1
|
Xu J, Zhou Z, Zheng Y, Yang S, Huang K, Li H. Roles of inflammasomes in viral myocarditis. Front Cell Infect Microbiol 2023; 13:1149911. [PMID: 37256114 PMCID: PMC10225676 DOI: 10.3389/fcimb.2023.1149911] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/28/2023] [Indexed: 06/01/2023] Open
Abstract
Viral myocarditis (VMC), characterized by viral infection-induced inflammation, is a life-threatening disease associated with dilated cardiomyopathy or heart failure. Innate immunity plays a crucial role in the progression of inflammation, in which inflammasomes provide a platform for the secretion of cytokines and mediate pyroptosis. Inflammasomes are rising stars gaining increasing attention. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, the caspase recruitment domain-containing protein 8 (CARD8) inflammasome, and the caspase-11 inflammasome are three inflammasomes that were reported to affect the process and prognosis of VMC. These inflammasomes can be activated by a wide range of cellular events. Accumulating evidence has suggested that inflammasomes are involved in different stages of VMC, including the trigger and progression of myocardial injury and remodeling after infection. In this review, we summarized the pathways involving inflammasomes in VMC and discussed the potential therapies targeting inflammasomes and related pathways.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yidan Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sai Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huili Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Li B, Cao X, Ai G, Liu Y, Lv C, Jin L, Xu R, Zhao G, Yuan H. Interleukin-37 alleviates myocardial injury induced by coxsackievirus B3 via inhibiting neutrophil extracellular traps formation. Int Immunopharmacol 2022; 113:109343. [DOI: 10.1016/j.intimp.2022.109343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
3
|
Muacevic A, Adler JR. Efficacy of Anakinra in Pericarditis: A Systematic Review. Cureus 2022; 14:e29862. [PMID: 36212270 PMCID: PMC9531702 DOI: 10.7759/cureus.29862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022] Open
Abstract
Inflammation of the pericardium is referred to as pericarditis, which can cause sharp chest pain and has a high chance of recurrence even after treatment. This review will explore anakinra, which is an interleukin-1 receptor antagonist, as a potential new treatment for pericarditis. The systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines by searching PubMed and GoogleScholar from the years 2012 to 2022. After applying inclusion and exclusion criteria, thorough screening, and quality appraisal, a total of eleven studies were included in the review; eight case reports and three clinical trials. All studies showed that 100 mg/day of anakinra caused a remarkable improvement in patient outcomes. In addition, the pericarditis resolved quicker and had a lower chance of recurrence in comparison to conventional therapy.
Collapse
|
4
|
Kwon KW, Kim LH, Kang SM, Lee JM, Choi E, Park J, Hong JJ, Shin SJ. Host-directed antimycobacterial activity of colchicine, an anti-gout drug, via strengthened host innate resistance reinforced by the IL-1β/PGE 2 axis. Br J Pharmacol 2022; 179:3951-3969. [PMID: 35301712 DOI: 10.1111/bph.15838] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE To diversify and expand possible tuberculosis (TB) drug candidates and maximize limited global resources, we investigated the effect of colchicine, an FDA-approved anti-gout drug, against Mycobacterium tuberculosis (Mtb) infection because of its immune-modulating effect. EXPERIMENTAL APPROACH We evaluated the intracellular anti-Mtb activity of different concentrations of colchicine in murine bone marrow-derived macrophages (BMDMs). To elucidate the underlying mechanism, RNA sequencing, biological and chemical inhibition assays, and Western blot, quantitative real-time PCR, enzyme-linked immunosorbent assay (ELISA) and immunohistochemical analyses were employed. Finally, type I interferon-dependent highly TB-susceptible A/J mice were challenged with virulent Mtb H37Rv, and the host-directed therapeutic effect of oral colchicine administration on bacterial burdens and lung inflammation was assessed 30 days post-infection (2.5 mg·kg-1 every two days). KEY RESULTS Colchicine reinforced the anti-Mtb activity of BMDMs without affecting cell viability, indicating that colchicine facilitated macrophage immune activation upon Mtb infection. The results from RNA sequencing, NLRP3 knockout BMDM, IL-1 receptor blockade, and immunohistochemistry analyses revealed that this unexpected intracellular anti-Mtb activity of colchicine was mediated through NLRP3-dependent IL-1β signalling and Cox-2-regulated PGE2 production in macrophages. Consequently, the TB-susceptible A/J mouse model showed remarkable protection, with decreased bacterial loads in both the lungs and spleens of oral colchicine-treated mice, with significantly elevated Cox-2 expression at infection sites. CONCLUSIONS AND IMPLICATIONS The repurposing of colchicine against Mtb infection in this study highlights its unique function in macrophages upon Mtb infection and its novel potential use in treating TB as host-directed or adjunctive therapy.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology and Institute for Immunology and Immunological Disease, Brain Korea 21 Project for the Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Lee-Han Kim
