51
|
Quader M, Mezzaroma E, Kenning K, Toldo S. Targeting the NLRP3 inflammasome to reduce warm ischemic injury in donation after circulatory death heart. Clin Transplant 2020; 34:e14044. [PMID: 32654189 DOI: 10.1111/ctr.14044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/15/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022]
Abstract
While the donation after circulatory death (DCD) heart transplantation is an emerging clinical practice, the primary source of donor hearts for transplantation remains donation after brain death (DBD) donors. DCD process induces formation of NOD-like receptor family pyrin domain containing-3 (NLRP3) inflammasome, a key mediator of inflammation-driven damage to heart. Inhibition of NLRP3 inflammasome formation could be protective to DCD hearts. Five groups (n = 8 each) of mice were studied-control beating heart donor (CBD) wild-type (WT), DCD WT, CBD NLRP3 knockout (KO), DCD NLRP3 KO, and DCD WT NLRP3 inhibitor group. Hearts were procured and reanimated on a Langendorff system to assess physiologic parameters and then for molecular assays. NLRP3 inhibitor (50 µmol/L) was administered to the DCD-NLRP3 inhibitor group at reanimation. Tissue NLRP3 levels were 80% higher in the DCD WT group compared with the CBD-WT group. Caspase-1 activity was significantly elevated in DCD WT but not in KO or NLRP3 inhibitor groups. The developed pressures and ±dP/dt were significantly impaired in the DCD WT group compared with the CBD-WT group, P < .05, but were well preserved in DCD-NLRP3 inhibitor group. The DCD process activates the NLRP3 inflammasome, contributing to myocardial damage and dysfunction. NLRP3 inflammasome inhibition limits myocardial injury and preserves DCD heart function.
Collapse
Affiliation(s)
- Mohammed Quader
- Division of Thoracic and Cardiovascular Surgery, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eleonora Mezzaroma
- School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Kristine Kenning
- Division of Thoracic and Cardiovascular Surgery, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Stefano Toldo
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
52
|
Wang J, Zhang W, Wu L, Mei Y, Cui S, Feng Z, Chen X. New insights into the pathophysiological mechanisms underlying cardiorenal syndrome. Aging (Albany NY) 2020; 12:12422-12431. [PMID: 32561688 PMCID: PMC7343447 DOI: 10.18632/aging.103354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
Communication between the heart and kidney occurs through various bidirectional pathways. The heart maintains continuous blood flow through the kidney while the kidney regulates blood volume thereby allowing the heart to pump effectively. Cardiorenal syndrome (CRS) is a pathologic condition in which acute or chronic dysfunction of the heart or kidney induces acute or chronic dysfunction of the other organ. CRS type 3 (CRS-3) is defined as acute kidney injury (AKI)-mediated cardiac dysfunction. AKI is common among critically ill patients and correlates with increased mortality and morbidity. Acute cardiac dysfunction has been observed in over 50% of patients with severe AKI and results in poorer clinical outcomes than heart or renal dysfunction alone. In this review, we describe the pathophysiological mechanisms responsible for AKI-induced cardiac dysfunction. Additionally, we discuss current approaches in the management of patients with CRS-3 and the development of targeted therapeutics. Finally, we summarize current challenges in diagnosing mild cardiac dysfunction following AKI and in understanding CRS-3 etiology.
Collapse
Affiliation(s)
- Jin Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Weiguang Zhang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Lingling Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Yan Mei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Shaoyuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Zhe Feng
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| |
Collapse
|
53
|
Abbate A, Toldo S, Marchetti C, Kron J, Van Tassell BW, Dinarello CA. Interleukin-1 and the Inflammasome as Therapeutic Targets in Cardiovascular Disease. Circ Res 2020; 126:1260-1280. [PMID: 32324502 DOI: 10.1161/circresaha.120.315937] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intracellular sensing protein termed NLRP3 (for NACHT, LRR, and PYD domains-containing protein 3) forms a macromolecular structure called the NLRP3 inflammasome. The NLRP3 inflammasome plays a major role in inflammation, particularly in the production of IL (interleukin)-1β. IL-1β is the most studied of the IL-1 family of cytokines, including 11 members, among which are IL-1α and IL-18. Here, we summarize preclinical and clinical findings supporting the key pathogenetic role of the NLRP3 inflammasome and IL-1 cytokines in the formation, progression, and complications of atherosclerosis, in ischemic (acute myocardial infarction), and nonischemic injury to the myocardium (myocarditis) and the progression to heart failure. We also review the clinically available IL-1 inhibitors, although not currently approved for cardiovascular indications, and discuss other IL-1 inhibitors, not currently approved, as well as oral NLRP3 inflammasome inhibitors currently in clinical development. Canakinumab, IL-1β antibody, prevented the recurrence of ischemic events in patients with prior acute myocardial infarction in a large phase III clinical trial, including 10 061 patients world-wide. Phase II clinical trials show promising data with anakinra, recombinant IL-1 receptor antagonist, in patients with ST-segment-elevation acute myocardial infarction or heart failure with reduced ejection fraction. Anakinra also improved outcomes in patients with pericarditis, and it is now considered standard of care as second-line treatment for patients with recurrent/refractory pericarditis. Rilonacept, a soluble IL-1 receptor chimeric fusion protein neutralizing IL-1α and IL-1β, has also shown promising results in a phase II study in recurrent/refractory pericarditis. In conclusion, there is overwhelming evidence linking the NLRP3 inflammasome and the IL-1 cytokines with the pathogenesis of cardiovascular diseases. The future will likely include targeted inhibitors to block the IL-1 isoforms, and possibly oral NLRP3 inflammasome inhibitors, across a wide spectrum of cardiovascular diseases.
Collapse
Affiliation(s)
- Antonio Abbate
- From the VCU Pauley Heart Center, Virginia Commonwealth University, Richmond (A.A., S.T., J.K.)
| | - Stefano Toldo
- From the VCU Pauley Heart Center, Virginia Commonwealth University, Richmond (A.A., S.T., J.K.)
| | - Carlo Marchetti
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Richmond, VA (C.M., C.A.D.)
| | - Jordana Kron
- From the VCU Pauley Heart Center, Virginia Commonwealth University, Richmond (A.A., S.T., J.K.)
| | | | - Charles A Dinarello
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Richmond, VA (C.M., C.A.D.)
| |
Collapse
|
54
|
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality in developed countries, and it is the primary cause of mortality in the elderly worldwide. The processes of inflammatory response activation, production and release of pro-inflammatory cytokines, activation of the complement system, synthesis of autoantibodies, and overexpression of Class II major histocompatibility complex molecules contribute to the HF development and progression. High levels of circulating cytokines correlate with the severity of HF, measured with the use of New York Heart Association's classification, and prognosis of the disease. In HF, there is an imbalance between pro-inflammatory and anti-inflammatory cytokines. Concentrations of several interleukins are increased in HF, including IL-1β, IL-6, IL-8, IL-9, IL-10, IL-13, IL-17, and IL-18, whereas the levels of IL-5, IL-7, or IL-33 are down-regulated. Concentrations of inflammatory mediators are associated with cardiac function and can be HF markers and predictors of adverse outcomes or mortality. This review presents the role of interleukins, which contribute to the HF initiation and progression, the importance of their pathways in transition from myocardial injury to HF, and the role of interleukins as markers of disease severity and outcome predictors.
Collapse
|
55
|
Abstract
The observation that heart failure with reduced ejection fraction is associated with elevated circulating levels of pro-inflammatory cytokines opened a new area of research that has revealed a potentially important role for the immune system in the pathogenesis of heart failure. However, until the publication in 2019 of the CANTOS trial findings on heart failure outcomes, all attempts to target inflammation in the heart failure setting in phase III clinical trials resulted in neutral effects or worsening of clinical outcomes. This lack of positive results in turn prompted questions on whether inflammation is a cause or consequence of heart failure. This Review summarizes the latest developments in our understanding of the role of the innate and adaptive immune systems in the pathogenesis of heart failure, and highlights the results of phase III clinical trials of therapies targeting inflammatory processes in the heart failure setting, such as anti-inflammatory and immunomodulatory strategies. The most recent of these studies, the CANTOS trial, raises the exciting possibility that, in the foreseeable future, we might be able to identify those patients with heart failure who have a cardio-inflammatory phenotype and will thus benefit from therapies targeting inflammation.
Collapse
|
56
|
Wang B, Wang ZM, Ji JL, Gan W, Zhang A, Shi HJ, Wang H, Lv L, Li Z, Tang T, Du J, Wang XH, Liu BC. Macrophage-Derived Exosomal Mir-155 Regulating Cardiomyocyte Pyroptosis and Hypertrophy in Uremic Cardiomyopathy. JACC Basic Transl Sci 2020; 5:148-166. [PMID: 32140622 PMCID: PMC7046511 DOI: 10.1016/j.jacbts.2019.10.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 02/08/2023]
Abstract
miR-155 was synthesized and loaded into exosomes in increased infiltration of macrophages in a uremic heart. The released exosomal fusion with the plasma membrane leads to the release of miR-155 into the cytosol and translational repression of forkhead transcription factors of the O class in cardiomyocytes. Macrophage-derived miR-155–containing exosomes promoted cardiomyocyte pyroptosis and uremic cardiomyopathy changes (cardiac hypertrophy and fibrosis) by directly targeting FoxO3a in uremic mice. Inhibiting secretion from macrophage-derived miR-155–containing exosomes represents a novel therapeutic strategy for the management of uremic cardiomyopathy.
miR-155 was synthesized and loaded into exosomes in increased infiltration of macrophages in a uremic heart. The released exosomal fusion with the plasma membrane leads to the release of miR-155 into the cytosol and translational repression of forkhead transcription factors of the O class (FoxO3a) in cardiomyocytes. Finally, macrophage-derived miR-155–containing exosomes promoted cardiomyocyte pyroptosis and uremic cardiomyopathy changes (cardiac hypertrophy and fibrosis) by directly targeting FoxO3a in uremic mice.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Ze-Mu Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jia-Ling Ji
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weihua Gan
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Aiqing Zhang
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao-Jie Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linli Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Zuolin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Taotao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaonan H Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
57
|
Liu D, Dong Z, Xiang F, Liu H, Wang Y, Wang Q, Rao J. Dendrobium Alkaloids Promote Neural Function After Cerebral Ischemia-Reperfusion Injury Through Inhibiting Pyroptosis Induced Neuronal Death in both In Vivo and In Vitro Models. Neurochem Res 2019; 45:437-454. [PMID: 31865520 DOI: 10.1007/s11064-019-02935-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 12/10/2019] [Accepted: 12/15/2019] [Indexed: 12/13/2022]
Abstract
Pyroptosis is a newly identified lytic form of programmed cell death which is characterized by plasma membrane blebbing and rupture. Pyroptosis occurs in cerebral ischemia injury, and contributes to the activation and secretion of the inflammatory cytokines interleukin (IL)-1β, IL-18, and IL-6. Previous reports have found that Dendrobium alkaloids (DNLA) can exert neuroprotective effects against oxygen-glucose deprivation/reperfusion (OGD/R) damage in vitro, but the mechanisms underlying these effects remain elusive. In this study, we investigated whether DNLA exerted therapeutic benefits against cerebral ischemia-reperfusion (CIR) damage via ameliorating pyroptosis and inflammation. OGD/R damage was induced in HT22 cells pretreated with DNLA (0.03, 0.3, or 3 mg/ml, 24 h prior to OGD/R), MCC950 (10 ng/ml, 1 h prior), and VX765 (10 ng/ml, 1 h prior). Neuronal apoptosis, necrosis, pyroptosis, and pathological changes were analyzed 24 h following OGD/R. Further to this, male C57/BL mice pretreated with different concentrations of DNLA (0.5 or 5 mg/kg, ip.) for 24 h and VX765 (50 mg/kg, ip., 1 h before CIR) underwent transient middle cerebral artery occlusion and reperfusion. We found that DNLA pretreatment resulted in a lower neurologic deficit score, a reduced infarct volume, fewer pyroptotic cells, and reduced levels of inflammatory factors 24 h after CIR. Furthermore, DNLA administration also reduced the levels of the pyroptosis-associated proteins Caspase-1 and gasdermin-D, particularly in the hippocampal CA1 region. Similar decreases were observed in the levels of the inflammatory factors IL-1β, IL-6, and IL-18. OGD/R-associated ultrastructural damage was seen to improve following DNLA administration, likely due to the regulation of the tight junction protein Pannexin-1 by DNLA. Overall, these findings demonstrate that DNLA can protect against CIR damage through inhibiting pyroptosis-induced neuronal death, providing new therapeutic insights for CIR injury.
