Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(4): 886-892, 2024 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-39170023

ABSTRACT

Objective: To investigate the effect of the loss of myeloid-derived growth factor (Mydgf) on the transformation of cardiac fibroblasts into myofibroblasts after myocardial infarction (MI). Methods: Two adult mouse groups, including a wild-type (WT) group and another group with Mydgf knockout (Mydgf-KO), were examined in the study. The mice in these two groups were tested for their cardiac function by measuring left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) (n=10). Quantitative real-time PCR (qRT-PCR) (n=3) was performed to determine the mRNA expression levels of myofibroblast markers, including α-smooth muscle actin (α-SMA), periostin (postn), type Ⅷ collagen (col8al), and connective tissue growth factor (ctgf). Western blot (n=3) was performed to verify the protein expression levels of α-SMA. MI modeling was performed on the WT and the Mydgf-KO mice. Postoperative LVEF and LVFS (n=10) were then measured. The hearts were harvested and Masson staining was performed to determine the infarcted area (n=10). The heart samples of Mydgf-KO and WT mice were collected at d 7 and d 14 after MI, respectively, to verify the expression of myofibroblast markers (n=3). Results: Compared with WT mice, LVEF and LVFS in adult Mydgf-KO mice showed no significant changes (all P>0.05). However, the mRNA levels of α-SMA and postn were upregulated, and α-SMA protein expression was also increased (all P<0.05). After MI, compared with WT mice, LVEF and LVFS in Mydgf-KO mice decreased, and the infarcted area increased significantly (all P<0.05). Furthermore, mRNA levels of α-SMA, col8al, postn, and ctgf were increased in Mydgf-KO mice. In addition, the α-SMA protein expression level was upregulated and α-SMA-positive fibroblasts were increased (P<0.05). Conclusion: Mydgf deletion promotes the transformation of cardiac fibroblasts into myofibroblasts and aggravates myocardial fibrosis after MI.


Subject(s)
Connective Tissue Growth Factor , Fibrosis , Mice, Knockout , Myocardial Infarction , Myofibroblasts , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Mice , Myofibroblasts/metabolism , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Actins/metabolism , Actins/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Myocardium/metabolism , Myocardium/pathology , Mice, Inbred C57BL , Male , Fibroblasts/metabolism
2.
Cell Transplant ; 33: 9636897241266725, 2024.
Article in English | MEDLINE | ID: mdl-39126321

ABSTRACT

Many studies support the idea that long noncoding RNAs (lncRNAs) are significantly involved in the process of cardiomyocyte (CM) regeneration following a myocardial infarction (MI). This study aimed to systematically review the emerging role of lncRNAs in cardiac regeneration by promoting CM proliferation after MI. Furthermore, the review summarized potential targets and the underlying mechanisms of lncRNAs to induce heart regeneration, suggesting utilizing lncRNAs as innovative therapeutic targets for mitigating MI injuries. We searched the PubMed, Scopus, and Web of Science databases for studies on lncRNAs that play a role in heart regeneration after MI. We used search terms that included MI, lncRNAs, CM, and proliferation. Relevant English articles published until June 11, 2023, were systematically reviewed based on inclusion and exclusion criteria. A total of 361 publications were initially identified, and after applying the inclusion and exclusion criteria, nine articles were included in this systematic review. These studies investigated the role of critical lncRNAs in cardiac regeneration after MI, including five upregulated and four downregulated lncRNAs. Acting as a competitive endogenous RNA is one of the main roles of lncRNAs in regulating genes involved in CM proliferation through binding to target microRNAs. The main molecular processes that greatly increase CM proliferation are those that turn on the Hippo/YAP1, PI3K/Akt, JAK2-STAT3, and E2F1-ECRAR-ERK1/2 signaling pathways. This systematic review highlights the significant role of lncRNAs in heart regeneration after MI and their impact on CM proliferation. The findings suggest that lncRNAs could serve as potential targets for therapeutic interventions aiming to enhance cardiac function.


Subject(s)
Cell Proliferation , Myocardial Infarction , Myocytes, Cardiac , RNA, Long Noncoding , Regeneration , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Myocytes, Cardiac/metabolism , Cell Proliferation/genetics , Myocardial Infarction/therapy , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Humans , Animals
3.
BMC Cardiovasc Disord ; 24(1): 406, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39098896

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is a major disease with high morbidity and mortality worldwide. However, existing treatments are far from satisfactory, making the exploration of potent molecular targets more imperative. The E3 ubiquitin ligase RING finger protein 5 (RNF5) has been previously reported to be involved in several diseases by regulating ubiquitination-mediated protein degradation. Nevertheless, few reports have focused on its function in cardiovascular diseases, including MI. METHODS: In this study, we established RNF5 knockout mice through precise CRISPR-mediated genome editing and utilized left anterior descending coronary artery ligation in 9-11-week-old male C57BL/6 mice. Subsequently, serum biochemical analysis and histopathological examination of heart tissues were performed. Furthermore, we engineered adenoviruses for modulating RNF5 expression and subjected neonatal rat cardiomyocytes to oxygen-glucose deprivation (OGD) to mimic ischemic conditions, demonstrating the impact of RNF5 manipulation on cellular viability. Gene and protein expression analysis provided insights into the molecular mechanisms. Statistical methods were rigorously employed to assess the significance of experimental findings. RESULTS: We found RNF5 was downregulated in infarcted heart tissue of mice and NRCMs subjected to OGD treatment. RNF5 knockout in mice resulted in exacerbated heart dysfunction, more severe inflammatory responses, and increased apoptosis after MI surgery. In vitro, RNF5 knockdown exacerbated the OGD-induced decline in cell activity, increased apoptosis, while RNF5 overexpression had the opposite effect. Mechanistically, it was proven that the kinase cascade initiated by apoptosis signal-regulating kinase 1 (ASK1) activation was closely regulated by RNF5 and mediated RNF5's protective function during MI. CONCLUSIONS: We demonstrated the protective effect of RNF5 on myocardial infarction and its function was dependent on inhibiting the activation of ASK1, which adds a new regulatory component to the myocardial infarction associated network and promises to enable new therapeutic strategy.


