regulating immune infiltration in AMI. Therefore, by integrating cuproptosis-related genes and GEO database-related microarray data, this study analyzed the association between cuproptosis genes and immune infiltration and built a risk model. METHODS The GSE59867 was used to extract cuproptosis gene expression profile. The R limma package was used to analyze the differentially expressed genes associated with AMI-Cuproptosis. The risk model was constructed according to AMI-cuproptosis differentially expressed genes. Prediction of AMI-cuproptosis-related gene drugs through Coremine Medical database. The upstream miRNAs were predicted using miRWalk, TargetScan, and miRDB libraries, and a miRNA-mRNA network was constructed. RESULTS Cuproptosis-related genes (DLST, LIAS, DBT, ATP7A, LIPT1, PDHB, GCSH, DLD, DLAT) were down-regulated in AMI patients. One (ATP7B) gene was up-regulated in AMI patients (P<0.05). These 10 Cuproptosis-related genes were significantly associated with immune cell infiltration. Based on these 10 differential genes, the AMI risk prediction model was constructed, and the AUC value was 0.825, among which the abnormal expression of DLST was a risk factor for AMI. Additionally, we also predicted DLAT upstream miRNAs and associated drug targets, finding that 9 miRNAs were upstream of DLST. CONCLUSIONS DLST is a potential cuproptosis gene associated with AMI, but its specific mechanism remains unclear and requires further investigation in future studies.
Collapse
Affiliation(s)
- Chunyun Fang
- Emergency Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shiling Sun
- Emergency Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenjing Chen
- Department of Gastroenterology, Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dongling Huang
- Emergency Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fan Wang
- Emergency Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wanxia Wei
- Emergency Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei Wang
- Emergency Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China -
| |
Collapse
|
4
|
Kosyakovsky LB, de Boer RA, Ho JE. Screening for Heart Failure: Biomarkers to Detect Heightened Risk in the General Population. Curr Heart Fail Rep 2024; 21:591-603. [PMID: 39287754 DOI: 10.1007/s11897-024-00686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE OF REVIEW Heart failure (HF) represents a growing global burden of morbidity and mortality. Identifying individuals at risk for HF development is increasingly important, particularly given the advent of disease-modifying therapies for HF as well as its major risk factors such as obesity actalnd diabetes. We aim to review the key circulating biomarkers associated with future HF which may contribute to HF risk prediction. RECENT FINDINGS While current guidelines recommend the use of natriuretic peptides and cardiac troponins in HF risk stratification, there are a diverse array of other emerging protein, metabolic, transcriptomic, and genomic biomarkers of future HF development. These biomarkers not only lend insight into the underlying pathophysiology of HF, which spans inflammation to cardiac fibrosis, but also offer an opportunity to further refine HF risk in addition to established biomarkers. As evolving techniques in molecular biology enable an increased understanding of the complex biologic contributions to HF pathophysiology, there is an important opportunity to construct integrated clinical and multi-omic models to best capture HF risk. Moving forward, future studies should seek to understand the contributions of sex differences, underlying comorbidity burden, and HF subtypes to an individual's HF risk. Further studies are necessary to fully define the clinical utility of biomarker screening approaches to refine HF risk assessment, as well as to link risk assessment directly to preventive strategies for HF.
Collapse
Affiliation(s)
- Leah B Kosyakovsky
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA
| | - Rudolf A de Boer
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, the Netherlands
| | - Jennifer E Ho
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA.
| |
Collapse
|
5
|
Zhou Q, Shi R, Liu J, Liu Z. Identification and characterization of novel ferroptosis-related genes in acute myocardial infarction. Hum Genomics 2024; 18:123. [PMID: 39538299 PMCID: PMC11562590 DOI: 10.1186/s40246-024-00693-7] [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: 08/26/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a leading cause of death and morbidity worldwide. Ferroptosis, a form of regulated cell death, plays a critical role in modulating immune functions during AMI. This study aimed to identify ferroptosis-related hub genes that could serve as potential therapeutic targets in the progression of AMI. METHODS Bioinformatics was used to identify overlapping genes associated with ferroptosis and the infiltration of 22 immune cells by Cell-type Identification by Estimating Relative Subsets of RNA Transcript (CIBERSORT) analysis. The expression of ferroptosis-related genes in AMI was validated across independent datasets, clinical samples, and in vitro cellular experiments. The predictive value for heart failure was evaluated in the first dimension of principal component analysis (PCA) using receiver operating characteristic (ROC) analysis. RESULTS The study identified 11 key ferroptosis-related genes significantly correlated with immune cell abundance. CIBERSORT analysis highlighted immune dysregulation in AMI. JDP2, DUSP1, TLR4, NFS1, and SLC1A5 were identified as potential biomarkers for AMI progression. Additionally, JDP2, DUSP1, and DDIT4 demonstrated strong predictive value for long-term heart failure. CONCLUSION This study highlights the potential association of ferroptosis-related genes with the pathogenesis of AMI, suggesting a role in the molecular mechanisms that may underlie acute coronary events.
Collapse
Affiliation(s)
- Qiaoyu Zhou
- Department of Cardiovascular Medicine, Postdoctoral Station of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruizheng Shi
- Department of Cardiovascular Medicine, The Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Liu
- Department of Cardiovascular Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoya Liu
- Department of Geriatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
6
|
Kosanam S, Pasupula R. Effect of Methyl Glycoside on Apoptosis and Oxidative Stress in Hypoxia Induced-Reoxygenated H9C2 Cell Lines. Cell Biochem Biophys 2024:10.1007/s12013-024-01539-8. [PMID: 39292425 DOI: 10.1007/s12013-024-01539-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
This study focuses on key genes (Caspase-3, JAK2, BCL2L1 and MAPK8) and their modulation in response to hypoxia-induced stress using Methyl Glycoside (MG), a small molecule spectroscopically screened from Aganosma dichotoma. Hypoxia/reoxygenation (H/R) induced H9C2 cells, pre- treated with MG, were subjected to cell viability assay, free radical scavenging activities (catalase, GST, GSH-Px, SOD), caspase activity, mitochondrial membrane potential, and gene expression profiling through standard assays and molecular techniques. Results indicated that MG treatment, has potential protective effects against H/R induced stress in H9C2 cell lines. Cell viability assays showed that MG maintained cellular viability with significant protection (P < 0.05) observed from 10 µM. Free radical scavenging assays revealed that MG, enhanced detoxification mechanisms and exhibited potential antioxidant effect in a significantly (P < 0.05) in a dose dependant manner. MG pre-treatment in H9C2 cells protected cellular damage from caspase activity, cells exhibited high mitochondrial membrane potential, and gene expression profiles, including upregulation of anti-apoptotic BCL2L1 and modulation of stress-responsive genes like CASP3, JAK2 and MAPK8. Hence, MG exhibited concentration-dependent protective effects on viability, oxidative stress, and apoptosis-related pathways, laying the foundation for further exploration and translational applications in cardiovascular interventions.
Collapse
Affiliation(s)
- Sreya Kosanam
- Department. of Pharmacology, College of Pharmacy, Koneru Lakshmaiah Education Foundation, KL deemed to be University, Green Fields, Vaddeswaram, Andhra Pradesh, India
| | - Rajeshwari Pasupula
- Department. of Pharmacology, College of Pharmacy, Koneru Lakshmaiah Education Foundation, KL deemed to be University, Green Fields, Vaddeswaram, Andhra Pradesh, India.
| |
Collapse
|
7
|
Zhang Z, Zou Z, Zhang H, Zhang DM. Regulatory network analysis based on integrated miRNA-TF reveals key genes in heart failure. Sci Rep 2024; 14:13896. [PMID: 38886500 PMCID: PMC11183224 DOI: 10.1038/s41598-024-64732-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
The etiology and pathophysiology of heart failure are still unknown. Increasing evidence suggests that abnormal microRNAs (miRNAs) and transcription factors (TFs) expression may be associated with the development of heart failure. Therefore, this study aims to explore key miRNAs, TFs, and related genes in heart failure to gain a greater understanding of the pathogenesis of heart failure. To search and download the dataset of mRNA chips related to heart failure from the GEO database (GSE59867, GSE9128, and GSE134766), we analyzed differential genes and screened the common differentially expressed genes on two chips using R language software. The binary interactions and circuits among miRNAs, TFs, and corresponding genes were determined by Pearson correlation coefficient. A regulatory network of miRNAs, TFs, and target genes was constructed based on bioinformatics. By comparing the sequences of patients with and without heart failure, five downregulated genes with hypermethylated mRNA and three upregulated genes with hypomethylated mRNA were identified. The miRNA-TF gene regulatory network consisted of 26 miRNAs, 22 TFs and six genes. GO and KEGG analysis results revealed that BP terms like cellular response to organic substance, cellular response to cytokine stimulus, and KEGG pathways like osteoclast differentiation, MAPK signaling pathway, and legionellosis were enriched of the DEGs. TMEM87A, PPP2R2A, DUSP1, and miR-92a have great potential as biomarkers for heart failure. The integrated analysis of the mRNA expression spectrum and microRNA-transcription factor-gene revealed the regulatory network of heart failure, which may provide clues to its alternative treatment.
Collapse
Affiliation(s)
- Ziyue Zhang
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Road, Nanjing, 211112, Jiangsu, People's Republic of China
| | - Ziying Zou
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Road, Nanjing, 211112, Jiangsu, People's Republic of China
| | - Hui Zhang
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Road, Nanjing, 211112, Jiangsu, People's Republic of China
| | - Dai-Min Zhang
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Road, Nanjing, 211112, Jiangsu, People's Republic of China.
| |
Collapse
|
8
|
Gao Y, Chen B, Han Y, Lu J, Li X, Tian A, Zhang L, Wang B, Hong Y, Liu J, Li Y, Bilige W, Zhang H, Zheng X, Li J. Prognostic Value of a Multi-mRNA Signature for 1-Year All-Cause Death in Hospitalized Patients With Heart Failure With a Preserved Ejection Fraction. Circ Heart Fail 2024; 17:e011118. [PMID: 38847104 DOI: 10.1161/circheartfailure.123.011118] [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] [Received: 08/07/2023] [Accepted: 04/26/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Heart failure with preserved ejection fraction is a major global public health problem, while effective risk stratification tools are still lacking. We sought to construct a multi-mRNA signature to predict 1-year all-cause death. METHODS We selected 30 patients with heart failure with preserved ejection fraction who died during 1-year follow-up and 30 who survived in the discovery set. One hundred seventy-one and 120 patients with heart failure with preserved ejection fraction were randomly selected as a test set and a validation set, respectively. We performed mRNA microarrays in all patients. RESULTS We constructed a 5-mRNA signature for predicting 1-year all-cause death. The scores of the 5-mRNA signature were significantly associated with the 1-year risk of all-cause death in both the test set (hazard ratio, 2.72 [95% CI, 1.98-3.74]; P<0.001) and the validation set (hazard ratio, 3.95 [95% CI, 2.40-6.48]; P<0.001). Compared with a reference model, which included sex, ASCEND-HF (Acute Study of Clinical Effectiveness of Nesiritide in Decompensated Heart Failure) score, history of HF and NT-proBNP (N-terminal pro-B-type natriuretic peptide), the 5-mRNA signature had a better discrimination capability, with an increased area under the curve from 0.696 to 0.813 in the test set and from 0.712 to 0.848 in the validation set. A composite model integrating the 5-mRNA risk score and variables in the reference model demonstrated an excellent discrimination capability, with an area under the curve of 0.861 (95% CI, 0.784-0.939) in the test set and an area under the curve of 0.859 (95% CI, 0.755-0.963) in the validation set. The net reclassification improvement and integrated discrimination improvement indicated that the composite model significantly improved patient classification compared with the reference model in both sets (P<0.001). CONCLUSIONS The 5-mRNA signature is a promising predictive tool for 1-year all-cause death and shows improved prognostic power over the established risk scores and NT-proBNP in patients with heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Yan Gao
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Bowang Chen
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Yi Han
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Jiapeng Lu
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Xi Li
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Aoxi Tian
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Lihua Zhang
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Bin Wang
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Yun Hong
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Jiamin Liu
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Yan Li
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Wuhan Bilige
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Haibo Zhang
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Xin Zheng
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
| | - Jing Li
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China (Y.G., B.C., Y. Han, J. Lu, X. L., A.T., L.Z., B.W., Y. Hong, J. Liu, Y.L., W.B., H.Z., X.Z., J. Li)
- Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University (J. Li)
| |
Collapse
|
9
|
Zhao LZ, Liang Y, Yin T, Liao HL, Liang B. Identification of Potential Crucial Biomarkers in STEMI Through Integrated Bioinformatic Analysis. Arq Bras Cardiol 2024; 121:e20230462. [PMID: 38597542 DOI: 10.36660/abc.20230462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/14/2023] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND ST-segment elevation myocardial infarction (STEMI) is one of the leading causes of fatal cardiovascular diseases, which have been the prime cause of mortality worldwide. Diagnosis in the early phase would benefit clinical intervention and prognosis, but the exploration of the biomarkers of STEMI is still lacking. OBJECTIVES In this study, we conducted a bioinformatics analysis to identify potential crucial biomarkers in the progress of STEMI. METHODS We obtained GSE59867 for STEMI and stable coronary artery disease (SCAD) patients. Differentially expressed genes (DEGs) were screened with the threshold of |log2fold change| > 0.5 and p <0.05. Based on these genes, we conducted enrichment analysis to explore the potential relevance between genes and to screen hub genes. Subsequently, hub genes were analyzed to detect related miRNAs and DAVID to detect transcription factors for further analysis. Finally, GSE62646 was utilized to assess DEGs specificity, with genes demonstrating AUC results exceeding 75%, indicating their potential as candidate biomarkers. RESULTS 133 DEGs between SCAD and STEMI were obtained. Then, the PPI network of DEGs was constructed using String and Cytoscape, and further analysis determined hub genes and 6 molecular complexes. Functional enrichment analysis of the DEGs suggests that pathways related to inflammation, metabolism, and immunity play a pivotal role in the progression from SCAD to STEMI. Besides, related-miRNAs were predicted, has-miR-124, has-miR-130a/b, and has-miR-301a/b regulated the expression of the largest number of genes. Meanwhile, Transcription factors analysis indicate that EVI1, AML1, GATA1, and PPARG are the most enriched gene. Finally, ROC curves demonstrate that MS4A3, KLRC4, KLRD1, AQP9, and CD14 exhibit both high sensitivity and specificity in predicting STEMI. CONCLUSIONS This study revealed that immunity, metabolism, and inflammation are involved in the development of STEMI derived from SCAD, and 6 genes, including MS4A3, KLRC4, KLRD1, AQP9, CD14, and CCR1, could be employed as candidate biomarkers to STEMI.