- Department of Microbiology and Institute for Immunology and Immunological Disease, Brain Korea 21 Project for the Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Soon Myung Kang
- Department of Microbiology and Institute for Immunology and Immunological Disease, Brain Korea 21 Project for the Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ju Mi Lee
- Department of Microbiology and Institute for Immunology and Immunological Disease, Brain Korea 21 Project for the Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunsol Choi
- Department of Microbiology and Institute for Immunology and Immunological Disease, Brain Korea 21 Project for the Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jiyun Park
- Department of Microbiology and Institute for Immunology and Immunological Disease, Brain Korea 21 Project for the Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jung Joo Hong
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheongbuk-do, South Korea
| | - Sung Jae Shin
- Department of Microbiology and Institute for Immunology and Immunological Disease, Brain Korea 21 Project for the Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
5
|
Pappritz K, Lin J, El-Shafeey M, Fechner H, Kühl U, Alogna A, Spillmann F, Elsanhoury A, Schulz R, Tschöpe C, Van Linthout S. Colchicine prevents disease progression in viral myocarditis via modulating the NLRP3 inflammasome in the cardiosplenic axis. ESC Heart Fail 2022; 9:925-941. [PMID: 35178861 PMCID: PMC8934990 DOI: 10.1002/ehf2.13845] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 12/17/2021] [Accepted: 02/04/2022] [Indexed: 12/11/2022] Open
Abstract
Aim The acute phase of a coxsackievirus 3 (CVB3)‐induced myocarditis involves direct toxic cardiac effects and the systemic activation of the immune system, including the cardiosplenic axis. Consequently, the nucleotide‐binding oligomerization domain‐like receptor pyrin domain‐containing‐3 (NLRP3) inflammasome pathway is activated, which plays a role in disease pathogenesis and progression. The anti‐inflammatory drug colchicine exerts its effects, in part, via reducing NLRP3 activity, and has been shown to improve several cardiac diseases, including acute coronary syndrome and pericarditis. The aim of the present study was to evaluate the potential of colchicine to improve experimental CVB3‐induced myocarditis. Methods and results C57BL6/j mice were intraperitoneally injected with 1 × 105 plaque forming units of CVB3. After 24 h, mice were treated with colchicine (5 μmol/kg body weight) or phosphate‐buffered saline (PBS) via oral gavage (p.o.). Seven days post infection, cardiac function was haemodynamically characterized via conductance catheter measurements. Blood, the left ventricle (LV) and spleen were harvested for subsequent analyses. In vitro experiments on LV‐derived fibroblasts (FB) and HL‐1 cells were performed to further evaluate the anti‐(fibro)inflammatory and anti‐apoptotic effects of colchicine via gene expression analysis, Sirius Red assay, and flow cytometry. CVB3 + colchicine mice displayed improved LV function compared with CVB3 + PBS mice, paralleled by a 4.7‐fold (P < 0.01) and 1.7‐fold (P < 0.001) reduction in LV CVB3 gene expression and cardiac troponin‐I levels in the serum, respectively. Evaluation of components of the NLRP3 inflammasome revealed an increased percentage of apoptosis‐associated speck‐like protein containing a CARD domain (ASC)‐expressing, caspase‐1‐expressing, and interleukin‐1β‐expressing cells in the myocardium and in the spleen of CVB3 + PBS vs. control mice, which was reduced in CVB3 + colchicine compared with CVB3 + PBS mice. This was accompanied by 1.4‐fold (P < 0.0001), 1.7‐fold (P < 0.0001), and 1.7‐fold (P < 0.0001) lower numbers of cardiac dendritic cells, natural killer cells, and macrophages, respectively, in CVB3 + colchicine compared with CVB3 + PBS mice. A 1.9‐fold (P < 0.05) and 4.6‐fold (P < 0.001) reduced cardiac gene expression of the fibrotic markers, Col1a1 and lysyl oxidase, respectively, was detected in CVB3 + colchicine mice compared with CVB3 + PBS animals, and reflected by a 2.2‐fold (P < 0.05) decreased Collagen I/III protein ratio. Colchicine further reduced Col3a1 mRNA and collagen protein expression in CVB3‐infected FB and lowered apoptosis and viral progeny release in CVB3‐infected HL‐1 cells. In both CVB3 FB and HL‐1 cells, colchicine down‐regulated the NLRP3 inflammasome‐related components ASC, caspase‐1, and IL‐1β. Conclusions Colchicine improves LV function in CVB3‐induced myocarditis, involving a decrease in cardiac and splenic NLRP3 inflammasome activity, without exacerbation of CVB3 load.
