1
|
Wang X, Fu M, Wang W, Shu S, Zhang N, Zhao R, Chen X, Hua X, Wang X, Feng W, Wang X, Song J. Single-cell analysis reveals the loss of FABP4-positive proliferating valvular endothelial cells relates to functional mitral regurgitation. BMC Med 2024; 22:595. [PMID: 39707349 DOI: 10.1186/s12916-024-03791-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 11/20/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Functional mitral regurgitation (MR) is a common form of mitral valve dysfunction that often persists even after surgical intervention, requiring reoperation in some cases. To advance our understanding of the pathogenesis of functional MR, it is crucial to characterize the cellular composition of the mitral valve leaflet and identify molecular changes in each cell subtype within the mitral valves of MR patients. Therefore, we aimed to comprehensively examine the cellular and molecular components of mitral valves in patients with MR. METHODS We conducted a single-cell RNA sequencing (scRNA-seq) analysis of mitral valve leaflets extracted from six patients who underwent heart transplantation. The cohort comprised three individuals with moderate-to-severe functional MR (MR group) and three non-diseased controls (NC group). Bioinformatics was applied to identify cell types, delineate cell functions, and explore cellular developmental trajectories and interactions. Key findings from the scRNA-seq analysis were validated using pathological staining to visualize key markers in the mitral valve leaflets. Additionally, in vitro experiments with human primary valvular endothelial cells were conducted to further support our results. RESULTS Our study revealed that valve interstitial cells are critical for adaptive valve remodelling, as they secrete extracellular matrix proteins and promote fibrosis. We discovered an abnormal decrease in a subpopulation of FABP4 (fatty acid binding protein 4)-positive proliferating valvular endothelial cells. The trajectory analysis identifies this subcluster as the origin of VECs. Immunohistochemistry on the expanded cohort showed a reduction of FABP4-positive VECs in patients with functional MR. Intervention experiments with primary cells indicated that FABP4 promotes proliferation and migration in mitral valve VECs and enhances TGFβ-induced differentiation. CONCLUSIONS Our study presented a comprehensive assessment of the mitral valve cellular landscape of patients with MR and sheds light on the molecular changes occurring in human mitral valves during functional MR. We found a notable reduction in the proliferating endothelial cell subpopulation of valve leaflets, and FABP4 was identified as one of their markers. Therefore, FABP4 positive VECs served as proliferating endothelial cells relates to functional mitral regurgitation. These VECs exhibited high proliferative and differentiative properties. Their reduction was associated with the occurrence of functional MR.
Collapse
Affiliation(s)
- Xiaohu Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
| | - Mengxia Fu
- Galactophore Department, Galactophore Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Weiteng Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
| | - Songren Shu
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Zhang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruojin Zhao
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
| | - Xiao Chen
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiumeng Hua
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Feng
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianqiang Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China.
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jiangping Song
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China.
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
2
|
Begum A, Modumudi S, Subramani S, Khoont D, Vanaparti A, Master M, Khan J, Botticelli AL, Botticelli RW, Mian HS, Saad M, Abbas K. Novel putative biomarkers for infective endocarditis by serum proteomic analysis: a comprehensive review of literature. Ann Med Surg (Lond) 2023; 85:5497-5503. [PMID: 37915652 PMCID: PMC10617819 DOI: 10.1097/ms9.0000000000001249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/19/2023] [Indexed: 11/03/2023] Open
Abstract
Infective endocarditis (IE) is a challenging condition with high mortality. Prompt detection of IE has become essential for early and immediate management. The authors aimed to comprehensively review the existing literature on novel putative biomarkers for IE through serum proteomic analysis. The literature reveals high levels of N-terminal-pro-B-type natriuretic peptide (NT-proBNP) levels in IE with staphylococcal etiology, valvular lesions, and when combined with cardiac troponin I (cTnI), had a more significant value for risk stratification. A higher pro-ADM level, copeptin, NT-proBNP, and the monocyte-to-high-density lipoprotein cholesterol ratio (MHR) all impacted mortality during the hospital stay. The biomarker matrix metalloproteinase-9 was utilized to predict new-onset embolic events in patients, thus serving as a predictive marker. Procalcitonin was an important diagnostic marker in IE complicated with severe infection. Interleukin-6 (IL-6), Interleukin-8 (IL-8), Interferon-γ, cTnI, and NT-proBNP were also discovered to be useful as prognostic indicators. Early diagnosis and appropriate treatment are possible using antiphospholipid antibodies as a diagnostic test for definite IE. It is also concluded that antineutrophilic cytoplasmic antibody positive individuals with IE had a lengthier hospital stay. These noninvasive biomarkers can identify patients at risk and provide appropriate and early clinical management. NT-proBNP, Cystatin C, troponins, IL-6, IL-8, S100A11, and AQP9 are examples of possible markers that appear promising for further research. In conclusion, large-scale validation studies should study these biomarkers further to establish their use in clinical settings.