Collapse
Affiliation(s)
- Daohang Liu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, China
| | - Zhi Dong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, China.
| | - Fei Xiang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, China
| | - Hailin Liu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, China
| | - Yuchun Wang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, China
| | - Qian Wang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, China
| | - Jiangyan Rao
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, China
| |
Collapse
|
58
|
Shen Z, Lu J, Wei J, Zhao J, Wang M, Wang M, Shen X, Lü X, Zhou B, Zhao Y, Fu G. Investigation of the underlying hub genes and mechanisms of reperfusion injury in patients undergoing coronary artery bypass graft surgery by integrated bioinformatic analyses. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:664. [PMID: 31930065 DOI: 10.21037/atm.2019.10.43] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Although coronary artery bypass graft (CABG) surgery is the main method to revascularize the occluded coronary vessels in coronary artery diseases, the full benefits of the operation are mitigated by ischemia-reperfusion (IR) injury. Although many studies have been devoted to reducing IR injury in animal models, the translation of this research into the clinical field has been disappointing. Our study aimed to explore the underlying hub genes and mechanisms of IR injury. Methods A weighted gene co-expression network analysis (WGCNA) was executed based on the expression profiles in patients undergoing CABG surgery (GSE29396). Functional annotation and protein-protein interaction (PPI) network construction were executed within the modules of interest. Potential hub genes were predicted, combining both intramodular connectivity (IC) and degrees. Meanwhile, potential transcription factors (TFs) and microRNAs (miRNAs) were predicted by corresponding bioinformatics tools. Results A total of 336 differentially expressed genes (DEGs) were identified. DEGs were mainly enriched in neutrophil activity and immune response. Within the modules of interest, 5 upregulated hub genes (IL-6, CXCL8, IL-1β, MYC, PTGS-2) and 6 downregulated hub genes (C3, TIMP1, VSIG4, SERPING1, CD163, and HP) were predicted. Predicted miRNAs (hsa-miR-333-5p, hsa-miR-26b-5p, hsa-miR-124-3p, hsa-miR-16-5p, hsa-miR-98-5p, hsa-miR-17-5p, hsa-miR-93-5p) and TF (STAT1) might have regulated gene expression in the most positively related module, while hsa-miR-333-5p and HSF-1 were predicted to regulate the genes within the most negatively related module. Conclusions Our study illustrates an overview of gene expression changes in human atrial samples from patients undergoing CABG surgery and might help translate future research into clinical work.
Collapse
Affiliation(s)
- Zhida Shen
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jiangting Lu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jiejin Wei
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.,Department of Electrocardiogram, Shaoxing People's Hospital, Shaoxing 312000, China
| | - Juanjuan Zhao
- Biomedical Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Meihui Wang
- Biomedical Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Ming Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xiaohua Shen
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xue Lü
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Binquan Zhou
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yanbo Zhao
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| |
Collapse
|
59
|
Suetomi T, Miyamoto S, Brown JH. Inflammation in nonischemic heart disease: initiation by cardiomyocyte CaMKII and NLRP3 inflammasome signaling. Am J Physiol Heart Circ Physiol 2019; 317:H877-H890. [PMID: 31441689 PMCID: PMC6879920 DOI: 10.1152/ajpheart.00223.2019] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/09/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
There is substantial evidence that chronic heart failure in humans and in animal models is associated with inflammation. Ischemic interventions such as myocardial infarction lead to necrotic cell death and release of damage associated molecular patterns, factors that signal cell damage and induce expression of proinflammatory chemokines and cytokines. It has recently become evident that nonischemic interventions are also associated with increases in inflammatory genes and immune cell accumulation in the heart and that these contribute to fibrosis and ventricular dysfunction. How proinflammatory responses are elicited in nonischemic heart disease which is not, at least initially, associated with cell death is a critical unanswered question. In this review we provide evidence supporting the hypothesis that cardiomyocytes are an initiating site of inflammatory gene expression in response to nonischemic stress. Furthermore we discuss the role of the multifunctional Ca2+/calmodulin-regulated kinase, CaMKIIδ, as a transducer of stress signals to nuclear factor-κB activation, expression of proinflammatory cytokines and chemokines, and priming and activation of the NOD-like pyrin domain-containing protein 3 (NLRP3) inflammasome in cardiomyocytes. We summarize recent evidence that subsequent macrophage recruitment, fibrosis and contractile dysfunction induced by angiotensin II infusion or transverse aortic constriction are ameliorated by blockade of CaMKII, of monocyte chemoattractant protein-1/C-C chemokine receptor type 2 signaling, or of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Takeshi Suetomi
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Shigeki Miyamoto
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Joan Heller Brown
- Department of Pharmacology, University of California San Diego, La Jolla, California
| |
Collapse
|
60
|
Mühl H, Bachmann M. IL-18/IL-18BP and IL-22/IL-22BP: Two interrelated couples with therapeutic potential. Cell Signal 2019; 63:109388. [PMID: 31401146 DOI: 10.1016/j.cellsig.2019.109388] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
Abstract
Interleukin (IL)-18 and IL-22 are key components of cytokine networks that play a decisive role in (pathological) inflammation, host defense, and tissue regeneration. Tight regulation of cytokine-driven signaling, inflammation, and immunoactivation is supposed to enable nullification of a given deleterious trigger without mediating overwhelming collateral tissue damage or even activating a cancerous face of regeneration. In fact, feedback regulation by specific cytokine opponents is regarded as a major means by which the immune system is kept in balance. Herein, we shine a light on the interplay between IL-18 and IL-22 and their opponents IL-18 binding protein (IL-18BP) and IL-22BP in order to provide integrated information on their biology, pathophysiological significance, and prospect as targets and/or instruments of therapeutic intervention.
Collapse
Affiliation(s)
- Heiko Mühl
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany.
| | - Malte Bachmann
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
61
|
Pfeiler S, Winkels H, Kelm M, Gerdes N. IL-1 family cytokines in cardiovascular disease. Cytokine 2019; 122:154215. [DOI: 10.1016/j.cyto.2017.11.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022]
|
62
|
Suetomi T, Willeford A, Brand CS, Cho Y, Ross RS, Miyamoto S, Brown JH. Inflammation and NLRP3 Inflammasome Activation Initiated in Response to Pressure Overload by Ca 2+/Calmodulin-Dependent Protein Kinase II δ Signaling in Cardiomyocytes Are Essential for Adverse Cardiac Remodeling. Circulation 2019; 138:2530-2544. [PMID: 30571348 DOI: 10.1161/circulationaha.118.034621] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Inflammation is associated with cardiac remodeling and heart failure, but how it is initiated in response to nonischemic interventions in the absence of cell death is not known. We tested the hypothesis that activation of Ca2+/calmodulin-dependent protein kinase II δ (CaMKIIδ) in cardiomyocytes (CMs) in response to pressure overload elicits inflammatory responses leading to adverse remodeling. METHODS Mice in which CaMKIIδ was selectively deleted from CMs (cardiac-specific knockout [CKO]) and floxed control mice were subjected to transverse aortic constriction (TAC). The effects of CM-specific CaMKIIδ deletion on inflammatory gene expression, inflammasome activation, macrophage accumulation, and fibrosis were assessed by quantitative polymerase chain reaction, histochemistry, and ventricular remodeling by echocardiography. RESULTS TAC induced increases in cardiac mRNA levels for proinflammatory chemokines and cytokines in ≤3 days, and these responses were significantly blunted when CM CaMKIIδ was deleted. Apoptotic and necrotic cell death were absent at this time. CMs isolated from TAC hearts mirrored these robust increases in gene expression, which were markedly attenuated in CKO. Priming and activation of the NOD-like receptor pyrin domain-containing protein 3 inflammasome, assessed by measuring interleukin-1β and NOD-like receptor pyrin domain-containing protein 3 mRNA levels, caspase-1 activity, and interleukin-18 cleavage, were increased at day 3 after TAC in control hearts and in CMs isolated from these hearts. These responses were dependent on CaMKIIδ and associated with activation of Nuclear Factor-kappa B and reactive oxygen species. Accumulation of macrophages observed at days 7 to 14 after TAC was diminished in CKO and, by blocking Monocyte Chemotactic Protein-1 signaling, deletion of CM Monocyte Chemotactic Protein-1 or inhibition of inflammasome activation. Fibrosis was also attenuated by these interventions and in the CKO heart. Ventricular dilation and contractile dysfunction observed at day 42 after TAC were diminished in the CKO. Inhibition of CaMKII, Nuclear Factor-kappa B, inflammasome, or Monocyte Chemotactic Protein-1 signaling in the first 1 or 2 weeks after TAC decreased remodeling, but inhibition of CaMKII after 2 weeks did not. CONCLUSIONS Activation of CaMKIIδ in response to pressure overload triggers inflammatory gene expression and activation of the NOD-like receptor pyrin domain-containing protein 3 inflammasome in CMs. These responses provide signals for macrophage recruitment, fibrosis, and myocardial dysfunction in the heart. Our work suggests the importance of targeting early inflammatory responses induced by CM CaMKIIδ signaling to prevent progression to heart failure.