Collapse
Affiliation(s)
- Kathleen Pappritz
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité, Universitätmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Jie Lin
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Muhammad El-Shafeey
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité, Universitätmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Physiologisches Institut, Fachbereich Medizin der Justus-Liebig-Universität, Giessen, Germany.,Medical Biotechnology Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications, Alexandria, Egypt
| | - Henry Fechner
- Department of Applied Biochemistry, Institute for Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Uwe Kühl
- Department of Cardiology, Charité - Universitätsmedizin Berlin, CVK, Berlin, Germany
| | - Alessio Alogna
- Department of Cardiology, Charité - Universitätsmedizin Berlin, CVK, Berlin, Germany
| | - Frank Spillmann
- Department of Cardiology, Charité - Universitätsmedizin Berlin, CVK, Berlin, Germany
| | - Ahmed Elsanhoury
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité, Universitätmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Rainer Schulz
- Physiologisches Institut, Fachbereich Medizin der Justus-Liebig-Universität, Giessen, Germany
| | - Carsten Tschöpe
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité, Universitätmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité - Universitätsmedizin Berlin, CVK, Berlin, Germany
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité, Universitätmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| |
Collapse
|
6
|
Bellos I, Karageorgiou V, Viskin D. Myocarditis following mRNA Covid-19 vaccination: a pooled analysis. Vaccine 2022; 40:1768-1774. [PMID: 35153093 PMCID: PMC8818354 DOI: 10.1016/j.vaccine.2022.02.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/22/2022] [Accepted: 02/02/2022] [Indexed: 12/15/2022]
Abstract
Background Methods Results Conclusions
Collapse
|
7
|
Enayati A, Banach M, Jamialahmadi T, Sahebkar A. Protective role of nutraceuticals against myocarditis. Biomed Pharmacother 2021; 146:112242. [PMID: 34953630 DOI: 10.1016/j.biopha.2021.112242] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 01/22/2023] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium that mostly affects young adults. The disease is commonly caused by viral infection, medications, autoimmune disorders, and inflammatory conditions. Nearly 50% of the cases of myocarditis are due to post-viral immune response in a setting of an identifiable or non-identifiable infection. The clinical manifestation is nonspecific ranging from asymptomatic courses to sudden death in infants and young patients. This review describes the properties of phytochemicals as plant-derived active ingredients which can be used in the prevention and treatment of myocarditis and its associated risk factors. Meanwhile, it has illustrated epidemiological analyses, mechanism of action, and the metabolism of phytochemicals in animal and human clinical trials. We also mentioned the precise mechanism of action by which phytochemicals elicit their anti-viral, anti-inflammatory, antioxidant, and immunomodulatory effects and how they regulate signal transduction pathways. Nevertheless, comprehensive clinical trials are required to study the properties of phytochemicals in vivo, in vitro, and in silico for a proper management of myocarditis. Our findings indicate that phytochemicals function as potent adjunctive therapeutic drugs in myocarditis and its related complications.
Collapse
Affiliation(s)
- Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland.
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | |
Collapse
|
8
|
Viruses in the Heart: Direct and Indirect Routes to Myocarditis and Heart Failure. Viruses 2021; 13:v13101924. [PMID: 34696354 PMCID: PMC8537553 DOI: 10.3390/v13101924] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023] Open
Abstract
Viruses are an underappreciated cause of heart failure. Indeed, several types of viral infections carry cardiovascular risks. Understanding shared and unique mechanisms by which each virus compromises heart function is critical to inform on therapeutic interventions. This review describes how the key viruses known to lead to cardiac dysfunction operate. Both direct host-damaging mechanisms and indirect actions on the immune systems are discussed. As viral myocarditis is a key pathologic driver of heart failure in infected individuals, this review also highlights the role of cytokine storms and inflammation in virus-induced cardiomyopathy.
Collapse
|
9
|
Wu L, Woudstra L, Dam TA, Germans T, van Rossum AC, Niessen HWM, Krijnen PAJ. Electrocardiographic changes are strongly correlated with the extent of cardiac inflammation in mice with Coxsackievirus B3-induced viral myocarditis. Cardiovasc Pathol 2021; 54:107367. [PMID: 34245872 DOI: 10.1016/j.carpath.2021.107367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/11/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Viral myocarditis (VM) can induce changes in myocardial electrical conduction and arrhythmia. However, their relationship with myocarditis-associated arrhythmic substrates in the heart such as inflammation and fibrosis is relatively unknown. This we have analyzed in the present study. METHODS C3H mice were infected with 1×105 plaque-forming units Coxsackievirus B3 (CVB3, n=68) and were compared with uninfected control mice (n=10). Electrocardiograms (ECGs) were recorded in all conscious mice shortly before sacrifice and included heart rate; P-R interval; QRS duration; QTc interval and R-peak amplitude of lead II and aVF. Mice were sacrificed at 4, 7, 10, 21, 35 or 49 days post-infection. Cardiac lesion size, calcification, fibrosis and cellular infiltration of CD45+ lymphocytes, MAC3+ macrophages, Ly6G+ neutrophils and mast cells were quantitatively determined in cross-sections of the ventricles. Putative relations between ECG changes and lesion size and/or cardiac inflammation were then analyzed. RESULTS Significant transient reductions in QRS duration and R-peak amplitude occurred between 4 and 14 days post-infection and returned to baseline values thereafter. The magnitude of these ECG changes strongly correlated to the extent of lymphocyte (days 7 and 14), macrophage (days 7 and 10) and neutrophil (days 7) infiltration. The ECG changes did not significantly correlate with lesion size and fibrosis. CONCLUSION VM induces transient changes in myocardial electrical conduction that are strongly related to cellular inflammation of the heart. These data show that even in mild VM, with relatively little cardiac damage, the inflammatory infiltrate can form an important arrhythmogenic substrate.
