1
|
Xu G, Sun X, An J, Sun F, Zhang C, Williams JP. Ozone protects from myocardial ischemia-reperfusion injury via inhibition of the NLRP3 inflammasome. Eur J Pharmacol 2025; 997:177631. [PMID: 40246138 DOI: 10.1016/j.ejphar.2025.177631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Ischemic heart disease (IHD) is a leading cause of morbidity and mortality worldwide. Myocardial ischemia/reperfusion injury (MIRI) is the primary cause of myocardial injury triggered by post-myocardial infarction reperfusion therapy. Its pathogenesis involves Ca2+ overload, the production of large amounts of oxygen-free radicals, inflammation, and cell necrosis. Growing evidence suggests that the NLRP3 inflammasome significantly contributes to the sterile inflammatory response and pyroptosis in MIRI, linking damage sensing to the initiation and amplification of the inflammatory response. Reportedly, ozone exerts anti-inflammatory and anti-infection effects by activating the antioxidant system. Additional evidence suggests that ozone inhibits NLRP3 inflammasome expression to relieve ischemic injury. In this study, we aimed to explore whether pretreating the myocardium with ozone protects it from MIRI by inhibiting the NLRP3 inflammasome. Rats were subjected to rectal infusion of ozone for 5 consecutive days, followed by ligation of the left anterior descending coronary artery for 30 min and reperfusion for 120 min to induce MIRI. Experimental results were obtained using echocardiography, triphenyltetrazolium chloride and hematoxylin and eosin staining, western blotting, and enzyme-linked immunosorbent assay. The results showed that ozone significantly improved the diastolic function of the heart, reduced the area of myocardial infarction, and decreased the expression levels of NLRP3, pro-caspase-1, ASC, and the secretion of caspase-1, interleukin (IL)-1β, and IL-18. In summary, these findings reveal that ozone pretreatment can alleviate the damage that occurs during MIRI by inhibiting the NLRP3 Inflammasome.
Collapse
Affiliation(s)
- Guohao Xu
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China; Institute for Lnnovation Diagnosis & Treatment in Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China; Pain and Sleep Medicine Center, Rapid Anti-depression Center, The Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China
| | - Xiaotong Sun
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Jianxiong An
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China; Institute for Lnnovation Diagnosis & Treatment in Anesthesiology, Shandong Second Medical University, Weifang, Shandong Province, China; Pain and Sleep Medicine Center, Rapid Anti-depression Center, The Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China; Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Brain Disease Institute & Department of Anesthesiology for the Integrated Traditional Chinese and Western Medicine Hospital, Southern Medical University, Guangzhou, China.
| | - Fan Sun
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China
| | - Chengming Zhang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifang, Shandong, 261000, China.
| | - John P Williams
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Simats A, Sager HB, Liesz A. Heart-brain axis in health and disease: role of innate and adaptive immunity. Cardiovasc Res 2025; 120:2325-2335. [PMID: 39180327 DOI: 10.1093/cvr/cvae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 08/26/2024] Open
Abstract
The importance of the brain-heart interaction has been increasingly recognized as a critical physiological axis that is altered in disease. In this review, we explore the intricate relationship between the central nervous system and cardiovascular health, focusing particularly on immunological mechanisms that influence the course of both neurological and cardiovascular diseases. While previous studies have established a key role of the autonomic nervous system (ANS) in linking brain and the heart, more recent studies have expanded our understanding of the multifaceted inter-organ interactions. As such, circulating mediators include immune cells of the adaptive and innate immune system and their secreted immunogenic factors have come into the focus as mediators along this bidirectional communication. Hence, in this review we briefly discuss the contribution of the ANS and then focus on innate and adaptive immune mechanisms along the heart-to-brain and brain-to-heart axes, illustrating how cardiovascular diseases affect cognitive functions and how brain pathologies lead to cardiac complications.
Collapse
Affiliation(s)
- Alba Simats
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), c/Rosselló 161, 08036 Barcelona, Spain
| | - Hendrik B Sager
- DZHK (German Center for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Medical Center Munich, Feodor-Lynen-Straße 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| |
Collapse
|
3
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
4
|
Gigli M, Stolfo D, Merlo M, Sinagra G, Taylor MRG, Mestroni L. Pathophysiology of dilated cardiomyopathy: from mechanisms to precision medicine. Nat Rev Cardiol 2025; 22:183-198. [PMID: 39394525 PMCID: PMC12046608 DOI: 10.1038/s41569-024-01074-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/13/2024]
Abstract
Dilated cardiomyopathy (DCM) is a complex disease with multiple causes and various pathogenic mechanisms. Despite improvements in the prognosis of patients with DCM in the past decade, this condition remains a leading cause of heart failure and premature death. Conventional treatment for DCM is based on the foundational therapies for heart failure with reduced ejection fraction. However, increasingly, attention is being directed towards individualized treatments and precision medicine. The ability to confirm genetic causality is gradually being complemented by an increased understanding of genotype-phenotype correlations. Non-genetic factors also influence the onset of DCM, and growing evidence links genetic background with concomitant non-genetic triggers or precipitating factors, increasing the extreme complexity of the pathophysiology of DCM. This Review covers the spectrum of pathophysiological mechanisms in DCM, from monogenic causes to the coexistence of genetic abnormalities and triggering environmental factors (the 'two-hit' hypothesis). The roles of common genetic variants in the general population and of gene modifiers in disease onset and progression are also discussed. Finally, areas for future research are highlighted, particularly novel therapies, such as small molecules, RNA and gene therapy, and measures for the prevention of arrhythmic death.
Collapse
Affiliation(s)
- Marta Gigli
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Davide Stolfo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marco Merlo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luisa Mestroni
- Molecular Genetics Program, Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
5
|
Gillan JL, Jaeschke L, Kuebler WM, Grune J. Immune mediators in heart-lung communication. Pflugers Arch 2025; 477:17-30. [PMID: 39256247 PMCID: PMC11711577 DOI: 10.1007/s00424-024-03013-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
It is often the case that serious, end-stage manifestations of disease result from secondary complications in organs distinct from the initial site of injury or infection. This is particularly true of diseases of the heart-lung axis, given the tight anatomical connections of the two organs within a common cavity in which they collectively orchestrate the two major, intertwined circulatory pathways. Immune cells and the soluble mediators they secrete serve as effective, and targetable, messengers of signals between different regions of the body but can also contribute to the spread of pathology. In this review, we discuss the immunological basis of interorgan communication between the heart and lung in various common diseases, and in the context of organ crosstalk more generally. Gaining a greater understanding of how the heart and lung communicate in health and disease, and viewing disease progression generally from a more holistic, whole-body viewpoint have the potential to inform new diagnostic approaches and strategies for better prevention and treatment of comorbidities.
Collapse
Affiliation(s)
- Jonathan L Gillan
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Lara Jaeschke
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany.
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Ghiasi M. Investigating the NF-κB signaling pathway in heart failure: Exploring potential therapeutic approaches. Heliyon 2024; 10:e40812. [PMID: 39717608 PMCID: PMC11664283 DOI: 10.1016/j.heliyon.2024.e40812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Heart failure (HF) syndrome is of great interest as an emerging epidemic. Due to the increasing elderly population worldwide, the total number of HF patients is increasing every day. This disease places a significant economic burden on the healthcare and treatment systems of developing societies, and this situation is very concerning. Despite many advances in the diagnosis and treatment of cardiovascular diseases, HF is still the main cause of death worldwide. This clinical syndrome has many cellular and molecular complications, which are often aggravated by increased levels of pro-inflammatory cytokines, which lead to adverse clinical outcomes. Nuclear factor kappa B (NF-κB), a pivotal family of transcription factors, plays a crucial role in various biological processes, particularly in inflammation, immune response, cell proliferation, and cell survival. Studies show that the NF-κB signaling pathway plays a role in modulating cardiac regeneration, apoptosis, and myocardial fibrosis. It has been found that the NF-κB signaling pathway can affect heart function and HF through the regulation of matrix metalloproteinases and fibrotic mediators. Also, the NF-κB pathway regulates cell activities in cardiac cardiomyocytes and regulates the function of this organ by establishing a precise interaction between apoptosis and pyroptosis. However, the exact molecular mechanisms of this influence have not been well defined and there are many scientific gaps in this matter. This review tries to highlights potential therapeutic strategies to target NF-κB, including the use of anti-inflammatory agents and genetic modulation, which may provide new ways to reduce cardiac fibrosis and improve outcomes in HF patients. Certainly, increasing understanding of the multifaceted role of NF-κB in HF can lead to innovative treatments aimed at reducing the growing number of patients worldwide.
Collapse
Affiliation(s)
- Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
d'Aiello A, Filomia S, Brecciaroli M, Sanna T, Pedicino D, Liuzzo G. Targeting Inflammatory Pathways in Atherosclerosis: Exploring New Opportunities for Treatment. Curr Atheroscler Rep 2024; 26:707-719. [PMID: 39404934 PMCID: PMC11530513 DOI: 10.1007/s11883-024-01241-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/02/2024]
Abstract
PURPOSE OF THE REVIEW This review discusses the molecular mechanisms involved in the immuno-pathogenesis of atherosclerosis, the pleiotropic anti-inflammatory effects of approved cardiovascular therapies and the available evidence on immunomodulatory therapies for atherosclerotic cardiovascular disease (ACVD). We highlight the importance of clinical and translational research in identifying molecular mechanisms and discovering new therapeutic targets. RECENT FINDINGS The CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcomes Study) trial was the first to demonstrate a reduction in cardiovascular (CV) risk with anti-inflammatory therapy, irrespective of serum lipid levels. ACVD is the leading cause of death worldwide. Although targeting principal risk factors significantly reduces CV risk, residual risk remains unaddressed. The immunological mechanisms underlying atherosclerosis represent attractive therapeutic targets. Several commonly used and non-primarily anti-inflammatory drugs (i.e. SGLT2i, and PCSK9i) exhibit pleiotropic properties. Otherwise, recent trials have investigated the blockade of primarily inflammatory compounds, trying to lower the residual risk via low-dose IL-2, PTPN22 and CD31 pathway modulation. In the era of precision medicine, modern approaches may explore new pharmacological targets, identify new markers of vascular inflammation, and evaluate therapeutic responses.
Collapse
Affiliation(s)
- Alessia d'Aiello
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Simone Filomia
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| | - Mattia Brecciaroli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| | - Tommaso Sanna
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| | - Daniela Pedicino
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy.
| | - Giovanna Liuzzo
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| |
Collapse
|
8
|
Verra C, Paulmann MK, Wegener J, Marzani E, Ferreira Alves G, Collino M, Coldewey SM, Thiemermann C. Spleen tyrosine kinase: a novel pharmacological target for sepsis-induced cardiac dysfunction and multi-organ failure. Front Immunol 2024; 15:1447901. [PMID: 39559354 PMCID: PMC11570271 DOI: 10.3389/fimmu.2024.1447901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Sepsis is a systemic condition caused by a dysregulated host response to infection and often associated with excessive release of proinflammatory cytokines resulting in multi-organ failure (MOF), including cardiac dysfunction. Despite a number of effective supportive treatments (e.g. ventilation, dialysis), there are no specific interventions that prevent or reduce MOF in patients with sepsis. To identify possible intervention targets, we re-analyzed the publicly accessible Gene Expression Omnibus accession GSE131761 dataset, which revealed an increased expression of spleen tyrosine kinase (SYK) in the whole blood of septic patients compared to healthy volunteers. This result suggests a potential involvement of SYK in the pathophysiology of sepsis. Thus, we investigated the effects of the highly selective SYK inhibitor PRT062607 (15mg/kg; i.p.) on sepsis-induced cardiac dysfunction and MOF in a clinically-relevant, murine model of sepsis. PRT062607 or vehicle (saline) was administered to 10-weeks-old C57BL/6 mice at 1h after the onset of sepsis induced by cecal ligation and puncture (CLP). Antibiotics (imipenem/cilastatin; 2mg/kg; s.c.) and analgesic (buprenorphine; 0.05mg/kg; i.p.) were administered at 6h and 18h post-CLP. After 24h, cardiac function was assessed in vivo by echocardiography and, after termination of the experiments, serum and cardiac samples were collected to evaluate the effects of SYK inhibition on the systemic release of inflammatory mediators and the degree of organ injury and dysfunction. Our results show that treatment of CLP-mice with PRT062607 significantly reduces systolic and diastolic cardiac dysfunction, renal dysfunction and liver injury compared to CLP-mice treated with vehicle. In addition, the sepsis-induced systemic inflammation (measured as an increase in inflammatory cytokines and chemokines in the serum) and the cardiac activation of NF-kB (IKK) and the NLRP3 inflammasome were significantly reduced in CLP-mice treated with PRT062607. These results demonstrate, for the first time, that SYK inhibition 1h after the onset of sepsis reduces the systemic inflammation, cardiac dysfunction and MOF, suggesting a potential role of the activation of SYK in the pathophysiology of sepsis. Novel therapeutic strategies that inhibit SYK activity may be of benefit in patients with diseases associated with local or systemic inflammation including sepsis.
