Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 228
Filter
1.
PLoS One ; 19(5): e0303496, 2024.
Article in English | MEDLINE | ID: mdl-38739622

ABSTRACT

INTRODUCTION: Rheumatic heart disease (RHD), degenerative aortic stenosis (AS), and congenital valve diseases are prevalent in sub-Saharan Africa. Many knowledge gaps remain in understanding disease mechanisms, stratifying phenotypes, and prognostication. Therefore, we aimed to characterise patients through clinical profiling, imaging, histology, and molecular biomarkers to improve our understanding of the pathophysiology, diagnosis, and prognosis of RHD and AS. METHODS: In this cross-sectional, case-controlled study, we plan to recruit RHD and AS patients and compare them to matched controls. Living participants will undergo clinical assessment, echocardiography, CMR and blood sampling for circulatory biomarker analyses. Tissue samples will be obtained from patients undergoing valve replacement, while healthy tissues will be obtained from cadavers. Immunohistology, proteomics, metabolomics, and transcriptome analyses will be used to analyse circulatory- and tissue-specific biomarkers. Univariate and multivariate statistical analyses will be used for hypothesis testing and identification of important biomarkers. In summary, this study aims to delineate the pathophysiology of RHD and degenerative AS using multiparametric CMR imaging. In addition to discover novel biomarkers and explore the pathomechanisms associated with RHD and AS through high-throughput profiling of the tissue and blood proteome and metabolome and provide a proof of concept of the suitability of using cadaveric tissues as controls for cardiovascular disease studies.


Subject(s)
Aortic Valve Stenosis , Biomarkers , Rheumatic Heart Disease , Humans , Rheumatic Heart Disease/diagnostic imaging , Rheumatic Heart Disease/physiopathology , Rheumatic Heart Disease/metabolism , Aortic Valve Stenosis/diagnostic imaging , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/physiopathology , Biomarkers/metabolism , Case-Control Studies , Cross-Sectional Studies , Male , Female , Metabolomics/methods , Echocardiography/methods , Proteomics/methods , Magnetic Resonance Imaging/methods , Multiomics
2.
Arterioscler Thromb Vasc Biol ; 44(4): 807-821, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38269589

ABSTRACT

BACKGROUND: Rheumatic heart disease is the major cause of valvular heart disease in developing nations. Endothelial cells (ECs) are considered crucial contributors to rheumatic heart disease, but greater insight into their roles in disease progression is needed. METHODS: We used a Cdh5-driven EC lineage-tracing approach to identify and track ECs in the K/B.g7 model of autoimmune valvular carditis. Single-cell RNA sequencing was used to characterize the EC populations in control and inflamed mitral valves. Immunostaining and conventional histology were used to evaluate lineage tracing and validate single-cell RNA-sequencing findings. The effects of VEGFR3 (vascular endothelial growth factor receptor 3) and VEGF-C (vascular endothelial growth factor C) inhibitors were tested in vivo. The functional impact of mitral valve disease in the K/B.g7 mouse was evaluated using echocardiography. Finally, to translate our findings, we analyzed valves from human patients with rheumatic heart disease undergoing mitral valve replacements. RESULTS: Lineage tracing in K/B.g7 mice revealed new capillary lymphatic vessels arising from valve surface ECs during the progression of disease in K/B.g7 mice. Unsupervised clustering of mitral valve single-cell RNA-sequencing data revealed novel lymphatic valve ECs that express a transcriptional profile distinct from other valve EC populations including the recently identified PROX1 (Prospero homeobox protein 1)+ lymphatic valve ECs. During disease progression, these newly identified lymphatic valve ECs expand and upregulate a profibrotic transcriptional profile. Inhibiting VEGFR3 through multiple approaches prevented expansion of this mitral valve lymphatic network. Echocardiography demonstrated that K/B.g7 mice have left ventricular dysfunction and mitral valve stenosis. Valve lymphatic density increased with age in K/B.g7 mice and correlated with worsened ventricular dysfunction. Importantly, human rheumatic valves contained similar lymphatics in greater numbers than nonrheumatic controls. CONCLUSIONS: These studies reveal a novel mode of inflammation-associated, VEGFR3-dependent postnatal lymphangiogenesis in murine autoimmune valvular carditis, with similarities to human rheumatic heart disease.


Subject(s)
Heart Valve Diseases , Lymphatic Vessels , Myocarditis , Rheumatic Heart Disease , Humans , Mice , Animals , Rheumatic Heart Disease/genetics , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Lymphatic Vessels/metabolism , Heart Valve Diseases/pathology , Disease Progression , RNA
3.
Sci Rep ; 12(1): 5238, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35347173

ABSTRACT

Rheumatic heart disease (RHD) is often considered as a disease of developing countries and India is the home of about 40% of RHD patients. Environment seems to play a major role in its causation. Since gene environment interactions can lead to alterations of various metabolic pathways, identification of altered metabolites can help in understanding the various pathways leading to RHD. Blood plasma samples from 51 RHD and 49 healthy controls were collected for the study. Untargeted metabolomics approach was used to identify the metabolites that are altered in RHD patients. Data showed 25 altered metabolites among RHD patients. These altered metabolites were those involved in Purine, Glutamine, Glutamate, Pyrimidine, Arginine, Proline and Linoleic metabolism. Thus, the present study illuminates metabolic alterations among RHD patients which can help in determining the potential therapeutic targets.