Collapse
Affiliation(s)
- Takeshi Suetomi
- Department of Pharmacology (T.S., A.W., C.S.B., S.M., J.H.B.), University of California San Diego, La Jolla
| | - Andrew Willeford
- Department of Pharmacology (T.S., A.W., C.S.B., S.M., J.H.B.), University of California San Diego, La Jolla
| | - Cameron S Brand
- Department of Pharmacology (T.S., A.W., C.S.B., S.M., J.H.B.), University of California San Diego, La Jolla
| | - Yoshitake Cho
- Department of Medicine, Division of Cardiovascular Medicine (Y.C., R.S.R.), University of California San Diego, La Jolla
| | - Robert S Ross
- Department of Medicine, Division of Cardiovascular Medicine (Y.C., R.S.R.), University of California San Diego, La Jolla.,Veterans Administration Healthcare System, San Diego, CA (R.S.R.)
| | - Shigeki Miyamoto
- Department of Pharmacology (T.S., A.W., C.S.B., S.M., J.H.B.), University of California San Diego, La Jolla
| | - Joan Heller Brown
- Department of Pharmacology (T.S., A.W., C.S.B., S.M., J.H.B.), University of California San Diego, La Jolla
| |
Collapse
|
63
|
The IL-1 family of cytokines and receptors in rheumatic diseases. Nat Rev Rheumatol 2019; 15:612-632. [DOI: 10.1038/s41584-019-0277-8] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
64
|
Abstract
The natural history of heart failure (HF) is not linear, because changes in the heart structure and function start long before the disease becomes clinically evident. Many different cytokines originating from intracardiac tissues (cardiomyocytes, cardiac endothelial cells, cardiac fibroblasts, and cardiac infiltrated immune cells) or extracardiac tissues (adipose tissue, gut, and lymphoid organs) have been identified in HF. Because the levels of circulating cytokines correlate with the development and severity of HF, these mediators may have both pathophysiological importance, through their ability to modulate inflammation, myocyte stress/stretch, myocyte injury and apoptosis, fibroblast activation and extracellular matrix remodeling, and utility as clinical predictive biomarkers. A greater understanding of the mechanisms mediated by the multifaceted network of cytokines, leading to distinct HF phenotypes (HF with reduced or preserved ejection fraction), is urgently needed for the development of new treatment strategies. In this chapter, all these issues were thoroughly discussed, pointing on the practical considerations concerning the clinical use of the cytokines as prognostic biomarkers and potential therapeutic targets in HF.
Collapse
Affiliation(s)
- Adina Elena Stanciu
- Department of Carcinogenesis and Molecular Biology, Institute of Oncology Bucharest, Bucharest, Romania.
| |
Collapse
|
65
|
Gao R, Shi H, Chang S, Gao Y, Li X, Lv C, Yang H, Xiang H, Yang J, Xu L, Tang Y. The selective NLRP3-inflammasome inhibitor MCC950 reduces myocardial fibrosis and improves cardiac remodeling in a mouse model of myocardial infarction. Int Immunopharmacol 2019; 74:105575. [PMID: 31299609 DOI: 10.1016/j.intimp.2019.04.022] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIMS Early inflammatory responses after myocardial infarction (MI) are likely to increase myocardial fibrosis and subsequent cardiac remodeling. MCC950, a specific NLRP3 inhibitor, was previously found to effectively inhibit the release of inflammatory factors IL-18 and IL-1β. In this study, we evaluated the effect of MCC950, as a potential new treatment strategy for MI, on myocardial fibrosis and cardiac remodeling using an experimental mouse model. METHODS Male C57BL/6 mice were subjected to left coronary artery ligation to induce MI and then treated with MCC950 (10 mg/kg) or PBS for 14 days. After 30 days, echocardiography was performed to assess cardiac function and myocardial fibrosis was evaluated using H&E- and Masson's Trichrome-stained sections. Myocardial expression of inflammatory factors and fibrosis markers was analyzed by western blotting, immunofluorescence, ELISA, and real-time quantitative PCR. RESULTS The ejection fraction in the 10 mg/kg group (40.7 ± 4.2%; N = 6, p = 0.0029) was statistically preserved compared to that in the control group (14.0 ± 4.4%). Myocardial fibrosis was also reduced in MCC950-treated animals (MCC950, 23.2 ± 3.0 vs PBS, 36.2 ± 3.7; p < 0.05). Moreover, myocardial NLRP3, cleaved IL-1β, and IL-18 levels were reduced in MCC950-treated animals. H&E and molecular examination revealed decreases in inflammatory cell infiltration and inflammatory factor expression in the heart. In vitro, MCC950 inhibited NLRP3, reduced caspase-1 activity, and further downregulated IL-1β and IL-18. CONCLUSION MCC950, as a specific NLRP3 inhibitor, can alleviate fibrosis and improve cardiac function in a mouse model by suppressing early inflammatory responses post-MI.
Collapse
Affiliation(s)
- Rifeng Gao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China
| | - Huairui Shi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suchi Chang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Gao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyu Lv
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China
| | - Heng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China
| | - Haiyan Xiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China
| | - Juesheng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China
| | - Lei Xu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yanhua Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
66
|
Place DE, Kanneganti TD. Cell death-mediated cytokine release and its therapeutic implications. J Exp Med 2019; 216:1474-1486. [PMID: 31186281 PMCID: PMC6605758 DOI: 10.1084/jem.20181892] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
Targeting apoptosis to treat diseases has seen tremendous success over the past decades. More recently, alternative forms of regulated cell death, including pyroptosis and necroptosis, have been described. Understanding the molecular cascades regulating both pyroptosis and necroptosis will yield even more targets to treat diseases. These lytic forms of cell death are distinct from apoptosis due to their characteristic lysis and release of cellular components that promote disease or direct a beneficial immune response. In this review, we focus on how pyroptosis and necroptosis, which release potent immune cytokines such as IL-1 and IL-18, contribute to various diseases. We also consider the important role that the executioners of these cell death pathways, GSDMD and MLKL, play in the progression of inflammatory diseases. Crosstalk between the different cell death pathways likely plays a major role physiologically. New therapeutic strategies targeting these specific molecules hold enormous potential for managing inflammatory diseases.
Collapse
Affiliation(s)
- David E Place
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
67
|
Carbone S, Canada JM, Billingsley HE, Siddiqui MS, Elagizi A, Lavie CJ. Obesity paradox in cardiovascular disease: where do we stand? Vasc Health Risk Manag 2019; 15:89-100. [PMID: 31118651 PMCID: PMC6503652 DOI: 10.2147/vhrm.s168946] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
Obesity is associated with an increased risk of developing cardiovascular disease (CVD), particularly heart failure (HF) and coronary heart disease (CHD). The mechanisms through which obesity increases CVD risk involve changes in body composition that can affect hemodynamics and alters heart structure. Pro-inflammatory cytokines produced by the adipose tissue itself which can induce cardiac dysfunction and can promote the formation of atherosclerotic plaques. When obesity and HF or CHD coexist, individuals with class I obesity present a more favorable prognosis compared to individuals who are normal or underweight. This phenomenon has been termed the "obesity paradox." Obesity is defined as an excess fat mass (FM), but individuals with obesity typically also present with an increased amount of lean mass (LM). The increase in LM may explain part of the obesity paradox as it is associated with improved cardiorespiratory fitness (CRF), a major determinant of clinical outcomes in the general population, but particularly in those with CVD, including HF. While increased LM is a stronger prognosticator in HF compared to FM, in patients with CHD excess FM can exert protective effects particularly when not associated with increased systemic inflammation. In the present review, we discuss the mechanisms through which obesity may increase the risk for CVD, and how it may exert protective effects in the setting of established CVD, with a focus on body composition. We also highlight the importance of measuring or estimating CRF, including body composition-adjusted measures of CRF (ie, lean peak oxygen consumption) for an improved risk status stratification in patients with CVD and finally, we discuss the potential non-pharmacologic therapeutics, such as exercise training and dietary interventions, aimed at improving CRF and perhaps clinical outcomes.
Collapse
Affiliation(s)
- Salvatore Carbone
- VCU Pauley Heart Center, Department of Internal Medicine, Richmond, VA, USA
| | - Justin M Canada
- VCU Pauley Heart Center, Department of Internal Medicine, Richmond, VA, USA
| | - Hayley E Billingsley
- VCU Pauley Heart Center, Department of Internal Medicine, Richmond, VA, USA
- Kinesiology and Health Sciences, College of Humanities & Science, Richmond, VA, USA
| | - Mohammad S Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Elagizi
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-the University of Queensland School of Medicine, New Orleans, LA, USA
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-the University of Queensland School of Medicine, New Orleans, LA, USA
| |
Collapse
|
68
|
|
69
|
Cavalli G, Dinarello CA. Anakinra Therapy for Non-cancer Inflammatory Diseases. Front Pharmacol 2018; 9:1157. [PMID: 30459597 PMCID: PMC6232613 DOI: 10.3389/fphar.2018.01157] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/24/2018] [Indexed: 12/14/2022] Open
Abstract
Interleukin-1 (IL-1) is the prototypical inflammatory cytokine: two distinct ligands (IL-1α and IL-1β) bind the IL-1 type 1 receptor (IL-1R1) and induce a myriad of secondary inflammatory mediators, including prostaglandins, cytokines, and chemokines. IL-1α is constitutively present in endothelial and epithelial cells, whereas IL-1β is inducible in myeloid cells and released following cleavage by caspase-1. Over the past 30 years, IL-1-mediated inflammation has been established in a broad spectrum of diseases, ranging from rare autoinflammatory diseases to common conditions such as gout and rheumatoid arthritis (RA), type 2 diabetes, atherosclerosis, and acute myocardial infarction. Blocking IL-1 entered the clinical arena with anakinra, the recombinant form of the naturally occurring IL-1 receptor antagonist (IL-1Ra); IL-1Ra prevents the binding of IL-1α as well as IL-1β to IL-1R1. Quenching IL-1-mediated inflammation prevents the detrimental consequences of tissue damage and organ dysfunction. Although anakinra is presently approved for the treatment of RA and cryopyrin-associated periodic syndromes, off-label use of anakinra far exceeds its approved indications. Dosing of 100 mg of anakinra subcutaneously provides clinically evident benefits within days and for some diseases, anakinra has been used daily for over 12 years. Compared to other biologics, anakinra has an unparalleled record of safety: opportunistic infections, particularly Mycobacterium tuberculosis, are rare even in populations at risk for reactivation of latent infections. Because of this excellent safety profile and relative short duration of action, anakinra can also be used as a diagnostic tool for undefined diseases mediated by IL-1. Although anakinra is presently in clinical trials to treat cancer, this review focuses on anakinra treatment of acute as well as chronic inflammatory diseases.