Collapse
Affiliation(s)
- Li-Zhi Zhao
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou - China
- College of Integration of Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou - China
| | - Yi Liang
- Department of Geriatrics, Sichuan Second Hospital of T.C.M., Chengdu - China
| | - Ting Yin
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou - China
| | - Hui-Ling Liao
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou - China
- College of Integration of Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou - China
| | - Bo Liang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing - China
| |
Collapse
|
10
|
Yuan H, Zhang P, Xin Y, Liu Z, Gao B. Single cell RNA-seq identifies a FOS/JUN-related monocyte signature associated with clinical response of heart failure patients with mesenchymal stem cell therapy. Aging (Albany NY) 2024; 16:5651-5675. [PMID: 38517374 PMCID: PMC11006470 DOI: 10.18632/aging.205670] [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: 10/16/2023] [Accepted: 02/07/2024] [Indexed: 03/23/2024]
Abstract
Heart failure (HF) is a serious global health issue that demands innovative treatment approaches. In this study, we collected samples from 4 HF patients before and after MSC therapy and performed scRNA-seq. After the MSC therapy, the proportion of CD14+ monocytes decreased significantly in both the treatment response and non-response groups, with a more pronounced decrease in the treatment response group. The therapy-response and non-response group were clearly separated in the UMAP plot, while the CD14+ monocytes in the therapy-response group before and after MSC therapy were very similar, but there were significant differences in the non-response group. By further performing NMF analysis, we identified 11 subsets of CD14+ monocytes. More importantly, we identified a therapy-related CD14+ monocyte subpopulation. The predictive model based on CD14+ monocytes constructed by machine learning algorithms showed good performance. Moreover, genes such as FOS were highly enriched in the therapy-related CD14+ monocytes. The SCENIC analysis revealed potential regulatory factors for this treatment-responsive CD14+ monocytes, and FOS/JUN were identified as potential core indicators/regulators. Finally, HF patients were divided into three groups by NMF analysis, and the therapy-responsive CD14+ monocyte characteristics were differentially activated among the three groups. Together, this study identifies treatment-responsive CD14+ monocytes as a crucial biomarker for assessing the suitability of MSC therapy and determining which HF patients could benefit from it. This provides new clues for further investigating the therapeutic mechanisms of MSC therapy, offering beneficial insights for personalized treatment and improving prognosis in HF patients.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengfei Zhang
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuanfeng Xin
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Zhongmin Liu
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Bingren Gao
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
- Cardiopulmonary Vascular Center, Haikang Hospital, Xingguang Island, West Coast New Area, Qingdao 266400, Shandong, China
| |
Collapse
|
11
|
Chao P, Zhang X, Zhang L, Wang Y, Wusiman M, Aimaijiang G, Chen X, Yang Y. Characterization of the m 6A regulators' landscape highlights the clinical significance of acute myocardial infarction. Front Immunol 2024; 15:1308978. [PMID: 38571952 PMCID: PMC10987706 DOI: 10.3389/fimmu.2024.1308978] [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: 10/07/2023] [Accepted: 02/13/2024] [Indexed: 04/05/2024] Open
Abstract
Objective Acute myocardial infarction (AMI) is a severe cardiovascular disease that threatens human life and health globally. N6-methyladenosine (m6A) governs the fate of RNAs via m6A regulators. Nevertheless, how m6A regulators affect AMI remains to be deciphered. To solve this issue, an integrative analysis of m6A regulators in AMI was conducted. Methods We acquired transcriptome profiles (GSE59867, GSE48060) of peripheral blood samples from AMI patients and healthy controls. Key m6A regulators were used for LASSO, and consensus clustering was conducted. Next, the m6A score was also computed. Immune cell infiltration, ferroptosis, and oxidative stress were evaluated. In-vitro and in-vivo experiments were conducted to verify the role of the m6A regulator ALKBH5 in AMI. Results Most m6A regulators presented notable expression alterations in circulating cells of AMI patients versus those of controls. Based on key m6A regulators, we established a gene signature and a nomogram for AMI diagnosis and risk prediction. AMI patients were classified into three m6A clusters or gene clusters, respectively, and each cluster possessed the unique properties of m6A modification, immune cell infiltration, ferroptosis, and oxidative stress. Finally, the m6A score was utilized to quantify m6A modification patterns. Therapeutic targeting of ALKBH5 greatly alleviated apoptosis and intracellular ROS in H/R-induced H9C2 cells and NRCMs. Conclusion Altogether, our findings highlight the clinical significance of m6A regulators in the diagnosis and risk prediction of AMI and indicate the critical roles of m6A modification in the regulation of immune cell infiltration, ferroptosis, and oxidative stress.
Collapse
Affiliation(s)
- Peng Chao
- Department of Cardiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xueqin Zhang
- Department of Nephrology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Lei Zhang
- Department of Endocrinology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yong Wang
- Department of Cardiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Miriban Wusiman
- Department of Nephrology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Gulizere Aimaijiang
- Department of Nephrology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xiaoyang Chen
- Department of Cardiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yining Yang
- Department of Cardiology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
12
|
Li Y, Hu Y, Jiang F, Chen H, Xue Y, Yu Y. Combining WGCNA and machine learning to identify mechanisms and biomarkers of ischemic heart failure development after acute myocardial infarction. Heliyon 2024; 10:e27165. [PMID: 38455553 PMCID: PMC10918227 DOI: 10.1016/j.heliyon.2024.e27165] [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: 07/10/2023] [Revised: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Background Ischemic heart failure (IHF) is a serious complication after acute myocardial infarction (AMI). Understanding the mechanism of IHF after AMI will help us conduct early diagnosis and treatment. Methods We obtained the AMI dataset GSE66360 and the IHF dataset GSE57338 from the GEO database, and screened overlapping genes common to both diseases through WGCNA analysis. Subsequently, we performed GO and KEGG enrichment analysis on overlapping genes to elucidate the common mechanism of AMI and IHF. Machine learning algorithms are also used to identify key biomarkers. Finally, we performed immune cell infiltration analysis on the dataset to further evaluate immune cell changes in AMI and IHF. Results We obtained 74 overlapping genes of AMI and IHF through WGCNA analysis, and the enrichment analysis results mainly focused on immune and inflammation-related mechanisms. Through the three machine learning algorithms of LASSO, RF and SVM-RFE, we finally obtained the four Hub genes of IL1B, TIMP2, IFIT3, and P2RY2, and verified them in the IHF dataset GSE116250, and the diagnostic model AUC = 0.907. The results of immune infiltration analysis showed that 8 types of immune cells were significantly different in AMI samples, and 6 types of immune cells were significantly different in IHF samples. Conclusion We explored the mechanism of IHF after AMI by WGCNA, enrichment analysis, and immune infiltration analysis. Four potential diagnostic candidate genes and therapeutic targets were identified by machine learning algorithms. This provides a new idea for the pathogenesis, diagnosis, and treatment of IHF after AMI.
Collapse
Affiliation(s)
- Yan Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Ying Hu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Feng Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Haoyu Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| |
Collapse
|
13
|
Frisk C, Das S, Eriksson MJ, Walentinsson A, Corbascio M, Hage C, Kumar C, Ekström M, Maret E, Persson H, Linde C, Persson B. Cardiac biopsies reveal differences in transcriptomics between left and right ventricle in patients with or without diagnostic signs of heart failure. Sci Rep 2024; 14:5811. [PMID: 38461325 PMCID: PMC10924960 DOI: 10.1038/s41598-024-56025-1] [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: 12/18/2023] [Accepted: 02/29/2024] [Indexed: 03/11/2024] Open
Abstract
New or mild heart failure (HF) is mainly caused by left ventricular dysfunction. We hypothesised that gene expression differ between the left (LV) and right ventricle (RV) and secondly by type of LV dysfunction. We compared gene expression through myocardial biopsies from LV and RV of patients undergoing elective coronary bypass surgery (CABG). Patients were categorised based on LV ejection fraction (EF), diastolic function and NT-proBNP into pEF (preserved; LVEF ≥ 45%), rEF (reduced; LVEF < 45%) or normal LV function. Principal component analysis of gene expression displayed two clusters corresponding to LV and RV. Up-regulated genes in LV included natriuretic peptides NPPA and NPPB, transcription factors/coactivators STAT4 and VGLL2, ion channel related HCN2 and LRRC38 associated with cardiac muscle contraction, cytoskeleton, and cellular component movement. Patients with pEF phenotype versus normal differed in gene expression predominantly in LV, supporting that diastolic dysfunction and structural changes reflect early LV disease in pEF. DKK2 was overexpressed in LV of HFpEF phenotype, potentially leading to lower expression levels of β-catenin, α-SMA (smooth muscle actin), and enhanced apoptosis, and could be a possible factor in the development of HFpEF. CXCL14 was down-regulated in both pEF and rEF, and may play a role to promote development of HF.
Collapse
Affiliation(s)
- Christoffer Frisk
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, 751 24, Uppsala, Sweden
| | - Sarbashis Das
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, 751 24, Uppsala, Sweden
| | - Maria J Eriksson
- Department of Clinical Physiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Anna Walentinsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83, Gothenburg, Sweden
| | - Matthias Corbascio
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Thoracic Surgery, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Camilla Hage
- Department of Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Chanchal Kumar
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83, Gothenburg, Sweden
- Department of Medicine, Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, 141 57, Huddinge, Sweden
| | - Mattias Ekström
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, 182 88, Stockholm, Sweden
- Department of Cardiology, Danderyd Hospital, 182 88, Stockholm, Sweden
| | - Eva Maret
- Department of Clinical Physiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Hans Persson
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, 182 88, Stockholm, Sweden
- Department of Cardiology, Danderyd Hospital, 182 88, Stockholm, Sweden
| | - Cecilia Linde
- Department of Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Bengt Persson
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, 751 24, Uppsala, Sweden.
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
14
|
Tang X, Zhou Y, Chen Z, Liu C, Wu Z, Zhou Y, Zhang F, Lu X, Tang L. Identification of key biomarkers for predicting CAD progression in inflammatory bowel disease via machine-learning and bioinformatics strategies. J Cell Mol Med 2024; 28:e18175. [PMID: 38451044 PMCID: PMC10919158 DOI: 10.1111/jcmm.18175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/07/2024] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
The study aimed to identify the biomarkers for predicting coronary atherosclerotic lesions progression in patients with inflammatory bowel disease (IBD). Related transcriptome datasets were seized from Gene Expression Omnibus database. IBD-related modules were identified via Weighted Gene Co-expression Network Analysis. The 'Limma' was applied to screen differentially expressed genes between stable coronary artery disease (CAD) and acute myocardial infarction (AMI). Subsequently, we employed protein-protein interaction (PPI) network and three machine-learning strategies to further screen for candidate hub genes. Application of the receiver operating characteristics curve to quantitatively evaluate candidates to determine key diagnostic biomarkers, followed by a nomogram construction. Ultimately, we performed immune landscape analysis, single-gene GSEA and prediction of target-drugs. 3227 IBD-related module genes and 570 DEGs accounting for AMI were recognized. Intersection yielded 85 shared genes and mostly enriched in immune and inflammatory pathways. After filtering through PPI network and multi-machine learning algorithms, five candidate genes generated. Upon validation, CTSD, CEBPD, CYP27A1 were identified as key diagnostic biomarkers with a superior sensitivity and specificity (AUC > 0.8). Furthermore, all three genes were negatively correlated with CD4+ T cells and positively correlated with neutrophils. Single-gene GSEA highlighted the importance of pathogen invasion, metabolism, immune and inflammation responses during the pathogenesis of AMI. Ten target-drugs were predicted. The discovery of three peripheral blood biomarkers capable of predicting the risk of CAD proceeding into AMI in IBD patients. These identified biomarkers were negatively correlated with CD4+ T cells and positively correlated with neutrophils, indicating a latent therapeutic target.