Subject(s)
Rheumatic Heart Disease , Biomarkers , Chromatography, Liquid , Humans , Plasma/metabolism , Rheumatic Heart Disease/metabolism , Tandem Mass Spectrometry
4.
Circulation ; 145(7): 531-548, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35157519

ABSTRACT

BACKGROUND: Rheumatic heart valve disease (RHVD) is a leading cause of cardiovascular death in low- and middle-income countries and affects predominantly women. The underlying mechanisms of chronic valvular damage remain unexplored and regulators of sex predisposition are unknown. METHODS: Proteomics analysis of human heart valves (nondiseased aortic valves, nondiseased mitral valves [NDMVs], valves from patients with rheumatic aortic valve disease, and valves from patients with rheumatic mitral valve disease; n=30) followed by system biology analysis identified ProTα (prothymosin alpha) as a protein associated with RHVD. Histology, multiparameter flow cytometry, and enzyme-linked immunosorbent assay confirmed the expression of ProTα. In vitro experiments using peripheral mononuclear cells and valvular interstitial cells were performed using multiparameter flow cytometry and quantitative polymerase chain reaction. In silico analysis of the RHVD and Streptococcuspyogenes proteomes were used to identify mimic epitopes. RESULTS: A comparison of NDMV and nondiseased aortic valve proteomes established the baseline differences between nondiseased aortic and mitral valves. Thirteen unique proteins were enriched in NDMVs. Comparison of NDMVs versus valves from patients with rheumatic mitral valve disease and nondiseased aortic valves versus valves from patients with rheumatic aortic valve disease identified 213 proteins enriched in rheumatic valves. The expression of the 13 NDMV-enriched proteins was evaluated across the 213 proteins enriched in diseased valves, resulting in the discovery of ProTα common to valves from patients with rheumatic mitral valve disease and valves from patients with rheumatic aortic valve disease. ProTα plasma levels were significantly higher in patients with RHVD than in healthy individuals. Immunoreactive ProTα colocalized with CD8+ T cells in RHVD. Expression of ProTα and estrogen receptor alpha correlated strongly in circulating CD8+ T cells from patients with RHVD. Recombinant ProTα induced expression of the lytic proteins perforin and granzyme B by CD8+ T cells as well as higher estrogen receptor alpha expression. In addition, recombinant ProTα increased human leukocyte antigen class I levels in valvular interstitial cells. Treatment of CD8+ T cells with specific estrogen receptor alpha antagonist reduced the cytotoxic potential promoted by ProTα. In silico analysis of RHVD and Spyogenes proteomes revealed molecular mimicry between human type 1 collagen epitope and bacterial collagen-like protein, which induced CD8+ T-cell activation in vitro. CONCLUSIONS: ProTα-dependent CD8+ T-cell cytotoxicity was associated with estrogen receptor alpha activity, implicating ProTα as a potential regulator of sex predisposition in RHVD. ProTα facilitated recognition of type 1 collagen mimic epitopes by CD8+ T cells, suggesting mechanisms provoking autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Collagen Type I/metabolism , Estrogen Receptor alpha/metabolism , Heart Valve Diseases/etiology , Heart Valve Diseases/metabolism , Protein Precursors/metabolism , Thymosin/analogs & derivatives , Amino Acid Sequence , Collagen Type I/chemistry , Computational Biology/methods , Disease Susceptibility , Epitopes, T-Lymphocyte/immunology , Heart Valve Diseases/diagnosis , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Models, Biological , Models, Molecular , Protein Binding , Protein Precursors/chemistry , Protein Precursors/genetics , Proteome , Proteomics/methods , Rheumatic Heart Disease/diagnosis , Rheumatic Heart Disease/etiology , Rheumatic Heart Disease/metabolism , Structure-Activity Relationship , Thymosin/chemistry , Thymosin/genetics , Thymosin/metabolism
5.
Acta Biochim Biophys Sin (Shanghai) ; 53(11): 1450-1458, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34596216

ABSTRACT

Atrial fibrosis is a crucial mechanism responsible for atrial fibrillation (AF). Sex-determining region Y-box containing gene 9 (Sox9) plays a pivotal role in fibrosis of many organs such as the skin, kidney, and liver. However, there are few studies about the occurrence and maintenance of Sox9 in atrial fibrosis. In this study, we investigated the role of Sox9 in the fibrotic phenotype of human atrial tissues and rat atrial fibroblasts in vitro. In the human right atrial tissue, Masson's trichrome staining, immunofluorescence, real-time quantitative polymerase chain reaction, and western blot analysis were carried out to explore the relationship between Sox9 and atrial fibrosis at the morphological, functional, and molecular levels. In cultured atrial fibroblasts, Sox9 was overexpressed by adenovirus or depleted by siRNA, and then, recombinant human transforming growth factor (TGF)-ß1 was added. Immunofluorescence analysis, western blot analysis, Transwell assay, and scratch assay were used to analyze the cells. In patient atrial tissues, Sox9 was increased with worsened atrial fibrosis, and this increase was related to AF severity. In rat atrial fibroblasts, Sox9 was promoted by TGF-ß1, and the α-smooth muscle actin (α-SMA) protein level and the ability of cell migration were increased after Sox9 overexpression by adenovirus, while the α-SMA protein level and the cell migration ability were decreased after Sox9 depletion by siRNA. In conclusion, Sox9 is involved in the regulation of fibrosis in the atria and may be located downstream of TGF-ß1. Our findings may provide a new perspective to treat atrial fibrosis during AF.