Collapse
Affiliation(s)
- Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
- Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Charles A. Dinarello
- Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Medicine, University of Colorado Denver, Denver, CO, United States
| |
Collapse
|
70
|
Seropian IM, González GE, Maller SM, Berrocal DH, Abbate A, Rabinovich GA. Galectin-1 as an Emerging Mediator of Cardiovascular Inflammation: Mechanisms and Therapeutic Opportunities. Mediators Inflamm 2018; 2018:8696543. [PMID: 30524200 PMCID: PMC6247465 DOI: 10.1155/2018/8696543] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/30/2018] [Indexed: 01/07/2023] Open
Abstract
Galectin-1 (Gal-1), an evolutionarily conserved β-galactoside-binding lectin, controls immune cell homeostasis and tempers acute and chronic inflammation by blunting proinflammatory cytokine synthesis, engaging T-cell apoptotic programs, promoting expansion of T regulatory (Treg) cells, and deactivating antigen-presenting cells. In addition, this lectin promotes angiogenesis by co-opting the vascular endothelial growth factor receptor (VEGFR) 2 signaling pathway. Since a coordinated network of immunomodulatory and proangiogenic mediators controls cardiac homeostasis, this lectin has been proposed to play a key hierarchical role in cardiac pathophysiology via glycan-dependent regulation of inflammatory responses. Here, we discuss the emerging roles of Gal-1 in cardiovascular diseases including acute myocardial infarction, heart failure, Chagas cardiomyopathy, pulmonary hypertension, and ischemic stroke, highlighting underlying anti-inflammatory mechanisms and therapeutic opportunities. Whereas Gal-1 administration emerges as a potential novel treatment option in acute myocardial infarction and ischemic stroke, Gal-1 blockade may contribute to attenuate pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Ignacio M. Seropian
- Servicio de Hemodinamia y Cardiología Intervencionista, Instituto de Medicina Cardiovascular, Hospital Italiano de Buenos Aires, Buenos Aires C1199, Argentina
| | - Germán E. González
- Instituto de Biología y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiopatología Cardiovascular, Departamento de Patología, Universidad de Buenos Aires, Buenos Aires C1428, Argentina
| | - Sebastián M. Maller
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
| | - Daniel H. Berrocal
- Servicio de Hemodinamia y Cardiología Intervencionista, Instituto de Medicina Cardiovascular, Hospital Italiano de Buenos Aires, Buenos Aires C1199, Argentina
| | - Antonio Abbate
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Gabriel A. Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428, Argentina
| |
Collapse
|
71
|
Gomez I, Duval V, Silvestre JS. Cardiomyocytes and Macrophages Discourse on the Method to Govern Cardiac Repair. Front Cardiovasc Med 2018; 5:134. [PMID: 30333983 PMCID: PMC6175999 DOI: 10.3389/fcvm.2018.00134] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022] Open
Abstract
In response to pathophysiological stress, the cardiac tissue undergoes profound remodeling process that incorporates the elimination of dying resident cells, compensatory hypertrophy of functional cardiomyocytes, growth and remodeling of the vascular compartment and formation of a fibrotic scar. Accumulating evidences indicate that cardiac remodeling is, at least in part, controlled by a complex crosstalk between cardiomyocytes and macrophages. The strategic location of abundant macrophages to the proximity of cardiomyocytes suggest that they could regulate the fate of cardiomyocytes in the injured heart. As such, macrophages appear as critical support cells for cardiomyocytes and play central roles in cardiac hypertrophy, fibrosis and remodeling. Notably, the cardiac tissue expands heterogeneous population of cardiac macrophages through local proliferation of resident macrophage as well as recruitment and differentiation of blood-derived monocytes. It has also been suggested that cardiac-resident macrophages display distinct functional properties from that of monocyte-derived macrophages in cardiac tissue. Furthermore, macrophages are an overflowing source of biological entities with non-canonical roles on cardiac conduction or cardiomyocyte proliferation by regulating action potential diffusion or cardiac cell cycle reentry. Alternatively, stressed cardiomyocytes can trigger the release of a broad repertoire of instructive signals that can regulate macrophage number, skew their phenotype and therefore direct their beneficial or deleterious actions. In this review, we highlight recent discoveries describing how the intricate dialogue between cardiomyocytes and macrophages can shape the deleterious or healing signaling mechanisms in the injured cardiac tissue.
Collapse
Affiliation(s)
- Ingrid Gomez
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Vincent Duval
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
72
|
Butts B, Butler J, Dunbar SB, Corwin E, Gary RA. Effects of Exercise on ASC Methylation and IL-1 Cytokines in Heart Failure. Med Sci Sports Exerc 2018; 50:1757-1766. [PMID: 29683921 PMCID: PMC6095733 DOI: 10.1249/mss.0000000000001641] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION/PURPOSE Inflammation contributes to heart failure (HF) progression and the interleukin (IL)-1 cytokine IL-1β is implicated in this process. The adaptor protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) is necessary for inflammasome activation of IL-1β. Lower ASC methylation is associated with worse outcomes in HF. The purpose of this study was to examine the effects of exercise on changes in ASC methylation and activation of the IL-1 family cytokine IL-1β in persons with HF. METHODS Participants (N = 54) were randomized to receive exercise intervention (n = 38) or attention control (n = 16) for 3 months. Percent methylation of the ASC gene, plasma IL-1β, and ASC mRNA and were obtained at baseline, 3 months, and 6 months. RESULTS ASC methylation was higher in the exercise group as compared to control at 3 months (6.10% ± 0.5% vs 5.80% ± 0.4%; P = 0.04) and 6 months (6.07 ± 0.4 vs 5.82 ± 0.4; P = 0.04). Plasma IL-1β was lower in the exercise group at 3 months (1.43 ± 0.5 pg·mL vs 2.09 ± 1.3 pg·mL; P = 0.02) and 6 months (1.49 ± 0.5 pg·mL vs 2.13 ± 1.4 pg·mL; P = 0.004). ASC mRNA expression was negatively associated with ASC methylation at baseline (r = -0.97, P = 0.001), 3 months (r = -0.90, P = 0.001), and 6 months (r = -0.81, P = 0.001). ASC mRNA was lower than baseline at 3 months (P = 0.004) and 6 months (P = 0.002) among those in the exercise group. ASC methylation was positively associated with 6-min walk test at baseline (r = 0.517, P < 0.001), 3 months (r = 0.464, P = 0.004), and 6 months (r = 497, P = 0.05). CONCLUSIONS Exercise was related to increased mean percent ASC methylation and decreased IL-1β and ASC mRNA gene expression in HF. Epigenetic regulation of ASC can be a biological mechanism by which exercise can promote better outcomes in HF.
Collapse
Affiliation(s)
- Brittany Butts
- Division of Cardiovascular Disease, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Javed Butler
- Division of Cardiology, Stony Brook University, Stony Brook, NY
| | - Sandra B Dunbar
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | - Elizabeth Corwin
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | - Rebecca A Gary
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| |
Collapse
|
73
|
Van Tassell BW, Trankle CR, Canada JM, Carbone S, Buckley L, Kadariya D, Del Buono MG, Billingsley H, Wohlford G, Viscusi M, Oddi-Erdle C, Abouzaki NA, Dixon D, Biondi-Zoccai G, Arena R, Abbate A. IL-1 Blockade in Patients With Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2018; 11:e005036. [PMID: 30354558 PMCID: PMC6545106 DOI: 10.1161/circheartfailure.118.005036] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 07/13/2018] [Indexed: 02/05/2023]
Abstract
Background Enhanced inflammation may lead to exercise intolerance in heart failure with preserved ejection fraction. The aim of the current study was to determine whether IL (interleukin)-1 blockade with anakinra improved cardiorespiratory fitness in heart failure with preserved ejection fraction. Methods and Results Thirty-one patients with heart failure with preserved ejection fraction and CRP (C-reactive protein) >2 mg/L were randomized to anakinra (100 mg subcutaneously daily, N=21) or placebo (N=10) for 12 weeks. We measured peak oxygen consumption (Vo2), ventilatory efficiency (VE/Vco2 slope), and high-sensitivity CRP and NT-proBNP (N-terminal pro-B-type natriuretic peptide) at 4, 12, and 24 weeks. Twenty-eight patients completed ≥2 visits, 18 women (64%), 27 (96%) obese. There were no differences in peak Vo2 or VE/Vco2 slope between groups at baseline. Peak Vo2 was not changed after 12 weeks of anakinra (from 13.6 [11.8-18.0] to 14.2 [11.2-18.5] mL·kg-1·min-1, P=0.89), or placebo (14.9 [11.7-17.2] to 15.0 [13.8-16.9] mL·kg-1·min-1, P=0.40), without significant between-group differences in changes at 12 weeks (-0.4 [95% CI, -2.2 to +1.4], P=0.64). VE/Vco2 slope was also unchanged with anakinra (from 28.3 [27.2-33.0] to 30.5 [26.3-32.8], P=0.97) or placebo (from 31.6 [27.3-36.9] to 31.2 [27.8-33.4], P=0.78), without significant between-group differences in changes at 12 weeks (+1.2 [95% CI, -1.8 to +4.3], P=0.97). Within the anakinra-treated patients, high-sensitivity CRP and NT-proBNP levels were lower at 4 weeks compared with baseline ( P=0.026 and P=0.022 versus placebo [between-group analysis], respectively). Conclusions Treatment with anakinra for 12 weeks failed to improve peak Vo2 and VE/Vco2 slope in a group of obese heart failure with preserved ejection fraction patients. The favorable trends in high-sensitivity CRP and NT-proBNP with anakinra deserve exploration in future studies. Clinical Trial Registration URL: https://www.clinicaltrials.gov . Unique identifier: NCT02173548.
Collapse
Affiliation(s)
- Benjamin W Van Tassell
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond (B.W.V.T., L.B., G.W., D.D.)
| | - Cory R Trankle
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Justin M Canada
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Salvatore Carbone
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Leo Buckley
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond (B.W.V.T., L.B., G.W., D.D.)
| | - Dinesh Kadariya
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Marco G Del Buono
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Hayley Billingsley
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - George Wohlford
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond (B.W.V.T., L.B., G.W., D.D.)
| | - Michele Viscusi
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Claudia Oddi-Erdle
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Nayef A Abouzaki
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| | - Dave Dixon
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond (B.W.V.T., L.B., G.W., D.D.)
| | - Giuseppe Biondi-Zoccai
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.)
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (G.B.-Z.)
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago (R.A.)
| | - Antonio Abbate
- Division of Cardiology, Virginia Commonwealth University Pauley Heart Center, Richmond (B.W.V.T., C.R.T., J.C., S.C., D.K., M.G.D.B., H.B., G.W., M.V., C.O.-E., N.A.A., D.D., A.A.)
| |
Collapse
|
74
|
Abstract
Initially described as an interferon (IFN)γ‐inducing factor, interleukin (IL)‐18 is indeed involved in Th1 and NK cell activation, but also in Th2, IL‐17‐producing γδ T cells and macrophage activation. IL‐18, a member of the IL‐1 family, is similar to IL‐1β for being processed by caspase 1 to an 18 kDa‐biologically active mature form. IL‐18 binds to its specific receptor (IL‐18Rα, also known as IL‐1R7) forming a low affinity ligand chain. This is followed by recruitment of the IL‐18Rβ chain. IL‐18 then uses the same signaling pathway as IL‐1 to activate NF‐kB and induce inflammatory mediators such as adhesion molecules, chemokines and Fas ligand. IL‐18 also binds to the circulating high affinity IL‐18 binding protein (BP), such as only unbound free IL‐18 is active. IL‐18Rα may also bind IL‐37, another member of the IL‐1 family, but in association with the negative signaling chain termed IL‐1R8, which transduces an anti‐inflammatory signal. IL‐18BP also binds IL‐37 and this acts as a sink for the anti‐inflammatory properties of IL‐37. There is now ample evidence for a role of IL‐18 in various infectious, metabolic or inflammatory diseases such as influenza virus infection, atheroma, myocardial infarction, chronic obstructive pulmonary disease, or Crohn's disease. However, IL‐18 plays a very specific role in the pathogenesis of hemophagocytic syndromes (HS) also termed Macrophage Activation Syndrome. In children affected by NLRC4 gain‐of‐function mutations, IL‐18 circulates in the range of tens of nanograms/mL. HS is treated with the IL‐1 Receptor antagonist (anakinra) but also specifically with IL‐18BP. Systemic juvenile idiopathic arthritis or adult‐onset Still's disease are also characterized by high serum IL‐18 concentrations and are treated by IL‐18BP.