Subject(s)
Apoptosis , Disease Models, Animal , MAP Kinase Kinase Kinase 5 , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction , Myocytes, Cardiac , Signal Transduction , Ubiquitin-Protein Ligases , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Kinase Kinase 5/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Cells, Cultured , Mice , Ventricular Function, Left , Cell Hypoxia , Rats
4.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125899

ABSTRACT

Pericardial fluid (PF) has been suggested as a reservoir of molecular targets that can be modulated for efficient repair after myocardial infarction (MI). Here, we set out to address the content of this biofluid after MI, namely in terms of microRNAs (miRs) that are important modulators of the cardiac pathological response. PF was collected during coronary artery bypass grafting (CABG) from two MI cohorts, patients with non-ST-segment elevation MI (NSTEMI) and patients with ST-segment elevation MI (STEMI), and a control group composed of patients with stable angina and without previous history of MI. The PF miR content was analyzed by small RNA sequencing, and its biological effect was assessed on human cardiac fibroblasts. PF accumulates fibrotic and inflammatory molecules in STEMI patients, namely causing the soluble suppression of tumorigenicity 2 (ST-2), which inversely correlates with the left ventricle ejection fraction. Although the PF of the three patient groups induce similar levels of fibroblast-to-myofibroblast activation in vitro, RNA sequencing revealed that PF from STEMI patients is particularly enriched not only in pro-fibrotic miRs but also anti-fibrotic miRs. Among those, miR-22-3p was herein found to inhibit TGF-ß-induced human cardiac fibroblast activation in vitro. PF constitutes an attractive source for screening diagnostic/prognostic miRs and for unveiling novel therapeutic targets in cardiac fibrosis.


Subject(s)
Fibrosis , MicroRNAs , Myocardial Infarction , Pericardial Fluid , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Male , Pericardial Fluid/metabolism , Female , Myocardium/metabolism , Myocardium/pathology , Middle Aged , Fibroblasts/metabolism , Aged , Transforming Growth Factor beta/metabolism , ST Elevation Myocardial Infarction/metabolism , ST Elevation Myocardial Infarction/pathology , ST Elevation Myocardial Infarction/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics
5.
BMC Cardiovasc Disord ; 24(1): 408, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103773

ABSTRACT

BACKGROUND: Acute myocardial infarction (AMI) is a leading cause of death worldwide. Mitochondrial dysfunction is a key determinant of cell death post-AMI. Preventing mitochondrial dysfunction is thus a key therapeutic strategy. This study aimed to explore key genes and target compounds related to mitochondrial dysfunction in AMI patients and their association with major adverse cardiovascular events (MACE). METHODS: Differentially expressed genes in AMI were identified from the Gene Expression Omnibus (GEO) datasets (GSE166780 and GSE24519), and mitochondria-related genes were obtained from MitoCarta3.0 database. By intersection of the two gene groups, mitochondria-related genes in AMI were identified. Next, the identified genes related to mitochondria were subject to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses. Protein-protein interaction (PPI) network was constructed, and key genes were screened. Then, targeted drug screening and molecular docking were performed. Blood samples from AMI patients and healthy volunteers were analyzed for the key genes expressions using quantitative real time polymerase chain reaction (qRT-PCR). Later, receiver operating characteristic (ROC) curves assessed the diagnostic value of key genes, and univariate and multivariate COX analyses identified risk factors and protective factors for MACE in AMI patients. RESULTS: After screening and identification, 138 mitochondria-related genes were identified, mainly enriched in the processes and pathways of cellular respiration, redox, mitochondrial metabolism, apoptosis, amino acid and fatty acid metabolism. According to the PPI network, 5 key mitochondria-related genes in AMI were obtained: translational activator of cytochrome c oxidase I (TACO1), cytochrome c oxidase subunit Va (COX5A), PTEN-induced putative kinase 1 (PINK1), SURF1, and NDUFA11. Molecular docking showed that Cholic Acid, N-Formylmethionine interacted with COX5A, nicotinamide adenine dinucleotide + hydrogen (NADH) and NDUFA11. Subsequent basic experiments revealed that COX5A and NDUFA11 expressions were significantly lower in the blood of patients with AMI than those in the corresponding healthy volunteers; also, AMI patients with MACE had lower COX5A and NDUFA11 expressions in the blood than those without MACE (P < 0.01). ROC analysis also showed high diagnostic value for COX5A and NDUFA11 [area under the curve (AUC) > 0.85]. In terms of COX results, COX5A, NDUFA11 and left ventricular ejection fraction (LVEF) were protective factors for MACE in AMI, while C-reactive protein (CRP) was a risk factor. CONCLUSION: COX5A and NDUFA11, key mitochondria-related genes in AMI, may be used as biomarkers to diagnose AMI and predict MACE.