Collapse
Affiliation(s)
- Chiara Verra
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Kerstin Paulmann
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Jamila Wegener
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Enrica Marzani
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | | | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Sina Maren Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Christoph Thiemermann
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
9
|
Nayfeh M, DiGregorio H, Saad JM, Al-Mallah M, Al Rifai M. The Emerging Specialty of Cardio-Rheumatology. Curr Atheroscler Rep 2024; 26:499-509. [PMID: 38913292 DOI: 10.1007/s11883-024-01221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 06/25/2024]
Abstract
PURPOSE OF REVIEW In this review, we aimed to summarize the different aspects of the field of cardio-rheumatology, the role of the cardio-rheumatologist, and future research in the field. RECENT FINDINGS Cardio-rheumatology is an emerging subspecialty within cardiology that focuses on addressing the intricate relationship between systemic inflammation and cardiovascular diseases. It involves understanding the cardiovascular impact of immune-mediated inflammatory diseases on the heart and vascular system. A cardio-rheumatologist's role is multifaceted. First, they should understand the cardiac manifestations of rheumatological diseases. They should also be knowledgeable about the different immunotherapies available and side effects. Additionally, they should know how to utilize imaging modalities, either for diagnosis, prognosis, or treatment monitoring. This field is constantly evolving with new research on both treatment and imaging of the effects of inflammation on the cardiovascular system.
Collapse
Affiliation(s)
- Malek Nayfeh
- Houston Methodist Academic Institute, Houston Methodist DeBakey Heart & Vascular Center, 6550 Fannin Street, Smith Tower - Suite 1801, Houston, TX, 77030, USA
| | | | | | - Mouaz Al-Mallah
- Houston Methodist Academic Institute, Houston Methodist DeBakey Heart & Vascular Center, 6550 Fannin Street, Smith Tower - Suite 1801, Houston, TX, 77030, USA
| | - Mahmoud Al Rifai
- Houston Methodist Academic Institute, Houston Methodist DeBakey Heart & Vascular Center, 6550 Fannin Street, Smith Tower - Suite 1801, Houston, TX, 77030, USA.
- Weill Cornell Medicine, Houston Methodist DeBakey Heart and Vascular Center, 6550 Fannin Street, Smith Tower - Suite 1801, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Wang D, Yu X, Gao K, Li F, Li X, Pu H, Zhang P, Guo S, Wang W. Sweroside alleviates pressure overload-induced heart failure through targeting CaMKⅡδ to inhibit ROS-mediated NF-κB/NLRP3 in cardiomyocytes. Redox Biol 2024; 74:103223. [PMID: 38851078 PMCID: PMC11219961 DOI: 10.1016/j.redox.2024.103223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Ongoing inflammation in the heart is positively correlated with adverse remodeling, characterized by elevated levels of cytokines that stimulate activation of cardiac fibroblasts. It was found that CaMKIIδ response to Ang II or TAC triggers the accumulation of ROS in cardiomyocytes, which subsequently stimulates NF-κB/NLRP3 and leads to an increase in IL-6, IL-1β, and IL-18. This is an important causative factor in the occurrence of adverse remodeling in heart failure. Sweroside is a biologically active natural iridoids extracted from Lonicerae Japonicae Flos. It shows potent anti-inflammatory and antioxidant activity in various cardiovascular diseases. In this study, we found that sweroside inhibited ROS-mediated NF-κB/NLRP3 in Ang II-treated cardiomyocytes by directly binding to CaMKIIδ. Knockdown of CaMKⅡδ abrogated the effect of sweroside regulation on NF-κB/NLRP3 in cardiomyocytes. AAV-CaMKⅡδ induced high expression of CaMKⅡδ in the myocardium of TAC/Ang II-mice, and the inhibitory effect of sweroside on TAC/Ang Ⅱ-induced elevation of NF-κB/NLRP3 was impeded. Sweroside showed significant inhibitory effects on CaMKIIδ/NF-κB/NLRP3 in cardiomyocytes from TAC/Ang Ⅱ-induced mice. This would be able to mitigate the adverse events of myocardial remodeling and contractile dysfunction at 8 weeks after the onset of the inflammatory response. Taken together, our findings have revealed the direct protein targets and molecular mechanisms by which sweroside improves heart failure, thereby supporting the further development of sweroside as a therapeutic agent for heart failure.
Collapse
Affiliation(s)
- Dong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Kuo Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Fanghe Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haiyin Pu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Peng Zhang
- Wuhan Hospital of Traditional Chinese Medicine, Wuhan, 430014, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
11
|
Aromolaran KA, Corbin A, Aromolaran AS. Obesity Arrhythmias: Role of IL-6 Trans-Signaling. Int J Mol Sci 2024; 25:8407. [PMID: 39125976 PMCID: PMC11313575 DOI: 10.3390/ijms25158407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Obesity is a chronic disease that is rapidly increasing in prevalence and affects more than 600 million adults worldwide, and this figure is estimated to increase by at least double by 2030. In the United States, more than one-third of the adult population is either overweight or obese. The global obesity epidemic is a major risk factor for the development of life-threatening arrhythmias occurring in patients with long QT, particularly in conditions where multiple heart-rate-corrected QT-interval-prolonging mechanisms are simultaneously present. In obesity, excess dietary fat in adipose tissue stimulates the release of immunomodulatory cytokines such as interleukin (IL)-6, leading to a state of chronic inflammation in patients. Over the last decade, increasing evidence has been found to support IL-6 signaling as a powerful predictor of the severity of heart diseases and increased risk for ventricular arrhythmias. IL-6's pro-inflammatory effects are mediated via trans-signaling and may represent a novel arrhythmogenic risk factor in obese hearts. The first selective inhibitor of IL-6 trans-signaling, olamkicept, has shown encouraging results in phase II clinical studies for inflammatory bowel disease. Nevertheless, the connection between IL-6 trans-signaling and obesity-linked ventricular arrhythmias remains unexplored. Therefore, understanding how IL-6 trans-signaling elicits a cellular pro-arrhythmic phenotype and its use as an anti-arrhythmic target in a model of obesity remain unmet clinical needs.
Collapse
Affiliation(s)
- Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
| | - Andrea Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (K.A.A.); (A.C.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
12
|
Corbin A, Aromolaran KA, Aromolaran AS. STAT4 Mediates IL-6 Trans-Signaling Arrhythmias in High Fat Diet Guinea Pig Heart. Int J Mol Sci 2024; 25:7813. [PMID: 39063055 PMCID: PMC11277091 DOI: 10.3390/ijms25147813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is a major risk factor for the development of life-threatening malignant ventricular tachyarrhythmias (VT) and sudden cardiac death (SCD). Risks may be highest for patients with high levels of the proinflammatory cytokine interleukin (IL)-6. We used our guinea pig model of high-fat diet (HFD)-induced arrhythmias that exhibit a heightened proinflammatory-like pathology, which is also observed in human obesity arrhythmias, as well as immunofluorescence and confocal microscopy approaches to evaluate the pathological IL-6 trans-signaling function and explore the underlying mechanisms. Using blind-stick and electrocardiogram (ECG) techniques, we tested the hypothesis that heightened IL-6 trans-signaling would exhibit increased ventricular arrhythmia/SCD incidence and underlying arrhythmia substrates. Remarkably, compared to low-fat diet (LFD)-fed controls, HFD promoted phosphorylation of the IL-6 signal transducer and activator of transcription 4 (STAT4), leading to its activation and enhanced nuclear translocation of pSTAT4/STAT4 compared to LFD controls and pSTAT3/STAT3 nuclear expression. Overactivation of IL-6 trans-signaling in guinea pigs prolonged the QT interval, which resulted in greater susceptibility to arrhythmias/SCD with isoproterenol challenge, as also observed with the downstream Janus kinase (JAK) 2 activator. These findings may have potentially profound implications for more effective arrhythmia therapy in the vulnerable obese patient population.
Collapse
Affiliation(s)
- Andrea Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (A.C.); (K.A.A.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (A.C.); (K.A.A.)
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (A.C.); (K.A.A.)
- Department of Biomedical Engineering, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
13
|
Jiang L, Lunding LP, Webber WS, Beckmann K, Azam T, Falkesgaard Højen J, Amo-Aparicio J, Dinarello A, Nguyen TT, Pessara U, Parera D, Orlicky DJ, Fischer S, Wegmann M, Dinarello CA, Li S. An antibody to IL-1 receptor 7 protects mice from LPS-induced tissue and systemic inflammation. Front Immunol 2024; 15:1427100. [PMID: 38983847 PMCID: PMC11231367 DOI: 10.3389/fimmu.2024.1427100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
Introduction Interleukin-18 (IL-18), a pro-inflammatory cytokine belonging to the IL-1 Family, is a key mediator ofautoinflammatory diseases associated with the development of macrophage activation syndrome (MAS).High levels of IL-18 correlate with MAS and COVID-19 severity and mortality, particularly in COVID-19patients with MAS. As an inflammation inducer, IL-18 binds its receptor IL-1 Receptor 5 (IL-1R5), leadingto the recruitment of the co-receptor, IL-1 Receptor 7 (IL-1R7). This heterotrimeric complex subsequentlyinitiates downstream signaling, resulting in local and systemic inflammation. Methods We reported earlier the development of a novel humanized monoclonal anti-human IL-1R7 antibody whichspecifically blocks the activity of human IL-18 and its inflammatory signaling in human cell and wholeblood cultures. In the current study, we further explored the strategy of blocking IL-1R7 inhyperinflammation in vivo using animal models. Results We first identified an anti-mouse IL-1R7 antibody that significantly suppressed mouse IL-18 andlipopolysaccharide (LPS)-induced IFNg production in mouse splenocyte and peritoneal cell cultures. Whenapplied in vivo, the antibody reduced Propionibacterium acnes and LPS-induced liver injury and protectedmice from tissue and systemic hyperinflammation. Importantly, anti-IL-1R7 significantly inhibited plasma,liver cell and spleen cell IFNg production. Also, anti-IL-1R7 downregulated plasma TNFa, IL-6, IL-1b,MIP-2 production and the production of the liver enzyme ALT. In parallel, anti-IL-1R7 suppressed LPSinducedinflammatory cell infiltration in lungs and inhibited the subsequent IFNg production andinflammation in mice when assessed using an acute lung injury model. Discussion Altogether, our data suggest that blocking IL-1R7 represents a potential therapeutic strategy to specificallymodulate IL-18-mediated hyperinflammation, warranting further investigation of its clinical application intreating IL-18-mediated diseases, including MAS and COVID-19.