Collapse
Affiliation(s)
- Xiaoqi Tang
- School of MedicineShaoxing UniversityZhejiangChina
| | - Yufei Zhou
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan UniversityShanghaiChina
| | - Zhuolin Chen
- Department of OrthopedicsShaoxing People's Hospital (Zhejiang University School of Medicine)ShaoxingChina
| | - Chunjiang Liu
- Department of General Surgery, Division of Vascular SurgeryShaoxing People's HospitalShaoxingChina
| | - Zhifeng Wu
- School of MedicineShaoxing UniversityZhejiangChina
| | - Yue Zhou
- Department of General Surgery, Division of Vascular SurgeryShaoxing People's HospitalShaoxingChina
| | - Fan Zhang
- School of MedicineShaoxing UniversityZhejiangChina
| | - Xuanyuan Lu
- Department of OrthopedicsShaoxing People's Hospital (Zhejiang University School of Medicine)ShaoxingChina
| | - Liming Tang
- Department of General Surgery, Division of Vascular SurgeryShaoxing People's HospitalShaoxingChina
| |
Collapse
|
15
|
Hu Y, Chen X, Mei X, Luo Z, Wu H, Zhang H, Zeng Q, Ren H, Xu D. Identification of diagnostic immune-related gene biomarkers for predicting heart failure after acute myocardial infarction. Open Med (Wars) 2023; 18:20230878. [PMID: 38152337 PMCID: PMC10751901 DOI: 10.1515/med-2023-0878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/02/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023] Open
Abstract
Post-myocardial infarction heart failure (HF) is a major public health concern. Previous studies have reported the critical role of immune response in HF pathogenesis. However, limited studies have reported predictive immune-associated biomarkers for HF. So we attempted to identify potential immune-related indicators for HF early diagnosis and therapy guidance. This study identified two potential immune-related hub genes (IRHGs), namely CXCR5 and FOS, using bioinformatic approaches. The expression levels of CXCR5 and FOS and their ability to predict long-term HF were analyzed. Functional enrichment analysis revealed that the hub genes were enriched in immune system processes, including the interleukin-17 and nuclear factor-kappa B signaling pathways, which are involved in the pathogenesis of HF. Quantitative real-time polymerase chain reaction revealed that the Fos mRNA levels, but not the Cxcr5 mRNA levels, were downregulated in the mice of the HF group. This study successfully identified two IRHGs that were significantly and differentially expressed in the HF group and could predict long-term HF, providing novel insights for future studies on HF and developing novel therapeutic targets for HF.
Collapse
Affiliation(s)
- Yingchun Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaoyu Chen
- Department of Nephrology, Rheumatism and Immunology, Chongqing Jiulongpo People’s Hospital, Chongqing, 400050, China
| | - Xiyuan Mei
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hongguang Wu
- Department of Arrhythmic, Cardiovascular Medical Center, Shenzhen Hospital of University of Hong Kong, Shenzhen, 518040, Guangdong, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qingchun Zeng
- Department of Cardiology, Nanfang Hospital, Southern Medical University,
Guangzhou, 510515, Guangdong, China
| | - Hao Ren
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University,
Guangzhou, 510515, Guangdong, China
| |
Collapse
|
16
|
Hou Q, Sun Z, Zhao L, Liu Y, Zhang J, Huang J, Luo Y, Xiao Y, Hu Z, Shen A. Role of serum cytokines in the prediction of heart failure in patients with coronary artery disease. ESC Heart Fail 2023; 10:3102-3113. [PMID: 37608687 PMCID: PMC10567644 DOI: 10.1002/ehf2.14491] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/12/2023] [Accepted: 07/16/2023] [Indexed: 08/24/2023] Open
Abstract
AIMS Coronary artery disease (CAD) is the most common cause of heart failure (HF). This study aimed to identify cytokine biomarkers for predicting HF in patients with CAD. METHODS AND RESULTS Twelve patients with CAD without HF (CAD-non HF), 12 patients with CAD complicated with HF (CAD-HF), and 12 healthy controls were enrolled for Human Cytokine Antibody Array, which were used as the training dataset. Then, differentially expressed cytokines among the different groups were identified, and crucial characteristic proteins related to CAD-HF were screened using a combination of the least absolute shrinkage and selection operator, recursive feature elimination, and random forest methods. A support vector machine (SVM) diagnostic model was constructed based on crucial characteristic proteins, followed by receiver operating characteristic curve analysis. Finally, two validation datasets, GSE20681 and GSE59867, were downloaded to verify the diagnostic performance of the SVM model and expression of crucial proteins, as well as enzyme-linked immunosorbent assay was also used to verify the levels of crucial proteins in blood samples. In total, 12 differentially expressed proteins were overlapped in the three comparison groups, and then four optimal characteristic proteins were identified, including VEGFR2, FLRG, IL-23, and FGF-21. After that, the area under the receiver operating characteristic curve of the constructed SVM classification model for the training dataset was 0.944. The accuracy of the SVM classification model was validated using the GSE20681 and GSE59867 datasets, with area under the receiver operating characteristic curve values of 0.773 and 0.745, respectively. The expression trends of the four crucial proteins in the training dataset were consistent with those in the validation dataset and those determined by enzyme-linked immunosorbent assay. CONCLUSIONS The combination of VEGFR2, FLRG, IL-23, and FGF-21 can be used as a candidate biomarker for the prediction and prevention of HF in patients with CAD.
Collapse
Affiliation(s)
- Qingzhen Hou
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhuhua Sun
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Liqin Zhao
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Ye Liu
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Junfang Zhang
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Jing Huang
- Department of Laboratory Medicine, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Yifeng Luo
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Yan Xiao
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhaoting Hu
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Anna Shen
- Department of Cardiology, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
17
|
Yang J, Shangguan Q, Xie G, Yang M, Sheng G. M6A regulator methylation patterns and characteristics of immunity in acute ST-segment elevation myocardial infarction. Sci Rep 2023; 13:15688. [PMID: 37735234 PMCID: PMC10514189 DOI: 10.1038/s41598-023-42959-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/16/2023] [Indexed: 09/23/2023] Open
Abstract
M6A methylation is the most prevalent and abundant RNA modification in mammals. Although there are many studies on the regulatory role of m6A methylation in the immune response, the m6A regulators in the pathogenesis of acute ST-segment elevation myocardial infarction (STEMI) remain unclear. We comprehensively analysed the role of m6A regulators in STEMI and built a predictive model, revealing the relationship between m6A methylations and the immune microenvironment. Differential analysis revealed that 18 of 24 m6A regulators were significantly differentially expressed, and there were substantial interactions between the m6A regulator. Then, we established a classifier and nomogram model based on 6 m6A regulators, which can easily distinguish the STEMI and control samples. Finally, two distinct m6A subtypes were obtained and significantly differentially expressed in terms of infiltrating immunocyte abundance, immune reaction activity and human leukocyte antigen genes. Three hub m6A phenotype related genes (RAC2, RELA, and WAS) in the midnightblue module were identified by weighted gene coexpression network analysis, and were associated with immunity. These findings suggest that m6A modification and the immune microenvironment play a key role in the pathogenesis of STEMI.
Collapse
Affiliation(s)
- Jingqi Yang
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, China
| | - Qing Shangguan
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, China
| | - Guobo Xie
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, China
| | - Ming Yang
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, China.
| | - Guotai Sheng
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, 152 Aiguo Road, Nanchang, China
| |
Collapse
|
18
|
Zhang G, Cui X, Qin Z, Wang Z, Lu Y, Xu Y, Xu S, Tang L, Zhang L, Liu G, Wang X, Zhang J, Tang J. Atherosclerotic plaque vulnerability quantification system for clinical and biological interpretability. iScience 2023; 26:107587. [PMID: 37664595 PMCID: PMC10470306 DOI: 10.1016/j.isci.2023.107587] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 05/02/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Acute myocardial infarction dominates coronary artery disease mortality. Identifying bio-signatures for plaque destabilization and rupture is important for preventing the transition from coronary stability to instability and the occurrence of thrombosis events. This computational systems biology study enrolled 2,235 samples from 22 independent bulks cohorts and 14 samples from two single-cell cohorts. A machine-learning integrative program containing nine learners was developed to generate a warning classifier linked to atherosclerotic plaque vulnerability signature (APVS). The classifier displays the reliable performance and robustness for distinguishing ST-elevation myocardial infarction from chronic coronary syndrome at presentation, and revealed higher accuracy to 33 pathogenic biomarkers. We also developed an APVS-based quantification system (APVSLevel) for comprehensively quantifying atherosclerotic plaque vulnerability, empowering early-warning capabilities, and accurate assessment of atherosclerosis severity. It unraveled the multidimensional dysregulated mechanisms at high resolution. This study provides a potential tool for macro-level differential diagnosis and evaluation of subtle genetic pathological changes in atherosclerosis.
Collapse
Affiliation(s)
- Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Xiaolin Cui
- School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen 518172, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Zeyu Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Yongzheng Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Yanyan Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Shuai Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Laiyi Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Gangqiong Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Xiaofang Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| |
Collapse
|
19
|
Samadishadlou M, Rahbarghazi R, Piryaei Z, Esmaeili M, Avcı ÇB, Bani F, Kavousi K. Unlocking the potential of microRNAs: machine learning identifies key biomarkers for myocardial infarction diagnosis. Cardiovasc Diabetol 2023; 22:247. [PMID: 37697288 PMCID: PMC10496209 DOI: 10.1186/s12933-023-01957-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/10/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) play a crucial role in regulating adaptive and maladaptive responses in cardiovascular diseases, making them attractive targets for potential biomarkers. However, their potential as novel biomarkers for diagnosing cardiovascular diseases requires systematic evaluation. METHODS In this study, we aimed to identify a key set of miRNA biomarkers using integrated bioinformatics and machine learning analysis. We combined and analyzed three gene expression datasets from the Gene Expression Omnibus (GEO) database, which contains peripheral blood mononuclear cell (PBMC) samples from individuals with myocardial infarction (MI), stable coronary artery disease (CAD), and healthy individuals. Additionally, we selected a set of miRNAs based on their area under the receiver operating characteristic curve (AUC-ROC) for separating the CAD and MI samples. We designed a two-layer architecture for sample classification, in which the first layer isolates healthy samples from unhealthy samples, and the second layer classifies stable CAD and MI samples. We trained different machine learning models using both biomarker sets and evaluated their performance on a test set. RESULTS We identified hsa-miR-21-3p, hsa-miR-186-5p, and hsa-miR-32-3p as the differentially expressed miRNAs, and a set including hsa-miR-186-5p, hsa-miR-21-3p, hsa-miR-197-5p, hsa-miR-29a-5p, and hsa-miR-296-5p as the optimum set of miRNAs selected by their AUC-ROC. Both biomarker sets could distinguish healthy from not-healthy samples with complete accuracy. The best performance for the classification of CAD and MI was achieved with an SVM model trained using the biomarker set selected by AUC-ROC, with an AUC-ROC of 0.96 and an accuracy of 0.94 on the test data. CONCLUSIONS Our study demonstrated that miRNA signatures derived from PBMCs could serve as valuable novel biomarkers for cardiovascular diseases.