Subject(s)
Atrial Fibrillation/genetics , Fibroblasts/metabolism , Heart Defects, Congenital/genetics , Rheumatic Heart Disease/genetics , SOX9 Transcription Factor/genetics , Transforming Growth Factor beta1/genetics , Actins/genetics , Actins/metabolism , Adult , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/surgery , Cell Movement , Collagen Type I/genetics , Collagen Type I/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Female , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation , Heart Atria/metabolism , Heart Atria/pathology , Heart Atria/surgery , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Defects, Congenital/surgery , Humans , Male , Middle Aged , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/pathology , Rheumatic Heart Disease/surgery , SOX9 Transcription Factor/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism
6.
Bioengineered ; 12(1): 5904-5915, 2021 12.
Article in English | MEDLINE | ID: mdl-34482802

ABSTRACT

In the present study, mitral valve tissues from three mitral stenosis patients with RHD by valve replacement and two healthy donors were harvested and conducted DNA methylation signature on PRKCA by MeDIP-qPCR. The presence of hypomethylated CpG islands at promoter and 5' terminal of PRKCA was observed in RHD accompanied with highly expressed PRKCA and down-regulated antisense long non-coding RNA (lncRNA) PRKCA-AS1 compared to health control. Furthermore, the enrichments of DNMT1/3A/3B on PRKCA were detected by ChIP-qPCR assay in vivo and in human cardiomyocyte AC16 and RL-14 cells exposed to TNF-α in vitro, and both demonstrated that DNMT1 substantially contributed to DNA methylation. Additionally, PRKCA-AS1 was further determined to bind with promoter of PRKCA via 5' terminal and interact with DNMT1 via 3' terminal. Taken together, our results illuminated a novel regulatory mechanism of DNA methylation on regulating PRKCA transcription through lncRNA PRKCA-AS1, and shed light on the molecular pathogenesis of RHD occurrence.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , Protein Kinase C-alpha/genetics , RNA, Long Noncoding/genetics , Rheumatic Heart Disease , Aged , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/genetics , Female , Humans , Male , Middle Aged , Mitral Valve/metabolism , Promoter Regions, Genetic/genetics , Protein Kinase C-alpha/metabolism , RNA, Long Noncoding/metabolism , Rheumatic Heart Disease/genetics , Rheumatic Heart Disease/metabolism
7.
Cardiovasc Res ; 117(13): 2506-2524, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34432007

ABSTRACT

Calcific aortic valve disease (CAVD), and its clinical manifestation that is calcific aortic valve stenosis, is the leading cause for valve disease within the developed world, with no current pharmacological treatment available to delay or halt its progression. Characterized by progressive fibrotic remodelling and subsequent pathogenic mineralization of the valve leaflets, valve disease affects 2.5% of the western population, thus highlighting the need for urgent intervention. Whilst the pathobiology of valve disease is complex, involving genetic factors, lipid infiltration, and oxidative damage, the immune system is now being accepted to play a crucial role in pathogenesis and disease continuation. No longer considered a passive degenerative disease, CAVD is understood to be an active inflammatory process, involving a multitude of pro-inflammatory mechanisms, with both the adaptive and the innate immune system underpinning these complex mechanisms. Within the valve, 15% of cells evolve from haemopoietic origin, and this number greatly expands following inflammation, as macrophages, T lymphocytes, B lymphocytes, and innate immune cells infiltrate the valve, promoting further inflammation. Whether chronic immune infiltration or pathogenic clonal expansion of immune cells within the valve or a combination of the two is responsible for disease progression, it is clear that greater understanding of the immune systems role in valve disease is required to inform future treatment strategies for control of CAVD development.


Subject(s)
Adaptive Immunity , Aortic Valve Stenosis/immunology , Aortic Valve/immunology , Aortic Valve/pathology , Calcinosis/immunology , Hematopoietic System/immunology , Immune System/immunology , Immunity, Innate , Animals , Aortic Valve/metabolism , Aortic Valve/physiopathology , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/physiopathology , Calcinosis/metabolism , Calcinosis/physiopathology , Cytokines/metabolism , Hematopoiesis , Hematopoietic System/metabolism , Hematopoietic System/pathology , Humans , Immune System/metabolism , Immune System/physiopathology , Inflammation Mediators/metabolism , Lipid Metabolism , Rheumatic Heart Disease/immunology , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/physiopathology , Signal Transduction
8.
Mol Med Rep ; 23(1)2021 01.
Article in English | MEDLINE | ID: mdl-33179113