Collapse
Affiliation(s)
- Liqiong Jiang
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Lars P. Lunding
- Division of Lung Immunology, Priority Area of Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Airway Research Center North, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - William S. Webber
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | | | - Tania Azam
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Jesper Falkesgaard Højen
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Jesus Amo-Aparicio
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Alberto Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Tom T. Nguyen
- Mucosal Inflammation Program and Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado, Aurora, CO, United States
| | - Ulrich Pessara
- MAB Discovery GmbH, Polling, Germany
- IcanoMAB GmbH, Polling, Germany
| | - Daniel Parera
- MAB Discovery GmbH, Polling, Germany
- IcanoMAB GmbH, Polling, Germany
| | - David J. Orlicky
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Stephan Fischer
- MAB Discovery GmbH, Polling, Germany
- IcanoMAB GmbH, Polling, Germany
| | - Michael Wegmann
- Division of Lung Immunology, Priority Area of Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Airway Research Center North, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Suzhao Li
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
14
|
Golino M, Harding D, Del Buono MG, Fanti S, Mohiddin S, Toldo S, Smyth J, Sanna T, Marelli-Berg F, Abbate A. Innate and adaptive immunity in acute myocarditis. Int J Cardiol 2024; 404:131901. [PMID: 38403204 PMCID: PMC11450758 DOI: 10.1016/j.ijcard.2024.131901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
Acute myocarditis is an acute inflammatory cardiomyopathy associated with cardiac damage triggered by a virus or a pathological immune activation. It may present with a wide range of clinical presentations, ranging from mild symptoms to severe forms like fulminant myocarditis, characterized by hemodynamic compromise and cardiogenic shock. The immune system plays a central role in the pathogenesis of myocarditis. In fact, while its function is primarily protective, aberrant responses can be detrimental. In this context, both innate and adaptive immunity play pivotal roles; notably, the innate system offers a non-specific and immediate defense, while the adaptive provides specialized protection with immunological memory. However, dysregulation in these systems can misidentify cardiac tissue, triggering autoimmune reactions and possibly leading to significant cardiac tissue damage. This review highlights the importance of innate and adaptive immune responses in the progression and treatment of acute myocarditis.
Collapse
Affiliation(s)
- Michele Golino
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America; Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Daniel Harding
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Saidi Mohiddin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom; Barts Heart Centre, London, United Kingdom
| | - Stefano Toldo
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America
| | - James Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, United States of America; Virginia Tech Carilion School of Medicine, Roanoke, VA, United States of America; Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, United States of America
| | - Tommaso Sanna
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom.
| | - Antonio Abbate
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America.
| |
Collapse
|
15
|
Qin J, Yang Q, Wang Y, Shi M, Zhao X, Zhou Y. The role of pyroptosis in heart failure and related traditional chinese medicine treatments. Front Pharmacol 2024; 15:1377359. [PMID: 38868667 PMCID: PMC11168204 DOI: 10.3389/fphar.2024.1377359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/09/2024] [Indexed: 06/14/2024] Open
Abstract
Pyroptosis is a type of programmed cell death that is mediated by both typical and atypical pathways and ultimately leads to the lysis and rupture of cell membranes and the release of proinflammatory factors, triggering an intense inflammatory response. Heart failure (HF) is a serious and terminal stage of various heart diseases. Myocardial hypertrophy, myocardial fibrosis, ventricular remodeling, oxidative stress, the inflammatory response and cardiomyocyte ionic disorders caused by various cardiac diseases are all risk factors for and aggravate HF. Numerous studies have shown that pyroptosis can induce and exacerbate these reactions, causing progression to HF. Therefore, targeting pyroptosis is a promising strategy to treat HF. This paper summarizes the role of pyroptosis in the development of HF and the underlying mechanism involved. Recent research progress on the ability of traditional Chinese medicine (TCM) extracts and formulas to inhibit pyroptosis and treat HF was summarized, and some traditional Chinese medicine extracts and formulas can alleviate different types of HF, including heart failure with preserved ejection fraction (HFpEF), heart failure with reduced ejection fraction (HFrEF), and heart failure with midrange ejection fraction (HFmrEF), by targeting pyroptosis. These findings may provide new ideas and evidence for the treatment or adjuvant treatment of HF by targeting pyroptosis.
Collapse
Affiliation(s)
- Jie Qin
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Qianhe Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yan Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Mengdi Shi
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xin Zhao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yabin Zhou
- Department of Cardiovascular Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
16
|
Toldo S, Abbate A. The role of the NLRP3 inflammasome and pyroptosis in cardiovascular diseases. Nat Rev Cardiol 2024; 21:219-237. [PMID: 37923829 PMCID: PMC11550901 DOI: 10.1038/s41569-023-00946-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2023] [Indexed: 11/06/2023]
Abstract
An intense, stereotyped inflammatory response occurs in response to ischaemic and non-ischaemic injury to the myocardium. The NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is a finely regulated macromolecular protein complex that senses the injury and triggers and amplifies the inflammatory response by activation of caspase 1; cleavage of pro-inflammatory cytokines, such as pro-IL-1β and pro-IL-18, to their mature forms; and induction of inflammatory cell death (pyroptosis). Inhibitors of the NLRP3 inflammasome and blockers of IL-1β and IL-18 activity have been shown to reduce injury to the myocardium and pericardium, favour resolution of the inflammation and preserve cardiac function. In this Review, we discuss the components of the NLRP3 inflammasome and how it is formed and activated in various ischaemic and non-ischaemic cardiac pathologies (acute myocardial infarction, cardiac dysfunction and remodelling, atherothrombosis, myocarditis and pericarditis, cardiotoxicity and cardiac sarcoidosis). We also summarize current preclinical and clinical evidence from studies of agents that target the NLRP3 inflammasome and related cytokines.
Collapse
Affiliation(s)
- Stefano Toldo
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
17
|
Chisavu L, Ivan VM, Mihaescu A, Chisavu F, Schiller O, Marc L, Bob F, Schiller A. Novel Biomarkers in Evaluating Cardiac Function in Patients on Hemodialysis-A Pilot Prospective Observational Cohort Study. Diagnostics (Basel) 2024; 14:664. [PMID: 38535084 PMCID: PMC10969652 DOI: 10.3390/diagnostics14060664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 01/03/2025] Open
Abstract
Chronic kidney disease patients treated by hemodialysis present a high cardiovascular morbidity and mortality. There is an imperative need for novel biomarkers for identifying these patients and to offer possible therapeutically interventions. We performed a prospective observational cohort study on 77 patients in the period of October 2021-October 2023. We measured serum plasma levels of interleukin 1-beta, galectin 3, human suppression of tumorigenicity factor 2, bone morphogenetic protein 2 and fibroblastic growth factor 23 at the inclusion site. We evaluated the correlations of these biomarkers with cardiac function and structure evaluated by echocardiography. The mean age was 61.02 (±11.81) years, with 45 (56.2%) males and with a dialysis vintage of 4.95 (2.4-7.8) years. Median ejection fraction was 51 (43-54%), and more than two-thirds of the patients presented valvular calcifications. Overall mortality was 22%. Interleukin 1-beta was correlated positively with ejection fraction and global longitudinal strain and negatively with left atrium diameter and left ventricle telesystolic diameter. Galectin 3 values were negatively correlated with aortic valve fibrosis and mitral valve calcifications, and human suppression tumorigenicity factor 2 was negatively correlated with mitral valve calcifications. Some of these novel biomarkers could be used to better assess cardiovascular disease in patients on maintenance hemodialysis.
Collapse
Affiliation(s)
- Lazar Chisavu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Viviana Mihaela Ivan
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Cardiology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adelina Mihaescu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Flavia Chisavu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Louis Turcanu’ Emergency County Hospital for Children, 300011 Timisoara, Romania
| | | | - Luciana Marc
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Flaviu Bob
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adalbert Schiller
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine ‘Victor Babes’, 300041 Timisoara, Romania; (L.C.); (V.M.I.); (F.C.); (L.M.); (F.B.); (A.S.)
- Department of Internal Medicine II–Division of Nephrology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
18
|
Chan MJ, Liu KD. Acute Kidney Injury and Subsequent Cardiovascular Disease: Epidemiology, Pathophysiology, and Treatment. Semin Nephrol 2024; 44:151515. [PMID: 38849258 DOI: 10.1016/j.semnephrol.2024.151515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Cardiovascular disease poses a significant threat to individuals with kidney disease, including those affected by acute kidney injury (AKI). In the short term, AKI has several physiological consequences that can impact the cardiovascular system. These include fluid and sodium overload, activation of the renin-angiotensin-aldosterone system and sympathetic nervous system, and inflammation along with metabolic complications of AKI (acidosis, electrolyte imbalance, buildup of uremic toxins). Recent studies highlight the role of AKI in elevating long-term risks of hypertension, thromboembolism, stroke, and major adverse cardiovascular events, though some of this increased risk may be due to the impact of AKI on the course of chronic kidney disease. Current management strategies involve avoiding nephrotoxic agents, optimizing hemodynamics and fluid balance, and considering renin-angiotensin-aldosterone system inhibition or sodium-glucose cotransporter 2 inhibitors. However, future research is imperative to advance preventive and therapeutic strategies for cardiovascular complications in AKI. This review explores the existing knowledge on the cardiovascular consequences of AKI, delving into epidemiology, pathophysiology, and treatment of various cardiovascular complications following AKI.
Collapse
Affiliation(s)
- Ming-Jen Chan
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kathleen D Liu
- Divisions of Nephrology and Critical Care Medicine, Departments of Medicine and Anesthesia, University of California, San Francisco, CA.
| |
Collapse
|
19
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
20
|
Accogli T, Hibos C, Vegran F. Canonical and non-canonical functions of NLRP3. J Adv Res 2023; 53:137-151. [PMID: 36610670 PMCID: PMC10658328 DOI: 10.1016/j.jare.2023.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Since its discovery, NLRP3 is almost never separated from its major role in the protein complex it forms with ASC, NEK7 and Caspase-1, the inflammasome. This key component of the innate immune response mediates the secretion of proinflammatory cytokines IL-1β and IL-18 involved in immune response to microbial infection and cellular damage. However, NLRP3 has also other functions that do not involve the inflammasome assembly nor the innate immune response. These non-canonical functions have been poorly studied. Nevertheless, NLRP3 is associated with different kind of diseases probably through its inflammasome dependent function as through its inflammasome independent functions. AIM OF THE REVIEW The study and understanding of the canonical and non-canonical functions of NLRP3 can help to better understand its involvement in various pathologies. In parallel, the description of the mechanisms of action and regulation of its various functions, can allow the identification of new therapeutic strategies. KEY SCIENTIFIC CONCEPTS OF THE REVIEW NLRP3 functions have mainly been studied in the context of the inflammasome, in myeloid cells and in totally deficient transgenic mice. However, for several year, the work of different teams has proven that NLRP3 is also expressed in other cell types where it has functions that are independent of the inflammasome. If these studies suggest that NLRP3 could play different roles in the cytoplasm or the nucleus of the cells, the mechanisms underlying NLRP3 non-canonical functions remain unclear. This is why we propose in this review an inventory of the canonical and non-canonical functions of NLRP3 and their impact in different pathologies.
