Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
BMC Cardiovasc Disord ; 24(1): 346, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977948

ABSTRACT

OBJECTIVE: We aimed to investigate key differentially expressed immune related genes in persistent atrial fibrillation. METHODS: Gene expression profiles were downloaded from Gene Expression Omnibus (GEO) using "GEO query" package. "limma" package and "sva" package were used to conduct normalization and eliminate batch effects, respectively. We screened out differentially expressed genes (DEGs) based on "limma" package with the standard of |log fold change (FC)| ≥ 1.5 and false discovery rate (FDR) < 0.05. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of DEGs were performed by "clusterProfler" package. We further applied LASSO to select key DEGs, and intersected key DEGs with immune related genes from ImmPort database. The ROC curve of each DEIRG was constructed to evaluate its diagnostic efficiency for AF. RESULTS: A total of 103 DEGs we were screened out, of them, 48 genes were down-regulated and 55 genes were up-regulated. Result of functional enrichment analysis show that, most of DEGs were related to immune response, inflammation, and oxidative stress. Ultimately, CYBB, RORB, S100A12, and CHGB were determined as key DEIRGs, each of which displayed a favor efficiency for diagnosing persistent AF. CONCLUSION: CYBB, RORB, S100A12, and CHGB were identified as key DEIRGs in persistent AF, and future studies are needed to further explore the underlying roles of CYBB, RORB, S100A12, and CHGB in persistent AF.


Subject(s)
Atrial Fibrillation , Computational Biology , Databases, Genetic , Gene Expression Profiling , Transcriptome , Atrial Fibrillation/genetics , Atrial Fibrillation/immunology , Atrial Fibrillation/diagnosis , Humans , Gene Regulatory Networks , Predictive Value of Tests , Genetic Markers , Genetic Predisposition to Disease , Gene Expression Regulation
2.
BMC Cardiovasc Disord ; 24(1): 334, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38961330

ABSTRACT

BACKGROUND: Systemic inflammation markers have recently been identified as being associated with cardiac disorders. However, limited research has been conducted to estimate the pre-diagnostic associations between these markers and paroxysmal atrial fibrillation (PAF). Our aim is to identify potential biomarkers for early detection of PAF. METHODS: 91 participants in the PAF group and 97 participants in the non-PAF group were included in this study. We investigated the correlations between three systemic inflammation markers, namely the systemic immune inflammation index (SII), system inflammation response index (SIRI), and aggregate index of systemic inflammation (AISI), and PAF. RESULTS: The proportion of patients with PAF gradually increased with increasing logSII, logSIRI, and logAISI tertiles. Compared to those in the lowest tertiles, the PAF risks in the highest logSII and logSIRI tertiles were 3.2-fold and 2.9-fold, respectively. Conversely, there was no significant correlation observed between logAISI and PAF risk within the highest tertile of logAISI. The restricted cubic splines (RCS) analysis revealed a non-linear relationship between the elevation of systemic inflammation markers and PAF risk. Specifically, the incidence of PAF is respectively increased by 56%, 95%, and 150% for each standard deviation increase in these variables. The ROC curve analysis of logSII, logSIRI and logAISI showed that they had AUC of 0.6, 0.7 and 0.6, respectively. It also demonstrated favorable sensitivity and specificity of these systemic inflammation markers in detecting the presence of PAF. CONCLUSIONS: In conclusion, our study reveals significant positive correlations between SII, SIRI, and AISI with the incidence of PAF.


Subject(s)
Atrial Fibrillation , Biomarkers , Inflammation Mediators , Inflammation , Predictive Value of Tests , Humans , Atrial Fibrillation/diagnosis , Atrial Fibrillation/blood , Atrial Fibrillation/immunology , Atrial Fibrillation/epidemiology , Male , Female , Middle Aged , Biomarkers/blood , Inflammation/blood , Inflammation/diagnosis , Inflammation/immunology , Inflammation/epidemiology , Inflammation Mediators/blood , Aged , Risk Assessment , Risk Factors , Incidence , Case-Control Studies , Early Diagnosis
4.
Gen Physiol Biophys ; 43(3): 209-219, 2024 May.
Article in English | MEDLINE | ID: mdl-38774921