Collapse
Affiliation(s)
- Mehrdad Samadishadlou
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeynab Piryaei
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Mahdad Esmaeili
- Medical Bioengineering Department, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Çığır Biray Avcı
- Medical Biology Department, School of Medicine, Ege University, İzmir, Türkiye
| | - Farhad Bani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kaveh Kavousi
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
20
|
Liang WL, Liao HL, Liang B. Immune landscape and regulatory mechanisms in human atherosclerotic coronary plaques: Evidence from single-cell and bulk transcriptomics. Heliyon 2023; 9:e19392. [PMID: 37674826 PMCID: PMC10477495 DOI: 10.1016/j.heliyon.2023.e19392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/10/2023] [Accepted: 08/21/2023] [Indexed: 09/08/2023] Open
Abstract
Atherosclerosis is a chronic immuno-inflammatory disease, however, the immune landscape and regulatory mechanisms have not been clear. We detected seven principal immune cell clusters with distinct phenotypic and spatial characteristics using single-cell RNA-sequencing of aortic immune cells from patients with acute coronary syndrome and stable angina pectoris. Then we acquired 265 differentially expressed immune-related genes and the high scores were mainly found in T cells and monocytes, which were differentially regulated in atherosclerotic coronary plaques. The CCL signaling pathway was the most relevant pattern in the T cells and CCL5-CCR1 and CCL5-CCR5 ligand-receptor pairs played a vital role in the CCL signaling pathway. Further comparative analysis indicated MCH-I signaling was the most relevant pattern in the T cells and HLA ligand-related ligand-receptor pairs played a vital role. Functional analysis of the single-cell and bulk transcriptomics pointed to multiple pathways, such as antigen presentation and immune response. Nineteen common differentially expressed immune-related genes were found in both immune cells and the human peripheral blood mononuclear cells. Nine common differentially expressed transcription factors were differentially expressed in both T cell and monocyte clusters from the coronary plaques and human peripheral blood mononuclear cells and the network demonstrated that CEBPB might play an essential role in the transcriptional regulation of atherosclerosis as a hub transcription factor. The definition of immune cell diversity and heterogeneity by single-cell level analysis of aortic immune cell subsets not only unveils cell-type-specific pathways and new immune mechanisms but also discovers the functional correlation of immune cells in human atherosclerosis. Our findings provide great promise for the discovery of novel molecular mechanisms and precise therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Wei-Lin Liang
- Department of Cardiology, Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan, China
| | - Hui-Ling Liao
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- College of Integration of Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Bo Liang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
21
|
Liu W, Li Y, Zhang Y, Li S, Chen Y, Han B, Lu Y. Identification of biomarkers and immune infiltration in acute myocardial infarction and heart failure by integrated analysis. Biosci Rep 2023; 43:BSR20222552. [PMID: 37334672 PMCID: PMC10329185 DOI: 10.1042/bsr20222552] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/24/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023] Open
Abstract
The mortality of heart failure after acute myocardial infarction (AMI) remains high. The aim of the present study was to analyze hub genes and immune infiltration in patients with AMI and heart failure (HF). The study utilized five publicly available gene expression datasets from peripheral blood in patients with AMI who either developed or did not develop HF. The unbiased patterns of 24 immune cell were estimated by xCell algorithm. Single-cell RNA sequencing data were used to examine the immune cell infiltration in heart failure patients. Hub genes were validated by quantitative reverse transcription-PCR (RT-qPCR). In comparison with the coronary heart disease (CHD) group, immune infiltration analysis of AMI patients showed that macrophages M1, macrophages, monocytes, natural killer (NK) cells, and NKT cells were the five most highly activated cell types. Five common immune-related genes (S100A12, AQP9, CSF3R, S100A9, and CD14) were identified as hub genes associated with AMI. Using RT-qPCR, we confirmed FOS, DUSP1, CXCL8, and NFKBIA as the potential biomarkers to identify AMI patients at risk of HF. The study identified several transcripts that differentiate between AMI and CHD, and between HF and non-HF patients. These findings could improve our understanding of the immune response in AMI and HF, and allow for early identification of AMI patients at risk of HF.
Collapse
Affiliation(s)
- Wei Liu
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Institute of Cardiovascular Disease, Xuzhou Clinical School of Nanjing Medical University, No. 199 Jiefang South Road, Xuzhou 221009, PR China
| | - Yuling Li
- Department of Ultrasonography, Xuzhou Central Hospital, Xuzhou Clinical School of Nanjing Medical University, No. 199 Jiefang South Road, Xuzhou 221009, PR China
| | - Yan Zhang
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou Clinical School of Nanjing Medical University, No. 199 Jiefang South Road, Xuzhou 221009, PR China
| | - Su Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuqiong Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Bing Han
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Institute of Cardiovascular Disease, Xuzhou Clinical School of Nanjing Medical University, No. 199 Jiefang South Road, Xuzhou 221009, PR China
| | - Yao Lu
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Institute of Cardiovascular Disease, Xuzhou Clinical School of Nanjing Medical University, No. 199 Jiefang South Road, Xuzhou 221009, PR China
| |
Collapse
|
22
|
Zhang M, Li J, Hua C, Niu J, Liu P, Zhong G. Exploring an immune cells-related molecule in STEMI by bioinformatics analysis. BMC Med Genomics 2023; 16:151. [PMID: 37391746 PMCID: PMC10311814 DOI: 10.1186/s12920-023-01579-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/11/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND ST-elevated myocardial infarction (STEMI) is the leading cause of mortality worldwide. The mortality rate of heart attacks has decreased due to various preventive factors and the development of early diagnostic resuscitation measures, but the long-term prognosis remains poor. The present study aimed to identify novel serum biomarkers in STEMI patients and explored a possible new mechanism of STEMI from an immune molecular angle with bioinformatics analysis. METHODS Gene expression profiles were obtained from Gene Expression Omnibus (GEO) database. Differential gene analysis, machine learning algorithms, gene set enrichment analysis, and immune cell infiltration analysis were conducted using R software. RESULTS We identified 146 DEGs (differentially expressed genes) in the integrated dataset between the STEMI and CAD (coronary artery disease) groups. Immune infiltration analysis indicated that eleven cell types were differentially infiltrated. Through correlation analysis, we further screened 25 DEGs that showed a high correlation with monocytes and neutrophils. Afterwards, five genes consistently selected by all three machine learning algorithms were considered candidate genes. Finally, we identified a hub gene (ADM) as a biomarker of STEMI. AUC curves showed that ADM had more than 80% high accuracy in all datasets. CONCLUSIONS In this study, we explored a potentially new mechanism of STEMI from an immune molecular perspective, which might provide insights into the pathogenesis of STEMI. ADM positively correlated with monocytes and neutrophils, suggesting its potential role in the immune response during STEMI. Additionally, we validated the diagnostic performance of ADM in two external datasets, which could help to develop new diagnostic tools or therapeutic strategies.
Collapse
Affiliation(s)
- Min Zhang
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiaxing Li
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Cuncun Hua
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiayin Niu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Pengfei Liu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangzhen Zhong
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Yang Z, Sun L, Wang H. Identification of mitophagy-related genes with potential clinical utility in myocardial infarction at transcriptional level. Front Cardiovasc Med 2023; 10:1166324. [PMID: 37304955 PMCID: PMC10250750 DOI: 10.3389/fcvm.2023.1166324] [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: 02/16/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Background Myocardial infarction (MI) ranks among the most prevalent cardiovascular diseases. Insufficient blood flow to the coronary arteries always leads to ischemic necrosis of the cardiac muscle. However, the mechanism of myocardial injury after MI remains unclear. This article aims to explore the potential common genes between mitophagy and MI and to construct a suitable prediction model. Methods Two Gene Expression Omnibus (GEO) datasets (GSE62646 and GSE59867) were used to screen the differential expression genes in peripheral blood. SVM, RF, and LASSO algorithm were employed to find MI and mitophagy-related genes. Moreover, DT, KNN, RF, SVM and LR were conducted to build the binary models, and screened the best model to further external validation (GSE61144) and internal validation (10-fold cross validation and Bootstrap), respectively. The performance of various machine learning models was compared. In addition, immune cell infiltration correlation analysis was conducted with MCP-Counter and CIBERSORT. Results We finally identified ATG5, TOMM20, MFN2 transcriptionally differed between MI and stable coronary artery diseases. Both internal and external validation supported that these three genes could accurately predict MI withAUC = 0.914 and 0.930 by logistic regression, respectively. Additionally, functional analysis suggested that monocytes and neutrophils might be involved in mitochondrial autophagy after myocardial infarction. Conclusion The data showed that the transcritional levels of ATG5, TOMM20 and MFN2 in patients with MI were significantly different from the control group, which might be helpful to further accurately diagnose diseases and have potential application value in clinical practice.
Collapse
Affiliation(s)
- Zhikai Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Liang Sun
- The NHC Key Laboratory of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Hua Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Sun H, Kong X, Wei K, Hao J, Xi Y, Meng L, Li G, Lv X, Zou X, Gu X. Risk prediction model construction for post myocardial infarction heart failure by blood immune B cells. Front Immunol 2023; 14:1163350. [PMID: 37287974 PMCID: PMC10242647 DOI: 10.3389/fimmu.2023.1163350] [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: 02/10/2023] [Accepted: 04/27/2023] [Indexed: 06/09/2023] Open
Abstract
Background Myocardial infarction (MI) is a common cardiac condition with a high incidence of morbidity and mortality. Despite extensive medical treatment for MI, the development and outcomes of post-MI heart failure (HF) continue to be major factors contributing to poor post-MI prognosis. Currently, there are few predictors of post-MI heart failure. Methods In this study, we re-examined single-cell RNA sequencing and bulk RNA sequencing datasets derived from the peripheral blood samples of patients with myocardial infarction, including patients who developed heart failure and those who did not develop heart failure after myocardial infarction. Using marker genes of the relevant cell subtypes, a signature was generated and validated using relevant bulk datasets and human blood samples. Results We identified a subtype of immune-activated B cells that distinguished post-MI HF patients from non-HF patients. Polymerase chain reaction was used to confirm these findings in independent cohorts. By combining the specific marker genes of B cell subtypes, we developed a prediction model of 13 markers that can predict the risk of HF in patients after myocardial infarction, providing new ideas and tools for clinical diagnosis and treatment. Conclusion Sub-cluster B cells may play a significant role in post-MI HF. We found that the STING1, HSPB1, CCL5, ACTN1, and ITGB2 genes in patients with post-MI HF showed the same trend of increase as those without post-MI HF.
Collapse
Affiliation(s)
- HouRong Sun
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - XiangJin Kong
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - KaiMing Wei
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jie Hao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Xi
- Department of Reproductive Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - LingWei Meng
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - GuanNan Li
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xin Lv
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xin Zou
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - XingHua Gu
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
25
|
Sheikh Beig Goharrizi MA, Ghodsi S, Mokhtari M, Moravveji SS. Non-invasive STEMI-related biomarkers based on meta-analysis and gene prioritization. Comput Biol Med 2023; 161:106997. [PMID: 37216774 DOI: 10.1016/j.compbiomed.2023.106997] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 04/01/2023] [Accepted: 05/01/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND AND AIMS Acute ST-Segment Myocardial infarction (STEMI) is a common cardiovascular issue with a considerable burden of the disease. The underlying genetic basis and non-invasive markers were not well-established. METHODS Here, we implemented a systematic literature review and meta-analyses integration methods on 217 STEMI patients and 72 normal individuals to prioritize and detect the STEMI-related non-invasive markers. Five high-scored genes were experimentally assessed on 10 STEMI patients and 9 healthy controls. Finally, the presence of co-expressed nodes of top-score genes was explored. RESULTS The differential expression of ARGL, CLEC4E, and EIF3D were significant for Iranian patients. The ROC curve for gene CLEC4E revealed an AUC (95% CI) of 0.786 (0.686-0.886) in the prediction of STEMI. The Cox-PH model was fitted to stratify high/low risk heart failure progression (CI-index = 0.83, Likelihood-Ratio-Test = 3e-10). The SI00AI2 was a common biomarker between STEMI and NSTEMI patients. CONCLUSIONS In conclusion, the high-scored genes and prognostic model could be applicable for Iranian patients.
Collapse
Affiliation(s)
| | - Saeed Ghodsi
- Department of Cardiology, Sina Hospital, Tehran University of Medical Sciences Tehran, Iran
| | - Majid Mokhtari
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran; Laboratory of Personalized Precision Medicine, Bioinformatics Research Institute, Tehran, Iran
| | - Sayyed Sajjad Moravveji
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| |
Collapse
|
26
|
Fontaine MAC, Jin H, Gagliardi M, Rousch M, Wijnands E, Stoll M, Li X, Schurgers L, Reutelingsperger C, Schalkwijk C, van den Akker NMS, Molin DG, Gullestad L, Eritsland J, Hoffman P, Skjelland M, Andersen GØ, Aukrust P, Karel J, Smirnov E, Halvorsen B, Temmerman L, Biessen EA. Blood Milieu in Acute Myocardial Infarction Reprograms Human Macrophages for Trauma Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203053. [PMID: 36526599 PMCID: PMC9929255 DOI: 10.1002/advs.202203053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/06/2022] [Indexed: 06/17/2023]
Abstract
Acute myocardial infarction (AMI) is accompanied by a systemic trauma response that impacts the whole body, including blood. This study addresses whether macrophages, key players in trauma repair, sense and respond to these changes. For this, healthy human monocyte-derived macrophages are exposed to 20% human AMI (n = 50) or control (n = 20) serum and analyzed by transcriptional and multiparameter functional screening followed by network-guided data interpretation and drug repurposing. Results are validated in an independent cohort at functional level (n = 47 AMI, n = 25 control) and in a public dataset. AMI serum exposure results in an overt AMI signature, enriched in debris cleaning, mitosis, and immune pathways. Moreover, gene networks associated with AMI and with poor clinical prognosis in AMI are identified. Network-guided drug screening on the latter unveils prostaglandin E2 (PGE2) signaling as target for clinical intervention in detrimental macrophage imprinting during AMI trauma healing. The results demonstrate pronounced context-induced macrophage reprogramming by the AMI systemic environment, to a degree decisive for patient prognosis. This offers new opportunities for targeted intervention and optimized cardiovascular disease risk management.