Collapse
Affiliation(s)
- Théo Accogli
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE
| | - Christophe Hibos
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE; Université de Bourgogne Franche-Comté, Dijon 21000, FRANCE
| | - Frédérique Vegran
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE; Department of Biology and Pathology of Tumors - Centre anticancéreux GF Leclerc, Dijon 21000, FRANCE.
| |
Collapse
|
21
|
Verra C, Mohammad S, Alves GF, Porchietto E, Coldewey SM, Collino M, Thiemermann C. Baricitinib protects mice from sepsis-induced cardiac dysfunction and multiple-organ failure. Front Immunol 2023; 14:1223014. [PMID: 37781388 PMCID: PMC10536262 DOI: 10.3389/fimmu.2023.1223014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/07/2023] [Indexed: 10/03/2023] Open
Abstract
Sepsis is one of the major complications of surgery resulting in high morbidity and mortality, but there are no specific therapies for sepsis-induced organ dysfunction. Data obtained under Gene Expression Omnibus accession GSE131761 were re-analyzed and showed an increased gene expression of Janus Kinase 2 (JAK2) and Signal Transducer and Activator of Transcription 3 (STAT3) in the whole blood of post-operative septic patients. Based on these results, we hypothesized that JAK/STAT activation may contribute to the pathophysiology of septic shock and, hence, investigated the effects of baricitinib (JAK1/JAK2 inhibitor) on sepsis-induced cardiac dysfunction and multiple-organ failure (MOF). In a mouse model of post-trauma sepsis induced by midline laparotomy and cecal ligation and puncture (CLP), 10-week-old male (n=32) and female (n=32) C57BL/6 mice received baricitinib (1mg/kg; i.p.) or vehicle at 1h or 3h post-surgery. Cardiac function was assessed at 24h post-CLP by echocardiography in vivo, and the degree of MOF was analyzed by determination of biomarkers in the serum. The potential mechanism underlying both the cardiac dysfunction and the effect of baricitinib was analyzed by western blot analysis in the heart. Trauma and subsequent sepsis significantly depressed the cardiac function and induced multiple-organ failure, associated with an increase in the activation of JAK2/STAT3, NLRP3 inflammasome and NF- κβ pathways in the heart of both male and female animals. These pathways were inhibited by the administration of baricitinib post the onset of sepsis. Moreover, treatment with baricitinib at 1h or 3h post-CLP protected mice from sepsis-induced cardiac injury and multiple-organ failure. Thus, baricitinib may be repurposed for trauma-associated sepsis.
Collapse
Affiliation(s)
- Chiara Verra
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Shireen Mohammad
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Elisa Porchietto
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Sina Maren Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
22
|
Fujimura K, Karasawa T, Komada T, Yamada N, Mizushina Y, Baatarjav C, Matsumura T, Otsu K, Takeda N, Mizukami H, Kario K, Takahashi M. NLRP3 inflammasome-driven IL-1β and IL-18 contribute to lipopolysaccharide-induced septic cardiomyopathy. J Mol Cell Cardiol 2023; 180:58-68. [PMID: 37172930 DOI: 10.1016/j.yjmcc.2023.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
Sepsis is a life-threatening syndrome, and its associated mortality is increased when cardiac dysfunction and damage (septic cardiomyopathy [SCM]) occur. Although inflammation is involved in the pathophysiology of SCM, the mechanism of how inflammation induces SCM in vivo has remained obscure. NLRP3 inflammasome is a critical component of the innate immune system that activates caspase-1 (Casp1) and causes the maturation of IL-1β and IL-18 as well as the processing of gasdermin D (GSDMD). Here, we investigated the role of the NLRP3 inflammasome in a murine model of lipopolysaccharide (LPS)-induced SCM. LPS injection induced cardiac dysfunction, damage, and lethality, which was significantly prevented in NLRP3-/- mice, compared to wild-type (WT) mice. LPS injection upregulated mRNA levels of inflammatory cytokines (Il6, Tnfa, and Ifng) in the heart, liver, and spleen of WT mice, and this upregulation was prevented in NLRP3-/- mice. LPS injection increased plasma levels of inflammatory cytokines (IL-1β, IL-18, and TNF-α) in WT mice, and this increase was markedly inhibited in NLRP3-/- mice. LPS-induced SCM was also prevented in Casp1/11-/- mice, but not in Casp11mt, IL-1β-/-, IL-1α-/-, or GSDMD-/- mice. Notably, LPS-induced SCM was apparently prevented in IL-1β-/- mice transduced with adeno-associated virus vector expressing IL-18 binding protein (IL-18BP). Furthermore, splenectomy, irradiation, or macrophage depletion alleviated LPS-induced SCM. Our findings demonstrate that the cross-regulation of NLRP3 inflammasome-driven IL-1β and IL-18 contributes to the pathophysiology of SCM and provide new insights into the mechanism underlying the pathogenesis of SCM.
Collapse
Affiliation(s)
- Kenta Fujimura
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tadayoshi Karasawa
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Takanori Komada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Naoya Yamada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Yoshiko Mizushina
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Chintogtokh Baatarjav
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Takayoshi Matsumura
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Kinya Otsu
- The School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom; National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
23
|
Wang Y, Li Y, Zhang W, Yuan Z, Lv S, Zhang J. NLRP3 Inflammasome: a Novel Insight into Heart Failure. J Cardiovasc Transl Res 2023; 16:166-176. [PMID: 35697978 DOI: 10.1007/s12265-022-10286-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
Among numerous cardiovascular diseases, heart failure is a final and fatal stage, and its morbidity, mortality, and rehospitalization rate remain high, which reduces the exercise tolerance of patients and brings great medical burden and economic pressure to the society. Inflammation takes on a major influence in the occurrence, development, and prognosis of heart failure (HF). The NLRP3 inflammasome is a key node in a chronic inflammatory response, which can accelerate the production of pro-inflammatory cytokines IL-1β and IL-18, leading to the inflammatory response. Therefore, whether it is possible to suppress the downstream factors of NLRP3 inflammasome and its signaling path is expected to provide a new intervention mediator for the therapy of heart failure. This article synopsizes the research progress of NLRP3 inflammasome in heart failure, to provide a reference for clinical treatment. CLINICAL RELEVANCE: This study explored the downstream factors of NLRP3 inflammasome and its signal pathway. Targeted drug therapy for NLRP3 inflammasome is expected to provide a new intervention target for the treatment of heart failure.
Collapse
Affiliation(s)
- Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yanyang Li
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wanqin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Zhuo Yuan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
24
|
Liu H, Huang Y, Zhao Y, Kang GJ, Feng F, Wang X, Liu M, Shi G, Revelo X, Bernlohr D, Dudley SC. Inflammatory Macrophage Interleukin-1β Mediates High-Fat Diet-Induced Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2023; 8:174-185. [PMID: 36908663 PMCID: PMC9998610 DOI: 10.1016/j.jacbts.2022.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/02/2022]
Abstract
Diabetes mellitus (DM) is a main risk factor for diastolic dysfunction (DD) and heart failure with preserved ejection fraction. High-fat diet (HFD) mice presented with diabetes mellitus, DD, higher cardiac interleukin (IL)-1β levels, and proinflammatory cardiac macrophage accumulation. DD was significantly ameliorated by suppressing IL-1β signaling or depleting macrophages. Mice with macrophages unable to adopt a proinflammatory phenotype were low in cardiac IL-1β levels and were resistant to HFD-induced DD. IL-1β enhanced mitochondrial reactive oxygen species (mitoROS) in cardiomyocytes, and scavenging mitoROS improved HFD-induced DD. In conclusion, macrophage-mediated inflammation contributed to HFD-associated DD through IL-1β and mitoROS production.
Collapse
Key Words
- CCR2, C-C motif chemokine receptor 2
- CM, cardiomyocyte
- DD, diastolic dysfunction
- DM, diabetes mellitus
- EF, ejection fraction
- FABP4, fatty acid binding protein 4
- HF, heart failure
- HFD, high-fat diet
- HFpEF
- HFpEF, heart failure with preserved ejection fraction
- IL, interleukin
- IL-1β
- IL1RA, interleukin 1 receptor antagonist
- KO, knockout
- MCP, monocyte chemoattractant protein
- MyBP-C, myosin binding protein C
- TGF, transforming growth factor
- TNF, tumor necrosis factor
- Timd4, T cell immunoglobulin and mucin domain containing 4
- WT, wild-type
- diabetes
- diastolic dysfunction
- inflammation
- macrophage
- mitoROS, mitochondrial reactive oxygen species
- mitochondria
Collapse
Affiliation(s)
- Hong Liu
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yimao Huang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yang Zhao
- Division of Cardiology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu, China
| | - Gyeoung-Jin Kang
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Feng Feng
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xiaodan Wang
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Man Liu
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guangbin Shi
- Division of Cardiology, Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Xavier Revelo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - David Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Samuel C. Dudley
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
25
|
Zhang MQ, Wang CC, Pang XB, Shi JZ, Li HR, Xie XM, Wang Z, Zhang HD, Zhou YF, Chen JW, Han ZY, Zhao LL, He YY. Role of macrophages in pulmonary arterial hypertension. Front Immunol 2023; 14:1152881. [PMID: 37153557 PMCID: PMC10154553 DOI: 10.3389/fimmu.2023.1152881] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 03/27/2023] [Indexed: 05/09/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe cardiopulmonary vascular disease characterized by progressive pulmonary artery pressure elevation, increased pulmonary vascular resistance and ultimately right heart failure. Studies have demonstrated the involvement of multiple immune cells in the development of PAH in patients with PAH and in experimental PAH. Among them, macrophages, as the predominant inflammatory cells infiltrating around PAH lesions, play a crucial role in exacerbating pulmonary vascular remodeling in PAH. Macrophages are generally polarized into (classic) M1 and (alternative) M2 phenotypes, they accelerate the process of PAH by secreting various chemokines and growth factors (CX3CR1, PDGF). In this review we summarize the mechanisms of immune cell action in PAH, as well as the key factors that regulate the polarization of macrophages in different directions and their functional changes after polarization. We also summarize the effects of different microenvironments on macrophages in PAH. The insight into the interactions between macrophages and other cells, chemokines and growth factors may provide important clues for the development of new, safe and effective immune-targeted therapies for PAH.
Collapse
Affiliation(s)
- Meng-Qi Zhang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Chen-Chen Wang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Xiao-Bin Pang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Jun-Zhuo Shi
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Hao-Ran Li
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Xin-Mei Xie
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Zhe Wang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Hong-Da Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun-Feng Zhou
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Ji-Wang Chen
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Zhi-Yan Han
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yang-Yang He, ; Lu-Ling Zhao, ; Zhi-Yan Han,
| | - Lu-Ling Zhao
- School of Pharmacy, Henan University, Kaifeng, Henan, China
- *Correspondence: Yang-Yang He, ; Lu-Ling Zhao, ; Zhi-Yan Han,
| | - Yang-Yang He
- School of Pharmacy, Henan University, Kaifeng, Henan, China
- *Correspondence: Yang-Yang He, ; Lu-Ling Zhao, ; Zhi-Yan Han,
| |
Collapse
|
26
|
Bhattachar S, Chawla A, Sikri G, Patrikar S. Body fat content correlates with maximum aerobic capacity in healthy sedentary Indian males. Med J Armed Forces India 2023; 79:93-100. [PMID: 36605353 PMCID: PMC9807674 DOI: 10.1016/j.mjafi.2021.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/21/2021] [Indexed: 01/09/2023] Open
Abstract
Background Obesity is a multifactorial public health problem with varying effects on physical fitness determined by maximum aerobic capacity or VO2max. The relationship between body fat content and VO2max has shown varying results. The present study was planned as an experimental study to evaluate the relationship between body fat content and maximum aerobic capacity. Methods 104 healthy Indian males [Age: 21 (4.87)years; Height: 171.4 (6.14)cm; Weight: 64.1 (8.57)kg] were evaluated for body fat content using body mass index (BMI), bioimpedance, skinfold thickness (SFT), body girth (BG) measurements, waist circumference (WC), and waist-hip ratio (WHR). Maximum aerobic capacity or VO2max for all subjects was determined indirectly from maximum heart rate achieved using an incremental treadmill protocol using Astrand and Astrand nomogram. Results VO2max, when expressed in L/min, showed a statistically significant positive correlation with body fat irrespective of the method of estimation. VO2max, when expressed in ml/kg/min, showed negative correlation with five of the seven clinical parameters of fat estimation. Of these, a statistically significant negative correlation was seen with SFT. Conclusion VO2max (L/min) shows a significant positive correlation with all methods of body fat estimation. VO2max (ml/kg/min) shows a significant negative correlation with skinfold thickness. Monitoring of body fat content using skinfold thickness could be studied further for its use in the early identification of young, healthy adult Indian males with low aerobic fitness.