Collapse
Affiliation(s)
| | - Sravani Modumudi
- Department of Medicine, Kamineni Academy of Medical Sciences and Research Center, Hyderabad
| | - Sachin Subramani
- Department of Internal Medicine, ESIC Medical College and Hospital
| | - Dhruvi Khoont
- Department of Medicine, Narendra Modi Medical College
| | - Ankitha Vanaparti
- Department of Internal Medicine, Kakatiya Medical College, Warangal, Telangana State, India
| | - Mahima Master
- Department of Medicine, LG Hospital, Maninagar, Ahmedabad
| | - Javeria Khan
- Department of Adult Cardiology, National Institute of Cardiovascular Diseases
| | | | | | - Hafsa S. Mian
- Department of Medicine, Sheikh Zayed Hospital, Rahimyar Khan, Lahore, Pakistan
| | - Muhammad Saad
- Department of Medicine, FMH College of Medicine and Dentistry
| | - Kiran Abbas
- Department of Community Health Sciences, Aga Khan University, Karachi
| |
Collapse
|
3
|
Halawa S, Latif N, Tseng YT, Ibrahim AM, Chester AH, Moustafa A, Aguib Y, Yacoub MH. Profiling Genome-Wide DNA Methylation Patterns in Human Aortic and Mitral Valves. Front Cardiovasc Med 2022; 9:840647. [PMID: 35463757 PMCID: PMC9019152 DOI: 10.3389/fcvm.2022.840647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/11/2022] [Indexed: 12/05/2022] Open
Abstract
Cardiac valves exhibit highly complex structures and specialized functions that include dynamic interactions between cells, extracellular matrix (ECM) and their hemodynamic environment. Valvular gene expression is tightly regulated by a variety of mechanisms including epigenetic factors such as histone modifications, RNA-based mechanisms and DNA methylation. To date, methylation fingerprints of non-diseased human aortic and mitral valves have not been studied. In this work we analyzed the differential methylation profiles of 12 non-diseased aortic and mitral valve tissue samples (in matched pairs). Analysis of methylation data [reduced representation bisulfite sequencing (RRBS)] of 16,101 promoters genome-wide revealed 584 differentially methylated (DM) promoters, of which 13 were reported in endothelial mesenchymal trans-differentiation (EMT), 37 in aortic and mitral valve disease and 7 in ECM remodeling. Both functional classification as well as network analysis showed that the genes associated with the DM promoters were enriched for WNT-, Cadherin-, Endothelin-, PDGF-, HIF-1 and VEGF- signaling implicated in valvular physiology and pathophysiology. Additional enrichment was detected for TGFB-, NOTCH- and Integrin- signaling involved in EMT as well as ECM remodeling. This data provides the first insight into differential regulation of human aortic and mitral valve tissue and identifies candidate genes linked to DM promoters. Our work will improve the understanding of valve biology, valve tissue engineering approaches and contributes to the identification of relevant drug targets.