Collapse
|
27
|
Yang J, Yang M, Sheng G. Dysregulated lncRNAs are involved in the progress of myocardial infarction by constructing regulatory networks. Open Med (Wars) 2023; 18:20230657. [PMID: 36910851 PMCID: PMC9999115 DOI: 10.1515/med-2023-0657] [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: 09/25/2022] [Revised: 01/08/2023] [Accepted: 02/07/2023] [Indexed: 03/10/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) mediate important epigenetic regulation in a wide range of biological processes. However, the effect of all dysregulated lncRNAs in myocardial infarction (MI) is not clear. Whole transcriptome sequencing analysis was used to characterize the dynamic changes in lncRNA and mRNA expression. A gene network was constructed, and genes were classified into different modules using WGCNA. In addition, for all dysregulated lncRNAs, gene ontology analysis and cis-regulatory analysis were applied. The results demonstrated that a large number of the differentially co-expressed genes were primarily linked to the immune system process, inflammatory response, and innate immune response. The functional pathway analysis of the MEblue module included immune system process and apoptosis, and MEbrown included the T-cell receptor signal pathway by WGCNA. In addition, through cis-acting analysis of lncRNA regulation, the cis-regulated mRNAs were mainly enriched in immune system processes, innate immune responses, and VEGF signal pathways. We found that lncRNA regulation of mRNAs plays an important role in immune and inflammatory pathways. Our study provides a foundation to further understand the role and potential mechanism of dysregulated lncRNAs in the regulation of MI, in which many of them could be potential targets for MI.
Collapse
Affiliation(s)
- Jingqi Yang
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Ming Yang
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Guotai Sheng
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| |
Collapse
|
28
|
Wang Y, Chen Y, Zhang T. Integrated whole-genome gene expression analysis reveals an atlas of dynamic immune landscapes after myocardial infarction. Front Cardiovasc Med 2023; 10:1087721. [PMID: 36937942 PMCID: PMC10020602 DOI: 10.3389/fcvm.2023.1087721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Myocardial infarction (MI) is a deadly medical condition leading to irreversible damage to the inflicted cardiac tissue. Elevated inflammatory response marks the severity of MI and is associated with the development of heart failure (HF), a long-term adverse outcome of MI. However, the efficacy of anti-inflammatory therapies for MI remains controversial. Deciphering the dynamic transcriptional signatures in peripheral blood mononuclear cells (PBMCs) is a viable and translatable route to better understand post-MI inflammation, which may help guide post-MI anti-inflammatory treatments. Methods In this work, integrated whole-genome gene expression analysis was performed to explore dynamic immune landscapes associated with MI. Results GSEA and GSVA showed that pathways involved in the inflammatory response and metabolic reprogramming were significantly enriched in PBMCs from MI patients. Based on leukocyte profiles generated by xCell algorithm, the relative abundance of monocytes and neutrophils was significantly increased in PBMCs from MI patients and had positive correlations with typical inflammation-associated transcripts. Mfuzz clustering revealed temporal gene expression profiles of PBMCs during the 6-month post-MI follow-up. Analysis of DEGs and gene sets indicated that PBMCs from HF group were characterized by elevated and lasting expression of genes implicated in inflammation and coagulation. Consensus clustering generated 4 metabolic subtypes of PBMCs with molecular heterogeneity in HF patients. Discussion In summary, integrated whole-genome gene expression analysis here outlines a transcriptomic framework that may improve the understanding of dynamic signatures present in PBMCs, as well as the heterogeneity of PBMCs in MI patients with or without long-term clinical outcome of HF. Moreover, the work here uncovers the diversity and heterogeneity of PBMCs from HF patients, providing novel bioinformatic evidence supporting the mechanistic implications of metabolic reprogramming and mitochondrial dysfunction in the post-MI inflammation and HF. Therefore, our work here supports the notion that individualized anti-inflammatory therapies are needed to improve the clinical management of post-MI patients.
Collapse
Affiliation(s)
- Yujue Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Laboratory of Clinical and Molecular Pharmacology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yu Chen, ; Teng Zhang,
| | - Teng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yu Chen, ; Teng Zhang,
| |
Collapse
|
29
|
Chicco D, Shiradkar R. Ten quick tips for computational analysis of medical images. PLoS Comput Biol 2023; 19:e1010778. [PMID: 36602952 PMCID: PMC9815662 DOI: 10.1371/journal.pcbi.1010778] [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: 01/06/2023] Open
Abstract
Medical imaging is a great asset for modern medicine, since it allows physicians to spatially interrogate a disease site, resulting in precise intervention for diagnosis and treatment, and to observe particular aspect of patients' conditions that otherwise would not be noticeable. Computational analysis of medical images, moreover, can allow the discovery of disease patterns and correlations among cohorts of patients with the same disease, thus suggesting common causes or providing useful information for better therapies and cures. Machine learning and deep learning applied to medical images, in particular, have produced new, unprecedented results that can pave the way to advanced frontiers of medical discoveries. While computational analysis of medical images has become easier, however, the possibility to make mistakes or generate inflated or misleading results has become easier, too, hindering reproducibility and deployment. In this article, we provide ten quick tips to perform computational analysis of medical images avoiding common mistakes and pitfalls that we noticed in multiple studies in the past. We believe our ten guidelines, if taken into practice, can help the computational-medical imaging community to perform better scientific research that eventually can have a positive impact on the lives of patients worldwide.
Collapse
Affiliation(s)
- Davide Chicco
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Rakesh Shiradkar
- Department of Biomedical Engineering, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
30
|
Guo Y, Jiang H, Wang J, Li P, Zeng X, Zhang T, Feng J, Nie R, Liu Y, Dong X, Hu Q. 5mC modification patterns provide novel direction for early acute myocardial infarction detection and personalized therapy. Front Cardiovasc Med 2022; 9:1053697. [PMID: 36620624 PMCID: PMC9816341 DOI: 10.3389/fcvm.2022.1053697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background Most deaths from coronary artery disease (CAD) are due to acute myocardial infarction (AMI). There is an urgent need for early AMI detection, particularly in patients with stable CAD. 5-methylcytosine (5mC) regulatory genes have been demonstrated to involve in the progression and prognosis of cardiovascular diseases, while little research examined 5mC regulators in CAD to AMI progression. Method Two datasets (GSE59867 and GSE62646) were downloaded from Gene Expression Omnibus (GEO) database, and 21 m5C regulators were extracted from previous literature. Dysregulated 5mC regulators were screened out by "limma." The least absolute shrinkage and selection operator (LASSO) and support vector machine recursive feature elimination (SVM-RFE) algorithm were employed to identify hub 5mC regulators in CAD to AMI progression, and 43 clinical samples (Quantitative real-time PCR) were performed for expression validation. Then a logistic model was built to construct 5mC regulator signatures, and a series of bioinformatics algorithms were performed for model validation. Besides, 5mC-associated molecular clusters were studied via unsupervised clustering analysis, and correlation analysis between immunocyte and 5mC regulators in each cluster was conducted. Results Nine hub 5mC regulators were identified. A robust model was constructed, and its prominent classification accuracy was verified via ROC curve analysis (area under the curve [AUC] = 0.936 in the training cohort and AUC = 0.888 in the external validation cohort). Besides, the clinical effect of the model was validated by decision curve analysis. Then, 5mC modification clusters in AMI patients were identified, along with the immunocyte infiltration levels of each cluster. The correlation analysis found the strongest correlations were TET3-Mast cell in cluster-1 and TET3-MDSC in cluster-2. Conclusion Nine hub 5mC regulators (DNMT3B, MBD3, UHRF1, UHRF2, NTHL1, SMUG1, ZBTB33, TET1, and TET3) formed a diagnostic model, and concomitant results unraveled the critical impact of 5mC regulators, providing interesting epigenetics findings in AMI population vs. stable CAD.
Collapse
Affiliation(s)
- Yiqun Guo
- Department of Interventional Radiology and Vascular, Guangzhou Women and Children’s Medical Center, The Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hua Jiang
- Department of Interventional Radiology and Vascular, Guangzhou Women and Children’s Medical Center, The Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinlong Wang
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Ping Li
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaoquan Zeng
- Department of Cardiology, Xinfeng County People’s Hospital, Shaoguan, Guangdong, China
| | - Tao Zhang
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jianyi Feng
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Ruqiong Nie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yulong Liu
- Department of Intervention and Vascular Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China,*Correspondence: Yulong Liu,
| | - Xiaobian Dong
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China,Xiaobian Dong,
| | - Qingsong Hu
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China,Qingsong Hu,
| |
Collapse
|
31
|
Liu Z, Wang L, Xing Q, Liu X, Hu Y, Li W, Yan Q, Liu R, Huang N. Identification of GLS as a cuproptosis-related diagnosis gene in acute myocardial infarction. Front Cardiovasc Med 2022; 9:1016081. [PMID: 36440046 PMCID: PMC9691691 DOI: 10.3389/fcvm.2022.1016081] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
Acute myocardial infarction (AMI) has the characteristics of sudden onset, rapid progression, poor prognosis, and so on. Therefore, it is urgent to identify diagnostic and prognostic biomarkers for it. Cuproptosis is a new form of mitochondrial respiratory-dependent cell death. However, studies are limited on the clinical significance of cuproptosis-related genes (CRGs) in AMI. In this study, we systematically assessed the genetic alterations of CRGs in AMI by bioinformatics approach. The results showed that six CRGs (LIAS, LIPT1, DLAT, PDHB, MTF1, and GLS) were markedly differentially expressed between stable coronary heart disease (stable_CAD) and AMI. Correlation analysis indicated that CRGs were closely correlated with N6-methyladenosine (m6A)-related genes through R language “corrplot” package, especially GLS was positively correlated with FMR1 and MTF1 was negatively correlated with HNRNPA2B1. Immune landscape analysis results revealed that CRGs were closely related to various immune cells, especially GLS was positively correlated with T cells CD4 memory resting and negatively correlated with monocytes. Kaplan–Meier analysis demonstrated that the group with high DLAT expression had a better prognosis. The area under curve (AUC) certified that GLS had good diagnostic value, in the training set (AUC = 0.87) and verification set (ACU = 0.99). Gene set enrichment analysis (GSEA) suggested that GLS was associated with immune- and hypoxia-related pathways. In addition, Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, competing endogenous RNA (ceRNA) analysis, transcription factor (TF), and compound prediction were performed to reveal the regulatory mechanism of CRGs in AMI. Overall, our study can provide additional information for understanding the role of CRGs in AMI, which may provide new insights into the identification of therapeutic targets for AMI.
Collapse
Affiliation(s)
- Zheng Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Lei Wang
- Department of Cardiovascular Medicine, Xiangtan Center Hospital, Xiangtan, China
| | - Qichang Xing
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Xiang Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Yixiang Hu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Wencan Li
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Qingzi Yan
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Renzhu Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- Zhou Honghao Research Institute Xiangtan, Xiangtan, China
| | - Nan Huang
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, China
- *Correspondence: Nan Huang,
| |
Collapse
|
32
|
Jiang YH, Wu SY, Wang Z, Zhang L, Zhang J, Li Y, Liu C, Wu WZ, Xue YT. Bioinformatics analysis identifies ferroptosis‑related genes in the regulatory mechanism of myocardial infarction. Exp Ther Med 2022; 24:748. [PMID: 36561967 PMCID: PMC9748705 DOI: 10.3892/etm.2022.11684] [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: 07/11/2022] [Accepted: 09/22/2022] [Indexed: 11/11/2022] Open
Abstract
Since ferroptosis is considered to be a notable cause of cardiomyocyte death, inhibiting ferroptosis has become a novel strategy in reducing cardiac cell death and improving cardiopathic conditions. Therefore, the aim of the present study was to search for ferroptosis-related hub genes and determine their diagnostic value in myocardial infarction (MI) to aid in the diagnosis and treatment of the disease. A total of 10,286 DEGs were identified, including 6,822 upregulated and 3.464 downregulated genes in patients with MI compared with healthy controls. After overlapping with ferroptosis-related genes, 128 ferroptosis-related DEGs were obtained. WGCNA successfully identified a further eight functional modules, from which the blue module had the strongest correlation with MI. Blue module genes and ferroptosis-related differentially expressed genes were overlapped to obtain 20 ferroptosis-related genes associated with MI. Go and KEGG analysis showed that these genes were mainly enriched in cellular response to chemical stress, trans complex, transferring, phosphorus-containing groups, protein serine/threonine kinase activity, FoxO signaling pathway. Hub genes were obtained from 20 ferroptosis-related genes through the PPI network. The expression of hub genes was found to be down-regulated in the MI group. Finally, the miRNAs-hub genes and TFs-hub genes networks were constructed. The GSE141512 dataset and the use of RT-qPCR assays on patient blood samples were used to confirm these results. The results showed that ATM, PIK3CA, MAPK8, KRAS and SIRT1 may play key roles in the development of MI, and could therefore be novel markers or targets for the diagnosis or treatment of MI.