Subject(s)
Databases, Genetic , Gene Regulatory Networks , Mitochondria, Heart , Myocardial Infarction , Predictive Value of Tests , Protein Interaction Maps , Humans , Male , Female , Middle Aged , Myocardial Infarction/genetics , Myocardial Infarction/diagnosis , Myocardial Infarction/blood , Prognosis , Risk Assessment , Aged , Mitochondria, Heart/metabolism , Mitochondria, Heart/genetics , Molecular Docking Simulation , Case-Control Studies , Mitochondrial Proteins/genetics , Gene Expression Profiling , Transcriptome , Genetic Markers , Genetic Predisposition to Disease
6.
PLoS One ; 19(8): e0308081, 2024.
Article in English | MEDLINE | ID: mdl-39088580

ABSTRACT

BACKGROUND: Inflammatory cytokines play a major role in the pathogenesis of myocardial infarction (MI). Although information on the importance of interleukin 13 (IL13) in human MI is limited, it has been well documented in the mouse model. Genetic variation in the IL13 gene has been associated with the structure and expression of the IL13. In the present study, we hypothesized that IL13 common genetic variants would be associated with a predisposition to the development of MI. MATERIALS AND METHODS: The present study enrolled 305 MI patients and 310 matched healthy controls. Common genetic polymorphisms in the IL13 gene (rs20541, rs1881457, and rs1800925) were genotyped using the TaqMan SNP genotyping method. Plasma levels of IL13 were measured using an enzyme-linked immunosorbent assay (ELISA). RESULTS: In MI patients, minor alleles of the IL13 rs1881457 and rs1800925 polymorphisms were less common than in healthy controls [rs1881457: AC (P = 0.004, OR = 0.61), C (P = 0.001, OR = 0.66); rs1800925: CT (P = 0.006, OR = 0.59)]. Further haplotype analysis of three studied SNPs revealed a significant association with predisposition to MI. Interestingly, IL13 rs1881457 and rs1800925 were linked to plasma levels of IL13: the reference genotype had higher levels, heterozygotes were intermediate, and the alternate genotype had the lowest levels. CONCLUSIONS: In the Chinese population, IL13 (rs1881457 and rs180092) variants are associated with different plasma IL13 levels and offer protection against MI development. However, additional research is required to validate our findings in different populations, including descent samples.


Subject(s)
Asian People , Genetic Predisposition to Disease , Haplotypes , Interleukin-13 , Myocardial Infarction , Polymorphism, Single Nucleotide , Humans , Myocardial Infarction/genetics , Male , Female , Case-Control Studies , Interleukin-13/genetics , Interleukin-13/blood , Middle Aged , Asian People/genetics , Aged , China/epidemiology , Alleles , Genotype , East Asian People
7.
Nat Commun ; 15(1): 6480, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090108

ABSTRACT

Regulatory T cells (Tregs) are key immune regulators that have shown promise in enhancing cardiac repair post-MI, although the mechanisms remain elusive. Here, we show that rapidly increasing Treg number in the circulation post-MI via systemic administration of exogenous Tregs improves cardiac function in male mice, by limiting cardiomyocyte death and reducing fibrosis. Mechanistically, exogenous Tregs quickly home to the infarcted heart and adopt an injury-specific transcriptome that mediates repair by modulating monocytes/macrophages. Specially, Tregs lead to a reduction in pro-inflammatory Ly6CHi CCR2+ monocytes/macrophages accompanied by a rapid shift of macrophages towards a pro-repair phenotype. Additionally, exogenous Treg-derived factors, including nidogen-1 and IL-10, along with a decrease in cardiac CD8+ T cell number, mediate the reduction of the pro-inflammatory monocyte/macrophage subset in the heart. Supporting the pivotal role of IL-10, exogenous Tregs knocked out for IL-10 lose their pro-repair capabilities. Together, this study highlights the beneficial use of a Treg-based therapeutic approach for cardiac repair with important mechanistic insights that could facilitate the development of novel immunotherapies for MI.


Subject(s)
Interleukin-10 , Macrophages , Mice, Inbred C57BL , Myocardial Infarction , T-Lymphocytes, Regulatory , Animals , Myocardial Infarction/immunology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , T-Lymphocytes, Regulatory/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Interleukin-10/metabolism , Interleukin-10/genetics , Phenotype , Myocardium/pathology , Myocardium/immunology , Myocardium/metabolism , Monocytes/immunology , Monocytes/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/immunology , Fibrosis , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Mice, Knockout
8.
Front Immunol ; 15: 1398990, 2024.
Article in English | MEDLINE | ID: mdl-39086489