Collapse
Affiliation(s)
- Sarah Halawa
- Aswan Heart Centre, Aswan, Egypt
- Biotechnology Graduate Program, American University in Cairo, New Cairo, Egypt
- Sarah Halawa
| | - Najma Latif
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
| | - Yuan-Tsan Tseng
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
| | - Ayman M. Ibrahim
- Aswan Heart Centre, Aswan, Egypt
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Adrian H. Chester
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
| | - Ahmed Moustafa
- Biotechnology Graduate Program, American University in Cairo, New Cairo, Egypt
- Department of Biology, American University in Cairo, New Cairo, Egypt
| | - Yasmine Aguib
- Aswan Heart Centre, Aswan, Egypt
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
- Yasmine Aguib
| | - Magdi H. Yacoub
- Aswan Heart Centre, Aswan, Egypt
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- National Heart and Lung Institute (NHLI), Imperial College London, London, United Kingdom
- *Correspondence: Magdi H. Yacoub
| |
Collapse
|
4
|
Schumacher D, Liehn EA, Singh A, Curaj A, Wijnands E, Lira SA, Tacke F, Jankowski J, Biessen EA, van der Vorst EP. CCR6 Deficiency Increases Infarct Size after Murine Acute Myocardial Infarction. Biomedicines 2021; 9:1532. [PMID: 34829761 PMCID: PMC8614800 DOI: 10.3390/biomedicines9111532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 11/20/2022] Open
Abstract
Ischemia-reperfusion injury after the reopening of an occluded coronary artery is a major cause of cardiac damage and inflammation after acute myocardial infarction. The chemokine axis CCL20-CCR6 is a key player in various inflammatory processes, including atherosclerosis; however, its role in ischemia-reperfusion injury has remained elusive. Therefore, to gain more insight into the role of the CCR6 in acute myocardial infarction, we have studied cardiac injury after transient ligation of the left anterior descending coronary artery followed by reperfusion in Ccr6-/- mice and their respective C57Bl/6 wild-type controls. Surprisingly, Ccr6-/- mice demonstrated significantly reduced cardiac function and increased infarct sizes after ischemia/reperfusion. This coincided with a significant increase in cardiac inflammation, characterized by an accumulation of neutrophils and inflammatory macrophage accumulation. Chimeras with a bone marrow deficiency of CCR6 mirrored this adverse Ccr6-/- phenotype, while cardiac injury was unchanged in chimeras with stromal CCR6 deficiency. This study demonstrates that CCR6-dependent (bone marrow) cells exert a protective role in myocardial infarction and subsequent ischemia-reperfusion injury, supporting the notion that augmenting CCR6-dependent immune mechanisms represents an interesting therapeutic target.
Collapse
Affiliation(s)
- David Schumacher
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (D.S.); (A.C.); (J.J.); (E.A.L.B.)
- Department of Anesthesiology, University Hospital, RWTH Aachen University, 52074 Aachen, Germany
| | - Elisa A. Liehn
- Department of Intensive Care and Intermediate Care, University Hospital, RWTH Aachen University, 52074 Aachen, Germany;
- Department of Cardiology, Angiology and Intensive Medicine, University Hospital Aachen, 52074 Aachen, Germany
- National Institute for Pathology “Victor Babes”, 050096 Bucharest, Romania
- Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Anjana Singh
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands; (A.S.); (E.W.)
- Cognizant Technology Solutions, Phase II Hinjawadi, Pune 411 057, Maharashtra, India
| | - Adelina Curaj
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (D.S.); (A.C.); (J.J.); (E.A.L.B.)
| | - Erwin Wijnands
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands; (A.S.); (E.W.)
| | - Sergio A. Lira
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Frank Tacke
- Department of Hepatology and Gastroenterolgy, Campus Virchow-Klinikum and Campus Charité Mitte, Charité–Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (D.S.); (A.C.); (J.J.); (E.A.L.B.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands; (A.S.); (E.W.)
| | - Erik A.L. Biessen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (D.S.); (A.C.); (J.J.); (E.A.L.B.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands; (A.S.); (E.W.)