ABSTRACT

Atrial fibrillation (AF) is the most common cardiac arrhythmia and can cause serious complications. Several studies have shown that neutrophils may influence AF progression. However, the key genes related to neutrophils in AF have not been fully elucidated. Here, we downloaded microarray expression data of AF, and screened differentially expressed genes. Key immune cells in AF were identified by immune cell infiltration analysis. Weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) analysis were used to construct gene co-expression modules and identify hub genes. The association between key genes and neutrophils was then verified. Our results showed that 303 differentially expressed genes (DEGs) were screened in AF and sinus rhythm (SR), of which 194 were up-regulated and 109 were down-regulated. DEGs were mainly enriched in functions and pathways of neutrophil activation and biological functions of neutrophil activation-mediated immune response. Immune infiltration analysis revealed elevated levels of neutrophil infiltration in AF. WGCNA analysis revealed that the modules in dark red were associated with neutrophils. PPI analysis of these modules yielded 10 hub genes. S100A12, FCGR3B and S100A8 are 3 potential key genes related to neutrophils in AF, which are significantly positively correlated with neutrophils. These genes deserve further investigation and may be potential therapeutic targets for AF.


Subject(s)
Atrial Fibrillation , Neutrophils , Atrial Fibrillation/genetics , Atrial Fibrillation/immunology , Neutrophils/metabolism , Neutrophils/immunology , Humans , Protein Interaction Maps/genetics , Gene Regulatory Networks , Gene Expression Profiling
5.
Medicine (Baltimore) ; 103(19): e38079, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728471

ABSTRACT

Atrial fibrillation (AF) is a prevalent cardiac arrhythmia, with recent research indicating a correlation between immune system characteristics and the development of AF. However, it remains uncertain whether the immunological response is the primary underlying component or a secondary consequence of AF. Initially, we investigated the effect of immune cells on AF by performing forward Mendelian randomization (MR) analyses with immune cells as the exposure variable and their associated genetic variants as instrumental variables. Subsequently, we performed reverse MR analyses with AF as the exposure variable and immune cells as the outcome variable to exclude the interference of reverse causality, to distinguish between primary and secondary effects, and to further elucidate the causal relationship between the immune system and AF. We discovered that membrane proteins on specific immune cells, such as CD25 on memory B cells-which functions as a part of the interleukin-2 receptor-may be risk factors for AF development, with odds ratios of 1.0233 (95% confidence interval: 1.0012-1.0458, P = .0383). In addition, certain immune cell counts, such as the CD4 regulatory T cell Absolute Count, play a protective factor in the development of AF (odds ratio: 0.9513, 95% confidence interval: 0.9165-0.9874; P = .0086). More detailed results are elaborated in the main text. Our MR study has yielded evidence that substantiates a genetically inferred causal association between the immune system and AF. Identifying the risk factors associated with AF is vital to facilitate the development of innovative pharmaceutical treatments.


Subject(s)
Atrial Fibrillation , Mendelian Randomization Analysis , Atrial Fibrillation/genetics , Atrial Fibrillation/immunology , Atrial Fibrillation/epidemiology , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Risk Factors , B-Lymphocytes/immunology
6.
Science ; 381(6654): 231-239, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37440641

ABSTRACT

Atrial fibrillation disrupts contraction of the atria, leading to stroke and heart failure. We deciphered how immune and stromal cells contribute to atrial fibrillation. Single-cell transcriptomes from human atria documented inflammatory monocyte and SPP1+ macrophage expansion in atrial fibrillation. Combining hypertension, obesity, and mitral valve regurgitation (HOMER) in mice elicited enlarged, fibrosed, and fibrillation-prone atria. Single-cell transcriptomes from HOMER mouse atria recapitulated cell composition and transcriptome changes observed in patients. Inhibiting monocyte migration reduced arrhythmia in Ccr2-∕- HOMER mice. Cell-cell interaction analysis identified SPP1 as a pleiotropic signal that promotes atrial fibrillation through cross-talk with local immune and stromal cells. Deleting Spp1 reduced atrial fibrillation in HOMER mice. These results identify SPP1+ macrophages as targets for immunotherapy in atrial fibrillation.