ABSTRACT

Rheumatic heart disease (RHD) is an autoimmune disease caused by rheumatic fever following group A hemolytic streptococcal infection and primarily affects the mitral valve. RHD is currently a major global health problem. However, the exact pathological mechanisms associated with RHD­induced cardiac valve damage remain to be elucidated. The endothelial­mesenchymal transition (EndMT) serves a key role in a number of diseases with an important role in cardiac fibrosis and the activin/Smad2 and 3 signaling pathway is involved in regulating the EndMT. Nevertheless, there are no studies to date, to the best of the authors' knowledge, investigating the association between RHD and EndMT. Thus, the aim of the current study was to investigate the potential role of EndMT in cardiac valve damage and assess whether activin/Smad2 and 3 signaling was activated during RHD­induced valvular injury in a rat model of RHD induced by inactivated Group A streptococci and complete Freund's adjuvant. Inflammation and fibrosis were assessed by hematoxylin and eosin and Sirius red staining. Serum cytokine and rheumatoid factor levels were measured using ELISA kits. Expression levels of activin/Smad2 and 3 signaling pathway­related factors [activin A, Smad2, Smad3, phosphorylated (p­)Smad2 and p­Smad3], EndMT­related factors [lymphoid enhancer factor­1 (LEF­1), Snail1, TWIST, zinc finger E­box­binding homeobox (ZEB)1, ZEB2, α smooth muscle actin (α­SMA) and type I collagen α 1 (COL1A1)], apoptosis­related markers (BAX and cleaved caspase­3) and valvular inflammation markers (NF­κB and p­NF­κB) were detected using reverse transcription­quantitative PCR and western blot analyses. Compared with the control group, the degree of valvular inflammation and fibrosis, serum levels of IL­6, IL­17, TNF­α and expression of apoptosis­related markers (BAX and cleaved caspase­3) and valvular inflammation marker (p­NF­κB), activin/Smad2 and 3 signaling pathway­related factors (activin A, p­Smad2 and p­Smad3), EndMT­related factors (LEF­1, Snail1, TWIST, ZEB 1, ZEB2, α­SMA and COL1A1) were significantly increased in the RHD group. These results suggested that the activin/Smad2 and 3 signaling pathway was activated during the development of valvular damage caused by RHD and that the EndMT is involved in RHD­induced cardiac valve damage.


Subject(s)
Activins/metabolism , Mitral Valve/pathology , Rheumatic Heart Disease/pathology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Animals , Disease Models, Animal , Female , Fibrosis , Freund's Adjuvant/adverse effects , Mitral Valve/metabolism , Rats , Rheumatic Heart Disease/etiology , Rheumatic Heart Disease/metabolism , Signal Transduction , Streptococcus pyogenes/pathogenicity
9.
Immunol Lett ; 229: 27-31, 2021 01.
Article in English | MEDLINE | ID: mdl-33232720

ABSTRACT

Rheumatic fever (RF) and chronic rheumatic heart disease (RHD) are complications of oropharyngeal infection caused by Streptococcus pyogenes. Despite the importance of the complement system against infections and autoimmunity diseases, studies on the role of the lectin pathway in RF and RHD are scarce. Thus, our aim was to evaluate the association of ficolin-3 serum levels, FCN3 polymorphisms and haplotypes with the susceptibility to RF and RHD. We investigated 179 patients with a history of RF (126 RHD and 53 RF only) and 170 healthy blood donors as control group. Ficolin-3 serum concentrations were measured using enzyme-linked immunosorbent assay (ELISA). Three FCN3 single nucleotide polymorphisms (SNPs rs532781899, rs28362807 and rs4494157) were genotyped through the sequence-specific PCR method. Lower ficolin-3 serum levels were observed in RF patients when compared to controls (12.81 µg/mL vs. 18.14 µg/mL respectively, p < 0.0001, OR 1.22 [1.12-1.34]), and in RHD in comparison to RF only (RFo) (12.72 µg/mL vs. 14.29 µg/mL respectively, p = 0.016, OR 1.38 [1.06-1.80]). Low ficolin-3 levels (<10.7 µg/mL) were more common in patients (39.5 %, 30/76) than controls (20.6 %, 13/63, p = 0.018, OR = 2.51 [1.14-5.31]), and in RHD (44.4 %, 28/63) than RFo (15.4 %, 2/13, p = 0.007, OR = 3.08 [1.43-6.79]). On the other hand, FCN3 polymorphism/haplotypes were not associated with ficolin-3 serum levels or the disease. Low ficolin-3 levels might be associated with RF, being a potential marker of disease progression.