Collapse
Affiliation(s)
- Srinivasa Bhattachar
- Assistant Professor, Department of Physiology, Armed Forces Medical College, Pune, India
| | - Anuj Chawla
- Head, Department of Physiology, Army College of Medical Sciences, Delhi Cantt, India
| | - Gaurav Sikri
- Professor & Head, Department of Physiology, Armed Forces Medical College, Pune, India
| | - Seema Patrikar
- Statistician, Department of Community Medicine, Armed Forces Medical College, Pune, India
| |
Collapse
|
27
|
Zhang X, Qu H, Yang T, Liu Q, Zhou H. Astragaloside IV attenuate MI-induced myocardial fibrosis and cardiac remodeling by inhibiting ROS/caspase-1/GSDMD signaling pathway. Cell Cycle 2022; 21:2309-2322. [PMID: 35770948 DOI: 10.1080/15384101.2022.2093598] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Astragalus membranaceus is a traditional Chinese medicine and has been widely used in treating cardiovascular diseases (CVDs), such as asthma, edema, and chest tightness. Astragaloside IV (AS-IV), one of the major active components extracted from Astragalus membranaceus, has a series of pharmacological effects, including inhibiting inflammation, regulating energy metabolism, reducing oxidative stress and apoptosis. However, the effect of AS-IV on myocardial infarction (MI) and the underlying molecular mechanism remains unclear. The purpose of our study is to investigate the effects of AS-IV on MI-induced myocardial fibrosis and cardiac remodeling and to elucidate its underlying mechanisms. MI was induced by ligation of the left anterior descending (LAD) coronary artery. Echocardiography was used to evaluate cardiac function in mice. Pathological changes in cardiac tissues were analyzed with hematoxylin and eosin (H&E) staining, Masson staining, and wheat germ agglutinin (WGA) staining. Immunohistochemistry was used to detect the expression of fibrosis and inflammation-related proteins. Immunofluorescence and flow cytometry were used to detect ROS level. The expressions of α-SMA, Collagen I, NLRP3, cleaved cas-1, cleaved IL-18, cleaved IL-β, GSDMD-N, and cleaved caspase-1 were examined using western blot. The results of cardiac ultrasound showed that AS-IV could improve poor ventricular remodeling, myocardial pathological staining showed that AS-IV could significantly reduce the myocardial fibrosis and myocardial hypertrophy, ROS levels were also significantly reduced, and the protein expression of NLRP3/Caspase-1/GSDMD signaling pathway was remarkably decreased in the AS-IV group. Furthermore, immunohistochemical staining results showed that the expression of myocardial macrophages and neutrophils in AS-IV group decreased significantly, to further investigate whether the reduction of myocardial pyroptosis by AS-IV is related to the regulation of macrophages, in vitro, AS-IV was selected to stimulate bone marrow-derived macrophages (BMDMs). Our findings indicated that AS-IV protective effect of the heart might be related to the reduction of macrophage pyroptosis. These results demonstrate that AS-IV alleviated MI-induced myocardial fibrosis and cardiac remodeling by suppressing ROS/Caspase-1/GSDMD signaling pathway, AS-IV should be further studied in the future.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyan Qu
- Department of Cardiovascular Disease, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Yang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Disease, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
28
|
Yang W, Guo R, Pi A, Ding Q, Hao L, Song Q, Chen L, Dou X, Na L, Li S. Long non-coding RNA-EN_181 potentially contributes to the protective effects of N-acetylcysteine against non-alcoholic fatty liver disease in mice. Br J Nutr 2022; 129:1-15. [PMID: 35710106 DOI: 10.1017/s0007114522001829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
N-acetylcysteine (NAC) possesses a strong capability to ameliorate high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) in mice, but the underlying mechanism is still unknown. Our study aimed to clarify the involvement of long non-coding RNA (lncRNA) in the beneficial effects of NAC on HFD-induced NAFLD. C57BL/6J mice were fed a normal-fat diet (10 % fat), a HFD (45 % fat) or a HFD plus NAC (2 g/l). After 14-week of intervention, NAC rescued the deleterious alterations induced by HFD, including the changes in body and liver weights, hepatic TAG, plasma alanine aminotransferase, plasma aspartate transaminase and liver histomorphology (haematoxylin and eosin and Oil red O staining). Through whole-transcriptome sequencing, 52 167 (50 758 known and 1409 novel) hepatic lncRNA were detected. Our cross-comparison data revealed the expression of 175 lncRNA was changed by HFD but reversed by NAC. Five of those lncRNA, lncRNA-NONMMUT148902·1 (NO_902·1), lncRNA-XR_001781798·1 (XR_798·1), lncRNA-NONMMUT141720·1 (NO_720·1), lncRNA-XR_869907·1 (XR_907·1), and lncRNA-ENSMUST00000132181 (EN_181), were selected based on an absolute log2 fold change value of greater than 4, P-value < 0·01 and P-adjusted value < 0·01. Further qRT-PCR analysis showed the levels of lncRNA-NO_902·1, lncRNA-XR_798·1, and lncRNA-EN_181 were decreased by HFD but restored by NAC, consistent with the RNA sequencing. Finally, we constructed a ceRNA network containing lncRNA-EN_181, 3 miRNA, and 13 mRNA, which was associated with the NAC-ameliorated NAFLD. Overall, lncRNA-EN_181 might be a potential target in NAC-ameliorated NAFLD. This finding enhanced our understanding of the biological mechanisms underlying the beneficial role of NAC.
Collapse
Affiliation(s)
- Wenwen Yang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| | - Rui Guo
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Institute of Nutrition and Health, School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| | - Aiwen Pi
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| | - Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| | - Liuyi Hao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Institute of Nutrition and Health, School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| | - Qing Song
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| | - Lin Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Institute of Nutrition and Health, School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| | - Lixin Na
- Public Health College, Shanghai University of Medicine & Health Sciences, Shanghai201318, People's Republic of China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Institute of Nutrition and Health, School of Public Health, Zhejiang Chinese Medical University, Hangzhou310053, People's Republic of China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang310053, People's Republic of China
| |
Collapse
|
29
|
Van Tassell B, Mihalick V, Thomas G, Marawan A, Talasaz AH, Lu J, Kang L, Ladd A, Damonte JI, Dixon DL, Markley R, Turlington J, Federmann E, Del Buono MG, Biondi-Zoccai G, Canada JM, Arena R, Abbate A. Rationale and design of interleukin-1 blockade in recently decompensated heart failure (REDHART2): a randomized, double blind, placebo controlled, single center, phase 2 study. J Transl Med 2022; 20:270. [PMID: 35706006 PMCID: PMC9198622 DOI: 10.1186/s12967-022-03466-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/30/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Heart failure (HF) is a global leading cause of mortality despite implementation of guideline directed therapy which warrants a need for novel treatment strategies. Proof-of-concept clinical trials of anakinra, a recombinant human Interleukin-1 (IL-1) receptor antagonist, have shown promising results in patients with HF. METHOD We designed a single center, randomized, placebo controlled, double-blind phase II randomized clinical trial. One hundred and two adult patients hospitalized within 2 weeks of discharge due to acute decompensated HF with reduced ejection fraction (HFrEF) and systemic inflammation (high sensitivity of C-reactive protein > 2 mg/L) will be randomized in 2:1 ratio to receive anakinra or placebo for 24 weeks. The primary objective is to determine the effect of anakinra on peak oxygen consumption (VO2) measured at cardiopulmonary exercise testing (CPX) after 24 weeks of treatment, with placebo-corrected changes in peak VO2 at CPX after 24 weeks (or longest available follow up). Secondary exploratory endpoints will assess the effects of anakinra on additional CPX parameters, structural and functional echocardiographic data, noninvasive hemodynamic, quality of life questionnaires, biomarkers, and HF outcomes. DISCUSSION The current trial will assess the effects of IL-1 blockade with anakinra for 24 weeks on cardiorespiratory fitness in patients with recent hospitalization due to acute decompensated HFrEF. TRIAL REGISTRATION The trial was registered prospectively with ClinicalTrials.gov on Jan 8, 2019, identifier NCT03797001.
Collapse
Affiliation(s)
- Benjamin Van Tassell
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA.
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA.
| | - Virginia Mihalick
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Georgia Thomas
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Amr Marawan
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Azita H Talasaz
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Juan Lu
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, USA
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy Ladd
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Juan Ignacio Damonte
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Dave L Dixon
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Roshanak Markley
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Jeremy Turlington
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Emily Federmann
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Marco Giuseppe Del Buono
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Justin M Canada
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| | - Ross Arena
- Department of Physical Therapy, College of Applied Science, University of Illinois Chicago, Chicago, IL, USA
| | - Antonio Abbate
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Pauley Heart Center, Richmond, VA, USA
| |
Collapse
|
30
|
Liao Y, Liu K, Zhu L. Emerging Roles of Inflammasomes in Cardiovascular Diseases. Front Immunol 2022; 13:834289. [PMID: 35464402 PMCID: PMC9021369 DOI: 10.3389/fimmu.2022.834289] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/07/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases are known as the leading cause of morbidity and mortality worldwide. As an innate immune signaling complex, inflammasomes can be activated by various cardiovascular risk factors and regulate the activation of caspase-1 and the production and secretion of proinflammatory cytokines such as IL-1β and IL-18. Accumulating evidence supports that inflammasomes play a pivotal role in the progression of atherosclerosis, myocardial infarction, and heart failure. The best-known inflammasomes are NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes, among which NLRP3 inflammasome is the most widely studied in the immune response and disease development. This review focuses on the activation and regulation mechanism of inflammasomes, the role of inflammasomes in cardiovascular diseases, and the research progress of targeting NLRP3 inflammasome and IL-1β for related disease intervention.
Collapse
Affiliation(s)
- Yingnan Liao
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Kui Liu
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, China
| | - Liyuan Zhu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
31
|
Won T, Gilotra NA, Wood MK, Hughes DM, Talor MV, Lovell J, Milstone AM, Steenbergen C, Čiháková D. Increased Interleukin 18-Dependent Immune Responses Are Associated With Myopericarditis After COVID-19 mRNA Vaccination. Front Immunol 2022; 13:851620. [PMID: 35251049 PMCID: PMC8894592 DOI: 10.3389/fimmu.2022.851620] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/31/2022] [Indexed: 12/25/2022] Open
Abstract
Myocarditis and myopericarditis may occur after COVID-19 vaccination with an incidence of two to twenty cases per 100,000 individuals, but underlying mechanisms related to disease onset and progression remain unclear. Here, we report a case of myopericarditis following the first dose of the mRNA-1273 COVID-19 vaccine in a young man who had a history of mild COVID-19 three months before vaccination. The patient presented with chest pain, elevated troponin I level, and electrocardiogram abnormality. His endomyocardial biopsy revealed diffuse CD68+ cell infiltration. We characterized the immune profile of the patient using multiplex cytokine assay and flow cytometry analysis. Sex-matched vaccinated individuals and healthy individuals were used as controls. IL-18 and IL-27, Th1-type cytokines, were highly increased in the patient with COVID-19 vaccine-related myopericarditis compared with vaccinated controls who experienced no cardiac complications. In the patient, circulating NK cells and T cells showed an activated phenotype and mRNA profile, and monocytes expressed increased levels of IL-18 and its upstream NLRP3 inflammasome. We found that recombinant IL-18 administration into mice caused mild cardiac dysfunction and activation of NK cells and T cells in the hearts, similar to the findings in the patient with myopericarditis after COVID-19 mRNA vaccination. Collectively, myopericarditis following COVID-19 mRNA vaccination may be associated with increased IL-18-mediated immune responses and cardiotoxicity.