Subject(s)
Atrial Fibrillation , Macrophages , Osteopontin , Animals , Humans , Mice , Atrial Fibrillation/genetics , Atrial Fibrillation/immunology , Heart Atria , Macrophages/immunology , Mitral Valve Insufficiency/genetics , Osteopontin/genetics , Gene Deletion , Cell Movement , Single-Cell Gene Expression Analysis
7.
Comput Math Methods Med ; 2022: 4029840, 2022.
Article in English | MEDLINE | ID: mdl-35273648

ABSTRACT

Objective: To identify potential key biomarkers and characterize immune infiltration in atrial tissue of patients with atrial fibrillation (AF) through bioinformatics analysis. Methods: Differentially expressed genes (DEGs) were identified by the LIMMA package in Bioconductor, and functional and pathway enrichment analyses were undertaken using GO and KEGG. The LASSO logistic regression and BORUTA algorithm were employed to screen for potential novel key markers of AF from all DEGs. Gene set variation analysis was also performed. Single-sample gene set enrichment analysis was employed to quantify the infiltration levels for each immune cell type, and the correlation between hub genes and infiltrating immune cells was analyzed. Results: A total of 52 DEGs were identified, including of 26 downregulated DEGs and 26 upregulated DEGs. DEGs were primarily enriched in the Major Histocompatibility Complex class II protein complex, glucose homeostasis, protein tetramerization, regulation of synapse organization, cytokine activity, heart morphogenesis, and blood circulation. Three downregulated genes and three upregulated genes were screened by LASSO logistic regression and the BORUTA algorithm. Finally, immune infiltration analysis indicated that the atrial tissue of AF patients contained significant infiltration of APC_co_inhibition, Mast_cell, neutrophils, pDCs, T_cell_costimulation, and Th1_cells compared with paired sinus rhythm (SR) atrial tissue, and the three downregulated genes were negatively correlated with the six kinds of immune cells mentioned above. Conclusion: The hub genes identified in this study and the differences in immune infiltration of atrial tissue observed between AF and SR tissue might help to characterize the occurrence and progression of AF.


Subject(s)
Atrial Fibrillation/genetics , Atrial Fibrillation/immunology , Genetic Markers/immunology , Heart Atria/immunology , Heart Atria/pathology , Algorithms , Atrial Fibrillation/metabolism , Biomarkers/metabolism , Case-Control Studies , Computational Biology , Databases, Genetic , Down-Regulation , Gene Ontology , Gene Regulatory Networks , Heart Atria/metabolism , Humans , Immune System/immunology , Immune System/pathology , Leukocytes/classification , Leukocytes/immunology , Leukocytes/pathology , Logistic Models
8.
BMC Cardiovasc Disord ; 22(1): 45, 2022 02 13.
Article in English | MEDLINE | ID: mdl-35152878

ABSTRACT

BACKGROUND AND AIMS: Inflammation plays a key role in the initiation and progression of atrial fibrillation (AF). The systemic inflammation indexes are easily evaluated and predict AF development. However, it's role in prediction of recurrence of AF is unknown. We aim to explore the association between the systemic inflammation indexes and recurrence of AF in patients underwent cryoablation (CryoMaze) concomitant with mitral valve surgery. METHODS: We examined systemic inflammation indexes during perioperative period in 122 patients between 2015 and 2018. Systemic inflammation indexes were developed by systemic immune-inflammation index (SII), neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), and lymphocytes to monocytes ratio. Univariate and multivariate analyses were performed to examine the association of each markers with recurrence of AF. RESULTS: Of the 122 patients included in this study, 22 patients (18%) experienced AF recurrence after CryoMaze concomitant with mitral valve surgery. There is no significant difference between each systemic inflammation indexes before surgery and recurrence of AF. In univariate analysis, MLR after surgery 3 days, PLR, MPLR, NLR, SII after surgery 7 days were able to predict recurrence of AF. In multivariate analyses, SII ≥ 1696 independently predicted recurrence (OR, 3.719; 95% CI, 1.417-9.760). Interestingly, baseline SII showed no significant in prediction of recurrence. It was sharply elevated after surgery and dropped slowly. In patients of recurrence, SII after 7 days of surgery increased again. CONCLUSIONS: The raised SII again was associated with an increased risk of the postoperative recurrence of AF and independently predicted the late recurrence of AF after CryoMaze concomitant with mitral valve surgery.