Collapse
Affiliation(s)
- Gilles Kaplanski
- Assistance Publique-Hôpitaux de Marseille, Centre Hospitalier Universitaire Conception, Service de Médecine Interne et Immunologie Clinique, Aix-Marseille Université, Marseille, France.,Vascular Research Center Marseille, Faculté de Pharmacie, Aix-Marseille Université, INSERM UMR_S1076, Marseille, France
| |
Collapse
|
75
|
De Luca G, Cavalli G, Campochiaro C, Tresoldi M, Dagna L. Myocarditis: An Interleukin-1-Mediated Disease? Front Immunol 2018; 9:1335. [PMID: 29951067 PMCID: PMC6008311 DOI: 10.3389/fimmu.2018.01335] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital and Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
76
|
Dinarello CA, Kaplanski G. Indeed, IL-18 is more than an inducer of IFN-γ. J Leukoc Biol 2018; 104:237-238. [PMID: 29733453 DOI: 10.1002/jlb.ce0118-025rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 04/06/2018] [Accepted: 04/12/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Charles Anthony Dinarello
- Department of Medicine, University of Colorado, Aurora, Colorado, USA.,Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gilles Kaplanski
- Aix-Marseille Université, Centre Hospitalier Universitaire Conception, Service de Médecine Interne et Immunologie Clinique, Marseille, France
| |
Collapse
|
77
|
Valle Raleigh J, Mauro AG, Devarakonda T, Marchetti C, He J, Kim E, Filippone S, Das A, Toldo S, Abbate A, Salloum FN. Reperfusion therapy with recombinant human relaxin-2 (Serelaxin) attenuates myocardial infarct size and NLRP3 inflammasome following ischemia/reperfusion injury via eNOS-dependent mechanism. Cardiovasc Res 2018; 113:609-619. [PMID: 28073832 DOI: 10.1093/cvr/cvw246] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/01/2016] [Indexed: 11/13/2022] Open
Abstract
Aims The preconditioning-like infarct-sparing and anti-inflammatory effects of the peptide hormone relaxin following ischemic injury have been studied in the heart. Whether reperfusion therapy with recombinant human relaxin-2, serelaxin, reduces myocardial infarct size and attenuates the subsequent NLRP3 inflammasome activation leading to further loss of functional myocardium following ischemia/reperfusion (I/R) injury is unknown. Methods and results After baseline echocardiography, adult male wild-type C57BL or eNOS knockout mice underwent myocardial infarction (MI) by coronary artery ligation for 30 min followed by 24 h reperfusion. Mice were treated with either serelaxin (10 µg/kg; sc) or saline 1 h prior to ischemia or 5 min before reperfusion. In both pre-treatment and reperfusion therapy arms, serelaxin improved survival at 24 h post MI in wild-type mice (79% and 82%) as compared with controls (46% and 50%, P = 0.01), whereas there was no difference in survival between serelaxin- and saline-treated eNOS knockout mice. Moreover, serelaxin significantly reduced infarct size (64% and 67% reduction, P < 0.05), measured with TTC staining, and preserved LV fractional shortening (FS) and end-systolic diameter (LVESD) in wild-type mice as compared with controls (P < 0.05). Interestingly, caspase-1 activity in the heart tissue, a measure of inflammasome formation, was markedly reduced in serelaxin-treated wild-type mice compared with controls at 24 h post-MI in both treatment modalities (P < 0.05). Genetic deletion of eNOS abolished the infarct-sparing and anti-inflammatory effects of serelaxin as well as functional preservation. Serelaxin plasma levels assessed at 5 min and 1 h after treatment, using ELISA, approximated physiologic relaxin levels during pregnancy in mice and parallels that in humans. Conclusion Serelaxin attenuates myocardial I/R injury and the subsequent caspase-1 activation via eNOS-dependent mechanism.
Collapse
Affiliation(s)
- Juan Valle Raleigh
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Adolfo G Mauro
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Teja Devarakonda
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Carlo Marchetti
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Jun He
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Erica Kim
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Scott Filippone
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Anindita Das
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Stefano Toldo
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| | - Fadi N Salloum
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Richmond, VA 23298-0204, USA
| |
Collapse
|
78
|
van Hout GPJ, Bosch L, Ellenbroek GHJM, de Haan JJ, van Solinge WW, Cooper MA, Arslan F, de Jager SCA, Robertson AAB, Pasterkamp G, Hoefer IE. The selective NLRP3-inflammasome inhibitor MCC950 reduces infarct size and preserves cardiac function in a pig model of myocardial infarction. Eur Heart J 2018; 38:828-836. [PMID: 27432019 DOI: 10.1093/eurheartj/ehw247] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 04/29/2016] [Indexed: 01/15/2023] Open
Abstract
Aims Myocardial infarction (MI) triggers an intense inflammatory response that is associated with infarct expansion and is detrimental for cardiac function. Interleukin (IL)-1β and IL-18 are key players in this response and are controlled by the NLRP3-inflammasome. In the current study, we therefore hypothesized that selective inhibition of the NLRP3-inflammasome reduces infarct size and preserves cardiac function in a porcine MI model. Methods and results Thirty female landrace pigs were subjected to 75 min transluminal balloon occlusion and treated with the NLRP3-inflammasome inhibitor MCC950 (6 or 3 mg/kg) or placebo for 7 days in a randomized, blinded fashion. After 7 days, 3D-echocardiography was performed to assess cardiac function and Evans blue/TTC double staining was executed to assess the area at risk (AAR) and infarct size (IS). The IS/AAR was lower in the 6 mg/kg group (64.6 ± 8.8%, P = 0.004) and 3 mg/kg group (69.7 ± 7.2%, P = 0.038) compared with the control group (77.5 ± 6.3%). MCC950 treatment markedly preserved left ventricular ejection fraction in treated animals (6 mg/kg 47 ± 8%, P = 0.001; 3 mg/kg 45 ± 7%, P = 0.031; control 37 ± 6%). Myocardial neutrophil influx was attenuated in treated compared with non-treated animals (6 mg/kg 132 ± 72 neutrophils/mm2, P = 0.035; 3 mg/kg 207 ± 210 neutrophils/mm2, P = 0.5; control 266 ± 158 neutrophils/mm2). Myocardial IL-1β levels were dose-dependently reduced in treated animals. Conclusions NLRP3-inflammasome inhibition reduces infarct size and preserves cardiac function in a randomized, blinded translational large animal MI model. Hence, NLRP3-inflammasome inhibition may have therapeutic potential in acute MI patients.
Collapse
Affiliation(s)
- Gerardus P J van Hout
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands
| | - Lena Bosch
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands
| | - Guilielmus H J M Ellenbroek
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands
| | - Judith J de Haan
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands
| | - Wouter W van Solinge
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthew A Cooper
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Fatih Arslan
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands
| | - Saskia C A de Jager
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands
| | - Avril A B Robertson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Gerard Pasterkamp
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands.,Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Imo E Hoefer
- Experimental Cardiology Laboratory (Room G02.523), University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508 GA, The Netherlands.,Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
79
|
OLT1177, a β-sulfonyl nitrile compound, safe in humans, inhibits the NLRP3 inflammasome and reverses the metabolic cost of inflammation. Proc Natl Acad Sci U S A 2018; 115:E1530-E1539. [PMID: 29378952 PMCID: PMC5816172 DOI: 10.1073/pnas.1716095115] [Citation(s) in RCA: 400] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The NLRP3 inflammasome is an intracellular oligomer regulating the activation of caspase-1 for the processing and secretion of IL-1β and IL-18. Although there is growing evidence to substantiate inflammasome inhibition as a therapeutic option for the treatment of inflammatory diseases, to date, there are no approved humans agents. OLT1177, a β-sulfonyl nitrile molecule, shown to be safe in humans, is a selective inhibitor of the NLRP3 inflammasome, with unique properties to reverse the metabolic costs of inflammation and to treat IL-1β– and IL-18–mediated diseases. Activation of the NLRP3 inflammasome induces maturation of IL-1β and IL-18, both validated targets for treating acute and chronic inflammatory diseases. Here, we demonstrate that OLT1177, an orally active β-sulfonyl nitrile molecule, inhibits activation of the NLRP3 inflammasome. In vitro, nanomolar concentrations of OLT1177 reduced IL-1β and IL-18 release following canonical and noncanonical NLRP3 inflammasome activation. The molecule showed no effect on the NLRC4 and AIM2 inflammasomes, suggesting specificity for NLRP3. In LPS-stimulated human blood-derived macrophages, OLT1177 decreased IL-1β levels by 60% and IL-18 by 70% at concentrations 100-fold lower in vitro than plasma concentrations safely reached in humans. OLT1177 also reduced IL-1β release and caspase-1 activity in freshly obtained human blood neutrophils. In monocytes isolated from patients with cryopyrin-associated periodic syndrome (CAPS), OLT1177 inhibited LPS-induced IL-1β release by 84% and 36%. Immunoprecipitation and FRET analysis demonstrated that OLT1177 prevented NLRP3-ASC, as well as NLRP3-caspase-1 interaction, thus inhibiting NLRP3 inflammasome oligomerization. In a cell-free assay, OLT1177 reduced ATPase activity of recombinant NLRP3, suggesting direct targeting of NLRP3. Mechanistically, OLT1177 did not affect potassium efflux, gene expression, or synthesis of the IL-1β precursor. Steady-state levels of phosphorylated NF-κB and IkB kinase were significantly lowered in spleen cells from OLT1177-treated mice. We observed reduced IL-1β content in tissue homogenates, limited oxidative stress, and increased muscle oxidative metabolism in OLT1177-treated mice challenged with LPS. Healthy humans receiving 1,000 mg of OLT1177 daily for 8 d exhibited neither adverse effects nor biochemical or hematological changes.
Collapse
|
80
|
Abstract
The interleukin-1 (IL-1) family of cytokines and receptors is unique in immunology because the IL-1 family and Toll-like receptor (TLR) families share similar functions. More than any other cytokine family, the IL-1 family is primarily associated with innate immunity. More than 95% of living organisms use innate immune mechanisms for survival whereas less than 5% depend on T- and B-cell functions. Innate immunity is manifested by inflammation, which can function as a mechanism of host defense but when uncontrolled is detrimental to survival. Each member of the IL-1 receptor and TLR family contains the cytoplasmic Toll-IL-1-Receptor (TIR) domain. The 50 amino acid TIR domains are highly homologous with the Toll protein in Drosophila. The TIR domain is nearly the same and present in each TLR and each IL-1 receptor family. Whereas IL-1 family cytokine members trigger innate inflammation via IL-1 family of receptors, TLRs trigger inflammation via bacteria, microbial products, viruses, nucleic acids, and damage-associated molecular patterns (DAMPs). In fact, IL-1 family member IL-1a and IL-33 also function as DAMPs. Although the inflammatory properties of the IL-1 family dominate in innate immunity, IL-1 family member can play a role in acquired immunity. This overview is a condensed update of the IL-1 family of cytokines and receptors.