Collapse
Affiliation(s)
- Linghe Wu
- Department of Pathology, Amsterdam University Medical Centers, location VUmc and AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Linde Woudstra
- Department of Pathology, Amsterdam University Medical Centers, location VUmc and AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Tariq A Dam
- Department of Pathology, Amsterdam University Medical Centers, location VUmc and AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Tjeerd Germans
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands; Department of Cardiology, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
| | - Albert C van Rossum
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands; Department of Cardiology, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, Amsterdam University Medical Centers, location VUmc and AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands; Department of Cardiac Surgery, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, Amsterdam University Medical Centers, location VUmc and AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Khawaja A, Bromage DI. The innate immune response in myocarditis. Int J Biochem Cell Biol 2021; 134:105973. [PMID: 33831592 DOI: 10.1016/j.biocel.2021.105973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
Acute myocarditis is an inflammatory condition of the heart characterised by cellular injury and the influx of leucocytes, including neutrophils, monocytes, macrophages and lymphocytes. While this response is vital for tissue repair, excessive scar deposition and maladaptive ventricular remodelling can result in a legacy of heart failure. It is increasingly recognised as a clinical phenomenon due, in part, to increased availability of cardiac magnetic resonance imaging in patients presenting with chest pain in the absence of significant coronary artery disease. Emerging epidemiological evidence has associated myocarditis with poor outcomes in the context of left ventricular impairment, and even when the left ventricle is preserved outcomes are less benign than once thought. Despite this, our understanding of the contribution of the inflammatory response to the pathophysiology of acute myocarditis lags behind that of acute myocardial infarction, which is the vanguard cardiovascular condition for inflammation research. We recently reviewed monocyte and macrophage phenotype and function in acute myocardial infarction, concluding that their plasticity and heterogeneity might account for conflicting evidence from attempts to target specific leucocyte subpopulations. Here, we revise our understanding of myocardial inflammation, which is predominantly derived from myocardial infarction research, review experimental evidence for the immune response in acute myocarditis, focusing on innate immunity, and discuss potential future directions for immunotherapy research in acute myocarditis.
Collapse
Affiliation(s)
- Abdullah Khawaja
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
11
|
The Role of NLRP3 Inflammasome in Pericarditis: Potential for Therapeutic Approaches. JACC Basic Transl Sci 2021; 6:137-150. [PMID: 33665514 PMCID: PMC7907621 DOI: 10.1016/j.jacbts.2020.11.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 01/08/2023]
Abstract
Acute pericarditis is characterized by an intense inflammatory response involving the pericardium. Although mostly benign in its clinical course, 30% of patients may experience complications (recurrence, treatment failure, cardiac tamponade). The pathogenesis of pericarditis is poorly understood. The scarcity of animal models might justify the limited understanding of this syndrome and the lack of targeted therapies. Acute pericarditis is believed to represent a stereotypical response to an acute injury of the pericardium. The NLRP3 inflammasome, through its main product, IL-1β, could play a central role in the clinical manifestations. A mouse model of acute pericarditis was developed through the intrapericardial injection of zymosan A, leading to the classical features of the inflamed pericardium: pericardial effusion, pericardial thickening, and increased expression of the NLRP3 inflammasome. By inhibiting the NLRP3 inflammasome or IL-1β, the pericardial effusion and thickening and the NLRP3 inflammasome expression were greatly reduced compared with vehicle. Treatment with IL-1 trap, neutralizing both IL-1β and IL-1α, produced a powerful effect on pericardial inflammation in the experimental pericarditis model.
Human samples of patients with chronic pericarditis and appropriate control subjects were stained for the inflammasome components. A mouse model of pericarditis was developed through the intrapericardial injection of zymosan A. Different inflammasome blockers were tested in the mouse model. Patients with pericarditis presented an intensification of the inflammasome activation compared with control subjects. The experimental model showed the pathological features of pericarditis. Among inflammasome blockers, NLRP3 inflammasome inhibitor, anakinra, and interleukin-1 trap were found to significantly improve pericardial alterations. Colchicine partially improved the pericardial inflammation. An intense activation of the inflammasome in pericarditis was demonstrated both in humans and in mice.