ABSTRACT

Background: More and more evidence supports the association between myocardial infarction (MI) and osteoarthritis (OA). The purpose of this study is to explore the shared biomarkers and pathogenesis of MI complicated with OA by systems biology. Methods: Gene expression profiles of MI and OA were downloaded from the Gene Expression Omnibus (GEO) database. The Weighted Gene Co-Expression Network Analysis (WGCNA) and differentially expressed genes (DEGs) analysis were used to identify the common DEGs. The shared genes related to diseases were screened by three public databases, and the protein-protein interaction (PPI) network was built. GO and KEGG enrichment analyses were performed on the two parts of the genes respectively. The hub genes were intersected and verified by Least absolute shrinkage and selection operator (LASSO) analysis, receiver operating characteristic (ROC) curves, and single-cell RNA sequencing analysis. Finally, the hub genes differentially expressed in primary cardiomyocytes and chondrocytes were verified by RT-qPCR. The immune cell infiltration analysis, subtypes analysis, and transcription factors (TFs) prediction were carried out. Results: In this study, 23 common DEGs were obtained by WGCNA and DEGs analysis. In addition, 199 common genes were acquired from three public databases by PPI. Inflammation and immunity may be the common pathogenic mechanisms, and the MAPK signaling pathway may play a key role in both disorders. DUSP1, FOS, and THBS1 were identified as shared biomarkers, which is entirely consistent with the results of single-cell RNA sequencing analysis, and furher confirmed by RT-qPCR. Immune infiltration analysis illustrated that many types of immune cells were closely associated with MI and OA. Two potential subtypes were identified in both datasets. Furthermore, FOXC1 may be the crucial TF, and the relationship of TFs-hub genes-immune cells was visualized by the Sankey diagram, which could help discover the pathogenesis between MI and OA. Conclusion: In summary, this study first revealed 3 (DUSP1, FOS, and THBS1) novel shared biomarkers and signaling pathways underlying both MI and OA. Additionally, immune cells and key TFs related to 3 hub genes were examined to further clarify the regulation mechanism. Our study provides new insights into shared molecular mechanisms between MI and OA.


Subject(s)
Biomarkers , Gene Expression Profiling , Gene Regulatory Networks , Myocardial Infarction , Osteoarthritis , Protein Interaction Maps , Systems Biology , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Humans , Databases, Genetic , Transcriptome , Chondrocytes/metabolism , Chondrocytes/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Computational Biology/methods
9.
BMC Cardiovasc Disord ; 24(1): 365, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014329

ABSTRACT

BACKGROUND: M1 macrophages are closely associated with cardiac injury after myocardial infarction (MI). Increasing evidence shows that exosomes play a key role in pathophysiological regulation after MI, but the role of M1 macrophage-derived exosomes (M1-Exos) in myocardial regeneration remains unclear. In this study, we explored the impact of M1 macrophage-derived exosomes on cardiomyocytes regeneration in vitro and in vivo. METHODS: M0 macrophages were induced to differentiate into M1 macrophages with GM-CSF (50 ng/mL) and IFN-γ (20 ng/mL). Then M1-Exos were isolated and co-incubated with cardiomyocytes. Cardiomyocyte proliferation was detected by pH3 or ki67 staining. Quantitative real-time PCR (qPCR) was used to test the level of miR-155 in macrophages, macrophage-derived exosomes and exosome-treated cardiomyocytes. MI model was constructed and LV-miR-155 was injected around the infarct area, the proliferation of cardiomyocytes was counted by pH3 or ki67 staining. The downstream gene and pathway of miR-155 were predicted and verified by dual-luciferase reporter gene assay, qPCR and immunoblotting analysis. IL-6 (50 ng/mL) was added to cardiomyocytes transfected with miR-155 mimics, and the proliferation of cardiomyocytes was calculated by immunofluorescence. The protein expressions of IL-6R, p-JAK2 and p-STAT3 were detected by Western blot. RESULTS: The results showed that M1-Exos suppressed cardiomyocytes proliferation. Meanwhile, miR-155 was highly expressed in M1-Exos and transferred to cardiomyocytes. miR-155 inhibited the proliferation of cardiomyocytes and antagonized the pro-proliferation effect of interleukin 6 (IL-6). Furthermore, miR-155 targeted gene IL-6 receptor (IL-6R) and inhibited the Janus kinase 2(JAK)/Signal transducer and activator of transcription (STAT3) signaling pathway. CONCLUSION: M1-Exos inhibited cardiomyocyte proliferation by delivering miR-155 and inhibiting the IL-6R/JAK/STAT3 signaling pathway. This study provided new insight and potential treatment strategy for the regulation of myocardial regeneration and cardiac repair by macrophages.


Subject(s)
Cell Proliferation , Disease Models, Animal , Exosomes , Janus Kinase 2 , Macrophages , MicroRNAs , Myocardial Infarction , Myocytes, Cardiac , STAT3 Transcription Factor , Signal Transduction , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Exosomes/metabolism , Exosomes/transplantation , Exosomes/genetics , Animals , Cell Proliferation/drug effects , Macrophages/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/genetics , Janus Kinase 2/metabolism , Male , Regeneration , Rats, Sprague-Dawley , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Cells, Cultured , Phosphorylation , Coculture Techniques , Mice, Inbred C57BL , Interleukin-6/metabolism
10.
PLoS One ; 19(7): e0305532, 2024.
Article in English | MEDLINE | ID: mdl-39024234