Subject(s)
Disease Susceptibility , Lectins/genetics , Rheumatic Fever/etiology , Rheumatic Fever/metabolism , Rheumatic Heart Disease/etiology , Adult , Alleles , Biomarkers , Female , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Lectins/blood , Lectins/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide , Prognosis , Rheumatic Fever/diagnosis , Rheumatic Heart Disease/diagnosis , Rheumatic Heart Disease/metabolism
10.
Circulation ; 142(20): e337-e357, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33073615

ABSTRACT

The global burden of rheumatic heart disease continues to be significant although it is largely limited to poor and marginalized populations. In most endemic regions, affected patients present with heart failure. This statement will seek to examine the current state-of-the-art recommendations and to identify gaps in diagnosis and treatment globally that can inform strategies for reducing disease burden. Echocardiography screening based on World Heart Federation echocardiographic criteria holds promise to identify patients earlier, when prophylaxis is more likely to be effective; however, several important questions need to be answered before this can translate into public policy. Population-based registries effectively enable optimal care and secondary penicillin prophylaxis within available resources. Benzathine penicillin injections remain the cornerstone of secondary prevention. Challenges with penicillin procurement and concern with adverse reactions in patients with advanced disease remain important issues. Heart failure management, prevention, early diagnosis and treatment of endocarditis, oral anticoagulation for atrial fibrillation, and prosthetic valves are vital therapeutic adjuncts. Management of health of women with unoperated and operated rheumatic heart disease before, during, and after pregnancy is a significant challenge that requires a multidisciplinary team effort. Patients with isolated mitral stenosis often benefit from percutaneous balloon mitral valvuloplasty. Timely heart valve surgery can mitigate the progression to heart failure, disability, and death. Valve repair is preferable over replacement for rheumatic mitral regurgitation but is not available to the vast majority of patients in endemic regions. This body of work forms a foundation on which a companion document on advocacy for rheumatic heart disease has been developed. Ultimately, the combination of expanded treatment options, research, and advocacy built on existing knowledge and science provides the best opportunity to address the burden of rheumatic heart disease.


Subject(s)
American Heart Association , Rheumatic Heart Disease/diagnosis , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/prevention & control , Rheumatic Heart Disease/physiopathology , Cost of Illness , Female , Humans , Male , Practice Guidelines as Topic , United States
11.
Biosci Rep ; 40(11)2020 11 27.
Article in English | MEDLINE | ID: mdl-33073840

ABSTRACT

To determine whether up-regulation of miR-1183 targeting the gene for anti-apoptotic factor, B-cell lymphoma 2 (BCL-2) contributes to apoptosis in patients with rheumatic heart disease (RHD). Peripheral blood samples were isolated for miR-1183 characterization. The function of miRNA-1183 in RHD using miRNA mimic on PBMCs and THP-1 cell models. The binding of miR-1183 and Bcl-2 gene was confirmed by luciferase activity test. We also measured expression levels of BCL-2 in heart valve tissue from patients with RHD using ELISA and immunohistochemistry. In silico analysis and reporter gene assays indicated that miR-1183 directly targets the mRNA encoding BCL-2. It is found that miR-1183 binds directly to the 3'UTR of the BCL-2 mRNA and down-regulates the mRNA and protein levels of BCL-2. Overexpression of miR-1183 in RHD patients and cell lines down-regulated BCL-2 expression and induced apoptosis. With the progression of the disease, the expression of BCL-2 in the heart valve tissue of patients with RHD decreased. MiRNA-1183 is up-regulated in RHD and induces cardiac myocyte apoptosis through direct targeting and suppression of BCL-2, both of which might play important roles in RHD pathogenesis. During the compensatory period of RHD, up-regulated miR-1183 destroyed the balance of apoptosis proteins (Bax and BAK) in Bcl-2 family, enhance the apoptosis cascade reaction and reduce the anti apoptosis effect. The significantly higher expression levels of miR-1183 appear to play distinct roles in RHD pathogenesis by regulation BCL-2, possibly affecting myocardial apoptosis and remodeling in the context of RHD.


Subject(s)
Apoptosis , Heart Valves/metabolism , Leukocytes, Mononuclear/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rheumatic Heart Disease/metabolism , 3' Untranslated Regions , Adult , Binding Sites , Case-Control Studies , Female , Gene Expression Regulation , Heart Valves/pathology , Humans , Male , MicroRNAs/genetics , Middle Aged , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Rheumatic Heart Disease/genetics , Rheumatic Heart Disease/pathology , Signal Transduction , THP-1 Cells
12.
Circulation ; 142(20): e358-e368, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33070654

ABSTRACT

Rheumatic heart disease (RHD) affects ≈40 million people and claims nearly 300 000 lives each year. The historic passing of a World Health Assembly resolution on RHD in 2018 now mandates a coordinated global response. The American Heart Association is committed to serving as a global champion and leader in RHD care and prevention. Here, we pledge support in 5 key areas: (1) professional healthcare worker education and training, (2) technical support for the implementation of evidence-based strategies for rheumatic fever/RHD prevention, (3) access to essential medications and technologies, (4) research, and (5) advocacy to increase global awareness, resources, and capacity for RHD control. In bolstering the efforts of the American Heart Association to combat RHD, we hope to inspire others to collaborate, communicate, and contribute.