Collapse
Affiliation(s)
- Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
81
|
Abstract
Cardiovascular disease (CVD) is the number one cause of death worldwide. The pathogenesis of various disease entities that comprise the area of CVD is complex and multifactorial. Inflammation serves a central role in these complex aetiologies. The inflammasomes are intracellular protein complexes activated by danger-associated molecular patterns (DAMPs) present in CVD such as atherosclerosis and myocardial infarction (MI). After a two-step process of priming and activation, inflammasomes are responsible for the formation of pro-inflammatory cytokines interleukin-1β and interleukin-18, inducing a signal transduction cascade resulting in a strong immune response that culminates in disease progression. In the past few years, increased interest has been raised regarding the inflammasomes in CVD. Inflammasome activation is thought to be involved in the pathogenesis of various disease entities such as atherosclerosis, MI and heart failure (HF). Interference with inflammasome-mediated signalling could reduce inflammation and attenuate the severity of disease. In this chapter we provide an overview of the current literature available on the role of inflammasome inhibition as a therapeutic intervention and the possible clinical implications for CVD.
Collapse
Affiliation(s)
- Gerardus P J van Hout
- Department of Cardiology, Utrecht University Medical Center, Utrecht, The Netherlands.
| | - Lena Bosch
- Department of Experimental Cardiology, Utrecht University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
82
|
Butts B, Butler J, Dunbar SB, Corwin EJ, Gary RA. ASC Methylation and Interleukin-1β Are Associated with Aerobic Capacity in Heart Failure. Med Sci Sports Exerc 2017; 49:1072-1078. [PMID: 28072632 DOI: 10.1249/mss.0000000000001200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Aerobic capacity, as measured by peak oxygen uptake (V˙O2), is one of the most powerful predictors of prognosis in heart failure (HF). Inflammation is a key factor contributing to alterations in aerobic capacity, and interleukin (IL)-1 cytokines are implicated in this process. The adaptor protein ASC is necessary for inflammasome activation of IL-1β and IL-18. ASC expression is controlled through epigenetic modification; lower ASC methylation is associated with worse outcomes in HF. The purpose of this study is to examine the relationships between ASC methylation, IL-1β, and IL-18 with V˙O2peak in persons with HF. METHODS This study examined the relationship between ASC methylation, IL-1β, and IL-18 with V˙O2peak in 54 stable outpatients with HF. All participants were NYHA class II or III, not engaged in an exercise program, and physically able to complete an exercise treadmill test. RESULTS Mean V˙O2peak was 16.68 ± 4.7 mL·kg·min. V˙O2peak was positively associated with mean percent ASC methylation (r = 0.47, P = 0.001) and negatively associated with IL-1β (r = -0.38, P = 0.007). Multiple linear regression models demonstrated that V˙O2peak increased by 2.30 mL·kg·min for every 1% increase in ASC methylation and decreased by 1.91 mL·kg·min for every 1 pg·mL increase in plasma IL-1β. CONCLUSIONS Mean percent ASC methylation and plasma IL-1β levels are associated with clinically meaningful differences in V˙O2peak in persons with HF. Inflammasome activation may play a mechanistic role in determining aerobic capacity. ASC methylation is a potentially modifiable mechanism for reducing the inflammatory response, thereby improving aerobic capacity in HF.
Collapse
Affiliation(s)
- Brittany Butts
- 1Division of Cardiology, University of Alabama at Birmingham, Birmingham, AL; 2Division of Cardiology, Stony Brook University, Stony Brook, NY; 3Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | | | | | | | | |
Collapse
|
83
|
Abstract
BACKGROUND Interleukin-1α (IL-1α) released by dying cells is an alarmin that activates the innate immunity. We hypothesized that after myocardial ischemia-reperfusion (I/R) injury, IL-1α amplifies the myocardial damage by activating the inflammasome and caspase-1. METHODS Adult male CD1 mice were used. The left anterior descending coronary artery was ligated for 30 minutes, after 24 hours of reperfusion. An IL-1α blocking antibody (15 μg/kg intraperitoneally) or matching vehicle was given after reperfusion. A subgroup of mice underwent sham surgery. We assessed the effects of IL-1α blockade on caspase-1 activity, infarct size, cardiac troponin I serum levels, and left ventricular fractional shortening, 24 hours after I/R. RESULTS I/R led to inflammasome formation, and IL-1α blockade significantly reduced inflammasome formation, reflected by a >50% reduction in caspase-1 activity versus vehicle (P = 0.03). IL-1α blockade also reduced the infarct size (-52% infarct expressed as percentage of area at risk, and -79% for cardiac troponin I serum levels, P < 0.001 vs. vehicle) and preserved the left ventricular fractional shortening (31 ± 3% vs. 25 ± 2%, P < 0.001 vs. vehicle). CONCLUSION IL-1α blockade after I/R reduces the inflammasome activation, decreases the infarct size, and preserves the left ventricular function. IL-1α blockade may therefore represent a novel therapeutic strategy to reduce I/R injury.
Collapse
|
84
|
Zhang Y, Huang Z, Li H. Insights into innate immune signalling in controlling cardiac remodelling. Cardiovasc Res 2017; 113:1538-1550. [PMID: 29088374 DOI: 10.1093/cvr/cvx130] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/29/2017] [Indexed: 01/03/2025] Open
Abstract
Canonical innate immune signalling involves complex cascades: multiple germline-encoded pattern recognition receptors rapidly recognize pathogen-associated or damage-associated molecular patterns to induce the production of cytokines, which bind to their corresponding receptors to orchestrate subsequent host defense phases. Inflammation is a healthy response to pathogenic signals, which are typically rapid and specific, and they terminate once the threat has passed. However, excessive activation or suppression of innate immune or inflammatory responses can lead to considerable human suffering, such as cardiac remodelling. Interestingly, recent studies have revealed that innate immune molecules in the parenchymal cells of the heart influence cardiac homeostasis not only by directly regulating innate immune responses but also through reprogrammed signalling pathways, which are independent of conventional innate immune signalling. Elucidating 'innate immune signalling reprogramming' events will help us better understand the functions of innate immune molecules and, moreover, the pathogenesis of cardiac diseases.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
| | - Zan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- College of Life Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People's Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People's Republic of China
| |
Collapse
|
85
|
Okuhara Y, Yokoe S, Iwasaku T, Eguchi A, Nishimura K, Li W, Oboshi M, Naito Y, Mano T, Asahi M, Okamura H, Masuyama T, Hirotani S. Interleukin-18 gene deletion protects against sepsis-induced cardiac dysfunction by inhibiting PP2A activity. Int J Cardiol 2017; 243:396-403. [PMID: 28526544 DOI: 10.1016/j.ijcard.2017.04.082] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/20/2017] [Accepted: 04/24/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Interleukin-18 (IL-18) neutralization protects against lipopolysaccharide (LPS)-induced injuries, including myocardial dysfunction. However, the mechanism is yet to be fully elucidated. The aim of the present study was to determine whether IL-18 gene deletion prevents sepsis-induced cardiac dysfunction and to elucidate the potential mechanisms underlying IL-18-mediated cardiotoxicity by LPS. METHODS AND RESULTS Ten-week-old male wild-type (WT) and IL-18 knockout (IL-18 KO) mice were intraperitoneally administered LPS. Serial echocardiography showed better systolic pump function and less left ventricular (LV) dilatation in LPS-treated IL-18 KO mice compared with those in LPS-treated WT mice. LPS treatment significantly decreased the levels of phospholamban (PLN) and Akt phosphorylation in WT mice compared with those in saline-treated WT mice, while the LPS-induced decrease in the phosphorylation levels was attenuated in IL-18 KO mice compared with that in WT mice. IL-18 gene deletion also attenuated an LPS-induced increase of type 2 protein phosphatase 2A (PP2A) activity, a molecule that dephosphorylates PLN and Akt. There was no difference in type 1 protein phosphatase (PP1) activity. To address whether IL-18 affects PLN and Akt phosphorylation via PP2A activation in cardiomyocytes, rat neonatal cardiac myocytes were cultured and stimulated using 100ng/ml of recombinant rat IL-18. Exogenous IL-18 decreased the level of PLN and Akt phosphorylation in cardiomyocytes. PP2A activity but not PP1 activity was increased by IL-18 stimulation in cardiomyocytes. CONCLUSIONS IL-18 plays a pivotal role in advancing sepsis-induced cardiac dysfunction, and the mechanisms underlying IL-18-mediated cardiotoxicity potentially involve the regulation of PLN and Akt phosphorylation through PP2A activity.
Collapse
Affiliation(s)
- Yoshitaka Okuhara
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shunichi Yokoe
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Toshihiro Iwasaku
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Akiyo Eguchi
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Koichi Nishimura
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Wen Li
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Makiko Oboshi
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Yoshiro Naito
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Toshiaki Mano
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tohru Masuyama
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shinichi Hirotani
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan.
| |
Collapse
|
86
|
Carbone S, Lee PJH, Mauro AG, Mezzaroma E, Buzzetti R, Van Tassell B, Abbate A, Toldo S. Interleukin-18 mediates cardiac dysfunction induced by western diet independent of obesity and hyperglycemia in the mouse. Nutr Diabetes 2017; 7:e258. [PMID: 28394363 PMCID: PMC5436096 DOI: 10.1038/nutd.2017.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/12/2016] [Accepted: 12/22/2016] [Indexed: 12/27/2022] Open
Abstract
Obesity and diabetes are independent risk factors for heart failure and are associated with the consumption of diet rich in saturated fat and sugar, Western diet (WD), known to induce cardiac dysfunction in the mouse through incompletely characterized inflammatory mechanisms. We hypothesized that the detrimental cardiac effects of WD are mediated by interleukin-18 (IL-18), pro-inflammatory cytokine linked to cardiac dysfunction. C57BL/6J wild-type male mice and IL-18 knockout male mice were fed high-saturated fat and high-sugar diet for 8 weeks. We measured food intake, body weight and fasting glycemia. We assessed left ventricular (LV) systolic and diastolic function by Doppler echocardiography and cardiac catheterization. In wild-type mice, WD induced a significant increase in isovolumetric relaxation time, myocardial performance index and left ventricular end-diastolic pressure, reflecting an impairment in diastolic function, paired with a mild reduction in LV ejection fraction. IL-18 KO mice had higher food intake and greater increase in body weight without significant differences in hyperglycemia. Despite displaying greater obesity, IL-18 knockout mice fed with WD for 8 weeks had preserved cardiac diastolic function and higher left ventricular ejection fraction. IL-18 mediates diet-induced cardiac dysfunction, independent of food intake and obesity, thus highlighting a disconnect between the metabolic and cardiac effects of IL-18.
Collapse
Affiliation(s)
- S Carbone
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA, USA
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - P J H Lee
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA, USA
| | - A G Mauro
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA, USA
| | - E Mezzaroma
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA, USA
- School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - R Buzzetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - B Van Tassell
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA, USA
- School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - A Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA, USA
| | - S Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
87
|
Li R, Lu K, Wang Y, Chen M, Zhang F, Shen H, Yao D, Gong K, Zhang Z. Triptolide attenuates pressure overload-induced myocardial remodeling in mice via the inhibition of NLRP3 inflammasome expression. Biochem Biophys Res Commun 2017; 485:69-75. [PMID: 28202417 DOI: 10.1016/j.bbrc.2017.02.021] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 02/05/2017] [Indexed: 01/19/2023]
Abstract
Triptolide is the predominant active component of the Chinese herb Tripterygium wilfordii Hook F (TwHF) that has been widely used to treat several chronic inflammatory diseases due to its immunosuppressive, anti-inflammatory, and anti-proliferative properties. In the present study, we elucidated the cardioprotective effects of triptolide against cardiac dysfunction and myocardial remodeling in chronic pressure-overloaded hearts. Furthermore, the potential mechanisms of triptolide were investigated. For this purpose, C57/BL6 mice were anesthetized and subjected to transverse aortic constriction (TAC) or sham operation. Six weeks after the operation, all mice were randomly divided into 4 groups: sham-operated with vehicle group, TAC with vehicle group, and TAC with triptolide (20 or 100 μg/kg/day intraperitoneal injection) groups. Our data showed that the levels of NLRP3 inflammasome were significantly increased in the TAC group and were associated with increased inflammatory mediators and profibrotic factor production, resulting in myocardial fibrosis, cardiomyocyte hypertrophy, and impaired cardiac function. Triptolide treatment attenuated TAC-induced myocardial remodeling, improved cardiac diastolic and systolic function, inhibited the NLRP3 inflammasome and downstream inflammatory mediators (IL-1β, IL-18, MCP-1, VCAM-1), activated the profibrotic TGF-β1 pathway, and suppressed macrophage infiltration in a dose-dependent manner. Our study demonstrated that the protective effect of triptolide against pressure overload in the heart may act by inhibiting the NLRP3 inflammasome-induced inflammatory response and activating the profibrotic pathway.
Collapse
Affiliation(s)
- Rujun Li
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Kuiying Lu
- Department of Clinical Laboratory, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Yao Wang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Mingxing Chen
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Fengyu Zhang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Hui Shen
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Deshan Yao
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Kaizheng Gong
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Zhengang Zhang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China.
| |
Collapse
|
88
|
Al-Mohanna F. The Cardiokines. ENDOCRINOLOGY OF THE HEART IN HEALTH AND DISEASE 2017:87-114. [DOI: 10.1016/b978-0-12-803111-7.00004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
89
|
Bugyei-Twum A, Abadeh A, Thai K, Zhang Y, Mitchell M, Kabir G, Connelly KA. Suppression of NLRP3 Inflammasome Activation Ameliorates Chronic Kidney Disease-Induced Cardiac Fibrosis and Diastolic Dysfunction. Sci Rep 2016; 6:39551. [PMID: 28000751 PMCID: PMC5175152 DOI: 10.1038/srep39551] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/23/2016] [Indexed: 01/28/2023] Open
Abstract
Cardiac fibrosis is a common finding in patients with chronic kidney disease. Here, we investigate the cardio-renal effects of theracurmin, a novel formulation of the polyphenolic compound curcumin, in a rat model of chronic kidney disease. Briefly, Sprague-Dawley rats were randomized to undergo sham or subtotal nephrectomy (SNx) surgery. At 3 weeks post surgery, SNx animals were further randomized to received theracurmin via once daily oral gavage or vehicle for 5 consecutive weeks. At 8 weeks post surgery, cardiac function was assessed via echocardiography and pressure volume loop analysis, followed by LV and renal tissue collection for analysis. SNx animals developed key hallmarks of renal injury including hypertension, proteinuria, elevated blood urea nitrogen, and glomerulosclerosis. Renal injury in SNx animals was also associated with significant diastolic dysfunction, macrophage infiltration, and cardiac NLRP3 inflammasome activation. Treatment of SNx animals with theracurmin improved structural and functional manifestations of cardiac injury associated with renal failure and also attenuated cardiac NLRP3 inflammasome activation and mature IL-1β release. Taken together, our findings suggest a significant role for the NLRP3 inflammasome in renal injury-induced cardiac dysfunction and presents inflammasome attenuation as a unique strategy to prevent adverse cardiac remodeling in the setting of chronic kidney disease.
Collapse
Affiliation(s)
- Antoinette Bugyei-Twum
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Armin Abadeh
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Kerri Thai
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Yanling Zhang
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Melissa Mitchell
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Golam Kabir
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's hospital, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Division of Cardiology, St. Michael's hospital, Toronto, Ontario, Canada
| |
Collapse
|
90
|
Slaats J, ten Oever J, van de Veerdonk FL, Netea MG. IL-1β/IL-6/CRP and IL-18/ferritin: Distinct Inflammatory Programs in Infections. PLoS Pathog 2016; 12:e1005973. [PMID: 27977798 PMCID: PMC5158075 DOI: 10.1371/journal.ppat.1005973] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The host inflammatory response against infections is characterized by the release of pro-inflammatory cytokines and acute-phase proteins, driving both innate and adaptive arms of the immune response. Distinct patterns of circulating cytokines and acute-phase responses have proven indispensable for guiding the diagnosis and management of infectious diseases. This review discusses the profiles of acute-phase proteins and circulating cytokines encountered in viral and bacterial infections. We also propose a model in which the inflammatory response to viral (IL-18/ferritin) and bacterial (IL-6/CRP) infections presents with specific plasma patterns of immune biomarkers.
Collapse
Affiliation(s)
- Jeroen Slaats
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| | - Jaap ten Oever
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
91
|
Lim JS, Park S, Park SI, Oh YT, Choi E, Kim JY, Rhee Y. Cardiac Dysfunction in Association with Increased Inflammatory Markers in Primary Aldosteronism. Endocrinol Metab (Seoul) 2016; 31:567-576. [PMID: 27834080 PMCID: PMC5195834 DOI: 10.3803/enm.2016.31.4.567] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 08/29/2016] [Accepted: 09/08/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Oxidative stress in primary aldosteronism (PA) is thought to worsen aldosterone-induced damage by activating proinflammatory processes. Therefore, we investigated whether inflammatory markers associated with oxidative stress is increased with negative impacts on heart function as evaluated by echocardiography in patients with PA. METHODS Thirty-two subjects (mean age, 50.3±11.0 years; 14 males, 18 females) whose aldosterone-renin ratio was more than 30 among patients who visited Severance Hospital since 2010 were enrolled. Interleukin-1β (IL-1β), IL-6, IL-8, monocyte chemoattractant protein 1, tumor necrosis factor α (TNF-α), and matrix metalloproteinase 2 (MMP-2), and MMP-9 were measured. All patients underwent adrenal venous sampling with complete access to both adrenal veins. RESULTS Only MMP-2 level was significantly higher in the aldosterone-producing adenoma (APA) group than in the bilateral adrenal hyperplasia (BAH). Patients with APA had significantly higher left ventricular (LV) mass and A velocity, compared to those with BAH. IL-1β was positively correlated with left atrial volume index. Both TNF-α and MMP-2 also had positive linear correlation with A velocity. Furthermore, MMP-9 showed a positive correlation with LV mass, whereas it was negatively correlated with LV end-systolic diameter. CONCLUSION These results suggest the possibility that some of inflammatory markers related to oxidative stress may be involved in developing diastolic dysfunction accompanied by LV hypertrophy in PA. Further investigations are needed to clarify the role of oxidative stress in the course of cardiac remodeling.
Collapse
Affiliation(s)
- Jung Soo Lim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Institute of Evidence Based Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sungha Park
- Division of Cardiology, Severance Cardiovascular Hospital and Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Il Park
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea
| | - Young Taik Oh
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea
| | - Eunhee Choi
- Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jang Young Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Yumie Rhee
- Department of Internal Medicine, Severance Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
92
|
Duarte JD, Desai AA, Sysol JR, Abbasi T, Patel AR, Lang RM, Gupta A, Garcia JGN, Gordeuk VR, Machado RF. Genome-Wide Analysis Identifies IL-18 and FUCA2 as Novel Genes Associated with Diastolic Function in African Americans with Sickle Cell Disease. PLoS One 2016; 11:e0163013. [PMID: 27636371 PMCID: PMC5026353 DOI: 10.1371/journal.pone.0163013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/01/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diastolic dysfunction is common in sickle cell disease (SCD), and is associated with an increased risk of mortality. However, the molecular pathogenesis underlying this development is poorly understood. The aim of this study was to identify a gene expression profile that is associated with diastolic function in SCD, potentially elucidating molecular mechanisms behind diastolic dysfunction development. METHODS Diastolic function was measured via echocardiography in 65 patients with SCD from two independent study populations. Gene expression microarray data was compared with diastolic function in both study cohorts. Candidate genes that associated in both analyses were tested for validation in a murine SCD model. Lastly, genotyping array data from the replication cohort was used to derive cis-expression quantitative trait loci (cis-eQTLs) and genetic associations within the candidate gene regions. RESULTS Transcriptome data from both patient cohorts implicated 7 genes associated with diastolic function, and mouse SCD myocardial expression validated 3 of these genes. Genetic associations and eQTLs were detected in 2 of the 3 genes, FUCA2 and IL18. CONCLUSIONS FUCA2 and IL18 are associated with diastolic function in SCD patients, and may be involved in the pathogenesis of the disease. Genetic polymorphisms within the FUCA2 and IL18 gene regions are also associated with diastolic function in SCD, likely by affecting expression levels of the genes.
Collapse
Affiliation(s)
- Julio D. Duarte
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Ankit A. Desai
- Division of Cardiology, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America
| | - Justin R. Sysol
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Taimur Abbasi
- Department of Medicine, Mercy Hospital and Medical Center, Chicago, IL, United States of America
| | - Amit R. Patel
- Department of Medicine, University of Chicago, Chicago, IL United States of America
| | - Roberto M. Lang
- Department of Medicine, University of Chicago, Chicago, IL United States of America
| | - Akash Gupta
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Joe G. N. Garcia
- Department of Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Victor R. Gordeuk
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Roberto F. Machado
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| |
Collapse
|
93
|
Hamar P, Kerjaschki D. Blood capillary rarefaction and lymphatic capillary neoangiogenesis are key contributors to renal allograft fibrosis in an ACE inhibition rat model. Am J Physiol Heart Circ Physiol 2016; 311:H981-H990. [PMID: 27496878 DOI: 10.1152/ajpheart.00320.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/29/2016] [Indexed: 12/17/2022]
Abstract
Chronic allograft fibrosis is the major cause of graft loss in kidney transplantation. Progression can only be reduced by inhibition of the renin-angiotensin system (RAS). We tested the hypothesis that the protection provided by angiotensin-converting enzyme (ACE) inhibition also decreases capillary rarefaction, lymphangiogenesis, and podocyte injury in allograft fibrosis. Fisher kidneys were transplanted into bilaterally nephrectomized Lewis rats treated with enalapril (60 mg/kg per day) (ACE inhibitor, ACEi) or vehicle. Proteinuria, blood urea nitrogen, and plasma creatinine were regularly assessed, and grafts were harvested for morphological and immunohistological analysis at various times up to 32 wk. In the vehicle group, many new lymphatic capillaries and severe and diffuse mononuclear infiltration of allografts were observed already 1 wk after transplantation. Lymphangiogenesis increased until week 4, by which time inflammatory infiltration became focal. Lymphatic capillaries were often located at sites of inflammation. Progressive interstitial fibrosis, glomerulosclerosis, capillary rarefaction, and proteinuria appeared later, at weeks 4-12 The number of lymphatic capillary cross sections strongly correlated with the interstitial fibrosis score. Podoplanin immunostaining, a marker of healthy podocytes, disappeared from inflamed or sclerotic glomerular areas. ACEi protected from lymphangiogenesis and associated inflammation, preserved glomerular podoplanin protein expression, and reduced glomerulosclerosis, proteinuria, tubulointerstitial fibrosis, and blood capillary rarefaction at 32 wk. In conclusion, ACEi considerably decreased and/or delayed both glomerulosclerosis and tubulointerstitial injury. Prevention of glomerular podoplanin loss and proteinuria could be attributed to the known intraglomerular pressure-lowering effects of ACEi. Reduction of lymphangiogenesis could contribute to amelioration of tubulointerstitial fibrosis and inflammatory infiltration after ACEi.