Collapse
Affiliation(s)
- Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nisha Aggarwal Gilotra
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Megan Kay Wood
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - David Matthew Hughes
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, United States
| | - Monica Vladut Talor
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jana Lovell
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Aaron Michael Milstone
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
- *Correspondence: Daniela Čiháková, ; orcid.org/0000-0002-8713-2860
| |
Collapse
|
32
|
Wang S, Zhang J, Wang Y, Jiang X, Guo M, Yang Z. NLRP3 inflammasome as a novel therapeutic target for heart failure. Anatol J Cardiol 2022; 26:15-22. [PMID: 35191381 PMCID: PMC8878950 DOI: 10.5152/anatoljcardiol.2021.580] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 06/30/2024] Open
Abstract
Heart failure (HF) is a leading cause of mortality worldwide. The pathogenesis of HF is complex and has not yet been fully elucidated, which has slowed drug development and long-term treatments. Inflammasome-mediated responses occur during the progression of HF. It has been reported that energy metabolism and metabolites of intestinal flora are also involved in the process of HF, and they interact with each other to promote the progression of HF. NLR family pyrin domain containing 3 (NLRP3) inflammasome may be a key target in the relationship between inflammation-mediated energy metabolism and metabolites of intestinal flora. Elucidating the relationship among the above three factors may help to identify new molecular targets for the prevention and treatment of HF and ultimately affect the course of HF. In this study, we systematically summarize evidence regarding the relationship among NLRP3 inflammasome, energy metabolism, intestinal microflora metabolites, and inflammation, as well as highlight advantages of NLRP3 inflammasome in treating HF.
Collapse
Affiliation(s)
- Shuangcui Wang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Jiaqi Zhang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Yuli Wang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Xijuan Jiang
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Maojuan Guo
- Department of Integrative Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| | - Zhen Yang
- Department of Chinese Medicine, Tianjin University of Traditional Chinese Medicine; Tianjin-China
| |
Collapse
|
33
|
Olsen MB, Gregersen I, Sandanger Ø, Yang K, Sokolova M, Halvorsen BE, Gullestad L, Broch K, Aukrust P, Louwe MC. Targeting the Inflammasome in Cardiovascular Disease. JACC Basic Transl Sci 2022; 7:84-98. [PMID: 35128212 PMCID: PMC8807732 DOI: 10.1016/j.jacbts.2021.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/24/2021] [Accepted: 08/28/2021] [Indexed: 01/10/2023]
Abstract
The pathogenesis of cardiovascular disease (CVD) is complex and multifactorial, and inflammation plays a central role. Inflammasomes are multimeric protein complexes that are activated in a 2-step manner in response to infection or tissue damage. Upon activation the proinflammatory cytokines, interleukins-1β and -18 are released. In the last decade, the evidence that inflammasome activation plays an important role in CVD development became stronger. We discuss the role of different inflammasomes in the pathogenesis of CVD, focusing on atherosclerosis and heart failure. This review also provides an overview of existing experimental studies and clinical trials on inflammasome inhibition as a therapeutic target in these disorders.
Collapse
Key Words
- ACS, acute coronary syndrome
- AIM2, absent in melanoma 2
- ASC, apoptosis associated speck-like protein
- ATP, adenosine triphosphate
- CAD, coronary artery disease
- CRP, C-reactive protein
- CVD, cardiovascular disease
- DAMP, damage associated molecular pattern
- GSDMD, gasdermin-D
- GSDMD-NT, gasdermin-D N-terminal
- HF, heart failure
- HFpEF, HF with preserved ejection fraction
- HFrEF, HF with reduced ejection fraction
- IL, interleukin
- IL-1
- LDL, low-density lipoprotein
- LV, left ventricular
- LVEF, left ventricular ejection fraction
- MI, myocardial infarction
- NF-κB, nuclear factor κB
- NLR, NOD-like receptor
- NLRP3
- NLRP3, NOD-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- PRR, pattern recognition receptor
- STEMI, ST-elevation myocardial infarction
- TLR, toll-like receptor
- atherosclerosis
- cardiovascular disease
- heart failure
- inflammasome
Collapse
Affiliation(s)
- Maria Belland Olsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Øystein Sandanger
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Dermatology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Marina Sokolova
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Bente E. Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Cardiac Research Center, Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Cardiac Research Center, Center for Heart Failure Research, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Mieke C. Louwe
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
34
|
Abstract
The NACHT, leucine-rich repeat (LRR), and pyrin domain (PYD)-containing protein 3 (NLRP3) inflammasome is a macromolecular structure responsible for the inflammatory response to injury or infection. Several types of heart disease are linked to the activity of the NLRP3 inflammasome and its cytokines, interleukin-1β (IL-1β), and IL-18. Recent pieces of evidence collected from human samples, together with experimental animal models, demonstrate a causative role for the pathogenesis and progression of heart failure (HF). Preclinical research showed that NLRP3 inhibition is a viable strategy to reduce adverse cardiac remodeling and improve left ventricular function in HF. Early phase clinical studies proved to be safe and effective supporting the potential benefit of blocking the NLRP3 inflammasome pathway in patients with HF.
Collapse
Affiliation(s)
- Eleonora Mezzaroma
- Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, USA.,VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
35
|
Szabo TM, Frigy A, Nagy EE. Targeting Mediators of Inflammation in Heart Failure: A Short Synthesis of Experimental and Clinical Results. Int J Mol Sci 2021; 22:13053. [PMID: 34884857 PMCID: PMC8657742 DOI: 10.3390/ijms222313053] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 01/22/2023] Open
Abstract
Inflammation has emerged as an important contributor to heart failure (HF) development and progression. Current research data highlight the diversity of immune cells, proteins, and signaling pathways involved in the pathogenesis and perpetuation of heart failure. Chronic inflammation is a major cardiovascular risk factor. Proinflammatory signaling molecules in HF initiate vicious cycles altering mitochondrial function and perturbing calcium homeostasis, therefore affecting myocardial contractility. Specific anti-inflammatory treatment represents a novel approach to prevent and slow HF progression. This review provides an update on the putative roles of inflammatory mediators involved in heart failure (tumor necrosis factor-alpha; interleukin 1, 6, 17, 18, 33) and currently available biological and non-biological therapy options targeting the aforementioned mediators and signaling pathways. We also highlight new treatment approaches based on the latest clinical and experimental research.
Collapse
Affiliation(s)
- Timea Magdolna Szabo
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania;
| | - Attila Frigy
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania;
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Laboratory of Medical Analysis, Clinical County Hospital Mures, 540394 Targu Mures, Romania
| |
Collapse
|
36
|
Zhao YJ, Qiao H, Liu DF, Li J, Li JX, Chang SE, Lu T, Li FT, Wang D, Li HP, He XJ, Wang F. Lithium promotes recovery after spinal cord injury. Neural Regen Res 2021; 17:1324-1333. [PMID: 34782578 PMCID: PMC8643056 DOI: 10.4103/1673-5374.327348] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Lithium is associated with oxidative stress and apoptosis, but the mechanism by which lithium protects against spinal cord injury remains poorly understood. In this study, we found that intraperitoneal administration of lithium chloride (LiCl) in a rat model of spinal cord injury alleviated pathological spinal cord injury and inhibited expression of tumor necrosis factor α, interleukin-6, and interleukin 1 β. Lithium inhibited pyroptosis and reduced inflammation by inhibiting Caspase-1 expression, reducing the oxidative stress response, and inhibiting activation of the Nod-like receptor protein 3 inflammasome. We also investigated the neuroprotective effects of lithium intervention on oxygen/glucose-deprived PC12 cells. We found that lithium reduced inflammation, oxidative damage, apoptosis, and necrosis and up-regulated nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 in PC12 cells. All-trans retinoic acid, an Nrf2 inhibitor, reversed the effects of lithium. These results suggest that lithium exerts anti-inflammatory, anti-oxidant, and anti-pyroptotic effects through the Nrf2/heme oxygenase-1 pathway to promote recovery after spinal cord injury. This study was approved by the Animal Ethics Committee of Xi’an Jiaotong University (approval No. 2018-2053) on October 23, 2018.
Collapse
Affiliation(s)
- Ying-Jie Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Hao Qiao
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Dong-Fan Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Jie Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Jia-Xi Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Su-E Chang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Teng Lu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Feng-Tao Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Dong Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Hao-Peng Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| | - Xi-Jing He
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine; Department of Orthopedics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi Province, China
| | - Fang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi Province, China
| |
Collapse
|
37
|
Zhao M, Zhang J, Xu Y, Liu J, Ye J, Wang Z, Ye D, Feng Y, Xu S, Pan W, Wang M, Wan J. Selective Inhibition of NLRP3 Inflammasome Reverses Pressure Overload-Induced Pathological Cardiac Remodeling by Attenuating Hypertrophy, Fibrosis, and Inflammation. Int Immunopharmacol 2021; 99:108046. [PMID: 34435581 DOI: 10.1016/j.intimp.2021.108046] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/25/2021] [Accepted: 08/01/2021] [Indexed: 12/24/2022]
Abstract
Activation of the NLRP3 inflammasome promotes pathological cardiac remodeling induced by pressure overload. However, the therapeutic effects of NLRP3 inhibition after cardiac remodeling remain unknown. The present study aimed to investigate whether the selective NLRP3 inhibitor, MCC950, could reverse transverse aortic constriction (TAC)-induced cardiac remodeling. Mice were divided into four groups based on the treatment given: sham, sham + MCC950, TAC, and TAC + MCC950. MCC950 (10 mg/kg, intraperitoneal injection, once per day) was administered from two weeks after TAC or sham surgery for four weeks. Echocardiography, histological analysis, RT-PCR, and Western blotting were performed to explore the function of MCC950 after TAC. We found that MCC950 reversed cardiac dysfunction after TAC. MCC950 attenuated cardiac hypertrophy by down-regulating calcineurin expression and inhibiting MAPK activation. Further, it also alleviated cardiac fibrosis post-TAC by inhibiting the TGF-β/Smad4 pathway, and reduced cardiac inflammation and macrophage infiltration post-TAC, including both M1 and M2 macrophages. Taken together, MCC950 can attenuate cardiac remodeling due to pressure overload by inhibiting hypertrophy, fibrosis, and inflammation. Our study provides a basis for the clinical application of NLRP3 inhibitors in the treatment of non-ischemic heart failure.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| |
Collapse
|
38
|
Abstract
Purpose of Review Pericarditis is a generally benign disease, although complications and/or recurrences may occur in up to 30% of cases. New evidence on the pathophysiology of the disease has accumulated in recent years. Recent Findings Recently, it has been shown that the activation of the NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein 3) inflammasome is central in the pathophysiology of pericarditis. These findings derive from clinical data, an experimental animal model of acute pericarditis supporting a role for the NLRP3 inflammasome in pericarditis, and from indirect evidence of inhibitors of NLRP3 inflammasome in clinical trials. Summary Pericarditis is regarded as a stereotypical response to an acute damage of the mesothelial cells of the pericardial layers. NLRP3 inflammasome, a macromolecular structure sensing damage and releasing pro-inflammatory cytokines, is centrally involved as it releases interleukin (IL)-1β, whose auto-induction feeds an autoinflammatory disease, mostly responsible for recurrences. Colchicine, an inhibitor of NLRP3 inflammasome formation, and IL-1-targeted therapies, such as anakinra and rilonacept, were found to effectively blunt the acute inflammation and reduce the risk for recurrences.