Collapse
|
12
|
McEwan T, Robinson PC. A systematic review of the infectious complications of colchicine and the use of colchicine to treat infections. Semin Arthritis Rheum 2021; 51:101-112. [PMID: 33360321 PMCID: PMC7832726 DOI: 10.1016/j.semarthrit.2020.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Colchicine has been used historically as an anti-inflammatory agent for a wide range of diseases. Little is known regarding the relationship between colchicine use and infectious disease outcomes. The objective of this study was to systematically examine infectious adverse events associated with colchicine usage and the clinical use of colchicine for infectious diseases. METHODS A systematic review was conducted in accordance with PRISMA methodology. PubMed, EMBASE, Scopus and Cochrane Library databases were searched (up to 12th October, 2020) for interventional and observational studies that included colchicine usage associated with infectious adverse events or infectious disease outcomes. RESULTS A total of 9,237 studies were initially identified and after exclusions, 36 articles comprising 21 interventional studies and 15 observational studies were included in this systematic review. There were 19 studies that reported infectious adverse events and 17 studies that examined the efficacy of colchicine in treating infectious disease. Only two out of six studies reported a significant benefit using colchicine in the management of viral liver disease. There was some evidence colchicine is beneficial in managing COVID-19 by reducing time to deterioration, length of stay in hospital and mortality. Colchicine had some benefit in managing malaria, condyloma accuminata and verruca vulgaris, viral myocarditis and erythema nodosum leprosum based on case-series or small, pilot clinical studies. Two of the clinical trials and five of the observational studies reported significant associations between infections adverse events and colchicine usage. Risk of pneumonia was found in three studies and post-operative infections were reported in two studies. Risks of urinary tract infections, H. pylori and C.difficile were only reported by one study each. CONCLUSION There is a current lack of clinical evidence that colchicine has a role in treating or managing infectious diseases. Preliminary studies have demonstrated a possible role in the management of COVID-19 but results from more clinical trials are needed. There is inconclusive evidence that suggests colchicine is associated with increased risk of infections, particularly pneumonia.
Collapse
Affiliation(s)
- Timothy McEwan
- University of Queensland School of Clinical Medicine, Queensland, Australia
| | - Philip C Robinson
- University of Queensland School of Clinical Medicine, Queensland, Australia.
| |
Collapse
|
13
|
Blagova OV, Nedostup AV, Sedov VP, Kogan EA, Alijeva IN, Sorokin GY, Sarkisova ND. [Pericarditis in contemporary therapeutic clinic: nosological spectrum, approaches to diagnosis and treatment]. TERAPEVT ARKH 2020; 92:10-18. [PMID: 33720567 DOI: 10.26442/00403660.2020.12.200432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 02/07/2021] [Indexed: 11/22/2022]
Abstract
AIM To analyze the register of pericarditis in a therapeutic clinic, to evaluate their nosological spectrum, to optimize approaches to diagnosis and treatment. MATERIALS AND METHODS For the period 20072018, the register includes 76 patients with the diagnosis of pericarditis (average age 53.115.7 years, 2085 years, 46 female). Patients with hydropericardium were not included in the register. Diagnostic puncture of pericardium was carried out in 5 patients, pleural puncture in 11 patients. Morphological diagnostics included endomyocardial/ intraoperative biopsy of myocardium (n=4/2), thoracoscopic/intraoperative biopsy of pericardium (n=1/6), pleural puncture (n=5), transbronchial (n=1), thoracoscopic biopsy of intrathoracic lymph nodes (n=2), lung (n=1), supraclavicular lymph node biopsy (n=1), salivary gland (n=1), subcutaneous fat and rectum biopsy per amyloid (n=6/1). The genome of cardiotropic viruses, level of anti-heart antibodies, C-reactive protein, antinuclear factor, rheumatoid factor (antibodies to cyclic citrullinized peptide), antibodies to neutrophil cytoplasm were determined, extractable nuclear antigens (ENA), protein immunoelectrophoresis, diaskin test, computed tomography of lungs and heart, cardiac magnetic resonance imaging, oncologic search. RESULTS The following forms of pericarditis were verified: tuberculosis (14%, including in combination with hypertrophic cardiomyopathy HCM), acute / chronic viral (8%) and infectious immune (38%), including perimyocarditis in 77%, pericarditis associated with mediastinum lymphoma/sarcoma (4%), sarcoidosis (3%), diffuse diseases of connective tissue and vasculitis (systemic lupus erythematosus, rheumatoid arthritis, diseases of Horton, Takayasu, Shegren, Wegener, 12%), leukoclastic vasculitis, Loefflers endomyocarditis, AL-amyloidosis, thrombotic microangiopathy (1% each), HCM (8%), coronary heart disease (constriction after repeated punctures and suppuration; postinfection and immune, 4%), after radiofrequency catheter ablation and valve prosthetics (2%). Tuberculosis was the main causes of constrictive pericarditis (36%). Treatment included steroids (n=39), also in combination with cytostatics (n=12), anti-tuberculosis drugs (n=9), acyclovir/ganclovir (n=14), hydroxychloroquine (n=23), colchicine (n=13), non-steroidal anti-inflammatory drugs (n=21), L-tyroxine (n=5), chemotherapy (n=1). In 36 patients different types of therapy were combined. Treatment results observed in 55 patients. Excellent and stable results were achieved in 82% of them. Pericardiectomy/pericardial resection was successfully performed in 8 patients. Lethality was 13.2% (10 patients) with an average follow-up 9 [2; 29.5] months (up to 10 years). Causes of death were chronic heart failure, surgery for HCM, pulmonary embolism, tumor. CONCLUSION During a special examination, the nature of pericarditis was established in 97% of patients. Morphological and cytological diagnostics methods play the leading role. Tuberculosis pericarditis, infectious-immune and pericarditis in systemic diseases prevailed. Infectious immune pericarditis is characterized by small and medium exudate without restriction and accompanying myocarditis. Steroids remain the first line of therapy in most cases. Hydroxychloroquine as well as colchicine can be successfully used in moderate / low activity of immune pericarditis and as a long-term maintenance therapy after steroid stop.