| | - Emiel P.C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; (D.S.); (A.C.); (J.J.); (E.A.L.B.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands; (A.S.); (E.W.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
5
|
Huang S, Yue Y, Liang M, Feng K, Hou J, Li H, Huang X, Huang L, Chen G, Wu Z. Neutrophil gelatinase-associated lipocalin: a potential predictor of embolic events in endocarditis. Ann Thorac Surg 2021; 113:1215-1222. [PMID: 33961821 DOI: 10.1016/j.athoracsur.2021.04.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND As the complication of infective endocarditis (IE), embolic events (EE) are associated with increased mortality and morbidity. However, there are no reliable indicators to predict embolism. The aim of this study was to evaluate neutrophil gelatinase-associated lipocalin (NGAL) as a biomarker to identify IE patients at high risk of embolism. METHODS We conducted a prospective observational study of IE patients from January 2018 to December 2020. Plasma NGAL levels were measured in 88 IE patients (37 patients with EE and 51 patients without EE), 91 noninfectious heart valve disease (HVD) patients, and 20 healthy blood donors by ELISA. Native valve tissue was obtained from 16 IE patients and 16 HVD patients. Western blot and immunohistochemical staining were performed to detect NGAL and matrix metalloproteinas-9 (MMP-9). RESULTS Higher levels of NGAL were observed in IE patients than HVD patients (P<0.001) or healthy blood donors (P<0.01). In addition, NGAL levels were higher in IE patients with EE compared to those without EE (P<0.001). Receiver operating characteristic analysis demonstrated that NGAL acted as a potential EE predictor with the cutoff value of 166.78 ng/ml. IE patients with higher NGAL levels were significantly related to more severe native valve morphologic changes. NGAL was co-localized with MMP-9, and their expression in the valves of IE patients was higher than those of HVD patients. CONCLUSIONS NGAL is a potential predictor of EE in IE. This may be attributed to its potency of increasing the proteolytic activity of MMP-9, which leads to valve morphologic impairment.
Collapse
Affiliation(s)
- Suiqing Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yuan Yue
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Mengya Liang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Kangni Feng
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jian Hou
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Huayang Li
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xiaolin Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Lin Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Guangxian Chen
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
6
|
Abdallah AM, Abu-Madi M. The Genetic Control of the Rheumatic Heart: Closing the Genotype-Phenotype Gap. Front Med (Lausanne) 2021; 8:611036. [PMID: 33842495 PMCID: PMC8024521 DOI: 10.3389/fmed.2021.611036] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/07/2021] [Indexed: 12/20/2022] Open
Abstract
Rheumatic heart disease (RHD) is a heritable inflammatory condition characterized by carditis, arthritis, and systemic disease. Although remaining neglected, the last 3 years has seen some promising advances in RHD research. Whilst it is clear that RHD can be triggered by recurrent group A streptococcal infections, the mechanisms driving clinical progression are still poorly understood. This review summarizes our current understanding of the genetics implicated in this process and the genetic determinants that predispose some people to RHD. The evidence demonstrating the importance of individual cell types and cellular states in delineating causal genetic variants is discussed, highlighting phenotype/genotype correlations where possible. Genetic fine mapping and functional studies in extreme phenotypes, together with large-scale omics studies including genomics, transcriptomics, epigenomics, and metabolomics, are expected to provide new information not only on RHD but also on the mechanisms of other autoimmune diseases and facilitate future clinical translation.
Collapse
Affiliation(s)
- Atiyeh M Abdallah
- Biomedical and Pharmaceutical Research Unit, Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Marawan Abu-Madi
- Biomedical and Pharmaceutical Research Unit, Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Neutrophil-Lymphocyte Ratio in Predicting Infective Endocarditis: A Case-Control Retrospective Study. Mediators Inflamm 2020; 2020:8586418. [PMID: 33354163 PMCID: PMC7735837 DOI: 10.1155/2020/8586418] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 10/27/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background Infective endocarditis (IE) is a complex infectious disease with high morbidity and mortality. The inflammation mechanism of IE is a complex network including interactions of inflammatory cytokines and other components of host response. As an important inflammation marker, the prediction ability of neutrophil-to-lymphocyte ratio (NLR) in IE deserves further investigation. Methods NLR values were measured and compared between IE patients and healthy controls, good and bad clinical outcome groups. The receiver operating characteristic curves (ROCs) of NLR and cut-off values were measured in IE patients, pathogen-subgroups, and different clinical outcome groups. Results There were 678 IE patients and 2520 healthy controls enrolled in our study. The number of good and bad clinical outcome patients was 537 and 141, respectively. The value of NLR was significantly higher in IE patients than healthy controls (6.29 ± 9.36 vs. 1.87 ± 0.34, p < 0.001), and the area under the ROC (AUC) was 0.817 (95% CI (0.794, 0.839), p < 0.001). The critical value of NLR for diagnosis of IE was 2.68, with a sensitivity of 69%, and a specificity of 88%. The value of NLR was significantly higher in bad clinical outcome patients than in good clinical outcome patients (5.8 ± 6.02 vs. 3.62 ± 2.61, p < 0.001). The critical value of NLR to predict the outcome of IE was 5.557, with a sensitivity of 39.0% and a specificity of 85.3%. Conclusions NLR is a predictive marker for IE patients, especially in Gram-negative bacteria and Gram-positive bacteria-infected IE patients. NLR also can predict the outcome of IE. Early detecting NLR upon admission may assist in early diagnosis and risk stratification of patients with IE.