Collapse
|
39
|
Interleukin 1 receptor antagonism abrogates acute pressure-overload induced murine heart failure. Ann Thorac Surg 2021; 114:98-107. [PMID: 34419440 DOI: 10.1016/j.athoracsur.2021.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recent clinical trials have suggested that blockade of interleukin-1 can favorably impact patients with myocardial infarction and heart failure. However, the mechanism of how antagonism of this specific cytokine in mediating cardiac disease remains unclear. Hence, we sought to determine the influence of IL-1 blockade on acute hypertensive remodeling. METHODS Transverse aortic constriction (TAC) was performed in C57BL mice with or without intraperitoneal administration of interleukin 1 receptor antagonism (IL-1ra). Function, structure, and molecular diagnostics were subsequently performed and analyzed. RESULTS Six weeks after TAC, a progressive decline of ejection fraction and increases in LV mass and dimensions was effectively mitigated with IL-1ra. TAC resulted in an expected profile of hypertrophic markers including myosin heavy chain, atrial natriuretic peptide, and skeletal muscle actin which were all significantly lower in IL-1ra treated mice. While trichrome staining 2-weeks post TAC demonstrated similar levels of fibrosis, IL-1ra reduced expression of collagen-1, TIMP1, and periostin. Investigating the angiogenic response to pressure overload, similar levels of VEGF were observed, but IL-1ra was associated with more SDF-1. Immune cell infiltration (macrophages and lymphocytes) was also decreased in IL-1ra treated mice. Similarly, cytokine concentrations of IL-1, IL-18, and IL-6 were all reduced in IL-1ra-treated animals. CONCLUSIONS IL-1ra prevents the progression towards heart failure associated with acute pressure overload. This functional response was associated with reductions in mediators of fibrosis, cellular infiltration, and cytokine production. These results provide mechanistic insight into recent clinical trials and could springboard future investigations in patients with pressure-overload based cardiomyopathies.
Collapse
|
40
|
Luo Y, Li Z, Ge P, Guo H, Li L, Zhang G, Xu C, Chen H. Comprehensive Mechanism, Novel Markers and Multidisciplinary Treatment of Severe Acute Pancreatitis-Associated Cardiac Injury - A Narrative Review. J Inflamm Res 2021; 14:3145-3169. [PMID: 34285540 PMCID: PMC8286248 DOI: 10.2147/jir.s310990] [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: 03/23/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is one of the common acute abdominal inflammatory diseases in clinic with acute onset and rapid progress. About 20% of the patients will eventually develop into severe acute pancreatitis (SAP) characterized by a large number of inflammatory cells infiltration, gland flocculus flaky necrosis and hemorrhage, finally inducing systemic inflammatory response syndrome (SIRS) and multiple organ dysfunction syndrome (MODS). Pancreatic enzyme activation, intestinal endotoxemia (IETM), cytokine activation, microcirculation disturbance, autonomic nerve dysfunction and autophagy dysregulation all play an essential role in the occurrence and progression of SAP. Organ dysfunction is the main cause of early death in SAP. Acute kidney injury (AKI) and acute lung injury (ALI) are common, while cardiac injury (CI) is not, but the case fatality risk is high. Many basic studies have observed obvious ultrastructure change of heart in SAP, including myocardial edema, cardiac hypertrophy, myocardial interstitial collagen deposition. Moreover, in clinical practice, patients with SAP often presented various abnormal electrocardiogram (ECG) and cardiac function. Cases complicated with acute myocardial infarction and pericardial tamponade have also been reported and even result in stress cardiomyopathy. Due to the molecular mechanisms underlying SAP-associated cardiac injury (SACI) remain poorly understood, and there is no complete, unified treatment and sovereign remedy at present, this article reviews reports referring to the pathogenesis, potential markers and treatment methods of SACI in recent years, in order to improve the understanding of cardiac injury in severe pancreatitis.
Collapse
Affiliation(s)
- YaLan Luo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - ZhaoXia Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Peng Ge
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - HaoYa Guo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - GuiXin Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - CaiMing Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - HaiLong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
41
|
Abstract
Neurohormones and inflammatory mediators have effects in both the heart and the peripheral vasculature. In patients with heart failure (HF), neurohormonal activation and increased levels of inflammatory mediators promote ventricular remodeling and development of HF, as well as vascular dysfunction and arterial stiffness. These processes may lead to a vicious cycle, whereby arterial stiffness perpetuates further ventricular remodeling leading to exacerbation of symptoms. Although significant advances have been made in the treatment of HF, currently available treatment strategies slow, but do not halt, this cycle. The current treatment for HF patients involves the inhibition of neurohormonal activation, which can reduce morbidity and mortality related to this condition. Beyond benefits associated with neurohormonal blockade, other strategies have focused on inhibition of inflammatory pathways implicated in the pathogenesis of HF. Unfortunately, attempts to target inflammation have not yet been successful to improve prognosis of HF. Further work is required to interrupt key maladaptive mechanisms involved in disease progression.
Collapse
|
42
|
A novel anti-human IL-1R7 antibody reduces IL-18-mediated inflammatory signaling. J Biol Chem 2021; 296:100630. [PMID: 33823154 PMCID: PMC8018910 DOI: 10.1016/j.jbc.2021.100630] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
Unchecked inflammation can result in severe diseases with high mortality, such as macrophage activation syndrome (MAS). MAS and associated cytokine storms have been observed in COVID-19 patients exhibiting systemic hyperinflammation. Interleukin-18 (IL-18), a proinflammatory cytokine belonging to the IL-1 family, is elevated in both MAS and COVID-19 patients, and its level is known to correlate with the severity of COVID-19 symptoms. IL-18 binds its specific receptor IL-1 receptor 5 (IL-1R5, also known as IL-18 receptor alpha chain), leading to the recruitment of the coreceptor, IL-1 receptor 7 (IL-1R7, also known as IL-18 receptor beta chain). This heterotrimeric complex then initiates downstream signaling, resulting in systemic and local inflammation. Here, we developed a novel humanized monoclonal anti-IL-1R7 antibody to specifically block the activity of IL-18 and its inflammatory signaling. We characterized the function of this antibody in human cell lines, in freshly obtained peripheral blood mononuclear cells (PBMCs) and in human whole blood cultures. We found that the anti-IL-1R7 antibody significantly suppressed IL-18-mediated NFκB activation, reduced IL-18-stimulated IFNγ and IL-6 production in human cell lines, and reduced IL-18-induced IFNγ, IL-6, and TNFα production in PBMCs. Moreover, the anti-IL-1R7 antibody significantly inhibited LPS- and Candida albicans–induced IFNγ production in PBMCs, as well as LPS-induced IFNγ production in whole blood cultures. Our data suggest that blocking IL-1R7 could represent a potential therapeutic strategy to specifically modulate IL-18 signaling and may warrant further investigation into its clinical potential for treating IL-18-mediated diseases, including MAS and COVID-19.
Collapse
|
43
|
De Luca G, Cavalli G, Campochiaro C, Bruni C, Tomelleri A, Dagna L, Matucci-Cerinic M. Interleukin-1 and Systemic Sclerosis: Getting to the Heart of Cardiac Involvement. Front Immunol 2021; 12:653950. [PMID: 33833766 PMCID: PMC8021854 DOI: 10.3389/fimmu.2021.653950] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is rare, severe connective tissue disease characterized by endothelial and vascular damage, immune activation, and resulting in inflammation and fibrosis of skin and internal organs, including the heart. SSc is associated with high morbidity and mortality. Cardiac involvement is frequent in SSc patients, even though often asymptomatic at early stages, and represents one of the major causes of SSc-related mortality. Heart involvement has a variable clinical presentation, and its pathogenesis is not completely understood. Myocardial fibrosis is traditionally considered the immunopathologic hallmark of heart involvement in SSc. This unique histological feature is paralleled by distinctive clinical and prognostic features. The so-called "vascular hypothesis" represents the most credited hypothesis to explain myocardial fibrosis. More recently, the prominent role of an inflammatory myocardial process has been identified as a cardinal event in the evolution to fibrosis, thus also delineating an "inflammation-driven pathway to fibrosis". The pro-inflammatory cytokine interleukin (IL)-1 has an apical and cardinal role in the myocardial inflammatory cascade and in cardiac dysfunction. The primary aim of this perspective article is: to present the emerging evidence on the role of IL-1 and inflammasome in both SSc and heart inflammation, to review the complex interplay between cellular metabolism and inflammasome activation, and to discuss the rationale for targeted inhibition of IL-1 for the treatment of SSc-heart involvement, providing preliminary experimental and clinical data to support this hypothesis.
Collapse
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology AOUC, Florence, Italy
| | - Alessandro Tomelleri
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, and Division of Rheumatology AOUC, Florence, Italy
| |
Collapse
|
44
|
Mauro AG, Bonaventura A, Vecchié A, Mezzaroma E, Carbone S, Narayan P, Potere N, Cannatà A, Paolini JF, Bussani R, Montecucco F, Sinagra G, Van Tassel BW, Abbate A, Toldo S. 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: 54] [Impact Index Per Article: 13.5] [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
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
Affiliation(s)
- Adolfo G. Mauro
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Aldo Bonaventura
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Alessandra Vecchié
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eleonora Mezzaroma
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Salvatore Carbone
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pratyush Narayan
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Nicola Potere
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Antonio Cannatà
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste, University of Trieste, Trieste, Italy
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Department of Cardiovascular Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | | | - Rossana Bussani
- Institute of Pathological Anatomy, Riuniti Hospital, University of Trieste, Trieste, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova, Italian Cardiovascular Network, Genoa, Italy
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria-Universitaria Integrata Trieste, University of Trieste, Trieste, Italy
| | - Benjamin W. Van Tassel
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Stefano Toldo
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Johnson Research Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Cardiothoracic Surgery, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
45
|
Quader M, Mezzaroma E, Kenning K, Toldo S. Modulation of Interleukin-1 and -18 Mediated Injury in Donation after Circulatory Death Mouse Hearts. J Surg Res 2021; 257:468-476. [PMID: 32896815 PMCID: PMC8687874 DOI: 10.1016/j.jss.2020.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/14/2020] [Accepted: 08/02/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Donation after circulatory death donors (DCD) can expand the donor pool for heart transplantation, which primarily depends on brain death donors. Ischemia and reperfusion injury are inherent to the DCD process. We hypothesize that pharmacologic inhibition of interleukin-1 (IL-1) and/or IL-18 is protective to DCD hearts. MATERIALS AND METHODS Following clinical protocol, in-situ ischemia time in control beating-heart donor (CBD) and DCD groups was less than 5 and 40 min, respectively. Wild type (WT) C57Bl6/j, IL-1 receptor type I knockout (IL-1RI-KO), and IL-18 KO mice were used. Hearts were reanimated for 90 min on a Langendorff system with Krebs-Henseleit buffer at 37°C, to assess physiologic parameters. Recombinant IL-1 receptor antagonist (IL-1Ra) and/or IL-18 binding protein (IL-18BP) were added to the Krebs-Henseleit buffer to inhibit IL-1 and/or the IL-18 signaling, respectively. RESULTS Developed pressure and ± dP/dt were significantly impaired in the DCD-WT group compared to CBD-WT (P ≤ 0.05). Troponin release was higher in DCD-WT groups. Functional parameters were preserved, and troponin release was significantly less in the DCD knockout groups. Heart function was improved in DCD groups treated with IL-1Ra or IL-18BP compared to the DCD-WT group. CONCLUSIONS Heart function was significantly impaired in the DCD-WT group compared to CBD-WT. Genetic deletion or pharmacologic blockade of IL-1 or IL-18 was protective to DCD hearts.