Collapse
Affiliation(s)
- O V Blagova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - A V Nedostup
- Sechenov First Moscow State Medical University (Sechenov University)
| | - V P Sedov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - E A Kogan
- Sechenov First Moscow State Medical University (Sechenov University)
| | - I N Alijeva
- Sechenov First Moscow State Medical University (Sechenov University)
| | - G Y Sorokin
- Sechenov First Moscow State Medical University (Sechenov University)
| | - N D Sarkisova
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
14
|
Colchicine as a Potential Therapeutic Agent Against Cardiovascular Complications of COVID-19: an Exploratory Review. ACTA ACUST UNITED AC 2020; 2:1419-1429. [PMID: 32838182 PMCID: PMC7398860 DOI: 10.1007/s42399-020-00421-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease-19 (COVID-19) may result in serious complications involving several organ systems, including myocardial tissue. An exaggerated host inflammatory response, described as a cytokine storm, has been linked to play a major role in these complications. Colchicine and other pharmaceutical agents have been proposed to counter the cytokine storm and improve outcomes. In this exploratory review, we utilized a PubMed and Cochrane Database search aiming to identify the biochemical characteristics of the cytokine storm as well as to identify the potential effect of colchicine on these inflammatory biomarkers. The research yielded 30 reports describing the characteristics of the cytokine storm and 44 reports describing the effect of colchicine on various inflammatory biomarkers. According to our research, colchicine may be an agent of interest in the treatment of COVID-19 via its anti-inflammatory properties. However, there are potential drug interactions with cytochrome P450 3A4 inhibitors resulting in acute colchicine toxicities. Additionally, there is scarce evidence regarding the efficacy of colchicine in the acute phase of disease, since most trials evaluated its effect in chronic conditions. In this direction, our team proposes three different hypotheses for evaluating the place of colchicine in the treatment of COVID-19.
Collapse
|
15
|
Abstract
Myocarditis is generally a mild and self-limited consequence of systemic infection of cardiotropic viruses. However, patients can develop a temporary or permanent impairment of cardiac function including acute cardiomyopathy with hemodynamic compromise or severe arrhythmias. In this setting, specific causes of inflammation are associated with variable risks of death and transplantation. Recent translational studies suggest that treatments tailored to specific causes of myocarditis may impact clinical outcomes when added to guideline-directed medical care. This review summarizes recent advances in translational research that influence the utility of endomyocardial biopsy for the management of inflammatory cardiomyopathies. Emerging therapies for myocarditis based on these mechanistic hypotheses are entering clinical trials and may add to the benefits of established heart failure treatment.
Collapse
Affiliation(s)
- Carsten Tschöpe
- From the Charité, University Medicine Berlin, Campus Virchow Klinikum (CVK), Department of Cardiology, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin-Brandenburg Centrum für Regenerative Therapien, Germany (C.T., S.V.L.).,Deutsches Zentrum für Herz Kreislauf Forschung (DZHK)-Standort Berlin/Charité, Germany (C.T., S.V.L.)
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL (L.T.C.)
| | - Guillermo Torre-Amione
- Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (G.T.-A.).,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico (G.T.-A.)
| | - Sophie Van Linthout
- From the Charité, University Medicine Berlin, Campus Virchow Klinikum (CVK), Department of Cardiology, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin-Brandenburg Centrum für Regenerative Therapien, Germany (C.T., S.V.L.).,Deutsches Zentrum für Herz Kreislauf Forschung (DZHK)-Standort Berlin/Charité, Germany (C.T., S.V.L.)
| |
Collapse
|
16
|
Chiabrando JG, Bonaventura A, Vecchié A, Wohlford GF, Mauro AG, Jordan JH, Grizzard JD, Montecucco F, Berrocal DH, Brucato A, Imazio M, Abbate A. Management of Acute and Recurrent Pericarditis. J Am Coll Cardiol 2020; 75:76-92. [DOI: 10.1016/j.jacc.2019.11.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022]
|
17
|
Tsai TL, Wei JCC, Wu YT, Ku YH, Lu KL, Wang YH, Chiou JY. The Association Between Usage of Colchicine and Pneumonia: A Nationwide, Population-Based Cohort Study. Front Pharmacol 2019; 10:908. [PMID: 31474864 PMCID: PMC6706461 DOI: 10.3389/fphar.2019.00908] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/18/2019] [Indexed: 12/22/2022] Open
Abstract
Objectives: A previous study suggested that colchicine may cause leukopenia and increase the risk of infection, such as pneumonia. Thus, we investigated the potential relationship between colchicine use and risk of developing pneumonia. Methods: Data were collected from Taiwan's National Health Insurance Research Database (NHIRD), a nationwide, population-based database. A 13-year retrospective cohort study was conducted, and all investigated subjects were identified by International Classification of Disease, Ninth Revision, Clinical Modification, codes between 2000 and 2012. Propensity score matching was applied to adjust for potential confounding variables, and then Cox proportional hazard model was used to evaluate the hazard ratio (HR) of pneumonia in gout patients and its associations with colchicine use, colchicine dosage, and days of colchicine use. Results: A total of 24,410 gout patients were enrolled in this study, including 12,205 cases who were treated with colchicine (colchicine group) and 12,205 cases who did not receive colchicine (non-colchicine group). The overall incidence rates of pneumonia in the colchicine group and non-colchicine group were 18.6 and 12.6 per 1,000 person-years, respectively. The colchicine group had a higher risk of pneumonia as compared with the non-colchicine group [adjusted HR, 1.42; 95% confidence interval (CI), 1.32 to 1.53; P < 0.05]. High cumulative dose and days of colchicine use notably increased the risk of contracting pneumonia. Conclusion: This nationwide population-based cohort study reveals that gout patients taking colchicine are at increased risk of developing pneumonia compared with gout patients who do not use colchicine. Therefore, it is crucial that gout patients being treated with colchicine be given the minimally effective dosage for the shortest possible duration to minimize their risk of pneumonia.