ABSTRACT

OBJECTIVE: Acute myocardial infarction (AMI) is a severe condition with high morbidity and mortality rates. This study aimed to identify hub genes potentially associated with AMI and assess their clinical utility in predicting AMI occurrence. METHODS: Gene microarray data were obtained from the Gene Expression Omnibus (GEO) database. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were conducted on samples from patients with AMI and control samples to identify modules significantly associated with AMI. GO and KEGG analyses were applied to investigate the potential functions of these hub genes. Lastly, the mendelian randomization (MR) method was applied to analyze the causal relationship between the hub gene TNF and AMI. RESULTS: 285 differentially expressed genes (DEGs) were identified through WCGNA and were clustered into 6 modules. The yellow module appeared most relevant to AMI. Further exploration through GO and KEGG pathway enrichment showed that key hub genes in the yellow module were linked to positive regulation of cytokine production, cytokine receptor binding, NF-kappa B signaling pathway, IL-17 signaling pathway, and TNF signaling pathway. The top 10 genes identified through Cytoscape software analysis were IL1B, TNF, TLR4, TLR2, FCGR3B, MMP9, CXCL8, TLR8, ICAM1, and JUK. Utilizing inverse variance weighting (IVW) analysis, we discovered a significant association between TNF and AMI risk, with an OR of 0.946 (95% CI = 0.911-0.984, p = 0.005). CONCLUSIONS: The result of this study indicated that TNF, TLR2, TLR4, IL1B and FCGR3B may be potential biodiagnostic markers for AMI. TNF can inhibit inflammatory and oxidative stress responses in AMI, exerting a protective role in the heart.


Subject(s)
Gene Regulatory Networks , Mendelian Randomization Analysis , Myocardial Infarction , Humans , Myocardial Infarction/genetics , Gene Expression Profiling , Tumor Necrosis Factor-alpha/genetics , Signal Transduction/genetics
11.
Medicine (Baltimore) ; 103(29): e38781, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39029016

ABSTRACT

The objective of this study was to explore changes in miRNA133 levels as a basis for clinical diagnostic markers in patients with acute myocardial infarction (AMI). A total of 100 chest pain patient cases admitted to a hospital from June 2021 to December 2022 were used. The study involved the selection of 50 patients: 25 patients with unstable undetermined heart pain and 25 healthy subjects were included in the control group of 50 patients with non-AMI patients. Meanwhile, 50 patients with AMI were designated as the experimental group. Changes in miRNA133 levels in patients' plasma were analyzed for expression using quantitative fluorescence analysis. When the serum TPI, plasma NT-ProBNP, glycosylated hemoglobin, and plasma D-dimer index values were compared between the control and experimental groups, there was a statistically significant difference (P < .05). mi-RNA-133 had a mean plasma level value of 2.60 ±â€…1.01, the mean level value of mi-RNA-133 in patients with non-AMI was 1.34 ±â€…1.18, and the patients in the AMI group showed significantly high values of the mean plasma level of mi-RNA-133. The relative expression level value of cTnl in patients with AMI was 10.84 ±â€…12.64. Of the specificity and sensitivity diagnostics, mi-RNA-133 had the best diagnostic effect. The area under mi-RNA-133 in the regression curve was 95.4%, the specificity of the whole combination of indicators was 89.4% and the sensitivity was 100%. Finally, the correlation between mi-RNA-133 and white blood cell count (WBC) and TG was statistically significant (P < .05). In conclusion, changes in the level of mi-RNA-133 may be an important marker for diagnosing the status of patients with AMI, while a faster and more accurate method will emerge along with the improvement of the detection technology, and at the same time, due to the variability of the study cases and other limitations, further research will be carried out subsequently.


Subject(s)
Biomarkers , MicroRNAs , Myocardial Infarction , Humans , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Male , Female , MicroRNAs/blood , Middle Aged , Biomarkers/blood , Aged , Case-Control Studies , Sensitivity and Specificity , Natriuretic Peptide, Brain/blood , Adult , Peptide Fragments/blood , Fibrin Fibrinogen Degradation Products/analysis
12.
Medicine (Baltimore) ; 103(27): e38877, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968466

ABSTRACT

BACKGROUND: Both ischemic stroke (IS) and myocardial infarction (MI) are caused by vascular occlusion that results in ischemia. While there may be similarities in their mechanisms, the potential relationship between these 2 diseases has not been comprehensively analyzed. Therefore, this study explored the commonalities in the pathogenesis of IS and MI. METHODS: Datasets for IS (GSE58294, GSE16561) and MI (GSE60993, GSE61144) were downloaded from the Gene Expression Omnibus database. Transcriptome data from each of the 4 datasets were analyzed using bioinformatics, and the differentially expressed genes (DEGs) shared between IS and MI were identified and subsequently visualized using a Venn diagram. A protein-protein interaction (PPI) network was constructed using the Interacting Gene Retrieval Tool database, and identification of key core genes was performed using CytoHubba. Gene Ontology (GO) term annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the shared DEGs were conducted using prediction and network analysis methods, and the functions of the hub genes were determined using Metascape. RESULTS: The analysis revealed 116 and 1321 DEGs in the IS and MI datasets, respectively. Of the 75 DEGs shared between IS and MI, 56 were upregulated and 19 were downregulated. Furthermore, 15 core genes - S100a12, Hp, Clec4d, Cd163, Mmp9, Ormdl3, Il2rb, Orm1, Irak3, Tlr5, Lrg1, Clec4e, Clec5a, Mcemp1, and Ly96 - were identified. GO enrichment analysis of the DEGs showed that they were mainly involved in the biological functions of neutrophil degranulation, neutrophil activation during immune response, and cytokine secretion. KEGG analysis showed enrichment in pathways pertaining to Salmonella infection, Legionellosis, and inflammatory bowel disease. Finally, the core gene-transcription factor, gene-microRNA, and small-molecule relationships were predicted. CONCLUSION: These core genes may provide a novel theoretical basis for the diagnosis and treatment of IS and MI.