Collapse
Affiliation(s)
- Mohammed Quader
- Cardiovascular Surgery, Virginia Commonwealth University, Richmond, Virginia; Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.
| | - Eleonora Mezzaroma
- Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Kristine Kenning
- Cardiovascular Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Stefano Toldo
- Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
46
|
Gao RF, Li X, Xiang HY, Yang H, Lv CY, Sun XL, Chen HZ, Gao Y, Yang JS, Luo W, Yang YQ, Tang YH. The covalent NLRP3-inflammasome inhibitor Oridonin relieves myocardial infarction induced myocardial fibrosis and cardiac remodeling in mice. Int Immunopharmacol 2021; 90:107133. [PMID: 33168408 DOI: 10.1016/j.intimp.2020.107133] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/18/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myocardial infarction (MI) triggers a strong inflammatory response that is associated with myocardial fibrosis and cardiac remodeling. Interleukin (IL)-1β and IL-18 are key players in this response and are controlled by NLRP3-inflammatory bodies. Oridonin is a newly reported NLRP3 inhibitor with strong anti-inflammatory activity. We hypothesized that the covalent NLRP3 inhibitor Oridonin could reduce IL-1β and IL-18 expression and ameliorate myocardial fibrosis after myocardial infarction in mice, improve poor heart remodeling, and preserve heart function. METHODS Male C57BL/6 mice were subjected to left coronary artery ligation to induce MI and then treated with Oridonin (1, 3, or 6 mg/kg), MCC950 (10 mg/kg), CY-09 (5 mg/kg) or saline three times a week for two weeks. Four weeks after MI, cardiac function and myocardial fibrosis were assessed. In addition, myocardial expressions of inflammatory factors and fibrotic markers were analyzed by western blot, immunofluorescence, enzyme-linked immunosorbent assay, and quantitative real-time polymerase chain reaction. RESULTS Oridonin treatment preserved left ventricular ejection fraction and fractional shortening, and markedly limited the myocardial infarct size in treated mice. The myocardial fibrosis was lower in the 1 mg/kg group (15.98 ± 1.64)%, 3 mg/kg group (17.39 ± 2.45)%, and 6 mg/kg group (16.76 ± 3.06)% compared to the control group (23.38 ± 1.65)%. Moreover, similar with the results of Oridonin, MCC950 and CY-09 also preserved cardiac function and reduced myocardial fibrosis. The expression levels of NLRP3, IL-1β and IL-18 were decreased in the Oridonin treatment group compared to non-treated group. In addition, myocardial macrophage and neutrophil influxes were attenuated in the Oridonin treated group. CONCLUSIONS The covalent NLRP3-inflammasome inhibitor Oridonin reduces myocardial fibrosis and preserves cardiac function in a mouse MI model, which indicates potential therapeutic effect of Oridonin on acute MI patients.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Cells, Cultured
- Disease Models, Animal
- Diterpenes, Kaurane/pharmacology
- Fibrosis
- Furans
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Indenes
- Inflammasomes/antagonists & inhibitors
- Inflammasomes/metabolism
- Interleukin-1beta/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Myocardial Infarction/drug therapy
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Neutrophil Infiltration/drug effects
- Neutrophils/drug effects
- Neutrophils/metabolism
- Receptors, Interleukin-18/metabolism
- Signal Transduction
- Stroke Volume/drug effects
- Sulfonamides
- Sulfones/pharmacology
- Thiazolidines/pharmacology
- Thiones/pharmacology
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Ri-Feng Gao
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xiao Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 20032, China
| | - Hai-Yan Xiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Heng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Chun-Yu Lv
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Lei Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 20032, China
| | - Hong-Zhang Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yang Gao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 20032, China
| | - Jue-Sheng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Wei Luo
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| | - Yan-Hua Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang 330006, Jiangxi Province, China.
| |
Collapse
|
47
|
Cavalli G, Farina N, Campochiaro C, Baldissera E, Dagna L. Current treatment options and safety considerations when treating adult-onset Still's disease. Expert Opin Drug Saf 2020; 19:1549-1558. [PMID: 33078630 DOI: 10.1080/14740338.2020.1839411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Adult onset Still disease (AOSD) is a rare systemic inflammatory condition. The clinical spectrum of this disease ranges from self-limiting forms with mild symptoms to life-threatening cases. Glucocorticoids and non-steroidal anti-inflammatory drugs (NSAIDs) represent the first line of therapy for AOSD, with add-on therapy with second-line drug reserved to steroid-dependent patients and in life-threatening cases. Currently, early treatment with conventional disease modifying anti-rheumatic drugs (DMARDs) and biologic agents blocking causal cytokines is advocated in patients with severe and recalcitrant clinical manifestations. AREAS COVERED This review analyzes the available controlled evidence and observational data regarding the efficacy and safety of conventional and biological pharmacological agents in the treatment of AOSD. EXPERT OPINION Non-steroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids are effective in controlling clinical manifestations in the majority of AOSD patients. Conventional DMARDs can be 20 effective in some severe and steroid-dependent cases of AOSD; however, anti-cytokine agents represent an effective and overall more suitable alternative in this specific subset of patients. IL-1 and IL-6 blockade are effective in treating systemic and articular inflammation of AOSD patients. IL-1 blockade also has an excellent safety profile and therefore represent the first choice of biologic treatment in this clinical scenario.
Collapse
Affiliation(s)
- Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital , Milan, Italy.,Unitof Immunology, Rheumatology, Allergy and Rare Diseases, Vita-Salute San Raffaele University , Milan, Italy
| | - Nicola Farina
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital , Milan, Italy.,Unitof Immunology, Rheumatology, Allergy and Rare Diseases, Vita-Salute San Raffaele University , Milan, Italy
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital , Milan, Italy.,Unitof Immunology, Rheumatology, Allergy and Rare Diseases, Vita-Salute San Raffaele University , Milan, Italy
| | - Elena Baldissera
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital , Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital , Milan, Italy.,Unitof Immunology, Rheumatology, Allergy and Rare Diseases, Vita-Salute San Raffaele University , Milan, Italy
| |
Collapse
|
48
|
Aleksova A, Ferro F, Gagno G, Cappelletto C, Santon D, Rossi M, Ippolito G, Zumla A, Beltrami AP, Sinagra G. COVID-19 and renin-angiotensin system inhibition: role of angiotensin converting enzyme 2 (ACE2) - Is there any scientific evidence for controversy? J Intern Med 2020; 288:410-421. [PMID: 32459372 PMCID: PMC7283873 DOI: 10.1111/joim.13101] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022]
Abstract
Renin-angiotensin system (RAS) blockers are extensively used worldwide to treat many cardiovascular disorders, where they are effective in reducing both mortality and morbidity. These drugs are known to induce an increased expression of angiotensin-converting enzyme 2 (ACE2). ACE2 acts as receptor for the novel SARS coronavirus-2 (SARS-CoV-2) which raising the important issue of possible detrimental effects that RAS blockers could exert on the natural history and pathogenesis of the coronavirus disease-19 (COVID-19) and associated excessive inflammation, myocarditis and cardiac arrhythmias. We review the current knowledge on the interaction between SARS-CoV-2 infection and RAS blockers and suggest a scientific rationale for continuing RAS blockers therapy in patients with COVID-19 infection.
Collapse
Affiliation(s)
- A Aleksova
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - F Ferro
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - G Gagno
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - C Cappelletto
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - D Santon
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - M Rossi
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - G Ippolito
- National Institute for Infectious Diseases Lazzaro Spallanzani - IRCCS, Rome, Italy
| | - A Zumla
- Division of Infection and Immunity, University College London, London, UK.,National Institute of Health Research, Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | | | - G Sinagra
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| |
Collapse
|
49
|
Shimada BK, Yang Y, Zhu J, Wang S, Suen A, Kronstadt SM, Jeyaram A, Jay SM, Zou L, Chao W. Extracellular miR-146a-5p Induces Cardiac Innate Immune Response and Cardiomyocyte Dysfunction. Immunohorizons 2020; 4:561-572. [PMID: 32958516 PMCID: PMC7754174 DOI: 10.4049/immunohorizons.2000075] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 08/28/2020] [Indexed: 11/19/2022] Open
Abstract
Previous studies have demonstrated that transient myocardial ischemia leads to release of cellular nucleic acids such as RNA. Extracellular RNA reportedly plays a pivotal role in myocardial inflammation and ischemic injury in animals. RNA profiling has identified that numerous microRNA (miRNAs), such as ss-miR-146a-5p, are upregulated in plasma following myocardial ischemia, and certain uridine-rich miRNAs exhibit strong proinflammatory effects in immune cells via ssRNA-sensing mechanism. However, the effect of extracellular miRNAs on myocardial inflammation and cardiac cell function remains unknown. In this study, we treated adult mouse cardiomyocytes with miR-146a-5p loaded in extracellular vesicles and observed a dose- and TLR7-dependent production of CXCL-2, IL-6, and TNF-α. In vivo, a single dose of myocardial injection of miR-146a-5p induced both cytokine expression (CXCL2, IL-6, and TNF-α) and innate immune cell activation (CD45+ leukocytes, Ly6Cmid+ monocytes, Ly6G+ neutrophils), which was significantly attenuated in the hearts of TLR7 KO mice. We discovered that conditioned media from miR-146a-treated macrophages stimulated proinflammatory cytokine production in adult cardiomyocytes and significantly inhibited their sarcomere shortening. Finally, using an electric cell impedance-sensing assay, we found that the conditioned media from miR-146a-treated cardiac fibroblasts or cardiomyocytes impaired the barrier function of coronary artery endothelial cells. Taken together, these data demonstrate that extracellular miR-146a-5p activates multiple cardiac cells and induces myocardial inflammation and cardiomyocyte dysfunction via intercellular interaction and innate immune TLR7 nucleic acid sensing.
Collapse
Affiliation(s)
- Briana K Shimada
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Yang Yang
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Jing Zhu
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Sheng Wang
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Andrew Suen
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Stephanie M Kronstadt
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20740
| | - Anjana Jeyaram
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20740
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20740
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201; and
| |
Collapse
|
50
|
Vecchié A, Bonaventura A, Toldo S, Dagna L, Dinarello CA, Abbate A. IL-18 and infections: Is there a role for targeted therapies? J Cell Physiol 2020; 236:1638-1657. [PMID: 32794180 DOI: 10.1002/jcp.30008] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-18 is a pro-inflammatory cytokine belonging to the IL-1 family, first identified for its interferon-γ-inducing properties. IL-18 regulates both T helper (Th) 1 and Th2 responses. It acts synergistically with IL-12 in the Th1 paradigm, whereas with IL-2 and without IL-12 it can induce Th2 cytokine production from cluster of differentation (CD)4+ T cells, natural killer (NK cells, NKT cells, as well as from Th1 cells. IL-18 also plays a role in the hemophagocytic lymphohistiocytosis, a life-threatening condition characterized by a cytokine storm that can be secondary to infections. IL-18-mediated inflammation was largely studied in animal models of bacterial, viral, parasitic, and fungal infections. These studies highlight the contribution of either IL-18 overproduction by the host or overresponsiveness of the host to IL-18 causing an exaggerated inflammatory burden and leading to tissue injury. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19). The damage in the later phase of the disease appears to be driven by a cytokine storm, including interleukin IL-1 family members and secondary cytokines like IL-6. IL-18 may participate in this hyperinflammation, as it was previously found to be able to cause injury in the lung tissue of infected animals. IL-18 blockade has become an appealing therapeutic target and has been tested in some IL-18-mediated rheumatic diseases and infantile-onset macrophage activation syndrome. Given its role in regulating the immune response to infections, IL-18 blockade might represent a therapeutic option for COVID-19, although further studies are warranted to investigate more in detail the exact role of IL-18 in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Alessandra Vecchié
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Aldo Bonaventura
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Stefano Toldo
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Charles A Dinarello
- Department of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonio Abbate
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|