Collapse
Affiliation(s)
- Yong-Hao Jiang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Su-Ying Wu
- Foreign Language College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Zhen Wang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Lei Zhang
- Foreign Language College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Juan Zhang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Yan Li
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Chenglong Liu
- Foreign Language College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China
| | - Wen-Zhe Wu
- Cardiovascular Department, Dezhou Municipal Hospital, Dezhou, Shandong 253000, P.R. China,Correspondence to: Professor Wen-Zhe Wu, Cardiovascular Department, Dezhou Municipal Hospital, 1766 Sanba Zhong Road, Decheng, Dezhou, Shandong 253000, P.R. China
| | - Yi-Tao Xue
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, P.R. China,Correspondence to: Professor Wen-Zhe Wu, Cardiovascular Department, Dezhou Municipal Hospital, 1766 Sanba Zhong Road, Decheng, Dezhou, Shandong 253000, P.R. China
| |
Collapse
|
33
|
Chen H, Jiang R, Huang W, Chen K, Zeng R, Wu H, Yang Q, Guo K, Li J, Wei R, Liao S, Tse HF, Sha W, Zhuo Z. Identification of energy metabolism-related biomarkers for risk prediction of heart failure patients using random forest algorithm. Front Cardiovasc Med 2022; 9:993142. [PMID: 36304554 PMCID: PMC9593065 DOI: 10.3389/fcvm.2022.993142] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Energy metabolism plays a crucial role in the improvement of heart dysfunction as well as the development of heart failure (HF). The current study is designed to identify energy metabolism-related diagnostic biomarkers for predicting the risk of HF due to myocardial infarction. Methods Transcriptome sequencing data of HF patients and non-heart failure (NF) people (GSE66360 and GSE59867) were obtained from gene expression omnibus (GEO) database. Energy metabolism-related differentially expressed genes (DEGs) were screened between HF and NF samples. The subtyping consistency analysis was performed to enable the samples to be grouped. The immune infiltration level among subtypes was assessed by single sample gene set enrichment analysis (ssGSEA). Random forest algorithm (RF) and support vector machine (SVM) were applied to identify diagnostic biomarkers, and the receiver operating characteristic curves (ROC) was plotted to validate the accuracy. Predictive nomogram was constructed and validated based on the result of the RF. Drug screening and gene-miRNA network were analyzed to predict the energy metabolism-related drugs and potential molecular mechanism. Results A total of 22 energy metabolism-related DEGs were identified between HF and NF patients. The clustering analysis showed that HF patients could be classified into two subtypes based on the energy metabolism-related genes, and functional analyses demonstrated that the identified DEGs among two clusters were mainly involved in immune response regulating signaling pathway and lipid and atherosclerosis. ssGSEA analysis revealed that there were significant differences in the infiltration levels of immune cells between two subtypes of HF patients. Random-forest and support vector machine algorithm eventually identified ten diagnostic markers (MEF2D, RXRA, PPARA, FOXO1, PPARD, PPP3CB, MAPK14, CREB1, MEF2A, PRMT1) for risk prediction of HF patients, and the proposed nomogram resulted in good predictive performance (GSE66360, AUC = 0.91; GSE59867, AUC = 0.84) and the clinical usefulness in HF patients. More importantly, 10 drugs and 15 miRNA were predicted as drug target and hub miRNA that associated with energy metabolism-related genes, providing further information on clinical HF treatment. Conclusion This study identified ten energy metabolism-related diagnostic markers using random forest algorithm, which may help optimize risk stratification and clinical treatment in HF patients.
Collapse
Affiliation(s)
- Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,School of Medicine, South China University of Technology, Guangzhou, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China,*Correspondence: Hao Chen
| | - Rui Jiang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,School of Medicine, South China University of Technology, Guangzhou, China
| | - Wentao Huang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Kequan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruijie Zeng
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huihuan Wu
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,School of Medicine, South China University of Technology, Guangzhou, China
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kehang Guo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jingwei Li
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Rui Wei
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Songyan Liao
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China,Hung-Fat Tse
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,School of Medicine, South China University of Technology, Guangzhou, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China,Weihong Sha
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,Zewei Zhuo
| |
Collapse
|
34
|
Yip HF, Chowdhury D, Wang K, Liu Y, Gao Y, Lan L, Zheng C, Guan D, Lam KF, Zhu H, Tai X, Lu A. ReDisX, a machine learning approach, rationalizes rheumatoid arthritis and coronary artery disease patients uniquely upon identifying subpopulation differentiation markers from their genomic data. Front Med (Lausanne) 2022; 9:931860. [PMID: 36072953 PMCID: PMC9441882 DOI: 10.3389/fmed.2022.931860] [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: 05/03/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
Diseases originate at the molecular-genetic layer, manifest through altered biochemical homeostasis, and develop symptoms later. Hence, symptomatic diagnosis is inadequate to explain the underlying molecular-genetic abnormality and individual genomic disparities. The current trends include molecular-genetic information relying on algorithms to recognize the disease subtypes through gene expressions. Despite their disposition toward disease-specific heterogeneity and cross-disease homogeneity, a gap still exists in describing the extent of homogeneity within the heterogeneous subpopulation of different diseases. They are limited to obtaining the holistic sense of the whole genome-based diagnosis resulting in inaccurate diagnosis and subsequent management. Addressing those ambiguities, our proposed framework, ReDisX, introduces a unique classification system for the patients based on their genomic signatures. In this study, it is a scalable machine learning algorithm deployed to re-categorize the patients with rheumatoid arthritis and coronary artery disease. It reveals heterogeneous subpopulations within a disease and homogenous subpopulations across different diseases. Besides, it identifies granzyme B (GZMB) as a subpopulation-differentiation marker that plausibly serves as a prominent indicator for GZMB-targeted drug repurposing. The ReDisX framework offers a novel strategy to redefine disease diagnosis through characterizing personalized genomic signatures. It may rejuvenate the landscape of precision and personalized diagnosis and a clue to drug repurposing.
Collapse
Affiliation(s)
- Hiu F. Yip
- Computational Medicine Laboratory, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Department of Mathematics, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Debajyoti Chowdhury
- Computational Medicine Laboratory, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Kexin Wang
- National Key Clinical Specialty, Engineering Technology Research Center of Education Ministry of China, Guangzhou, China
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Neurosurgery Institute, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yujie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yao Gao
- Department of Psychiatry, First Hospital, First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Liang Lan
- Department of Communication Studies, School of Communication, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Chaochao Zheng
- Department of Mathematics, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Daogang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Kei F. Lam
- Department of Mathematics, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Hailong Zhu
- Computational Medicine Laboratory, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Xuecheng Tai
- Department of Mathematics, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Aiping Lu
- Computational Medicine Laboratory, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
35
|
Gorica E, Mohammed SA, Ambrosini S, Calderone V, Costantino S, Paneni F. Epi-Drugs in Heart Failure. Front Cardiovasc Med 2022; 9:923014. [PMID: 35911511 PMCID: PMC9326055 DOI: 10.3389/fcvm.2022.923014] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Unveiling the secrets of genome's flexibility does not only foster new research in the field, but also gives rise to the exploration and development of novel epigenetic-based therapies as an approach to alleviate disease phenotypes. A better understanding of chromatin biology (DNA/histone complexes) and non-coding RNAs (ncRNAs) has enabled the development of epigenetic drugs able to modulate transcriptional programs implicated in cardiovascular diseases. This particularly applies to heart failure, where epigenetic networks have shown to underpin several pathological features, such as left ventricular hypertrophy, fibrosis, cardiomyocyte apoptosis and microvascular dysfunction. Targeting epigenetic signals might represent a promising approach, especially in patients with heart failure with preserved ejection fraction (HFpEF), where prognosis remains poor and breakthrough therapies have yet to be approved. In this setting, epigenetics can be employed for the development of customized therapeutic approaches thus paving the way for personalized medicine. Even though the beneficial effects of epi-drugs are gaining attention, the number of epigenetic compounds used in the clinical practice remains low suggesting that more selective epi-drugs are needed. From DNA-methylation changes to non-coding RNAs, we can establish brand-new regulations for drug targets with the aim of restoring healthy epigenomes and transcriptional programs in the failing heart. In the present review, we bring the timeline of epi-drug discovery and development, thus highlighting the emerging role of epigenetic therapies in heart failure.
Collapse
Affiliation(s)
- Era Gorica
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Shafeeq A. Mohammed
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
| | | | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, Zurich, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Toma A, Dos Santos C, Burzyńska B, Góra M, Kiliszek M, Stickle N, Kirsten H, Kosyakovsky L, Wang B, van Diepen S, Epelman S, Szekely Y, Marshall JC, Billia F, Lawler PR. Diversity in the Expressed Genomic Host Response to Myocardial Infarction. Circ Res 2022; 131:106-108. [PMID: 35534922 DOI: 10.1161/circresaha.121.318391] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Augustin Toma
- Peter Munk Cardiac Centre, Toronto General Hospital, Canada (A.T., B.W., S.E., Y.S., F.B., P.R.L.).,Department of Medicine (A.T., C.d.S., B.W., S.E., Y.S., F.B., P.R.L.)
| | - Claudia Dos Santos
- Department of Medicine (A.T., C.d.S., B.W., S.E., Y.S., F.B., P.R.L.).,Interdepartmental Division of Critical Care Medicine, University of Toronto, Canada. (C.d.S., J.C.M., P.R.L.).,St Michael's Hospital, Toronto, Canada (C.d.S., J.C.M.)
| | - Beata Burzyńska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland (B.B., M.G.)
| | - Monika Góra
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland (B.B., M.G.)
| | - Marek Kiliszek
- Department of Cardiology and Internal Diseases, Military Institute of Medicine, Warsaw, Poland (M.K.)
| | | | - Holger Kirsten
- LIFE - Leipzig Research Center for Civilization Diseases and Institute for Medical Informatics, Statistics and Epidemiology, Universität Leipzig, Germany (H.K.)
| | - Leah Kosyakovsky
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.K.)
| | - Bo Wang
- Peter Munk Cardiac Centre, Toronto General Hospital, Canada (A.T., B.W., S.E., Y.S., F.B., P.R.L.).,Department of Medicine (A.T., C.d.S., B.W., S.E., Y.S., F.B., P.R.L.)
| | - Sean van Diepen
- Division of Cardiology, Department of Critical Care, Department of Medicine, University of Alberta, Edmonton, Canada (S.v.D.)
| | - Slava Epelman
- Peter Munk Cardiac Centre, Toronto General Hospital, Canada (A.T., B.W., S.E., Y.S., F.B., P.R.L.).,Department of Medicine (A.T., C.d.S., B.W., S.E., Y.S., F.B., P.R.L.).,Ted Rogers Centre for Heart Research, Toronto, Canada (S.E., F.B., P.R.L.)
| | - Yishay Szekely
- Peter Munk Cardiac Centre, Toronto General Hospital, Canada (A.T., B.W., S.E., Y.S., F.B., P.R.L.).,Department of Medicine (A.T., C.d.S., B.W., S.E., Y.S., F.B., P.R.L.)
| | - John C Marshall
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Canada. (C.d.S., J.C.M., P.R.L.).,St Michael's Hospital, Toronto, Canada (C.d.S., J.C.M.)
| | - Filio Billia
- Peter Munk Cardiac Centre, Toronto General Hospital, Canada (A.T., B.W., S.E., Y.S., F.B., P.R.L.).,Department of Medicine (A.T., C.d.S., B.W., S.E., Y.S., F.B., P.R.L.).,Ted Rogers Centre for Heart Research, Toronto, Canada (S.E., F.B., P.R.L.)
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, Toronto General Hospital, Canada (A.T., B.W., S.E., Y.S., F.B., P.R.L.).,Department of Medicine (A.T., C.d.S., B.W., S.E., Y.S., F.B., P.R.L.).,Interdepartmental Division of Critical Care Medicine, University of Toronto, Canada. (C.d.S., J.C.M., P.R.L.).,Ted Rogers Centre for Heart Research, Toronto, Canada (S.E., F.B., P.R.L.)
| |
Collapse
|
37
|
Extracellular vesicles derived from human bone marrow mesenchymal stem cells protect rats against acute myocardial infarction-induced heart failure. Cell Tissue Res 2022; 389:23-40. [PMID: 35524813 DOI: 10.1007/s00441-022-03612-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 03/09/2022] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) derived from human bone marrow mesenchymal stem cells (BMSCs) are suggested to promote angiogenesis in a rat model of acute myocardial infarction (AMI). This study aimed to explore the underlying mechanism of BMSCs-EVs in AMI-induced heart failure (HF). BMSCs were isolated and verified, and EVs were purified and identified. After establishment of AMI-induced HF models, rats were treated with BMSCs-EVs and/or overexpressing (ov)/knocking down (kd) bone morphogenetic protein 2 (BMP2). Cardiac function, myocardial histopathological changes, angiogenesis, and vascular regeneration density were measured. Levels of pro-angiogenesis factors and cardiomyocyte apoptosis were detected. The viability and angiogenesis of hypoxic human umbilical vein endothelial cells (HUVECs) were measured. After BMSCs-EV treatment, the cardiac function of HF rats was improved, myocardial fibrosis and inflammatory cell infiltration were decreased, angiogenesis was increased, and cardiomyocyte apoptosis was inhibited. BMP2 was significantly upregulated in the myocardium. Ov-BMP2-BMSCs-EVs alleviated myocardial fibrosis and inflammatory cell infiltration, and promoted angiogenesis of HF rats, and improved the activity and angiogenesis of hypoxic HUVECs, while kd-BMP2-BMSCs-EVs showed limited protection against AMI-induced HF. BMSCs-EVs deliver BMP2 to promote angiogenesis and improve cardiac function of HF rats.
Collapse
|
38
|
Chen Q, Su L, Liu C, Gao F, Chen H, Yin Q, Li S. PRKAR1A and SDCBP Serve as Potential Predictors of Heart Failure Following Acute Myocardial Infarction. Front Immunol 2022; 13:878876. [PMID: 35592331 PMCID: PMC9110666 DOI: 10.3389/fimmu.2022.878876] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/01/2022] [Indexed: 12/20/2022] Open
Abstract
Background and Objectives Early diagnosis of patients with acute myocardial infarction (AMI) who are at a high risk of heart failure (HF) progression remains controversial. This study aimed at identifying new predictive biomarkers of post-AMI HF and at revealing the pathogenesis of HF involving these marker genes. Methods and Results A transcriptomic dataset of whole blood cells from AMI patients with HF progression (post-AMI HF, n = 16) and without progression (post-AMI non-HF, n = 16) was analyzed using the weighted gene co-expression network analysis (WGCNA). The results indicated that one module consisting of 720 hub genes was significantly correlated with post-AMI HF. The hub genes were validated in another transcriptomic dataset of peripheral blood mononuclear cells (post-AMI HF, n = 9; post-AMI non-HF, n = 8). PRKAR1A, SDCBP, SPRED2, and VAMP3 were upregulated in the two datasets. Based on a single-cell RNA sequencing dataset of leukocytes from heart tissues of normal and infarcted mice, PRKAR1A was further verified to be upregulated in monocytes/macrophages on day 2, while SDCBP was highly expressed in neutrophils on day 2 and in monocytes/macrophages on day 3 after AMI. Cell-cell communication analysis via the "CellChat" package showed that, based on the interaction of ligand-receptor (L-R) pairs, there were increased autocrine/paracrine cross-talk networks of monocytes/macrophages and neutrophils in the acute stage of MI. Functional enrichment analysis of the abovementioned L-R genes together with PRKAR1A and SDCBP performed through the Metascape platform suggested that PRKAR1A and SDCBP were mainly involved in inflammation, apoptosis, and angiogenesis. The receiver operating characteristic (ROC) curve analysis demonstrated that PRKAR1A and SDCBP, as well as their combination, had a promising prognostic value in the identification of AMI patients who were at a high risk of HF progression. Conclusion This study identified that PRKAR1A and SDCBP may serve as novel biomarkers for the early diagnosis of post-AMI HF and also revealed their potentially regulatory mechanism during HF progression.
Collapse
Affiliation(s)
- Qixin Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People’s Hospital, Beijing, China
| | - Lina Su
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People’s Hospital, Beijing, China
| | - Chuanfen Liu
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People’s Hospital, Beijing, China
| | - Fu Gao
- Department of Cardiac Surgery, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Hong Chen
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People’s Hospital, Beijing, China
| | - Qijin Yin
- Ministry of Education Key Laboratory of Bioinformatics, Research Department of Bioinformatics at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Sufang Li
- Department of Cardiology, Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Center for Cardiovascular Translational Research, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
39
|
Feng L, Tian R, Mu X, Chen C, Zhang Y, Cui J, Song Y, Liu Y, Zhang M, Shi L, Sun Y, Li L, Yi W. Identification of Genes Linking Natural Killer Cells to Apoptosis in Acute Myocardial Infarction and Ischemic Stroke. Front Immunol 2022; 13:817377. [PMID: 35432334 PMCID: PMC9012496 DOI: 10.3389/fimmu.2022.817377] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/11/2022] [Indexed: 12/27/2022] Open
Abstract
Natural killer (NK) cells are a type of innate lymphoid cell that are involved in the progression of acute myocardial infarction and ischemic stroke. Although multiple forms of programmed cell death are known to play important roles in these diseases, the correlation between NK cells and apoptosis-related genes during acute myocardial infarction and ischemic stroke remains unclear. In this study, we explored the distinct patterns of NK cell infiltration and apoptosis during the pathological progression of acute myocardial infarction and ischemic stroke using mRNA expression microarrays from the Gene Expression Omnibus database. Since the abundance of NK cells correlated positively with apoptosis in both diseases, we further examined the correlation between NK cell abundance and the expression of apoptosis-related genes. Interestingly, APAF1 and IRAK3 expression correlated negatively with NK cell abundance in both acute myocardial infarction and ischemic stroke, whereas ATM, CAPN1, IL1B, IL1R1, PRKACA, PRKACB, and TNFRSF1A correlated negatively with NK cell abundance in acute myocardial infarction. Together, these findings suggest that these apoptosis-related genes may play important roles in the mechanisms underlying the patterns of NK cell abundance and apoptosis in acute myocardial infarction and ischemic stroke. Our study, therefore, provides novel insights for the further elucidation of the pathogenic mechanism of ischemic injury in both the heart and the brain, as well as potential useful therapeutic targets.
Collapse
Affiliation(s)
- Lele Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Ruofei Tian
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi’an, China
| | - Xingdou Mu
- Department of Breast and Thyroid Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Cheng Chen
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- Department of Internal Medicine, Central Health Center of Huilong Town, Shangluo, China
| | - Yuxi Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yujie Song
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yingying Liu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- College of Life Science, Northwest University, Xi’an, China
| | - Miao Zhang
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- The Second Clinical Medicine College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lei Shi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Yang Sun, ; Ling Li, ; Wei Yi,
| | - Ling Li
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi’an, China
- *Correspondence: Yang Sun, ; Ling Li, ; Wei Yi,
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Yang Sun, ; Ling Li, ; Wei Yi,
| |
Collapse
|
40
|
Chen X, Li Q, Zhang Z, Yang M, Wang E. Identification of Potential Diagnostic Biomarkers From Circulating Cells During the Course of Sleep Deprivation-Related Myocardial Infarction Based on Bioinformatics Analyses. Front Cardiovasc Med 2022; 9:843426. [PMID: 35369343 PMCID: PMC8969017 DOI: 10.3389/fcvm.2022.843426] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 02/22/2022] [Indexed: 01/01/2023] Open
Abstract
Background Myocardial infarction (MI) is the leading cause of death from non-infectious diseases worldwide and results in rapid deterioration due to the sudden rupture of plaques associated with atherosclerosis, a chronic inflammatory disease. Sleep is a key factor that regulates immune homeostasis of the body. The imbalance in circulating immune cells caused by sleep deprivation (SD) may represent a risk factor leading to the rapid deterioration of plaques and MI. Therefore, it is of profound significance to identify diagnostic biomarkers for preventing SD-related MI. Methods In the present study, we identified coexpressed differentially expressed genes (co-DEGs) between peripheral blood mononuclear cells from MI and SD samples (compared to controls) from a public database. LASSO regression analysis was applied to identify significant diagnostic biomarkers from co-DEGs. Moreover, receiver operating characteristic (ROC) curve analysis was performed to test biomarker accuracy and diagnostic ability. We further analyzed immune cell enrichment in MI and SD samples using the CIBERSORT algorithm, and the correlation between biomarkers and immune cell composition was assessed. We also investigated whether diagnostic biomarkers are involved in immune cell signaling pathways in SD-related MI processes. Results A total of 10 downregulated co-DEGs from the sets of MI-DEGs and SD-DEGs were overlapped. After applying LASSO regression analysis, SYTL2, KLRD1, and C12orf75 were selected and validated as diagnostic biomarkers using ROC analysis. Next, we found that resting NK cells were downregulated in both the MI samples and SD samples, which is similar to the changes noted for SYTL2. Importantly, SYTL2 was strongly positively correlated not only with resting NK cells but also with most genes related to NK cell markers in the MI and SD datasets. Moreover, SYTL2 was highly associated with genes in NK cell signaling pathways, including the MAPK signaling pathway, cytotoxic granule movement and exocytosis, and NK cell activation. Furthermore, GSEA and KEGG analyses provided evidence that the DEGs identified from MI samples with low vs. high SYTL2 expression exhibited a strong association with the regulation of the immune response and NK cell-mediated cytotoxicity. Conclusion In conclusion, SYTL2, KLRD1, and C12orf75 represent potential diagnostic biomarkers of MI. The association between SYTL2 and resting NK cells may be critically involved in SD-related MI development and occurrence.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Xiangya Hospital Central South University, Changsha, China
| | - Qian Li
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Zhong Zhang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Minjing Yang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - E. Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Xiangya Hospital Central South University, Changsha, China
- *Correspondence: E. Wang
| |
Collapse
|
41
|
Identifying Potential New Gene Expression-Based Biomarkers in the Peripheral Blood Mononuclear Cells of Hepatitis B-Related Hepatocellular Carcinoma. Can J Gastroenterol Hepatol 2022; 2022:9541600. [PMID: 35265561 PMCID: PMC8901362 DOI: 10.1155/2022/9541600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/13/2021] [Accepted: 01/22/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE The analysis of the gene expression of peripheral blood mononuclear cells (PBMCs) is important to clarify the pathogenesis of hepatocellular carcinoma (HCC) and the detection of suitable biomarkers. The purpose of this investigation was to use RNA-sequencing to screen the appropriate differentially expressed genes (DEGs) in the PBMCs for the HCC. METHODS The comprehensive transcriptome of extracted RNA of PBMC (n = 20) from patients with chronic hepatitis B (CHB), liver cirrhosis, and early stage of HCC (5 samples per group) was carried out using RNA-sequencing. All raw RNA-sequencing data analyses were performed using conventional RNA-sequencing analysis tools. Next, gene ontology (GO) analyses were carried out to elucidate the biological processes of DEGs. Finally, relative transcript abundance of selected DEGs was verified using qRT-PCR on additional validation groups. RESULTS Specifically, 13, 1262, and 1450 DEGs were identified for CHB, liver cirrhosis, and HCC, when compared with the healthy controls. GO enrichment analysis indicated that HCC is closely related to the immune response. Seven DEGs (TYMP, TYROBP, CD14, TGFBI, LILRA2, GNLY, and GZMB) were common to HCC, cirrhosis, and CHB when compared to healthy controls. The data revealed that the expressions of these 7 DEGs were consistent with those from the RNA-sequencing results. Also, the expressions of 7 representative genes that had higher sensitivity were obtained by receiver operating characteristic analysis, which indicated their important diagnostic accuracy for HBV-HCC. CONCLUSION This study provides us with new horizons into the biological process and potential prospective clinical diagnosis and prognosis of HCC in the near future.
Collapse
|
42
|
Xu J, Yang Y. Integrated Gene Expression Profiling Analysis Reveals Potential Molecular Mechanisms and Candidate Biomarkers for Early Risk Stratification and Prediction of STEMI and Post-STEMI Heart Failure Patients. Front Cardiovasc Med 2021; 8:736497. [PMID: 34957234 PMCID: PMC8702808 DOI: 10.3389/fcvm.2021.736497] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/16/2021] [Indexed: 12/27/2022] Open
Abstract
Objective: To explore the molecular mechanism and search for the candidate differentially expressed genes (DEGs) with the predictive and prognostic potentiality that is detectable in the whole blood of patients with ST-segment elevation (STEMI) and those with post-STEMI HF. Methods: In this study, we downloaded GSE60993, GSE61144, GSE66360, and GSE59867 datasets from the NCBI-GEO database. DEGs of the datasets were investigated using R. Gene ontology (GO) and pathway enrichment were performed via ClueGO, CluePedia, and DAVID database. A protein interaction network was constructed via STRING. Enriched hub genes were analyzed by Cytoscape software. The least absolute shrinkage and selection operator (LASSO) logistic regression algorithm and receiver operating characteristics analyses were performed to build machine learning models for predicting STEMI. Hub genes for further validated in patients with post-STEMI HF from GSE59867. Results: We identified 90 upregulated DEGs and nine downregulated DEGs convergence in the three datasets (|log2FC| ≥ 0.8 and adjusted p < 0.05). They were mainly enriched in GO terms relating to cytokine secretion, pattern recognition receptors signaling pathway, and immune cells activation. A cluster of eight genes including ITGAM, CLEC4D, SLC2A3, BST1, MCEMP1, PLAUR, GPR97, and MMP25 was found to be significant. A machine learning model built by SLC2A3, CLEC4D, GPR97, PLAUR, and BST1 exerted great value for STEMI prediction. Besides, ITGAM and BST1 might be candidate prognostic DEGs for post-STEMI HF. Conclusions: We reanalyzed the integrated transcriptomic signature of patients with STEMI showing predictive potentiality and revealed new insights and specific prospective DEGs for STEMI risk stratification and HF development.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital and National Center for Cardiovascular Diseases, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital and National Center for Cardiovascular Diseases, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
43
|
Predictive Biomarkers for Postmyocardial Infarction Heart Failure Using Machine Learning: A Secondary Analysis of a Cohort Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2903543. [PMID: 34938340 PMCID: PMC8687817 DOI: 10.1155/2021/2903543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
Background There are few biomarkers with an excellent predictive value for postacute myocardial infarction (MI) patients who developed heart failure (HF). This study aimed to screen candidate biomarkers to predict post-MI HF. Methods This is a secondary analysis of a single-center cohort study including nine post-MI HF patients and eight post-MI patients who remained HF-free over a 6-month follow-up. Transcriptional profiling was analyzed using the whole blood samples collected at admission, discharge, and 1-month follow-up. We screened differentially expressed genes and identified key modules using weighted gene coexpression network analysis. We confirmed the candidate biomarkers using the developed external datasets on post-MI HF. The receiver operating characteristic curves were created to evaluate the predictive value of these candidate biomarkers. Results A total of 6,778, 1,136, and 1,974 genes (dataset 1) were differently expressed at admission, discharge, and 1-month follow-up, respectively. The white and royal blue modules were most significantly correlated with post-MI HF (dataset 2). After overlapping dataset 1, dataset 2, and external datasets (dataset 3), we identified five candidate biomarkers, including FCGR2A, GSDMB, MIR330, MED1, and SQSTM1. When GSDMB and SQSTM1 were combined, the area under the curve achieved 1.00, 0.85, and 0.89 in admission, discharge, and 1-month follow-up, respectively. Conclusions This study demonstrates that FCGR2A, GSDMB, MIR330, MED1, and SQSTM1 are the candidate predictive biomarker genes for post-MI HF, and the combination of GSDMB and SQSTM1 has a high predictive value.
Collapse
|
44
|
Abreu MT, Baptista R, Girão H. Immune cell subsets as a marker of development of heart failure: The application of bioinformatics tools. Rev Port Cardiol 2021; 40:849-851. [PMID: 34857157 DOI: 10.1016/j.repce.2021.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Mónica Teresa Abreu
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| | - Rui Baptista
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Cardiology Department, Centro Hospitalar Entre Douro e Vouga, Santa Maria da Feira, Portugal
| | - Henrique Girão
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| |
Collapse
|
45
|
Lin B, Zheng W, Jiang X. Crosstalk between Circulatory Microenvironment and Vascular Endothelial Cells in Acute Myocardial Infarction. J Inflamm Res 2021; 14:5597-5610. [PMID: 34744446 PMCID: PMC8565985 DOI: 10.2147/jir.s316414] [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: 07/02/2021] [Accepted: 09/29/2021] [Indexed: 12/03/2022] Open
Abstract
Background The reason of high mortality of acute myocardial infarction (AMI) was the lack of exploring the cellular and molecular mechanism of AMI. Therefore, we explored the crosstalk among cells, as well as its potential molecular mechanism of mediating AMI. Methods The gene expression profile of peripheral blood, endothelial, platelets and mononuclear cells were applied to differentially expressed genes (DEGs) analysis. ClusterProfiler and the package of gene set enrichment analysis (GSEA) were applied to explore the potential functional pathways of DEGs in 3 types of intravascular cells (endothelial, platelets and mononuclear cells) and peripheral blood. Subsequently, we extracted the surface receptors, secreted proteins and extracellular matrix from the up-regulated DEGs to explore their potential interactions mechanism of AMI by crosstalk and pivot analysis. Findings A total 11 common regulated DEGs (CDEGs) were identified, which might be potential biomarkers for AMI diagnosis. The abnormal pathways involved in DEGs of 3 types of intravascular cells and peripheral blood were shown, which also verified by GSEA. Afterwards, it was found that there was crosstalk in 3 types of intravascular cells and peripheral blood. Furthermore, we constructed a cell–cell interaction map among cells in AMI regulated by exosome lncRNA, which was involved in the development of AMI. Finally, we identified 8 hub genes, which might be potential biomarkers of AMI. Interpretation The result of this study can not only be used as a reference for subsequent experiments and further exploration, but also contribute to the development of novel cell and molecular therapies.
Collapse
Affiliation(s)
- Beiyou Lin
- Department of Cardiology, Zhuhai People's Hospital, (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, People's Republic of China
| | - Weiwei Zheng
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital & Zhengzhou University People's Hospital & Henan University People's Hospital, Zhengzhou, 450003, Henan, People's Republic of China
| | - Xiaofei Jiang
- Department of Cardiology, Zhuhai People's Hospital, (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, 519000, People's Republic of China
| |
Collapse
|
46
|
Abreu MT, Baptista R, Girão H. Immune cell subsets as a marker of development of heart failure: The application of bioinformatics tools. Rev Port Cardiol 2021. [DOI: 10.1016/j.repc.2021.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
47
|
Chicco D, Oneto L. An Enhanced Random Forests Approach to Predict Heart Failure From Small Imbalanced Gene Expression Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:2759-2765. [PMID: 33259306 DOI: 10.1109/tcbb.2020.3041527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Myocardial infarctions and heart failure are the cause of more than 17 million deaths annually worldwide. ST-segment elevation myocardial infarctions (STEMI) require timely treatment, because delays of minutes have serious clinical impacts. Machine learning can provide alternative ways to predict heart failure and identify genes involved in heart failure. For these scopes, we applied a Random Forests classifier enhanced with feature elimination to microarray gene expression of 111 patients diagnosed with STEMI, and measured the classification performance through standard metrics such as the Matthews correlation coefficient (MCC) and area under the receiver operating characteristic curve (ROC AUC). Afterwards, we used the same approach to rank all genes by importance, and to detect the genes more strongly associated with heart failure. We validated this ranking by literature review and gene set enrichment analysis. Our classifier employed to predict heart failure achieved MCC = +0.87 and ROC AUC = 0.918, and our analysis identified KLHL22, WDR11, OR4Q3, GPATCH3, and FAH as top five protein-coding genes related to heart failure. Our results confirm the effectiveness of machine learning feature elimination in predicting heart failure from gene expression, and the top genes found by our approach will be able to help biologists and cardiologists further our understanding of heart failure.
Collapse
|
48
|
Chen JH, Wang LL, Tao L, Qi B, Wang Y, Guo YJ, Miao L. Identification of MYH6 as the potential gene for human ischaemic cardiomyopathy. J Cell Mol Med 2021; 25:10736-10746. [PMID: 34697898 PMCID: PMC8581323 DOI: 10.1111/jcmm.17015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/23/2021] [Accepted: 09/30/2021] [Indexed: 11/29/2022] Open
Abstract
The present study aimed to explore the potential hub genes and pathways of ischaemic cardiomyopathy (ICM) and to investigate the possible associated mechanisms. Two microarray data sets (GSE5406 and GSE57338) were downloaded from the Gene Expression Omnibus (GEO) database. The limma package was used to analyse the differentially expressed genes (DEGs). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, Disease Ontology (DO) and Gene Ontology (GO) annotation analyses were performed. A protein‐protein interaction (PPI) network was set up using Cytoscape software. Significant modules and hub genes were identified by the Molecular Complex Detection (MCODE) app. Then, further functional validation of hub genes in other microarrays and survival analysis were performed to judge the prognosis. A total of 1065 genes were matched, with an adjusted p < 0.05, and 17 were upregulated and 25 were downregulated with|log2 (fold change)|≥1.2. After removing the lengthy entries, GO identified 12 items, and 8 pathways were enriched at adjusted p < 0.05 (false discovery rate, FDR set at <0.05). Three modules with a score >8 after MCODE analysis and MYH6 were ultimately identified. When validated in GSE23561, MYH6 expression was lower in patients with CAD than in healthy controls (p < 0.05). GSE60993 data suggested that MYH6 expression was also lower in AMI patients (p < 0.05). In the GSE59867 data set, MYH6 expression was lower in CAD patients than in AMI patients and lower in heart failure (HF) patients than in non‐HF patients. However, there was no difference at different periods within half a year, and HF was increased when MYH6 expression was low (p < 0.05–0.01). We performed an integrated analysis and validation and found that MYH6 expression was closely related to ICM and HF. However, whether this marker can be used as a predictor in blood samples needs further experimental verification.
Collapse
Affiliation(s)
- Jian-Hong Chen
- Department of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Lei-Li Wang
- Department of Oncology, Liuzhou People's Hospital, Liuzhou, China
| | - Lin Tao
- Department of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Bin Qi
- Department of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Yong Wang
- Department of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Yu-Jie Guo
- Department of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Liu Miao
- Department of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| |
Collapse
|
49
|
Lee T, Lee H. Identification of Disease-Related Genes That Are Common between Alzheimer's and Cardiovascular Disease Using Blood Genome-Wide Transcriptome Analysis. Biomedicines 2021; 9:biomedicines9111525. [PMID: 34829754 PMCID: PMC8614900 DOI: 10.3390/biomedicines9111525] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
Accumulating evidence has suggested a shared pathophysiology between Alzheimer’s disease (AD) and cardiovascular disease (CVD). Based on genome-wide transcriptomes, specifically those of blood samples, we identify the shared disease-related signatures between AD and CVD. In addition to gene expressions in blood, the following prior knowledge were utilized to identify several candidate disease-related gene (DRG) sets: protein–protein interactions, transcription factors, disease–gene relationship databases, and single nucleotide polymorphisms. We selected the respective DRG sets for AD and CVD that show a high accuracy for disease prediction in bulk and single-cell gene expression datasets. Then, gene regulatory networks (GRNs) were constructed from each of the AD and CVD DRG sets to identify the upstream regulating genes. Using the GRNs, we identified two common upstream genes (GPBP1 and SETDB2) between the AD and CVD GRNs. In summary, this study has identified the potential AD- and CVD-related genes and common hub genes between these sets, which may help to elucidate the shared mechanisms between these two diseases.
Collapse
Affiliation(s)
- Taesic Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
- Department of Family Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Hyunju Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
- Correspondence: ; Tel.: +82-62-715-2213
| | | |
Collapse
|
50
|
Wang S, Wang E, Chen Q, Yang Y, Xu L, Zhang X, Wu R, Hu X, Wu Z. Uncovering Potential lncRNAs and mRNAs in the Progression From Acute Myocardial Infarction to Myocardial Fibrosis to Heart Failure. Front Cardiovasc Med 2021; 8:664044. [PMID: 34336943 PMCID: PMC8322527 DOI: 10.3389/fcvm.2021.664044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/15/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Morbidity and mortality of heart failure (HF) post-myocardial infarction (MI) remain elevated. The aim of this study was to find potential long non-coding RNAs (lncRNAs) and mRNAs in the progression from acute myocardial infarction (AMI) to myocardial fibrosis (MF) to HF. Methods: Firstly, blood samples from AMI, MF, and HF patients were used for RNA sequencing. Secondly, differentially expressed lncRNAs and mRNAs were obtained in MF vs. AMI and HF vs. MF, followed by functional analysis of shared differentially expressed mRNAs between two groups. Thirdly, interaction networks of lncRNA-nearby targeted mRNA and lncRNA-co-expressed mRNA were constructed in MF vs. AMI and HF vs. MF. Finally, expression validation and diagnostic capability analysis of selected lncRNAs and mRNAs were performed. Results: Several lncRNA-co-expressed/nearby targeted mRNA pairs including AC005392.3/AC007278.2-IL18R1, AL356356.1/AL137145.2-PFKFB3, and MKNK1-AS1/LINC01127-IL1R2 were identified. Several signaling pathways including TNF and cytokine–cytokine receptor interaction, fructose and mannose metabolism and HIF-1, hematopoietic cell lineage and fluid shear stress, and atherosclerosis and estrogen were selected. IL1R2, IRAK3, LRG1, and PLAC4 had a potential diagnostic value for both AMI and HF. Conclusion: Identified AC005392.3/AC007278.2-IL18R1, AL356356.1/AL137145.2-PFKFB3, and MKNK1-AS1/LINC01127-IL1R2 lncRNA-co-expressed/nearby targeted mRNA pairs may play crucial roles in the development of AMI, MF, and HF.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Enmao Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Qincong Chen
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Yan Yang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Lei Xu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaolei Zhang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Rubing Wu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xitian Hu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhihong Wu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| |
Collapse
|