Subject(s)
Atrial Fibrillation/surgery , Cryosurgery/adverse effects , Decision Support Techniques , Heart Valve Diseases/surgery , Inflammation/diagnosis , Maze Procedure/adverse effects , Mitral Valve/surgery , Aged , Atrial Fibrillation/diagnosis , Atrial Fibrillation/immunology , Atrial Fibrillation/physiopathology , Blood Platelets/immunology , Female , Heart Valve Diseases/diagnosis , Heart Valve Diseases/immunology , Heart Valve Diseases/physiopathology , Humans , Inflammation/immunology , Lymphocyte Count , Lymphocytes/immunology , Male , Middle Aged , Mitral Valve/physiopathology , Monocytes/immunology , Platelet Count , Predictive Value of Tests , Recurrence , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
9.
J Clin Lab Anal ; 36(2): e24217, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34970783

ABSTRACT

BACKGROUND: Inflammation plays a key role in the initiation and progression of atrial fibrillation (AF). Lymphocyte-to-monocyte ratio (LMR) has been proved to be a reliable predictor of many inflammation-associated diseases, but little data are available on the relationship between LMR and AF. We aimed to evaluate the predictive value of LMR in predicting all-cause mortality among AF patients. METHODS: Data of patients diagnosed with AF were retrieved from the Medical Information Mart for Intensive Care-III (MIMIC-III) database. X-tile analysis was used to calculate the optimal cutoff value for LMR. The Cox regression model was used to assess the association of LMR and 28-day, 90-day, and 1-year mortality. Additionally, a propensity score matching (PSM) method was performed to minimize the impact of potential confounders. RESULTS: A total of 3567 patients hospitalized with AF were enrolled in this study. The X-tile software indicated that the optimal cutoff value of LMR was 2.67. A total of 1127 pairs were generated, and all the covariates were well balanced after PSM. The Cox proportional-hazards model showed that patients with the low LMR (≤2.67) had a higher 1-year all-cause mortality than those with the high LMR (>2.67) in the study cohort before PSM (HR = 1.640, 95% CI: 1.437-1.872, p < 0.001) and after PSM (HR = 1.279, 95% CI: 1.094-1.495, p = 0.002). The multivariable Cox regression analysis for 28-day and 90-day mortality yielded similar results. CONCLUSIONS: The lower LMR (≤2.67) was associated with a higher risk of 28-day, 90-day, and 1-year all-cause mortality, which might serve as an independent predictor in AF patients.


Subject(s)
Atrial Fibrillation/immunology , Lymphocytes , Monocytes , Propensity Score , Aged , Aged, 80 and over , Atrial Fibrillation/mortality , Female , Humans , Leukocyte Count , Male , Prognosis , Proportional Hazards Models
10.
Clin Res Cardiol ; 111(1): 96-104, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34559294

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is the most common age-related cardiac arrhythmia. The etiology underlying AF is still largely unknown. At the intersection of the innate immune system and hemostasis, immunothrombosis may be a possible cause of atrial remodeling, and therefore be an underlying cause of AF. METHODS: From 1990 to 2014, we followed participants aged 55 and over, free from AF at inclusion. Immunothrombosis factors fibrinogen, von Willebrand factor, ADAMTS13, and neutrophil extracellular traps (NETs) levels were measured at baseline. Participants were followed until either onset of AF, loss-to-follow-up, or reaching the end-date of 01-01-2014. Cox proportional hazard modelling was used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs), adjusted for cardiovascular risk factors. RESULTS: We followed 6174 participants (mean age 69.1 years, 57% women) for a median follow-up time of 12.8 years. 364 men (13.7%, incidence rate 13.0/1000 person-years) and 365 women (10.4%, incidence rate 8.9/1000 person-years) developed AF. We found no significant association between markers of immunothrombosis and new-onset AF after adjusting for cardiovascular risk factors [HR 1.00 (95% CI 0.93-1.08) for fibrinogen, 1.04 (0.97-1.12) for von Willebrand factor, 1.00 (1.00-1.01) for ADAMTS13, and 1.01 (0.94-1.09) for NETs]. In addition, we found no differences in associations between men and women. CONCLUSION: We found no associations between markers of immunothrombosis and new-onset AF in the general population. Inflammation and immunothrombosis may be associated with AF through other cardiovascular risk factors or predisposing conditions of AF. Our findings challenge the added value of biomarkers in AF risk prediction.