Collapse
|
8
|
Liu Z, Wang Y, Shi J, Chen S, Xu L, Li F, Dong N. IL-21 promotes osteoblastic differentiation of human valvular interstitial cells through the JAK3/STAT3 pathway. Int J Med Sci 2020; 17:3065-3072. [PMID: 33173427 PMCID: PMC7646116 DOI: 10.7150/ijms.49533] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Objectives: This study amied to whether IL-21 promotes osteoblast transdifferentiation of cultured human Valvular interstitial cells (VICs). Methods: We first confirmed that IL-21 alters gene expression between CAVD aortic valve tissue and normal samples by immunohistochemistry, qPCR, and western blotting. VICs were cultured and treated with IL-21. Gene and protein expression levels of the osteoblastic markers ALP and Runx2, which can be blocked by specific JAK3 inhibitors and/or siRNA of STAT3, were measured. Results: IL-21 expression was upregulated in calcified aortic valves and promotes osteogenic differentiation of human VICs. IL-21 accelerated VIC calcification through the JAK3/STAT3 pathway. Conclusion: Our data suggest that IL-21 is a key factor in valve calcification and a promising candidate for targeted therapeutics for CAVD.
Collapse
Affiliation(s)
- Zongtao Liu
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Si Chen
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Li
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Lerche CJ, Christophersen LJ, Goetze JP, Nielsen PR, Thomsen K, Enevold C, Høiby N, Jensen PØ, Bundgaard H, Moser C. Adjunctive dabigatran therapy improves outcome of experimental left-sided Staphylococcus aureus endocarditis. PLoS One 2019; 14:e0215333. [PMID: 31002679 PMCID: PMC6474597 DOI: 10.1371/journal.pone.0215333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background Staphylococcus aureus is the most frequent and fatal cause of left-sided infective endocarditis (IE). New treatment strategies are needed to improve the outcome. S. aureus coagulase promotes clot and fibrin formation. We hypothesized that dabigatran, could reduce valve vegetations and inflammation in S. aureus IE. Methods We used a rat model of severe aortic valve S. aureus IE. All infected animals were randomized to receive adjunctive dabigatran (10 mg/kg b.i.d., n = 12) or saline (controls, n = 11) in combination with gentamicin. Valve vegetation size, bacterial load, cytokine, cell integrins expression and peripheral platelets and neutrophils were assessed 3 days post-infection. Results Adjunctive dabigatran treatment significantly reduced valve vegetation size compared to controls (p< 0.0001). A significant reduction of the bacterial load in aortic valves was seen in dabigatran group compared to controls (p = 0.02), as well as expression of key pro-inflammatory markers keratinocyte-derived chemokine, IL-6, ICAM-1, TIMP-1, L-selectin (p< 0.04). Moreover, the dabigatran group had a 2.5-fold increase of circulating platelets compared to controls and a higher expression of functional and activated platelets (CD62p+) unbound to neutrophils. Conclusion Adjunctive dabigatran reduced the vegetation size, bacterial load, and inflammation in experimental S. aureus IE.