Subject(s)
American Heart Association , Cost of Illness , Education, Medical, Continuing , Rheumatic Heart Disease , Humans , Practice Guidelines as Topic , Rheumatic Heart Disease/diagnosis , Rheumatic Heart Disease/epidemiology , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/prevention & control , United States/epidemiology
13.
Circ Arrhythm Electrophysiol ; 13(7): e007588, 2020 07.
Article in English | MEDLINE | ID: mdl-32538131

ABSTRACT

BACKGROUND: Scientific research on atrial fibrosis in atrial fibrillation (AF) has mainly focused on quantitative or molecular features. The purpose of this study was to perform a clinicoarchitectural/structural investigation of fibrosis to provide one key to understanding the electrophysiological/clinical aspects of AF. METHODS: We characterized the fibrosis (amount, architecture, cellular components, and ultrastructure) in left atrial biopsies from 121 patients with persistent/long-lasting persistent AF (group 1; 59 males; 60±11 years; 91 mitral disease-related AF, 30 nonmitral disease-related AF) and from 39 patients in sinus rhythm with mitral valve regurgitation (group 2; 32 males; 59±12 years). Ten autopsy hearts served as controls. RESULTS: Qualitatively, the fibrosis exhibited the same characteristics in all cases and displayed particular architectural scenarios (which we arbitrarily subdivided into 4 stages) ranging from isolated foci to confluent sclerotic areas. The percentage of fibrosis was larger and at a more advanced stage in group 1 versus group 2 and, within group 1, in patients with rheumatic disease versus nonrheumatic cases. In patients with AF with mitral disease and no rheumatic disease, the percentage of fibrosis and the fibrosis stages correlated with both left atrial volume index and AF duration. The fibrotic areas mainly consisted of type I collagen with only a minor cellular component (especially fibroblasts/myofibroblasts; average value range 69-150 cells/mm2, depending on the areas in AF biopsies). A few fibrocytes-circulating and bone marrow-derived mesenchymal cells-were also detectable. The fibrosis-entrapped cardiomyocytes showed sarcolemmal damage and connexin 43 redistribution/internalization. CONCLUSIONS: Atrial fibrosis is an evolving and inhomogeneous histological/architectural change that progresses through different stages ranging from isolated foci to confluent sclerotic zones which-seemingly-constrain impulse conduction across restricted regions of electrotonically coupled cardiomyocytes. The fibrotic areas mainly consist of type I collagen extracellular matrix and, only to a lesser extent, mesenchymal cells.


Subject(s)
Atrial Fibrillation/pathology , Heart Atria/pathology , Heart Valve Diseases/pathology , Myocardium/pathology , Rheumatic Heart Disease/pathology , Adult , Aged , Aged, 80 and over , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Fibrillation/therapy , Atrial Function, Left , Atrial Remodeling , Biopsy , Collagen Type I/analysis , Connexin 43/analysis , Female , Fibrosis , Heart Atria/chemistry , Heart Atria/physiopathology , Heart Valve Diseases/metabolism , Heart Valve Diseases/physiopathology , Heart Valve Diseases/therapy , Humans , Male , Middle Aged , Myocardium/chemistry , Retrospective Studies , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/physiopathology , Rheumatic Heart Disease/therapy
14.
Sci Rep ; 9(1): 13069, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506454

ABSTRACT

Platelet-leukocyte aggregate (PLA) is implicated in the etiology of both vascular lesions and cardiovascular events. This prospective cohort study aimed to examine the prognostic value of PLA for major adverse cardiac and cerebrovascular events (MACCE) and perioperative adverse events (AEs) in patients with rheumatic heart disease undergoing surgical intervention by Cox proportional hazard regression and logistic regression. A total of 244 patients were included, of whom 7 were lost to follow-up. Among the analyzed 237 subjects who completed 3-year follow-up, 30 experienced MACCE and 38 experienced perioperative AEs. Preoperative PLA was higher in subjects who developed MACCE (13.32%) than in those who did not (8.69%, p = 0.040). In multivariate regression, elevated PLA was associated with increased MACCE (hazard ratio 1.51 for each quartile, 95% CI 1.07-2.13; p = 0.020), and perioperative AEs (odds ratio 1.61, 95% CI 1.14-2.26; p = 0.007). The optimal PLA cut-off for predicting MACCE was 6.8%. Subjects with PLA > 6.8% had a higher prevalence of MACCE (17.1% vs. 5.5%, p = 0.009) and perioperative AEs (19.9% vs. 8.6%, p = 0.018). Kaplan-Meier analysis showed shorter MACCE-free survival in patients with PLA > 6.8% (p = 0.007, log rank). Elevated preoperative PLA is associated with increased MACCE and perioperative AEs in patients with rheumatic valve disease undergoing surgical intervention.


Subject(s)
Blood Platelets/metabolism , Leukocytes/metabolism , Platelet Aggregation , Postoperative Complications/etiology , Postoperative Complications/metabolism , Rheumatic Heart Disease/complications , Rheumatic Heart Disease/metabolism , Adult , Aged , Disease Susceptibility , Female , Humans , Incidence , Male , Middle Aged , Proportional Hazards Models , Rheumatic Heart Disease/pathology , Rheumatic Heart Disease/surgery , Risk Factors
15.
Biotech Histochem ; 94(6): 398-403, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30712389