Subject(s)
Atrial Fibrillation/etiology , Atrial Fibrillation/immunology , Thromboinflammation/complications , Thromboinflammation/immunology , Aged , Atrial Fibrillation/epidemiology , Biomarkers/blood , Female , Humans , Immunity, Innate , Incidence , Male , Middle Aged , Netherlands/epidemiology , Prospective Studies , Risk Factors , Thromboinflammation/epidemiology
11.
Front Immunol ; 12: 744254, 2021.
Article in English | MEDLINE | ID: mdl-34721413

ABSTRACT

Patients with rheumatoid arthritis (RA) have a significantly high risk of atrial fibrillation (AF). This study aimed to compare the absolute and relative changes in peripheral T cells in patients with RA who were also affected with and without AF. To help make an early diagnosis and prevent the initiation and progression of AF, the changes in the lymphocyte subsets were assessed in RA patients with and without AF. A propensity score matching (PSM) system (1:3) was used to perform a matched case-control study with 40 RA-AF cases and 120 RA controls. Changes in the erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), anti-citrullinated peptide antibody (ACPA), and rheumatoid factor (RF) were examined. The percentage and absolute number of T, B, natural killer (NK), T helper (Th)1, Th2, Th17, and T-regulatory (Treg) cells in the peripheral blood of patients with and without RA-AF were determined using flow cytometry. Univariate and multivariate analyses were performed to determine the association between peripheral lymphocytes and RA-AF. Demographic data, ESR, CRP, ACPA, and the percentage, as well as the absolute value of B, NK, Th2, and Treg cells, showed no significant differences between the propensity score-matched groups of RA and RA-AF. Meanwhile, the absolute number and percentage of Th1 cells, the absolute number of Th17 cells, the ratio of Th1/Treg, Th17/Treg, and RF were significantly higher in patients with RA-AF than those in the control groups (P < 0.05). Univariate and multivariate logistic regression analyses also revealed that the percentage of Th1 cells, the absolute number of Th17 cells, and the ratio of Th1/Treg were associated with a significantly higher risk of AF. This PSM study demonstrated that the incidence of AF was higher in RA patients with Th cell immunological derangements.


Subject(s)
Arthritis, Rheumatoid/immunology , Atrial Fibrillation/epidemiology , Atrial Fibrillation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Aged , Arthritis, Rheumatoid/complications , Case-Control Studies , Female , Humans , Incidence , Male , Middle Aged , Propensity Score , Retrospective Studies
12.
Eur J Pharmacol ; 913: 174622, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34748769

ABSTRACT

Sepsis is a known risk factor for new-onset atrial fibrillation (AF), and previous studies have demonstrated that ferroptosis participates in sepsis-induced organ injury development. Nevertheless, the role of ferroptosis in new-onset AF with sepsis remains largely unknown. This study aims to investigate the underlying mechanisms linking ferroptosis and AF caused by sepsis. LPS-induced endotoxemia is often used to model the acute inflammatory response associated with sepsis. Herein, we reported that ferroptosis was significantly activated in LPS-induced endotoxemia rat model. We also observed that ferroportin (Fpn), the only identified mammalian non-heme iron exporter, was downregulated in the atrium of endotoxemia model. Vulnerability to AF was also significantly increased in a endotoxemia rat model. Additionally, Fpn knockdown by shFpn further increased intracellular iron concentration and oxidative stress and exaggerated the AF vulnerability, which was alleviated by ferroptosis inhibition. Mechanistically, silencing Fpn worsened the alterations in calcium handling proteins expression in a endotoxemia rat model. These findings suggest that Fpn-mediated ferroptosis is involved in the new-onset AF with LPS-induced endotoxemia via worsening the calcium handling proteins dysregulation and provides a novel and promising strategy for preventing AF development in sepsis.