Collapse
Affiliation(s)
- Tsung-Lin Tsai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Yue-Ting Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yueh-Han Ku
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Kun-Lin Lu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jeng-Yuan Chiou
- School of Health Policy and Management, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
18
|
Zhang YY, Zhang CX, Li Y, Jiang X, Wang YF, Sun Y, Wang J, Ji WY, Liu Y. Development of a novel rat model of heterogeneous hepatic injury by injection with colchicine via the splenic vein. World J Gastroenterol 2018; 24:5005-5012. [PMID: 30510375 PMCID: PMC6262251 DOI: 10.3748/wjg.v24.i44.5005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/20/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To develop a novel rat model of heterogeneous hepatic injury.
METHODS Seventy male Sprague-Dawley rats were randomly divided into a control group (n = 10) and a colchicine group (n = 60). A 0.25% colchicine solution (0.4 mL/kg) was injected via the splenic vein in the colchicine group to develop a rat model of heterogeneous hepatic injury. An equal volume of normal saline was injected via the splenic vein in the control group. At days 3, 7, and 14 and weeks 4, 8, and 12 after the operation, at least seven rats of the colchicine group were selected randomly for magnetic resonance imaging (MRI) examinations, and then they were euthanized. Ten rats of the control group underwent MRI examinations at the same time points, and then were euthanized at week 12. T2-weighted images (T2WI) and diffusion weighted imaging (DWI) were used to evaluate the heterogeneous hepatic injury. The heterogeneous injury between the left and right hepatic lobes was assessed on liver sections according to the histological scoring criteria, and correlated with the results of MRI study.
RESULTS Obvious pathological changes occurred in the hepatic parenchyma in the colchicine group. Hepatic injury scores were significantly different between the left and right lobes at each time point (P < 0.05). There was a significant difference in apparent diffusion coefficient (ADC) of DWI and liver-to-muscle ratio (LMR) of T2WI between the left and right lobes of rats in the colchicine group (P < 0.05) at each time point, and similar results were observed between the colchicine and control groups. Besides, there was a significant correlation between hepatic injury scores and ADC values or LMR (r = -0.682, P = 0.000; r = -0.245, P = 0.018).
CONCLUSION Injection with colchicine via the splenic vein can be used to successfully develop a rat model of heterogeneous hepatic injury. DWI and T2WI may help evaluate the heterogeneous injury among liver lobes.
Collapse
Affiliation(s)
- Yan-Yan Zhang
- Medical Imaging Center, The Affiliated Hospital of Liaoning Traditional Chinese Medical University, Shenyang 110032, Liaoning Province, China
- Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Chao-Xu Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yu Li
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Xuan Jiang
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yong-Fang Wang
- Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yang Sun
- Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Jun Wang
- Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Wan-Ying Ji
- Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yi Liu
- Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
19
|
Xu D, Wang P, Yang J, Qian Q, Li M, Wei L, Xu W. Gr-1+ Cells Other Than Ly6G+ Neutrophils Limit Virus Replication and Promote Myocardial Inflammation and Fibrosis Following Coxsackievirus B3 Infection of Mice. Front Cell Infect Microbiol 2018; 8:157. [PMID: 29868513 PMCID: PMC5962688 DOI: 10.3389/fcimb.2018.00157] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022] Open
Abstract
Coxsackievirus B3 (CVB3) is the primary cause of viral myocarditis. An early and abundant neutrophil accumulation in the myocardium is a hallmark of early CVB3 infection. Yet the relative contribution of neutrophils to host susceptibility to CVB3 myocarditis remains largely unknown. Herein, peripheral neutrophil depletion was implemented in a BALB/c mouse model of acute CVB3 myocarditis using the specific 1A-8 (anti-Ly6G) or a RB6-8C5 (anti-Gr-1) mAb covering a wide range. Anti-Ly6G treatment led to systemic neutropenia throughout the disease, but did not alter virus replication, disease susceptibility and histopathological changes in the heart and pancreas of mice. In contrast, depletion of both neutrophils and monocytes/macrophages by anti-Gr-1 mAb prior to and after infection significantly promoted susceptibility of mice to CVB3 infection which was associated with exacerbated cardiac and pancreatic viral load. However, depletion of Gr1+ cells significantly suppressed acute myocarditis and pancreatic acini destruction at day 7 post infection via reducing Ly6Chigh monocyte population in the circulation. Additionally, cardiac interstitial fibrosis was not affected by neutrophil depletion, whereas Gr-1+ cells other than neutrophils increased cardiac fibrosis at day 21 p.i. by increasing cardiac expression of profibrotic cytokine TNF-α and TGF-β. Thus, Neutrophil function is most likely not essential for CVB3 control and peripheral neutrophils play dispensable role in the pathogenesis of acute myocarditis and pancreatitis during CVB3 infection. Whereas Gr-1+ cells other than neutrophils play a major role in limiting viral replication while promoting myocardial and pancreatic inflammatory injury and fibrosis.