ABSTRACT

Rheumatic heart disease (RHD) is a public health burden in developing countries. Th17 cell-associated cytokines might play a role in the pathogenesis and development of RHD, but the specific molecular mechanism is not completely understood. We investigated the potential role of sphingosine-1-phosphate receptor 1 (S1PR1)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in cardiac valve damage in a rat model of RHD. We used 20 Lewis rats divided randomly into control and RHD groups. The RHD model was constructed by injecting inactivated group A Streptococci and complete Freund's adjuvant (CFA). The rats in the control group were injected with normal saline and CFA. Th17 cell-related cytokines were measured by ELISA. Fibrosis was assessed by histological examination. RT-qPCR and western blot were used to detect the expression of S1PR1 and STAT3/phosphorylated STAT3 (p-STAT3). The S1PR1/STAT3 signaling pathway was activated in the RHD model. Compared to the control group, serum levels of IL-17 and IL-21 cytokines associated with Th17 cells were increased significantly in the RHD group; the collagen volume fraction also was substantially increased. The S1PR1/STAT3 signaling pathway might be involved in RHD induced cardiac valve damage by regulating Th17 cells.


Subject(s)
Heart Valves/injuries , Rheumatic Heart Disease/metabolism , STAT3 Transcription Factor/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Heart Valves/physiopathology , Rats , Signal Transduction/physiology
16.
J Cell Mol Med ; 23(4): 3032-3039, 2019 04.
Article in English | MEDLINE | ID: mdl-30697920

ABSTRACT

OBJECTIVE: This study aimed to investigate the effects of transforming growth factor ß1 (TGF ß1) and hepatocyte growth factor (HGF) on the expression of connective tissue growth factor (CTGF) in human atrial fibroblasts, and to explore the relationship of these factors in atrial fibrosis and atrial anatomical remodelling (AAR) of patients with atrial fibrillation (AF). METHODS: Fresh right auricular appendix tissue of 20 patients with rheumatic heart disease undergoing valve replacement surgery was collected during surgeries, 10 patients had sinus rhythm(SR), and 10 patients had chronic atrial fibrillation (CAF). Atrial fibroblasts were then cultured from the tissues with differential attachment technique and treated with either TGFß1 (10 ng/mL) or HGF (100 ng/mL). CTGF mRNA levels were measured by RT-PCR, and CTGF protein content was determined using immunofluorescence and Western blotting assays. RESULTS: CAF group had higher left atrial diameters (LADs) and higher CTGF mRNA expression in atrial fibroblasts compared with SR group. The CTGF protein content in CAF group was higher than that of SR group and positively correlated with LAD and AF duration. After CAF group was treated with TGFß1, CTGF mRNA and protein expression were significantly down-regulated, whereas when treated with HGF, expression was up-regulated compared with SR group. CONCLUSIONS: Increased CTGF expression was associated with enlarged LAD, atrial fibrosis and AAR in patients with AF. TGFß1 and HGF regulate CTGF expression in human atrial fibroblasts with up-regulation of mRNA and down-regulation of protein, therefore, either promote or inhibit atrial fibrosis, which could be related to the incidence and persistence of AF.


Subject(s)
Atrial Remodeling , Connective Tissue Growth Factor/metabolism , Fibroblasts/pathology , Fibrosis/etiology , Hepatocyte Growth Factor/metabolism , Rheumatic Heart Disease/complications , Transforming Growth Factor beta1/metabolism , Adult , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Cells, Cultured , Connective Tissue Growth Factor/genetics , Female , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Hepatocyte Growth Factor/genetics , Humans , Male , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/pathology , Transforming Growth Factor beta1/genetics
17.
Genes Genet Syst ; 94(1): 23-34, 2019 Apr 09.
Article in English | MEDLINE | ID: mdl-30584185

ABSTRACT

MicroRNA (miRNA) is associated with the development and pathology of atrial fibrillation (AF). In this study, we performed miRNA profiling of left and right atrium samples from individuals with AF-associated rheumatic mitral valve disease (RMVD) to identify miRNAs that are differentially expressed between RMVD patients with AF and RMVD with sinus rhythm (SR) as controls, as well as between left and right atrium samples from RMVD with AF patients. We performed hematoxylin and eosin staining as well as scanning and transmission electron microscopy to examine in detail any morphological and physiological changes in cardiomyocytes from RMVD patients with AF or SR. Raman spectroscopy was performed to identify molecular and structural information of left and right atrium samples from RMVD with AF and SR. We also performed miRNA array profiling to separately profile miRNA expression patterns of right and left atrium samples from three independent RMVD patients with AF and in a mixed pool of 10 RMVD patients with SR. Morphological and physiological analysis showed distinct shapes and structures of cardiomyocytes from the left and right atria of RMVD patients with AF or SR. The intensity of Raman spectroscopy of atrial tissues from RMVD patients with AF and with SR was different. miRNA profiling showed differential miRNA expression between RMVD patients with AF or SR, and between the left and right atria of RMVD patients with AF. Importantly, miRNAs showed consistent expression changes among all three patients, suggesting that these miRNAs have potential as markers for AF pathology. Our results revealed potential biomarker miRNAs for atrial fibrillation pathology in patients with RMVD. Meanwhile, our data suggested that miR-10b and miR-138-2, which were both significantly increased in the left atrium, are responsible for morphological and physiological phenotype differences between the left and right atria.


Subject(s)
Atrial Fibrillation/genetics , Heart Atria/metabolism , Heart Valve Diseases/genetics , MicroRNAs/genetics , Mitral Valve/metabolism , Rheumatic Heart Disease/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Female , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Humans , Male , MicroRNAs/metabolism , Middle Aged , Myocytes, Cardiac/metabolism , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/pathology
18.
PLoS One ; 13(9): e0203756, 2018.
Article in English | MEDLINE | ID: mdl-30261069

ABSTRACT

AIMS: Left atrial (LA) structural remodelling develops in rheumatic heart disease (RHD) according to the disease severity of the mitral valve and the presence of atrial fibrillation. Sustained active inflammation has been previously reported in the LA of patients with RHD, suggesting a direct role of cell-mediated immunity in the pathogenesis of LA remodelling. Dendritic cells (DCs) have a major antigen-presenting role, and are known as crucial modulators of innate and adaptive immunity. We investigated whether DCs are involved in the pathogenesis of LA remodelling in RHD. METHODS AND RESULTS: Immunohistochemical analyses were performed using antibodies to CD11c, CD209 and CD80 as markers of myeloid DCs, migratory-active DCs, mature DCs and infiltrated inflammatory cells including T lymphocytes (CD3) and M1 (CD68; pro-inflammatory profile) and M2 (CD163; pro-resolution profile) macrophages. Furthermore, tenascin-C, an extracellular matrix (ECM) protein that appears during ECM remodelling and inflammatory response, was examined. Infiltrated myeloid DCs, migratory-active DCs, mature DCs and other inflammatory infiltrates including T lymphocytes and M1 and M2 macrophages, were significantly higher in the RHD group than the non-RHD group. The positive area fraction for tenascin-C was significantly higher in the RHD group than in the non-RHD group. CONCLUSION: Our histological findings suggest that inflammation may persist long after a bout of rheumatic fever, ultimately leading to ECM remodelling. We identified and quantitatively assessed several subsets of DCs and other immunocompetent cells, and our results indicated that activation of DCs has some role in persistence of LA inflammation in patients with chronic RHD.


Subject(s)
Heart Atria/pathology , Rheumatic Heart Disease/pathology , Aged , Aged, 80 and over , Atrial Remodeling , Dendritic Cells/pathology , Female , Heart Atria/metabolism , Humans , Immunity, Cellular , Immunohistochemistry , Male , Middle Aged , Rheumatic Fever/complications , Rheumatic Fever/immunology , Rheumatic Fever/pathology , Rheumatic Heart Disease/immunology , Rheumatic Heart Disease/metabolism , T-Lymphocytes/pathology , Tenascin/metabolism
20.
J Biol Chem ; 293(10): 3780-3792, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29358327

ABSTRACT

The most frequently used oral anti-coagulant warfarin has been implicated in inducing calcification of aortic valve interstitial cells (AVICs), whereas the mechanism is not fully understood. The low-level activation of p53 is found to be involved in osteogenic transdifferentiation and calcification of AVICs. Whether p53 participates in warfarin-induced AVIC calcification remains unknown. In this study, we investigated the role of low-level p53 overexpression in warfarin-induced porcine AVIC (pAVIC) calcification. Immunostaining, quantitative PCR, and Western blotting revealed that p53 was expressed in human and pAVICs and that p53 expression was slightly increased in calcific human aortic valves compared with non-calcific valves. Terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling staining indicated that apoptosis slightly increased in calcific aortic valves than in non-calcific valves. Warfarin treatment led to a low-level increase of p53 mRNA and protein in both pAVICs and mouse aortic valves. Low-level overexpression of p53 in pAVICs via an adenovirus vector did not affect pAVIC apoptosis but promoted warfarin-induced calcium deposition and expression of osteogenic markers. shRNA-mediated p53 knockdown attenuated the pAVIC calcium deposition and osteogenic marker expression. Moreover, ChIP and luciferase assays showed that p53 was recruited to the slug promoter and activated slug expression in calcific pAVICs. Of note, overexpression of Slug increased osteogenic marker Runx2 expression, but not pAVIC calcium deposition, and Slug knockdown attenuated pAVIC calcification and p53-mediated pAVIC calcium deposition and expression of osteogenic markers. In conclusion, we found that p53 plays an important role in warfarin induced pAVIC calcification, and increased slug transcription by p53 is required for p53-mediated pAVIC calcification.


Subject(s)
Aortic Valve/metabolism , Calcinosis/metabolism , Disease Models, Animal , Gene Expression Regulation , Heart Valve Diseases/metabolism , Snail Family Transcription Factors/agonists , Tumor Suppressor Protein p53/metabolism , Animals , Anticoagulants/adverse effects , Antifibrinolytic Agents/adverse effects , Aortic Valve/drug effects , Aortic Valve/pathology , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Calcinosis/chemically induced , Calcinosis/pathology , Cells, Cultured , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Heart Valve Diseases/chemically induced , Heart Valve Diseases/pathology , Humans , Male , Mice, Inbred C57BL , Promoter Regions, Genetic/drug effects , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/pathology , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Sus scrofa , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Vitamin K 1/adverse effects , Warfarin/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...