Subject(s)
Atrial Fibrillation/immunology , Cation Transport Proteins/metabolism , Endotoxemia/complications , Animals , Atrial Fibrillation/pathology , Cation Transport Proteins/genetics , Disease Models, Animal , Down-Regulation/immunology , Endotoxemia/immunology , Ferroptosis/immunology , Gene Knockdown Techniques , Heart Atria/immunology , Heart Atria/pathology , Humans , Male , Oxidative Stress/immunology , Rats , Rats, Transgenic , Signal Transduction/immunology
13.
Inflamm Res ; 70(10-12): 1011-1014, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34536081

ABSTRACT

INTRODUCTION: Atrial fibrillation (AF) represents the most common cardiac arrhythmia in daily clinical practice and substantially impacts affected patients by elevation of both morbidity and mortality. Previous investigations proved that inflammatory processes are closely linked to this multifactorial pathogenesis-especially autoreactive CD4+CD28null T cells received in-depth attention. PURPOSE: Consequently, a potential pathophysiological pathway of the impact of CD4+CD28null T lymphocytes on the development and progression AF can be outlined. CONCLUSION: Considering the available data in the literature, it needs to be assumed that CD4+CD28null T lymphocytes are mainly involved in the development of AF and disease progression. Of utmost importance, it can be considered as the result of a T-cell-mediated auto-immune reaction among myocardial tissue. However, mechanisms which recruit CD4+CD28null cells in cardiac tissue remain unclear and need further investigation.


Subject(s)
Atrial Fibrillation/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Disease Progression , Humans , Myocardium/immunology , Myocardium/pathology
15.
Prog Cardiovasc Dis ; 68: 52-59, 2021.
Article in English | MEDLINE | ID: mdl-34274371

ABSTRACT

Exercise training is generally beneficial for cardiovascular health, improving stroke volume, cardiac output, and aerobic capacity. Despite these benefits, some evidence indicates that endurance training may increase the risk of atrial fibrillation (AF), particularly in highly trained individuals. Among multiple mechanisms, autonomic tone changes and atrial remodeling have been proposed as main contributors for exercise-induced AF. However, the contribution of local and systemic immunity is poorly understood in the development of atrial arrhythmogenic substrates. Here we aim to update the field of immunomodulation in the context of exercise and AF by compiling and reconciling the most recent evidence from preclinical and human studies and rationalize the applicability of "lone" AF terminology in athletes.


Subject(s)
Athletes , Atrial Fibrillation/etiology , Heart Atria/immunology , Heart Rate , Immune System/immunology , Immunity, Innate , Physical Exertion/immunology , Animals , Atrial Fibrillation/immunology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Cardiomegaly, Exercise-Induced , Cytokines/metabolism , Heart Atria/metabolism , Heart Atria/physiopathology , Humans , Immune System/metabolism , Immune System/physiopathology , Inflammation Mediators/metabolism , Risk Assessment , Risk Factors , Signal Transduction
16.
PLoS One ; 16(5): e0252030, 2021.
Article in English | MEDLINE | ID: mdl-34043672