Collapse
Affiliation(s)
- Dan Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Peijie Wang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Jie Yang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Qian Qian
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lin Wei
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
20
|
Stanković DM, Švorc Ľ, Mariano JFML, Ortner A, Kalcher K. Electrochemical Determination of Natural Drug Colchicine in Pharmaceuticals and Human Serum Sample and its Interaction with DNA. ELECTROANAL 2017. [DOI: 10.1002/elan.201700233] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Dalibor M. Stanković
- The Vinča Institute of Nuclear Sciences; University of Belgrade; POB 522 11001 Belgrade Serbia
- Department of Analytical Chemistry, Innovation Center of the Faculty of Chemistry; University of Belgrade; Studentski trg 12-16 Belgrade 11000 Serbia
| | - Ľubomir Švorc
- Institute of Analytical Chemistry, Faculty of Chemical and Food Technology; Slovak University of Technology in Bratislava; Radlinského 9 Bratislava SK-812 37 Slovak Republic
| | - José F. M. L. Mariano
- Department of Physics and CeFEMA, Faculty of Science and Technology; University of Algarve; Campus de Gambelas Faro 8005-139 Portugal
| | - Astrid Ortner
- Institute of Chemistry - Analytical Chemistry; Karl-Franzens University; Universitätsplatz 1 Graz A-8010 Austria
| | - Kurt Kalcher
- Institute of Chemistry - Analytical Chemistry; Karl-Franzens University; Universitätsplatz 1 Graz A-8010 Austria
| |
Collapse
|
21
|
Smilde BJ, Woudstra L, Fong Hing G, Wouters D, Zeerleder S, Murk JL, van Ham M, Heymans S, Juffermans LJM, van Rossum AC, Niessen HWM, Krijnen PAJ, Emmens RW. Reply to the letter to the editor "Is colchicine really harmful in viral myocarditis?". Int J Cardiol 2017; 229:43. [PMID: 27916345 DOI: 10.1016/j.ijcard.2016.11.292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Bernard J Smilde
- Department of Pathology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands
| | - Linde Woudstra
- Department of Pathology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands; Institute for Cardiovascular Research of the Vrije Universiteit (ICaR-VU), VU University Medical Center, De Postbox 7057, 1007 MB Amsterdam, The Netherlands
| | - Gene Fong Hing
- Department of Pathology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands
| | - Diana Wouters
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Postbox 9892, 1006 AN Amsterdam, The Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Postbox 9892, 1006 AN Amsterdam, The Netherlands; Department of Hematology, Academic Medical Center, Postbox 22660, 1100 DD Amsterdam, The Netherlands
| | - Jean-Luc Murk
- Department of Virology, University Medical Center Utrecht, Postbox 85500, 3508 GA Utrecht, The Netherlands
| | - Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Postbox 9892, 1006 AN Amsterdam, The Netherlands
| | - Stephane Heymans
- Department of Cardiology, Maastricht University Medical Center, Postbox 5800, 6202 AZ Maastricht, The Netherlands
| | - Lynda J M Juffermans
- Department of Cardiology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands; Institute for Cardiovascular Research of the Vrije Universiteit (ICaR-VU), VU University Medical Center, De Postbox 7057, 1007 MB Amsterdam, The Netherlands
| | - Albert C van Rossum
- Department of Cardiology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands; Institute for Cardiovascular Research of the Vrije Universiteit (ICaR-VU), VU University Medical Center, De Postbox 7057, 1007 MB Amsterdam, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands; Department of Cardiac Surgery, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands; Institute for Cardiovascular Research of the Vrije Universiteit (ICaR-VU), VU University Medical Center, De Postbox 7057, 1007 MB Amsterdam, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands; Institute for Cardiovascular Research of the Vrije Universiteit (ICaR-VU), VU University Medical Center, De Postbox 7057, 1007 MB Amsterdam, The Netherlands
| | - Reindert W Emmens
- Department of Pathology, VU University Medical Center, Postbox 7057, 1007 MB Amsterdam, The Netherlands; Institute for Cardiovascular Research of the Vrije Universiteit (ICaR-VU), VU University Medical Center, De Postbox 7057, 1007 MB Amsterdam, The Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Postbox 9892, 1006 AN Amsterdam, The Netherlands.
| |
Collapse
|
22
|
|