Subject(s)
Ischemic Stroke , Myocardial Infarction , Protein Interaction Maps , Humans , Myocardial Infarction/genetics , Ischemic Stroke/genetics , Protein Interaction Maps/genetics , Computational Biology/methods , Gene Expression Profiling , Databases, Genetic , Gene Regulatory Networks , Transcriptome/genetics , Gene Ontology
13.
J Transl Med ; 22(1): 612, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956669

ABSTRACT

BACKGROUND: Programmed cell death (PCD) has recently been implicated in modulating the removal of neutrophils recruited in acute myocardial infarction (AMI). Nonetheless, the clinical significance and biological mechanism of neutrophil-related PCD remain unexplored. METHODS: We employed an integrative machine learning-based computational framework to generate a predictive neutrophil-derived PCD signature (NPCDS) within five independent microarray cohorts from the peripheral blood of AMI patients. Non-negative matrix factorization was leveraged to develop an NPCDS-based AMI subtype. To elucidate the biological mechanism underlying NPCDS, we implemented single-cell transcriptomics on Cd45+ cells isolated from the murine heart of experimental AMI. We finally conducted a Mendelian randomization (MR) study and molecular docking to investigate the therapeutic value of NPCDS on AMI. RESULTS: We reported the robust and superior performance of NPCDS in AMI prediction, which contributed to an optimal combination of random forest and stepwise regression fitted on nine neutrophil-related PCD genes (MDM2, PTK2B, MYH9, IVNS1ABP, MAPK14, GNS, MYD88, TLR2, CFLAR). Two divergent NPCDS-based subtypes of AMI were revealed, in which subtype 1 was characterized as inflammation-activated with more vibrant neutrophil activities, whereas subtype 2 demonstrated the opposite. Mechanically, we unveiled the expression dynamics of NPCDS to regulate neutrophil transformation from a pro-inflammatory phase to an anti-inflammatory phase in AMI. We uncovered a significant causal association between genetic predisposition towards MDM2 expression and the risk of AMI. We also found that lidoflazine, isotetrandrine, and cepharanthine could stably target MDM2. CONCLUSION: Altogether, NPCDS offers significant implications for prediction, stratification, and therapeutic management for AMI.


Subject(s)
Apoptosis , Myocardial Infarction , Neutrophils , Myocardial Infarction/genetics , Myocardial Infarction/blood , Humans , Neutrophils/metabolism , Animals , Apoptosis/genetics , Machine Learning , Molecular Docking Simulation , Mice, Inbred C57BL , Transcriptome/genetics , Mice , Male
14.
Int J Med Sci ; 21(9): 1718-1729, 2024.
Article in English | MEDLINE | ID: mdl-39006833

ABSTRACT

Isoproterenol (ISO) administration is a well-established model for inducing myocardial injury, replicating key features of human myocardial infarction (MI). The ensuing inflammatory response plays a pivotal role in the progression of adverse cardiac remodeling, characterized by myocardial dysfunction, fibrosis, and hypertrophy. The Mst1/Hippo signaling pathway, a critical regulator of cellular processes, has emerged as a potential therapeutic target in cardiovascular diseases. This study investigates the role of Mst1 in ISO-induced myocardial injury and explores its underlying mechanisms. Our findings demonstrate that Mst1 ablation in cardiomyocytes attenuates ISO-induced cardiac dysfunction, preserving cardiomyocyte viability and function. Mechanistically, Mst1 deletion inhibits cardiomyocyte apoptosis, oxidative stress, and calcium overload, key contributors to myocardial injury. Furthermore, Mst1 ablation mitigates endoplasmic reticulum (ER) stress and mitochondrial fission, both of which are implicated in ISO-mediated cardiac damage. Additionally, Mst1 plays a crucial role in modulating the inflammatory response following ISO treatment, as its deletion suppresses pro-inflammatory cytokine expression and neutrophil infiltration. To further investigate the molecular mechanisms underlying ISO-induced myocardial injury, we conducted a bioinformatics analysis using the GSE207581 dataset. GO and KEGG pathway enrichment analyses revealed significant enrichment of genes associated with DNA damage response, DNA repair, protein ubiquitination, chromatin organization, autophagy, cell cycle, mTOR signaling, FoxO signaling, ubiquitin-mediated proteolysis, and nucleocytoplasmic transport. These findings underscore the significance of Mst1 in ISO-induced myocardial injury and highlight its potential as a therapeutic target for mitigating adverse cardiac remodeling. Further investigation into the intricate mechanisms of Mst1 signaling may pave the way for novel therapeutic interventions for myocardial infarction and heart failure.


Subject(s)
Hippo Signaling Pathway , Isoproterenol , Myocardial Infarction , Myocytes, Cardiac , Protein Serine-Threonine Kinases , Signal Transduction , Animals , Isoproterenol/adverse effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Signal Transduction/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mice , Humans , Myocardial Infarction/pathology , Myocardial Infarction/chemically induced , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Ventricular Remodeling/drug effects , Oxidative Stress/drug effects , Endoplasmic Reticulum Stress/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/genetics , Inflammation/pathology , Disease Models, Animal , Proto-Oncogene Proteins , Hepatocyte Growth Factor
15.
Article in English | MEDLINE | ID: mdl-39008646

ABSTRACT

Thromboembolic events are a common cause of morbidity and mortality with significant socioeconomic impact especially when young patients are affected. They are a rare medical event in young people and their clinical presentation can be mild or asymptomatic. The manifestation of symptoms and thrombotic events depends on both: the genetic mutations and the external risk factors that will induce the process. We present a case of a 34-year old young female, with three consecutive cerebrovascular insults in a period of ten years, and an acute myocardial infarction. There is a combination of gene mutations and polymorphism, with a predisposition to thromboembolic events. We emphasized the role of e-NOS (Endothelial nitric oxide synthase 786 T>C mutation) and the connection with smoking. The dual effect of the prolonged smoking and dysfunctional nitric oxide synthase in our young patient led to several thrombotic events. We discussed the various diagnostic tests and possible therapeutic and prophylactic strategies.