ABSTRACT

PURPOSE: Association of the neutrophil-to-lymphocyte ratio (NLR) with mortality has not been comprehensively explored in critical limb ischemia (CLI) patients. We investigated the association between the NLR and clinical outcomes in CLI. MATERIALS AND METHODS: We retrospectively enrolled consecutive CLI patients between 1/1/2013 and 12/31/2018. Receiver operating characteristic curve analysis determined NLR cutoffs for 1-year in-hospital, all-cause and cardiac-related mortality; major adverse cardiovascular events (MACEs); and major adverse limb events (MALEs). RESULTS: Among 195 patients (age, 74.0 years, SD: 11.5; 51.8% male; body mass index, 23.4 kg/m2, SD: 4.2), 14.4% exhibited acute limb ischemia. After 1 year, patients with NLR>8 had higher in-hospital mortality (21.1% vs. 3.6%, P<0.001), all-cause mortality (54.4% vs. 13.8%, P<0.001), cardiac-related mortality (28.1% vs. 6.5%, P<0.001), MACE (29.8% vs. 13.0%, P = 0.008), and MALE (28.1% vs. 13.0%, P = 0.021) rates than those with NLR<8. In multivariate logistic regression, NLR≥8 was significantly associated with all-cause (P<0.001) and cardiac-related (adjusted HR: 5.286, 95% CI: 2.075-13.47, P<0.001) mortality, and NLR≥6 was significantly associated with MALEs (adjusted HR: 2.804, 95% CI: 1.292-6.088, P = 0.009). Each increase in the NLR was associated with increases in all-cause (adjusted HR: 1.028, 95% CI: 1.008-1.049, P = 0.007) and cardiac-related (adjusted HR:1.027, 95% CI: 0.998-1.057, P = 0.073) mortality but not in-hospital mortality or MACEs. CONCLUSION: CLI patients with high NLRs had significantly higher risks of 1-year all-cause and cardiac-related mortality and MALEs. The NLR can be used for prognostic prediction in these patients.


Subject(s)
Amputation, Surgical/mortality , Atrial Fibrillation/diagnosis , Heart Failure/diagnosis , Ischemia/diagnosis , Lymphocytes/pathology , Myocardial Infarction/diagnosis , Neutrophils/pathology , Aged , Aged, 80 and over , Amputation, Surgical/statistics & numerical data , Atrial Fibrillation/immunology , Atrial Fibrillation/mortality , Atrial Fibrillation/pathology , Biomarkers/analysis , Body Mass Index , Female , Heart Failure/immunology , Heart Failure/mortality , Heart Failure/pathology , Humans , Ischemia/immunology , Ischemia/mortality , Ischemia/pathology , Leukocyte Count , Lower Extremity/pathology , Lymphocytes/immunology , Male , Middle Aged , Myocardial Infarction/immunology , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Neutrophils/immunology , Prognosis , Retrospective Studies , Risk Assessment , Survival Analysis
17.
BMC Cardiovasc Disord ; 21(1): 132, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33706714

ABSTRACT

BACKGROUND: The development of atrial fibrillation (AF) following valvular heart disease (VHD) remains a common disease and is associated with substantial adverse complications. However, valid molecular diagnostic and therapeutic tools for post-VHD AF have not been fully established. This study was conducted to discover the molecular mechanisms and immune microenvironment underlying AF following VHD. METHODS: Gene expression profiles of the GSE41177 dataset were assessed to construct a protein-protein interaction network, and then, autophagy-related hub genes were identified. In addition, to determine the functions of immune cell infiltration in valvular AF, we used the CIBERSORT algorithm to estimate the composition of 22 immune cell types in valvular heart disease. Finally, correlation analysis was carried out to identify the relationship between differentially expressed autophagy-related genes (DEARGs) and significant immune cell subpopulations to reveal potential regulatory pathways. RESULTS: A total of 153 DEARGs were identified in AF-VHD patients compared with controlled donors. Moreover, we screened the top ten hub nodes with the highest degrees through a network analysis. The ten hub nodes were considered hub genes related to AF genesis and progression. Then, we revealed six significant immune cell subpopulations through the CIBERSORT algorithm. Finally, correlation analysis was performed, and six DEARGs (BECN1, GAPDH, ATG7, MAPK3, BCL2L1, and MYC) and three immune cell subpopulations (T cells CD4 memory resting, T cells follicular helper, and neutrophils) were identified as the most significant potential regulators. CONCLUSION: The DEARGs and immune cells identified in our study may be critical in AF development following VHD and provide potential predictive markers and therapeutic targets for determining a treatment strategy for AF patients.