Collapse
Affiliation(s)
- Péter Hamar
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary; and
| | | |
Collapse
|
94
|
Lother A, Hein L. Pharmacology of heart failure: From basic science to novel therapies. Pharmacol Ther 2016; 166:136-49. [PMID: 27456554 DOI: 10.1016/j.pharmthera.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
95
|
Akar FG. Starve a fever to heal a heart? Interleukin-18 gives new meaning to an old adage. Am J Physiol Heart Circ Physiol 2016; 311:H311-2. [PMID: 27342879 DOI: 10.1152/ajpheart.00445.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Fadi G Akar
- The Cardiovascular Institute, Ichan School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
96
|
Joosten LAB, Crişan TO, Azam T, Cleophas MCP, Koenders MI, van de Veerdonk FL, Netea MG, Kim S, Dinarello CA. Alpha-1-anti-trypsin-Fc fusion protein ameliorates gouty arthritis by reducing release and extracellular processing of IL-1β and by the induction of endogenous IL-1Ra. Ann Rheum Dis 2016; 75:1219-27. [PMID: 26174021 DOI: 10.1136/annrheumdis-2014-206966] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 06/06/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVES In the present study, we generated a new protein, recombinant human alpha-1-anti-trypsin (AAT)-IgG1 Fc fusion protein (AAT-Fc), and evaluated its properties to suppress inflammation and interleukin (IL)-1β in a mouse model of gouty arthritis. METHODS A combination of monosodium urate (MSU) crystals and the fatty acid C16.0 (MSU/C16.0) was injected intra-articularly into the knee to induce gouty arthritis. Joint swelling, synovial cytokine production and histopathology were determined after 4 h. AAT-Fc was evaluated for inhibition of MSU/C16.0-induced IL-1β release from human blood monocytes and for inhibition of extracellular IL-1β precursor processing. RESULTS AAT-Fc markedly suppressed MSU/C16.0-induced joint inflammation by 85-91% (p<0.001). Ex vivo production of IL-1β and IL-6 from cultured synovia were similarly reduced (63% and 65%, respectively). The efficacy of 2.0 mg/kg AAT-Fc in reducing inflammation was comparable to 80 mg/kg of plasma-derived AAT. Injection of AAT-Fc into mice increased circulating levels of endogenous IL-1 receptor antagonist by fourfold. We also observed that joint swelling was reduced by 80%, cellular infiltration by 95% and synovial production of IL-1β by 60% in transgenic mice expressing low levels of human AAT. In vitro, AAT-Fc reduced MSU/C16.0-induced release of IL-1β from human blood monocytes and inhibited proteinase-3-mediated extracellular processing of the IL-1β precursor into active IL-1β. CONCLUSIONS A single low dose of AAT-Fc is highly effective in reducing joint inflammation in this model of acute gouty arthritis. Considering the long-term safety of plasma-derived AAT use in humans, subcutaneous AAT-Fc emerges as a promising therapy for gout attacks.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Gouty/drug therapy
- Arthritis, Gouty/immunology
- Arthritis, Gouty/pathology
- Cells, Cultured
- Dose-Response Relationship, Drug
- Drug Evaluation, Preclinical/methods
- Gout Suppressants/administration & dosage
- Gout Suppressants/pharmacology
- Gout Suppressants/therapeutic use
- Humans
- Immunoglobulin Fc Fragments/administration & dosage
- Immunoglobulin Fc Fragments/pharmacology
- Immunoglobulin Fc Fragments/therapeutic use
- Injections, Intra-Articular
- Injections, Intraperitoneal
- Interleukin 1 Receptor Antagonist Protein/biosynthesis
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/metabolism
- Lipopolysaccharide Receptors/analysis
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Monocytes/drug effects
- Monocytes/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/therapeutic use
- alpha 1-Antitrypsin/administration & dosage
- alpha 1-Antitrypsin/pharmacology
- alpha 1-Antitrypsin/therapeutic use
Collapse
Affiliation(s)
- Leo A B Joosten
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tania O Crişan
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tania Azam
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Maartje C P Cleophas
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije I Koenders
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Konkuk University, Seoul, Korea
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
97
|
Crea F, Liuzzo G. Anti-inflammatory treatment of acute coronary syndromes: the need for precision medicine. Eur Heart J 2016; 37:2414-6. [PMID: 27252447 DOI: 10.1093/eurheartj/ehw207] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Filippo Crea
- Institute of Cardiology, Catholic University, Rome, Italy
| | | |
Collapse
|
98
|
Jin Y, Wu Z, Wang N, Duan S, Wu Y, Wang J, Wu W, Feng F. Association of EGF Receptor and NLRs signaling with Cardiac Inflammation and Fibrosis in Mice Exposed to Fine Particulate Matter. J Biochem Mol Toxicol 2016; 30:429-37. [PMID: 27158778 DOI: 10.1002/jbt.21806] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/02/2016] [Accepted: 03/11/2016] [Indexed: 11/08/2022]
Abstract
ЄAmbient fine particulate matter (PM2.5 ) could induce cardiovascular diseases (CVD), but the mechanism remains unknown. To investigate the roles of epidermal growth factor receptor (EGFR) and NOD-like receptors (NLRs) in PM2.5 -induced cardiac injury, we set up a BALB/c mice model of PM2.5 -induced cardiac inflammation and fibrosis with intratracheal instillation of PM2.5 suspension (4.0 mg/kg b.w.) for 5 consecutive days (once per day). After exposure, we found that mRNA levels of CXCL1, interleukin (IL)-6, and IL-18 were elevated, but interestingly, mRNA level of NLRP12 was significant decreased in heart tissue from PM2.5 -induced mice compared with those of saline-treated mice using real-time PCR. Protein levels of phospho-EGFR (Tyr1068), phospho-Akt (Thr308), NLRP3, NF-κB-p52/p100, and NF-κB-p65 in heart tissue of PM2.5 -exposed mice were all significantly increased using immunohistochemistry or Western blotting. Therefore, PM2.5 exposure could induce cardiac inflammatory injury in mice, which may be involved with EGFR/Akt signaling, NLRP3, and NLRP12.
Collapse
Affiliation(s)
- Yuefei Jin
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Zhaoke Wu
- Department of Geriatrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, Henan, People's Republic of China
| | - Na Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Shuyin Duan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Jing Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China.
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
| |
Collapse
|
99
|
Marchetti C, Toldo S, Chojnacki J, Mezzaroma E, Liu K, Salloum FN, Nordio A, Carbone S, Mauro AG, Das A, Zalavadia AA, Halquist MS, Federici M, Van Tassell BW, Zhang S, Abbate A. Pharmacologic Inhibition of the NLRP3 Inflammasome Preserves Cardiac Function After Ischemic and Nonischemic Injury in the Mouse. J Cardiovasc Pharmacol 2016; 66:1-8. [PMID: 25915511 DOI: 10.1097/fjc.0000000000000247] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sterile inflammation resulting from myocardial injury activates the NLRP3 inflammasome and amplifies the inflammatory response mediating further damage. METHODS We used 2 experimental models of ischemic injury (acute myocardial infarction [AMI] with and without reperfusion) and a model of nonischemic injury due to doxorubicin 10 mg/kg to determine whether the NLRP3 inflammasome preserved cardiac function after injury. RESULTS Treatment with the NLRP3 inflammasome inhibitor in the reperfused AMI model caused a significant reduction in infarct size measured at pathology or as serum cardiac troponin I level (-56% and -82%, respectively, both P < 0.001) and preserved left ventricular fractional shortening (LVFS, 31 ± 2 vs. vehicle 26% ± 1%, P = 0.003). In the non-reperfused AMI model, treatment with the NLRP3 inhibitor significantly limited LV systolic dysfunction at 7 days (LVFS of 20 ± 2 vs. 14% ± 1%, P = 0.002), without a significant effect on infarct size. In the doxorubicin model, a significant increase in myocardial interstitial fibrosis and a decline in systolic function were seen in vehicle-treated mice, whereas treatment with the NLRP3 inhibitor significantly reduced fibrosis (-80%, P = 0.001) and preserved systolic function (LVFS 35 ± 2 vs. vehicle 27% ± 2%, P = 0.017). CONCLUSIONS Pharmacological inhibition of the NLRP3 inflammasome limits cell death and LV systolic dysfunction after ischemic and nonischemic injury in the mouse.
Collapse
Affiliation(s)
- Carlo Marchetti
- *VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA; †Victoria Johnson Research Laboratories, Richmond, VA; ‡Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Departments of §Medicinal Chemistry; ‖Pharmacotherapy and Outcome Studies, and ¶Pharmaceutics Virginia Commonwealth University, Richmond, VA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Bruns DR, Buttrick PM, Walker LA. Genetic ablation of interleukin-18 does not attenuate hypobaric hypoxia-induced right ventricular hypertrophy. Am J Physiol Lung Cell Mol Physiol 2016; 310:L542-50. [PMID: 26747780 DOI: 10.1152/ajplung.00166.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/29/2015] [Indexed: 11/22/2022] Open
Abstract
Interleukin-18 (IL-18), a proinflammatory cytokine, has been implicated in pathologic left ventricular hypertrophy and is elevated in plasma of heart failure patients. However, IL-18 blockade strategies have been conflicting. The purpose of these experiments was to determine whether genetic ablation of IL-18 would protect mice against hypobaric hypoxia (HH)-induced right ventricular (RV) hypertrophy, a condition in which chamber-specific inflammation is prominent. We hypothesized that IL-18 knockout (KO) mice would be protected while wild-type (WT) mice would demonstrate RV hypertrophy in response to HH exposure. KO and WT mice were exposed to HH for 7 wk, and control mice were exposed to normoxic ambient air. Following echocardiography, the RV was dissected and flash-frozen for biochemical analyses. HH exposure increased IL-18 mRNA (P = 0.08) in RV from WT mice. Genetic ablation of IL-18 mildly attenuated RV hypertrophy as assessed by myocyte size. However, IL-18 KO mice were not protected against HH-induced organ-level remodeling, as evidenced by higher RV weights, elevated RV systolic pressure, and increased RV anterior wall thickness compared with normoxic KO mice. These RV changes were similar to those seen in HH-exposed WT mice. Compensatory upregulation of other proinflammatory cytokines IL-2 and stromal cell-derived factor-1 was seen in the HH-KO animals, suggesting that activation of parallel inflammatory pathways might mitigate the effect of IL-18 KO. These data suggest targeted blockade of IL-18 alone is not a viable therapeutic strategy in this model.
Collapse
Affiliation(s)
- Danielle R Bruns
- Department of Medicine, Cardiology, University of Colorado-Denver, Aurora, Colorado
| | - Peter M Buttrick
- Department of Medicine, Cardiology, University of Colorado-Denver, Aurora, Colorado
| | - Lori A Walker
- Department of Medicine, Cardiology, University of Colorado-Denver, Aurora, Colorado
| |
Collapse
|