Subject(s)
Genetic Predisposition to Disease , Mutation , Nitric Oxide Synthase Type III , Thromboembolism , Humans , Female , Nitric Oxide Synthase Type III/genetics , Adult , Thromboembolism/genetics , Homozygote , Risk Factors , Smoking/adverse effects , Myocardial Infarction/genetics , Phenotype
16.
Medicine (Baltimore) ; 103(30): e38895, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058860

ABSTRACT

While observational studies suggest a connection between skipping breakfast and myocardial infarction (MI), the causal nature of this relationship is unclear. This study aims to investigate the genetic causal relationships between breakfast skipping and MI through Mendelian randomization (MR). Employing genetic data from a public genome-wide association study, this research focuses on genetic variations linked to breakfast skipping and MI. The primary analytical method was the inverse variance-weighted approach, complemented by additional methods like MR-Egger, weighted median, and mode analyses. It also includes heterogeneity and horizontal pleiotropy tests such as the Cochrane Q test, MR-Egger intercept, and MR-PRESSO tests, with a leave-one-out analysis for enhanced sensitivity assessment reliability. The study discovered a notable association between breakfast skipping and an increased risk of MI (odds ratios: 1.34, 95% confidence intervals: 1.03-1.76, P = .027). The test revealed no heterogeneity or multiplicity, and the sensitivity analysis confirmed the robustness of the results. Our MR analysis suggests that habitual breakfast skipping might elevate the likelihood of MI, underlining the importance of regular breakfast consumption in potentially mitigating heart attack risks.


Subject(s)
Breakfast , Genome-Wide Association Study , Mendelian Randomization Analysis , Myocardial Infarction , Humans , Myocardial Infarction/genetics , Myocardial Infarction/epidemiology , Feeding Behavior/physiology , Polymorphism, Single Nucleotide , Risk Factors , Intermittent Fasting
17.
Sci Rep ; 14(1): 15671, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977728

ABSTRACT

The study aims to explore the central genes that Kawasaki disease (KD) and Obesity (OB) may jointly contribute to coronary artery disease. Investigating single-cell datasets (GSE168732 and GSE163830) from a comprehensive gene expression database, we identified characteristic immune cell subpopulations in KD and OB. B cells emerged as the common immune cell characteristic subgroup in both conditions. Subsequently, we analyzed RNA sequencing datasets (GSE18606 and GSE87493) to identify genes associated with B-cell subpopulations in KD and OB. Lastly, a genome-wide association study and Mendelian randomization were conducted to substantiate the causal impact of these core genes on myocardial infarction. Quantitative real-time PCR (qRT-PCR) to validate the expression levels of hub genes in KD and OB. The overlapping characteristic genes of B cell clusters in both KD and OB yielded 70 shared characteristic genes. PPI analysis led to the discovery of eleven key genes that significantly contribute to the crosstalk. Employing receiver operating characteristic analysis, we evaluated the specificity and sensitivity of these core genes and scored them using Cytoscape software. The inverse variance weighting analysis suggested an association between TNFRSF17 and myocardial infarction risk, with an odds ratio of 0.9995 (95% CI = 0.9990-1.0000, p = 0.049). By employing a single-cell combined transcriptome data analysis, we successfully pinpointed central genes associated with both KD and OB. The implications of these findings extend to shedding light on the increased risk of coronary artery disease resulting from the co-occurrence of OB and KD.


Subject(s)
B-Lymphocytes , Genome-Wide Association Study , Mucocutaneous Lymph Node Syndrome , Pediatric Obesity , Transcriptome , Mucocutaneous Lymph Node Syndrome/genetics , Humans , Pediatric Obesity/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Child , Gene Expression Profiling , Male , Female , Mendelian Randomization Analysis , Coronary Artery Disease/genetics , Coronary Artery Disease/etiology , Child, Preschool , Myocardial Infarction/genetics , Single-Cell Analysis
18.
Clin Res Hepatol Gastroenterol ; 48(7): 102419, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992425

ABSTRACT

BACKGROUND AND AIMS: Primary biliary cholangitis (PBC) is an autoimmune disease often accompanied by multisystem damage. This study aimed to explore the causal association between genetically predicted PBC and diabetes, as well as multiple cardiovascular diseases (CVDs). METHODS: Genome-wide association studies (GWAS) summary data of PBC in 24,510 individuals of European ancestry from the European Association for the Study of the Liver was used to identify genetically predicted PBC. We conducted 2-sample single-variable Mendelian randomization (SVMR) and multivariable Mendelian randomization (MVMR) to estimate the impacts of PBC on diabetes (N = 17,685 to 318,014) and 20 CVDs from the genetic consortium (N = 171,875 to 1,030,836). RESULTS: SVMR provided evidence that genetically predicted PBC is associated with an increased risk of type 1 diabetes (T1D), type 2 diabetes (T2D), myocardial infarction (MI), heart failure (HF), hypertension, atrial fibrillation (AF), stroke, ischemic stroke, and small-vessel ischemic stroke. Additionally, there was no evidence of a causal association between PBC and coronary atherosclerosis. In the MVMR analysis, PBC maintained independent effects on T1D, HF, MI, and small-vessel ischemic stroke in most models. CONCLUSION: Our findings revealed the causal effects of PBC on diabetes and 7 CVDs, and no causal relationship was detected between PBC and coronary atherosclerosis.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 1 , Genome-Wide Association Study , Liver Cirrhosis, Biliary , Mendelian Randomization Analysis , Humans , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/complications , Cardiovascular Diseases/genetics , Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/complications , Atrial Fibrillation/genetics , Myocardial Infarction/genetics , Myocardial Infarction/epidemiology , Hypertension/complications , Hypertension/genetics , Heart Failure/genetics , Stroke/genetics , Stroke/etiology , Stroke/epidemiology
19.
J Am Heart Assoc ; 13(15): e031515, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39028008