Subject(s)
Atrial Fibrillation/genetics , Atrial Fibrillation/immunology , Autophagy-Related Proteins/genetics , Autophagy/genetics , Cellular Microenvironment/immunology , Heart Valve Diseases/genetics , Heart Valve Diseases/immunology , Transcriptome , Databases, Genetic , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunologic Memory , Immunophenotyping , Neutrophils/immunology , Phenotype , Protein Interaction Maps , T Follicular Helper Cells/immunology
19.
Sci Rep ; 11(1): 2972, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536523

ABSTRACT

Despite our expanding knowledge about the mechanism underlying atrial fibrillation (AF), the interplay between the biological events underlying AF remains incompletely understood. This study aimed to identify the functionally enriched gene-sets in AF and capture their interconnection via pivotal factors, that may drive or be driven by AF. Global abundance of the proteins in the left atrium of AF patients compared to control patients (n = 3/group), and the functionally enriched biological processes in AF were determined by mass-spectrometry and gene set enrichment analysis, respectively. The data were validated in an independent cohort (n = 19-20/group). In AF, the gene-sets of innate immune system, metabolic process, cellular component disassembly and ion homeostasis were up-regulated, while the gene-set of ciliogenesis was down-regulated. The innate immune system was over-represented by neutrophil degranulation, the components of which were extensively shared by other gene-sets altered in AF. In the independent cohort, an activated form of neutrophils was more present in the left atrium of AF patients with the increased gene expression of neutrophil granules. MYH10, required for ciliogenesis, was decreased in the atrial fibroblasts of AF patients. We report the increased neutrophil degranulation appears to play a pivotal role, and affects multiple biological processes altered in AF.


Subject(s)
Atrial Fibrillation/immunology , Cell Degranulation/immunology , Neutrophil Activation , Neutrophils/immunology , Atrial Fibrillation/pathology , Atrial Fibrillation/surgery , Case-Control Studies , Catheter Ablation , Fibroblasts/metabolism , Heart Atria/immunology , Heart Atria/pathology , Humans , Male , Myosin Heavy Chains/metabolism , Neutrophils/metabolism , Nonmuscle Myosin Type IIB/metabolism , Proteomics
20.
Sci Rep ; 11(1): 3563, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574429

ABSTRACT

Telocytes are interstitial cells with long, thin processes by which they contact each other and form a network in the interstitium. Myocardial remodeling of adult patients with different forms of atrial fibrillation (AF) occurs with an increase in fibrosis, age-related isolated atrial amyloidosis (IAA), cardiomyocyte hypertrophy and myolysis. This study aimed to determine the ultrastructural and immunohistochemical features of cardiac telocytes in patients with AF and AF + IAA. IAA associated with accumulation of atrial natriuretic factor was detected in 4.3-25% biopsies of left (LAA) and 21.7-41.7% of right (RAA) atrial appendage myocardium. Telocytes were identified at ultrastructural level more often in AF + IAA, than in AF group and correlated with AF duration and mitral valve regurgitation. Telocytes had ultrastructural signs of synthetic, proliferative, and phagocytic activity. Telocytes corresponded to CD117+, vimentin+, CD34+, CD44+, CD68+, CD16+, S100-, CD105- immunophenotype. No significant differences in telocytes morphology and immunophenotype were found in patients with various forms of AF. CD68-positive cells were detected more often in AF + IAA than AF group. We assume that in aged AF + IAA patients remodeling of atrial myocardium provoked transformation of telocytes into "transitional forms" combining the morphological and immunohistochemical features with signs of fibroblast-, histiocyte- and endotheliocyte-like cells.


Subject(s)
Amyloidosis/immunology , Atrial Fibrillation/immunology , Heart Defects, Congenital/immunology , Telocytes/immunology , Aged , Amyloidosis/complications , Amyloidosis/pathology , Atrial Appendage/immunology , Atrial Appendage/pathology , Atrial Fibrillation/complications , Atrial Fibrillation/pathology , Female , Heart Atria/immunology , Heart Atria/pathology , Heart Defects, Congenital/complications , Heart Defects, Congenital/pathology , Humans , Immunophenotyping , Male , Middle Aged , Mitral Valve Insufficiency/immunology , Mitral Valve Insufficiency/pathology , Myocardium/immunology , Myocardium/pathology , Telocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...