Collapse
Affiliation(s)
- Christian J. Lerche
- Department of Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Immunology and Microbiology, University of Copenhagen, Denmark
- * E-mail:
| | - Lars J. Christophersen
- Department of Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jens Peter Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Pia R. Nielsen
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Kim Thomsen
- Department of Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Christian Enevold
- Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Peter Ø. Jensen
- Department of Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Immunology and Microbiology, University of Copenhagen, Denmark
- Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Immunology and Microbiology, University of Copenhagen, Denmark
| |
Collapse
|
10
|
Lerche CJ, Christophersen LJ, Trøstrup H, Thomsen K, Jensen PØ, Hougen HP, Bundgaard H, Høiby N, Moser C. Low efficacy of tobramycin in experimental Staphylococcus aureus endocarditis. Eur J Clin Microbiol Infect Dis 2015; 34:2349-57. [PMID: 26440039 DOI: 10.1007/s10096-015-2488-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 09/03/2015] [Indexed: 12/31/2022]
Abstract
The empiric treatment of infective endocarditis (IE) varies widely and, in some places, a regimen of penicillin in combination with an aminoglycoside is administered. The increasing incidence of Staphylococcus aureus IE, poor tissue penetration by aminoglycosides and low frequency of penicillin-susceptible S. aureus may potentially lead to functional tobramycin monotherapy. Therefore, this study aimed to evaluate tobramycin monotherapy in an experimental S. aureus IE rat model. Catheter-induced IE at the aortic valves were established with S. aureus (NCTC 8325-4) and rats were randomised into untreated (n = 22) or tobramycin-treated (n = 13) groups. The treatment group received tobramycin once-daily. Animals were evaluated at 1 day post infection (DPI), 2 DPI or 3 DPI. Quantitative bacteriology and cytokine expression were measured for valves, myocardium and serum. A decrease of bacterial load was observed in valves and the spleens of the treated (n = 6) compared to the untreated group at 2 DPI (n = 8) (p ≤ 0.02 and p ≤ 0.01, respectively), but not at 3 DPI (n = 7). Quantitative bacteriology in the myocardium was not different between the groups. Keratinocyte-derived chemokine (KC) in the aortic valves was significantly reduced at 2 DPI in the tobramycin-treated group (p ≤ 0.03). However, the expression of interleukin (IL)-1b, IL-6 and granulocyte-colony stimulating factor (G-CSF) in the valves was not different between the two groups. In the myocardium, a significant reduction in IL-1b was observed at 2 DPI (p ≤ 0.001) but not at 3 DPI. Tobramycin as functional monotherapy only reduced bacterial load and inflammation transiently, and was insufficient in most cases of S. aureus IE.
Collapse
Affiliation(s)
- C J Lerche
- Department of Clinical Microbiology 9301, Copenhagen University Hospital, Rigshospitalet, Juliane Maries vej 22, 2100, Copenhagen, Denmark.
| | - L J Christophersen
- Department of Clinical Microbiology 9301, Copenhagen University Hospital, Rigshospitalet, Juliane Maries vej 22, 2100, Copenhagen, Denmark
| | - H Trøstrup
- Department of Clinical Microbiology 9301, Copenhagen University Hospital, Rigshospitalet, Juliane Maries vej 22, 2100, Copenhagen, Denmark
| | - K Thomsen
- Department of Clinical Microbiology 9301, Copenhagen University Hospital, Rigshospitalet, Juliane Maries vej 22, 2100, Copenhagen, Denmark
| | - P Ø Jensen
- Department of Clinical Microbiology 9301, Copenhagen University Hospital, Rigshospitalet, Juliane Maries vej 22, 2100, Copenhagen, Denmark
| | - H P Hougen
- Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark
| | - H Bundgaard
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - N Høiby
- Department of Clinical Microbiology 9301, Copenhagen University Hospital, Rigshospitalet, Juliane Maries vej 22, 2100, Copenhagen, Denmark.,Institute of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - C Moser
- Department of Clinical Microbiology 9301, Copenhagen University Hospital, Rigshospitalet, Juliane Maries vej 22, 2100, Copenhagen, Denmark
| |
Collapse
|