ABSTRACT

BACKGROUND: Gene therapy has been proposed as a strategy to induce cardiac regeneration following acute myocardial infarction (AMI). Given that Tbx20, a transcription factor of the T-box subfamily, stimulates cell proliferation and angiogenesis, we designed a baculovirus overexpressing Tbx20 (Bv-Tbx20) and evaluated its effects in cultured cardiomyocytes and in an ovine model of AMI. METHODS AND RESULTS: Cell proliferation and angiogenesis were measured in cardiomyocytes transduced with Bv-Tbx20 or Bv-Null (control). Subsequently, in sheep with AMI, Bv-Tbx20 or Bv-Null was injected in the infarct border. Cardiomyocyte cell cycle activity, angioarteriogenesis, left ventricular function, and infarct size were assessed. Cardiomyocytes transduced with BvTbx20 increased cell proliferation, cell cycle regulatory and angiogenic gene expression, and tubulogenesis. At 7 days posttreatment, sheep treated with Bv-Tbx20 showed increased Tbx20, promitotic and angiogenic gene expression, decreased levels of P21, increased Ki67- (17.09±5.73 versus 7.77±7.24 cardiomyocytes/mm2, P<0.05) and PHH3 (phospho-histone H3)-labeled cardiomyocytes (10.10±3.51 versus 5.23±2.87 cardiomyocytes/mm2, P<0.05), and increased capillary (2302.68±353.58 versus 1694.52±211.36 capillaries/mm2, P<0.001) and arteriolar (146.95±53.14 versus 84.06±16.84 arterioles/mm2, P<0.05) densities. At 30 days, Bv-Tbx20 decreased infarct size (9.89±1.92% versus 12.62±1.33%, P<0.05) and slightly improved left ventricular function. Baculoviral gene transfer-mediated Tbx20 overexpression exerted angiogenic and cardiomyogenic effects in vitro. CONCLUSIONS: In sheep with AMI, Bv-Tbx20 induced angioarteriogenesis, cardiomyocyte cell cycle activity, infarct size limitation, and a slight recovery of left ventricular function, suggesting that Bv-Tbx20 gene therapy may contribute to cardiac regeneration following AMI.


Subject(s)
Baculoviridae , Genetic Therapy , Myocardial Infarction , Myocytes, Cardiac , Neovascularization, Physiologic , T-Box Domain Proteins , Animals , Baculoviridae/genetics , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sheep , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Ventricular Function, Left
20.
Acta Physiol (Oxf) ; 240(9): e14202, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39016532

ABSTRACT

AIM: The transcriptional factor HIF-1α is recognized for its contribution to cardioprotection against acute ischemia/reperfusion injury. Adaptation to chronic hypoxia (CH) is known to stabilize HIF-1α and increase myocardial ischemic tolerance. However, the precise role of HIF-1α in mediating the protective effect remains incompletely understood. METHODS: Male wild-type (WT) mice and mice with partial Hif1a deficiency (hif1a +/-) were exposed to CH for 4 weeks, while their respective controls were kept under normoxic conditions. Subsequently, their isolated perfused hearts were subjected to ischemia/reperfusion to determine infarct size, while RNA-sequencing of isolated cardiomyocytes was performed. Mitochondrial respiration was measured to evaluate mitochondrial function, and western blots were performed to assess mitophagy. RESULTS: We demonstrated enhanced ischemic tolerance in WT mice induced by adaptation to CH compared with their normoxic controls and chronically hypoxic hif1a +/- mice. Through cardiomyocyte bulk mRNA sequencing analysis, we unveiled significant reprogramming of cardiomyocytes induced by CH emphasizing mitochondrial processes. CH reduced mitochondrial content and respiration and altered mitochondrial ultrastructure. Notably, the reduced mitochondrial content correlated with enhanced autophagosome formation exclusively in chronically hypoxic WT mice, supported by an increase in the LC3-II/LC3-I ratio, expression of PINK1, and degradation of SQSTM1/p62. Furthermore, pretreatment with the mitochondrial division inhibitor (mdivi-1) abolished the infarct size-limiting effect of CH in WT mice, highlighting the key role of mitophagy in CH-induced cardioprotection. CONCLUSION: These findings provide new insights into the contribution of HIF-1α to cardiomyocyte survival during acute ischemia/reperfusion injury by activating the selective autophagy pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia , Mitophagy , Myocardial Infarction , Animals , Mitophagy/physiology , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/genetics , Hypoxia/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Adaptation, Physiological/physiology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL