1
|
Laudani C, Occhipinti G, Greco A, Giacoppo D, Spagnolo M, Capodanno D. A pairwise and network meta-analysis of anti-inflammatory strategies after myocardial infarction: the TITIAN study. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2025; 11:218-229. [PMID: 39756386 PMCID: PMC12046582 DOI: 10.1093/ehjcvp/pvae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 10/18/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
BACKGROUND AND AIMS Multiple anti-inflammatory drugs have been tested for secondary prevention after myocardial infarction (MI), giving mixed results and questioning the efficacy of anti-inflammatory therapy. No head-to-head comparisons between anti-inflammatory drugs have been performed. This study aimed to compare the efficacy and safety of anti-inflammatory drugs for secondary prevention after MI and the relative merits of specific drugs and administration strategies. METHODS AND RESULTS Randomized trials of anti-inflammatory therapy for secondary prevention after MI were identified. Primary efficacy and safety endpoints were trial-defined major adverse cardiovascular events (MACEs) and serious adverse events. Secondary endpoints included all-cause death, individual MACE components, serious infection, cancer, and gastrointestinal adverse events. Pairwise meta-analyses were conducted with interaction analyses for drug type and timing of administration, in addition to network meta-analyses. Multiple sensitivity and meta-regression analyses were conducted to explore potential heterogeneity sources. Twenty-eight studies, involving 44 406 patients with a mean follow-up of 11 months, were included. Anti-inflammatory therapy reduced the incidence of MACEs [incidence rate ratio (IRR): 0.92; 95% confidence interval (CI): 0.86-0.98] compared to control, without increasing serious adverse events. However, it was associated with a higher incidence of gastrointestinal adverse events (IRR: 1.21; 95% CI: 1.07-1.36). No significant interaction was observed between the effects of anti-inflammatory therapy on MACE and the timing of administration. CONCLUSION In secondary prevention for MI, anti-inflammatory therapy significantly reduces MACE without increasing serious adverse events, but it is associated with an increased risk of gastrointestinal adverse events.
Collapse
Affiliation(s)
- Claudio Laudani
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Giovanni Occhipinti
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
- Institut Clinic Cardiovascular, Hospital Clínic de Barcelona, University of Barcelona, Carrer de Villarroel, 170, L'Eixample, 08036 Barcelona, Spain
| | - Antonio Greco
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Daniele Giacoppo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Marco Spagnolo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico “Rodolico—San Marco”, University of Catania, Via S. Sofia 76, 95125 Catania, Italy
| |
Collapse
|
2
|
Bonaventura A, Potere N, Liberale L, Kraler S, Weber BN, Abbate A. Colchicine in Coronary Artery Disease: Where Do We Stand? J Cardiovasc Pharmacol 2025; 85:243-247. [PMID: 39847601 DOI: 10.1097/fjc.0000000000001672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 12/28/2024] [Indexed: 01/25/2025]
Abstract
ABSTRACT Colchicine is an anti-inflammatory drug for different inflammatory conditions and is approved for secondary prevention of cardiovascular events in patients with coronary artery disease, mainly based on the results of the LODOCO2 and COLCOT trials. The recently published CLEAR SYNERGY trial reported neutral results for colchicine in patients with acute myocardial infarction undergoing percutaneous coronary intervention, challenging the previous reported benefits of colchicine. While colchicine appeared rather safe across the different studies, the variation in efficacy may suggest that the one-size-fits-all for the treatment of acute and chronic forms of coronary artery disease may not be appropriate, and that low-dose colchicine may be beneficial as an add-on therapy in patients who are stable or recovering from acute event, and not so helpful in patients with acute myocardial infarction already receiving intensive pharmaco-invasive therapies.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Italy
| | - Nicola Potere
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology and Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Brittany N Weber
- Heart and Vascular Center, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; and
| | - Antonio Abbate
- Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA
| |
Collapse
|
3
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
4
|
Denicolai M, Morello M, Golino M, Corna G, Del Buono MG, Agatiello CR, Van Tassell BW, Abbate A. Interleukin-1 Blockade in Patients With ST-Segment Elevation Myocardial Infarction Across the Spectrum of Coronary Artery Disease Complexity. J Cardiovasc Pharmacol 2025; 85:200-210. [PMID: 39531530 DOI: 10.1097/fjc.0000000000001652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
ABSTRACT Patients with ST-segment elevation myocardial infarction (STEMI) and complex coronary artery disease (CAD) face a poor prognosis, including increased heart failure (HF) risk. Phase 2 clinical trials of anakinra have shown inhibition of the acute inflammatory response and prevention of HF after STEMI, but data on its effects based on CAD complexity are lacking. We performed a pooled secondary analysis of 139 patients with STEMI. The SYNTAX (Synergy Between Percutaneous Coronary Intervention with Taxus and Cardiac Surgery), SYNTAX II, and Gensini scores were calculated, and patients were divided into 2 groups below and above the median. We evaluated the effect of anakinra on the area-under-the-curve of high-sensitivity C-reactive protein (hsCRP-AUC) at 14 days, and the composite endpoint of new-onset HF, HF hospitalization, or all-cause death at 1-year follow-up using Kaplan-Meier survival curves, Cox regression analysis for hazard ratios (HRs), and tested interactions between subgroups. All 3 CAD complexity scores (SYNTAX, SYNTAX II, and Gensini) were associated with an increased risk of adverse events (HR 1.02-1.06, all P-values ≤0.025). We found no statistically significant interactions between CAD extent, measured as single-vessel or multivessel CAD, SYNTAX score ≤9 or >9, SYNTAX II score ≤24 or >24, Gensini score ≤32 or >32, and treatment effect of anakinra on hsCRP-AUC or the composite clinical endpoint (all P - values for interaction >0.05). In conclusion, among patients with STEMI, IL-1 blockade with anakinra significantly attenuated the acute inflammatory response and reduced the risk of HF-related events regardless of the spectrum of CAD complexity.
Collapse
Affiliation(s)
- Martin Denicolai
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Matteo Morello
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
| | - Michele Golino
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Giuliana Corna
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Marco G Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy ; and
| | - Carla R Agatiello
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Benjamin W Van Tassell
- Department of Pharmacotherapy & Outcomes Sciences, Virginia Commonwealth University, Richmond, VA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Department of Medicine, University of Virginia, Charlottesville, VA
| |
Collapse
|
5
|
Yang YT, Zhen B, Cao X, Xia HY, Gong YZ, Yang YL. Machine Learning Algorithm-Based Discovery of Potential Regulators of Immune-Related Dilated Cardiomyopathy. ACTA CARDIOLOGICA SINICA 2025; 41:230-241. [PMID: 40123611 PMCID: PMC11923790 DOI: 10.6515/acs.202503_41(2).20241022a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 10/22/2024] [Indexed: 03/25/2025]
Abstract
Purpose Dilated cardiomyopathy (DCM) is considered a severe non-ischemic myocardial disease, and there is currently no effective method for the early detection of DCM. Therefore, we aimed to use machine learning algorithms to discover more accurate factors to guide clinical drug development and precision medicine diagnosis. Methods Two datasets containing patients with DCM and healthy controls were downloaded from the Gene Expression Omnibus database. After data preprocessing, differentially expressed genes (DEGs) between the DCM patients and normal samples were identified using the limma package. In addition, to screen for DEGs closely associated with immune inflammation, we collected immune-related genes and defined overlapping genes as differential immune genes (Immune-DEGs). Protein-protein interaction (PPI) network construction and functional enrichment analysis were then functionally validated for the differential immune genes. Subsequently, we further screened the immune-DEGs using the least absolute shrinkage and selection operator (LASSO) technique and support vector machine algorithm (SVM), resulting in the screening of five potential modulators closely associated with DCM. Finally, the diagnostic efficacy of the modifiers was assessed using subject operating characteristic curves based on independent external data, and the intrinsic pathological mechanisms of different differential immune genes were explored by immune infiltration analysis. Results A consensus of 184 differential immune genes were identified, and the functional enrichment results of their PPI network modules suggested that inflammation, immune disorders, and viral infections play an essential role in the pathogenesis of DCM. Five signature genes were then further screened using LASSO and SVM algorithms: KLRC4, CCL4, IGHV3-33, ITGAL, and inducible T-cell kinase. Conclusions This study constructed a gene set of potential DCM regulators with five immune-related genes, which could provide a new strategy for the diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Yi-Ting Yang
- Department of Cardiovascular Medicine, Songbei Branch of the Fourth Affiliated Hospital of Harbin Medical University
| | - Bao Zhen
- Harbin Fourth Hospital, Harbin, China
| | - Xue Cao
- Department of Cardiovascular Medicine, Songbei Branch of the Fourth Affiliated Hospital of Harbin Medical University
| | - Hong-Yuan Xia
- Department of Cardiovascular Medicine, Songbei Branch of the Fourth Affiliated Hospital of Harbin Medical University
| | - Ying-Zi Gong
- Department of Cardiovascular Medicine, Songbei Branch of the Fourth Affiliated Hospital of Harbin Medical University
| | - Yan-Li Yang
- Department of Cardiovascular Medicine, Songbei Branch of the Fourth Affiliated Hospital of Harbin Medical University
| |
Collapse
|
6
|
Mo B, Ding Y, Ji Q. NLRP3 inflammasome in cardiovascular diseases: an update. Front Immunol 2025; 16:1550226. [PMID: 40079000 PMCID: PMC11896874 DOI: 10.3389/fimmu.2025.1550226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of mortality worldwide. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is involved in numerous types of CVD. As part of innate immunity, the NLRP3 inflammasome plays a vital role, requiring priming and activation signals to trigger inflammation. The NLRP3 inflammasome leads both to the release of IL-1 family cytokines and to a distinct form of programmed cell death called pyroptosis. Inflammation related to CVD has been extensively investigated in relation to the NLRP3 inflammasome. In this review, we describe the pathways triggering NLRP3 priming and activation and discuss its pathogenic effects on CVD. This study also provides an overview of potential therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Binhai Mo
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yudi Ding
- First People’s Hospital of Nanning, Nanning, Guangxi, China
| | - Qingwei Ji
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
7
|
Muhs T, Ljubojevic-Holzer S, Sattler S. Anti-inflammatory Therapies for Ischemic Heart Disease. Curr Cardiol Rep 2025; 27:57. [PMID: 39969632 PMCID: PMC11839821 DOI: 10.1007/s11886-025-02211-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW The inclusion of immunomodulatory strategies as supportive therapies in ischemic heart disease (IHD) has garnered significant support over recent years. Several such approaches appear to be unified through their ultimate target, the NLRP3 inflammasome. This review presents a brief update on immunomodulatory strategies in the continuum of conditions constituting ischemic heart disease and emphasising on the seemingly unifying mechanism of NLRP3 activation as well as modulation across these conditions. RECENT FINDINGS The NLRP3 inflammasome is a multiprotein complex assembled upon inflammatory stimulation, causing the release of pro-inflammatory cytokines and initiating pyroptosis. The NLRP3 pathway is relevant in inflammatory signalling of cardiac immune cells as well as non-immune cells in the myocardium, including cardiomyocytes, fibroblasts and endothelial cells. In addition to a focus on clinical outcome and efficacy trials of targeting NLRP3-related pathways, the potential connection between immunomodulation in cardiology and the NLRP3 pathway is currently being explored in preclinical trials. Colchicine, cytokine-based approaches and SGLT2 inhibitors have emerged as promising agents. However, the conditions comprising IHD including atherosclerosis, coronary artery disease (CAD), myocardial infarction (MI) and ischemic cardiomyopathy/heart failure (iCMP/HF) are not equally amenable to immunomodulation with the respective drugs. Atherosclerosis, coronary artery disease and ischemic cardiomyopathy are affected by chronic inflammation, but the immunomodulatory approach to acute inflammation in the post-MI setting remains a pharmacological challenge, as detrimental and regenerative effects of myocardial inflammation are initiated in unison. The NLRP3 inflammasome lies at the center of cell mediated inflammation in IHD. Recent trial evidence has highlighted anti-inflammatory effects of colchicine, interleukin-based therapy as well as SGLT2i in IHD and that the respective drugs modulate the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Tillmann Muhs
- Department of Pharmacology, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, LKH Univ. Klinikum Graz, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Susanne Sattler
- Department of Pharmacology, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria.
- Department of Cardiology, LKH Univ. Klinikum Graz, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
8
|
Lian Y, Zhang H, Xing W, Li S, Lai X, Jia S, Shang J, Liu H. Global Research Trends and Focus on the Link Between Heart Failure and NLRP3 Inflammasome: A Bibliometric Analysis From 2010 to 2024. J Multidiscip Healthc 2025; 18:697-710. [PMID: 39949850 PMCID: PMC11822290 DOI: 10.2147/jmdh.s505356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Background Heart failure (HF) is characterized by elevated morbidity, mortality, and rehospitalization frequencies. This condition imposes a considerable medical burden and fiscal strain on society. Inflammation plays a crucial role in the inception, advancement, and outcome of HF. Despite mounting evidence demonstrating the pivotal function of the NLRP3 inflammasome in HF, a thorough bibliometric examination of research focal points and trajectories in this domain has yet to be undertaken. Methods Publications related to the NLRP3 inflammasome in HF were retrieved from the Web of Science database spanning 2010-2024. The acquired data were subsequently analyzed utilizing various visualization instruments, including Citespace, VOSviewer, Scimago Graphica, and Microsoft Office Excel 2021. Results A total of 282 papers were included in the analysis, authored by 2,130 researchers from 500 institutions across 34 nations/regions. China emerged as a significant contributor to this field, producing the highest number of outputs. Antonio Abbate was identified as the most prolific author. Virginia Commonwealth University and Wuhan University were the institutions with the highest publication output. INTERNATIONAL IMMUNOPHARMACOLOGY was the periodical with the most numerous publications in this sphere. CIRCULATION, however, received the highest number of citations, indicating its substantial influence on investigations in this field. Contemporary research focal points primarily concentrate on the activation and inhibition pathways of the NLRP3 inflammasome, the exploration of novel HF targets, and the association between HF and mitochondrial function. Future research trajectories are likely to encompass investigations into the relationship between HF and pyroptosis, as well as clinical studies on pharmaceuticals targeting the NLRP3 inflammasome as a therapeutic approach for HF. Conclusion This investigation provides a comprehensive bibliometric analysis and synopsis of NLRP3 inflammable-related studies in HF. The findings offer a conceptual foundation for further research on the NLRP3 inflammasome in HF and provide valuable guidance for future research directions in this domain.
Collapse
Affiliation(s)
- YanJie Lian
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
- Graduate School of Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Heyi Zhang
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
- Graduate School of Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Wenlong Xing
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Sinai Li
- Beijing Institute of Traditional Chinese Medicine, Beijing, 100010, People’s Republic of China
| | - Xiaolei Lai
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Sihan Jia
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - JuJu Shang
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Hongxu Liu
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| |
Collapse
|
9
|
DeBerge M, Glinton K, Lantz C, Ge ZD, Sullivan DP, Patil S, Lee BR, Thorp MI, Mullick A, Yeh S, Han S, van der Laan AM, Niessen HWM, Luo X, Sibinga NES, Thorp EB. Mechanical regulation of macrophage metabolism by allograft inflammatory factor 1 leads to adverse remodeling after cardiac injury. NATURE CARDIOVASCULAR RESEARCH 2025; 4:83-101. [PMID: 39747455 DOI: 10.1038/s44161-024-00585-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 11/12/2024] [Indexed: 01/04/2025]
Abstract
Myocardial infarction (MI) mobilizes macrophages, the central protagonists of tissue repair in the infarcted heart. Although necessary for repair, macrophages also contribute to adverse remodeling and progression to heart failure. In this context, specific targeting of inflammatory macrophage activation may attenuate maladaptive responses and enhance cardiac repair. Allograft inflammatory factor 1 (AIF1) is a macrophage-specific protein expressed in a variety of inflammatory settings, but its function after MI is unknown. Here we identify a maladaptive role for macrophage AIF1 after MI in mice. Mechanistic studies show that AIF1 increases actin remodeling in macrophages to promote reactive oxygen species-dependent activation of hypoxia-inducible factor (HIF)-1α. This directs a switch to glycolytic metabolism to fuel macrophage-mediated inflammation, adverse ventricular remodeling and progression to heart failure. Targeted knockdown of Aif1 using antisense oligonucleotides improved cardiac repair, supporting further exploration of macrophage AIF1 as a therapeutic target after MI.
Collapse
Affiliation(s)
- Matthew DeBerge
- Department of Anesthesiology, Critical Care, and Pain Medicine, The University of Texas Health Science Center, Houston, TX, USA.
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| | | | - Connor Lantz
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Zhi-Dong Ge
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - David P Sullivan
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Swapna Patil
- Department of Anesthesiology, Critical Care, and Pain Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Bo Ryung Lee
- Department of Anesthesiology, Critical Care, and Pain Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Minori I Thorp
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | | | - Steve Yeh
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Shuling Han
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Anja M van der Laan
- Department of Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Hans W M Niessen
- Department of Pathology and Cardiac Surgery, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU Medical Center, Amsterdam, The Netherlands
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Nicholas E S Sibinga
- Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
10
|
Rao SV, Lerner JBA. Inflammation in the Peri-ACS Period: Ready for Prime Time? Curr Atheroscler Rep 2024; 27:20. [PMID: 39714732 DOI: 10.1007/s11883-024-01263-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
PURPOSE Inflammation has been demonstrated to negatively impact patients in the peri-ACS period. This narrative review outlines the inflammatory response in ACS, highlighting the role of the NLRP3 inflammasome pathway following acute plaque rupture and coronary intervention and its potential as a pharmacologic target. RECENT: nvestigators have leveraged medications targeting the NLRP3 inflammasome currently used for other inflammatory pathologies, including colchicine, tocilizumab and anakinra. Investigation into these drugs in the peri-ACS period has yielded varying results, with the most encouraging findings in ACS patients treated with tocilizumab. More conflicting data exists for the role of colchicine and anakinra, with many studies limited in their power to detect clinical outcomes and heterogeneity in their patient populations and endpoints. Despite conflicting data, the NLRP3 remains an attractive therapeutic target in the peri-ACS period. Further investigation is required to prove benefit and safety with large clinical trials adequately powered for clinical outcomes.
Collapse
Affiliation(s)
- Sunil V Rao
- Department of Medicine, Division of Cardiology, New York University Langone Medical Center, NYU Langone Health System, 550 1st Ave, New York, NY, 10010, USA.
| | - Johanna Ben-Ami Lerner
- Department of Medicine, Division of Cardiology, New York University Langone Medical Center, NYU Langone Health System, 550 1st Ave, New York, NY, 10010, USA
| |
Collapse
|
11
|
Celeski M, Segreti A, Crisci F, Cricco R, Piscione M, Di Gioia G, Nusca A, Fossati C, Pigozzi F, Ussia GP, Solaro RJ, Grigioni F. The Role of Cardiac Troponin and Other Emerging Biomarkers Among Athletes and Beyond: Underlying Mechanisms, Differential Diagnosis, and Guide for Interpretation. Biomolecules 2024; 14:1630. [PMID: 39766337 PMCID: PMC11727179 DOI: 10.3390/biom14121630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025] Open
Abstract
Cardiovascular (CV) disease remains the leading cause of morbidity and mortality worldwide, highlighting the necessity of understanding its underlying molecular and pathophysiological pathways. Conversely, physical activity (PA) and exercise are key strategies in reducing CV event risks. Detecting latent CV conditions in apparently healthy individuals, such as athletes, presents a unique challenge. The early identification and treatment of CV disorders are vital for long-term health and patient survival. Cardiac troponin is currently the most commonly used biomarker for assessing CV changes in both athletes and the general population. However, there remains considerable debate surrounding the mechanisms underlying exercise-induced troponin elevations and its release in non-ischemic contexts. Thus, there is a pressing need to identify and implement more sensitive and specific biomarkers for CV disorders in clinical practice. Indeed, research continues to explore reliable biomarkers for evaluating the health of athletes and the effectiveness of physical exercise. It is essential to analyze current evidence on troponin release in non-ischemic conditions, post-strenuous exercise, and the complex biological pathways that influence its detection. Furthermore, this study summarizes current research on cytokines and exosomes, including their physiological roles and their relevance in various CV conditions, especially in athletes. In addition, this paper gives special attention to underlying mechanisms, potential biomarkers, and future perspectives.
Collapse
Affiliation(s)
- Mihail Celeski
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Andrea Segreti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Filippo Crisci
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Riccardo Cricco
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Mariagrazia Piscione
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Giuseppe Di Gioia
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
- Institute of Sports Medicine and Science, Italian National Olympic Committee, Largo Piero Gabrielli 1, 00197 Roma, Italy
| | - Annunziata Nusca
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Chiara Fossati
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Ross John Solaro
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Francesco Grigioni
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
12
|
Potere N, Bonaventura A, Abbate A. Novel Therapeutics and Upcoming Clinical Trials Targeting Inflammation in Cardiovascular Diseases. Arterioscler Thromb Vasc Biol 2024; 44:2371-2395. [PMID: 39387118 PMCID: PMC11602387 DOI: 10.1161/atvbaha.124.319980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cardiovascular disease (CVD) remains a major health burden despite significant therapeutic advances accomplished over the last decades. It is widely and increasingly recognized that systemic inflammation not only represents a major cardiovascular risk and prognostic factor but also plays key pathogenic roles in CVD development and progression. Despite compelling preclinical evidence suggesting large potential of anti-inflammatory pharmacological interventions across numerous CVDs, clinical translation remains incomplete, mainly due to (1) yet undefined molecular signaling; (2) challenges of safety and efficacy profile of anti-inflammatory drugs; and (3) difficulties in identifying optimal patient candidates and responders to anti-inflammatory therapeutics, as well as optimal therapeutic windows. Randomized controlled trials demonstrated the safety/efficacy of canakinumab and colchicine in secondary cardiovascular prevention, providing confirmation for the involvement of a specific inflammatory pathway (NLRP3 [NACHT, LRR, and PYD domain-containing protein 3] inflammasome/IL [interleukin]-1β) in atherosclerotic CVD. Colchicine was recently approved by the US Food and Drug Administration for this indication. Diverse anti-inflammatory drugs targeting distinct inflammatory pathways are widely used for the management of other CVDs including myocarditis and pericarditis. Ongoing research efforts are directed to implementing anti-inflammatory therapeutic strategies across a growing number of CVDs, through repurposing of available anti-inflammatory drugs and development of novel anti-inflammatory compounds, which are herein concisely discussed. This review also summarizes the main characteristics and findings of completed and upcoming randomized controlled trials directly targeting inflammation in CVDs, and discusses major challenges and future perspectives in the exciting and constantly expanding landscape of cardioimmunology.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Aldo Bonaventura
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Italy
| | - Antonio Abbate
- Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
13
|
d'Aiello A, Filomia S, Brecciaroli M, Sanna T, Pedicino D, Liuzzo G. Targeting Inflammatory Pathways in Atherosclerosis: Exploring New Opportunities for Treatment. Curr Atheroscler Rep 2024; 26:707-719. [PMID: 39404934 PMCID: PMC11530513 DOI: 10.1007/s11883-024-01241-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/02/2024]
Abstract
PURPOSE OF THE REVIEW This review discusses the molecular mechanisms involved in the immuno-pathogenesis of atherosclerosis, the pleiotropic anti-inflammatory effects of approved cardiovascular therapies and the available evidence on immunomodulatory therapies for atherosclerotic cardiovascular disease (ACVD). We highlight the importance of clinical and translational research in identifying molecular mechanisms and discovering new therapeutic targets. RECENT FINDINGS The CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcomes Study) trial was the first to demonstrate a reduction in cardiovascular (CV) risk with anti-inflammatory therapy, irrespective of serum lipid levels. ACVD is the leading cause of death worldwide. Although targeting principal risk factors significantly reduces CV risk, residual risk remains unaddressed. The immunological mechanisms underlying atherosclerosis represent attractive therapeutic targets. Several commonly used and non-primarily anti-inflammatory drugs (i.e. SGLT2i, and PCSK9i) exhibit pleiotropic properties. Otherwise, recent trials have investigated the blockade of primarily inflammatory compounds, trying to lower the residual risk via low-dose IL-2, PTPN22 and CD31 pathway modulation. In the era of precision medicine, modern approaches may explore new pharmacological targets, identify new markers of vascular inflammation, and evaluate therapeutic responses.
Collapse
Affiliation(s)
- Alessia d'Aiello
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Simone Filomia
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| | - Mattia Brecciaroli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| | - Tommaso Sanna
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| | - Daniela Pedicino
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy.
| | - Giovanna Liuzzo
- Department of Cardiovascular Sciences- CUORE, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University School of Medicine, Largo F. Vito 1, 00168, Rome, Italy
| |
Collapse
|
14
|
Abbate A, Van Tassell B, Bogin V, Markley R, Pevzner DV, Cremer PC, Meray IA, Privalov DV, Taylor A, Grishin SA, Egorova AN, Ponomar EG, Lavrovsky Y, Samsonov MY. Interleukin-1 Blockade With RPH-104 (Goflikicept) in Patients With ST-Segment Elevation Myocardial Infarction: Secondary End Points From an International, Double-Blind, Randomized, Placebo-Controlled, Phase 2a Study. J Cardiovasc Pharmacol 2024; 84:565-577. [PMID: 39642282 PMCID: PMC11617079 DOI: 10.1097/fjc.0000000000001635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 08/31/2024] [Indexed: 10/04/2024]
Abstract
ABSTRACT In a randomized double-blinded clinical trial of patients with ST segment elevation myocardial infarction (STEMI), goflikicept, an interleukin-1 blocker, significantly reduced systemic inflammation, measured as the area under the curve (AUC) for high-sensitivity C reactive protein at 14 days. We report secondary analyses of biomarkers at 28 days, and cardiac function and clinical end points at 1 year. Patients received a single administration of goflikicept 80 mg (n = 34), goflikicept 160 mg (n = 34), or placebo (n = 34). Both doses of goflikicept significantly reduced the AUC for high-sensitivity C reactive protein at 28 days compared with placebo, without statistically significant differences between the doses. There were no statistically significant differences between groups in the AUC for natriuretic peptides at 28 days. There were no significant differences between placebo, goflikicept 80 mg, and 160 mg groups in deaths (2.9%, 2.9%, and 0%), hospitalization for cardiovascular reasons (9.1%, 5.9%, and 0%), new-onset or progression of heart failure (9.1%, 5.9%, and 5.9%), and new or increased use of loop diuretics (24.2%, 14.7%, and 17.6%), nor in the number of patients with treatment emergent adverse events, with no treatment-related serious adverse events in any group. In conclusion, in patients with STEMI, interleukin-1 blockade with goflikicept 80 mg or 160 mg was well tolerated and associated with significant reduction of systemic inflammation. Further adequately powered studies are warranted to determine whether the reduction in systemic inflammation with goflikicept translates into a clinical benefit in patients with STEMI.
Collapse
Affiliation(s)
| | | | | | | | - Dmitry V. Pevzner
- “National Medical Research Center for Cardiology” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | | | - Imad A. Meray
- Peoples' Friendship University of Russia n.a Patrice Lumumba (RUDN University), Moscow, Russia
| | - Dmitry V. Privalov
- “City Clinical Hospital No. 51 of Moscow Healthcare Department”, Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
15
|
Yan J, Liu H, Shang J, Fang Q, Ye J, Lu X, Fan X. Protective effects of Shexiang-Tongxin dropping pill against acute myocardial infarction in rats through inhibition of apoptosis and ERK/MAPK signaling pathways. Heliyon 2024; 10:e39939. [PMID: 39553562 PMCID: PMC11565526 DOI: 10.1016/j.heliyon.2024.e39939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
Acute myocardial infarction (AMI) remains a significant health challenge globally, highlighting the ongoing need for effective treatments. Shexiang-Tongxin dropping pill (STDP) is widely utilized as a therapeutic option for AMI in China and Southeast Asia. However, the intricate mechanisms of action of STDP against AMI remain largely unknown. The pharmacodynamic effects of STDP in treating AMI were evaluated both in vitro and in vivo using human umbilical vein endothelial cell oxygen-glucose deprivation, RAW264.7 cell inflammatory injury, and rat left anterior descending surgery models. The whole transcriptome sequencing was performed to analyze gene expression changes in experimental rat hearts after left anterior descending surgery. An integrative approach combining network pharmacology and sequencing data was used to determine the multi-target and multi-pathway mechanisms underlying the action of STDP against AMI. Molecular docking was conducted to identify the primary anti-AMI ingredients in STDP. STDP treatment significantly resisted AMI in vivo and protected against inflammatory and hypoxic injuries in vitro. It resulted in 63 % (901 of 1430) of genes showing restorative regulation in the AMI disease network, relating to the TGF-β, PI3K, apoptosis, and MAPK pathways. Validation experiments indicated that inhibiting apoptosis and ERK/MAPK pathways by reducing Bax and p-ERK1/2 expression levels in rat hearts may be a crucial mechanism of STDP against AMI. Molecular target prediction indicated that tanshinone IIA, salvianolic acid A, salvianolic acid B, and resibufogenin were the essential pharmacodynamic substances of STDP in AMI treatment. This study sheds light on novel mechanisms by which STDP rebalances the AMI disease network through its multi-target and multi-pathway effects. The findings offer data support for the more precise clinical application of STDP.
Collapse
Affiliation(s)
- Jun Yan
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hanbing Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiaxin Shang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qianqian Fang
- Inner Mongolia Conba Pharmaceutical Co., Ltd., Ordos, 017000, China
| | - Jianfeng Ye
- Inner Mongolia Conba Pharmaceutical Co., Ltd., Ordos, 017000, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, 314100, Jiaxing, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Xiaohui Fan
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, 314100, Jiaxing, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| |
Collapse
|
16
|
Matter MA, Tschaikowsky T, Stähli BE, Matter CM. Acute-on-chronic inflammation in acute myocardial infarction. Curr Opin Cardiol 2024; 39:535-542. [PMID: 39195569 DOI: 10.1097/hco.0000000000001176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
PURPOSE OF REVIEW Acute myocardial infarction (AMI) is heralded by chronic inflammation and entails an excessive burst of acute-on-chronic inflammation (AoCI). This review describes the evolution from understanding atherosclerosis as a chronic inflammatory disease, to recent efforts in optimizing anti-inflammatory therapy to patients with AMI. It highlights the challenges and opportunities in selecting the optimal patient with AMI to derive maximal benefit from early anti-inflammatory therapy. RECENT FINDINGS The causal role of inflammation in atherosclerosis has been proven in large outcome trials. Since then, several smaller trials have sought to translate the concept of anti-inflammatory therapy targeting residual inflammatory risk to the dynamic early phase of AoCI after AMI. Current evidence highlights the importance of selecting patients with a high inflammatory burden. Surrogate criteria for large AMI (e.g., angiographic or electrocardiographic), as well as novel point-of-care biomarker testing may aid in selecting patients with particularly elevated AoCI. Additionally, patients presenting with AMI complicated by pro-inflammatory sequelae (e.g., atrial fibrillation, acute heart failure, left ventricular thrombosis) may dually profit from anti-inflammatory therapy. SUMMARY Improved understanding of the mechanisms and dynamics of acute and chronic inflammatory processes after AMI may aid the strive to optimize early anti-inflammatory therapy to patients with AMI.
Collapse
Affiliation(s)
- Michael A Matter
- Department of Cardiology, University Heart Center, University Hospital of Zurich
| | - Tristan Tschaikowsky
- Department of Cardiology, University Heart Center, University Hospital of Zurich
| | - Barbara E Stähli
- Department of Cardiology, University Heart Center, University Hospital of Zurich
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital of Zurich and University of Zurich, Zurich, Switzerland
| | - Christian M Matter
- Department of Cardiology, University Heart Center, University Hospital of Zurich
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital of Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Porsch F, Binder CJ. Autoimmune diseases and atherosclerotic cardiovascular disease. Nat Rev Cardiol 2024; 21:780-807. [PMID: 38937626 DOI: 10.1038/s41569-024-01045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Autoimmune diseases are associated with a dramatically increased risk of atherosclerotic cardiovascular disease and its clinical manifestations. The increased risk is consistent with the notion that atherogenesis is modulated by both protective and disease-promoting immune mechanisms. Notably, traditional cardiovascular risk factors such as dyslipidaemia and hypertension alone do not explain the increased risk of cardiovascular disease associated with autoimmune diseases. Several mechanisms have been implicated in mediating the autoimmunity-associated cardiovascular risk, either directly or by modulating the effect of other risk factors in a complex interplay. Aberrant leukocyte function and pro-inflammatory cytokines are central to both disease entities, resulting in vascular dysfunction, impaired resolution of inflammation and promotion of chronic inflammation. Similarly, loss of tolerance to self-antigens and the generation of autoantibodies are key features of autoimmunity but are also implicated in the maladaptive inflammatory response during atherosclerotic cardiovascular disease. Therefore, immunomodulatory therapies are potential efficacious interventions to directly reduce the risk of cardiovascular disease, and biomarkers of autoimmune disease activity could be relevant tools to stratify patients with autoimmunity according to their cardiovascular risk. In this Review, we discuss the pathophysiological aspects of the increased cardiovascular risk associated with autoimmunity and highlight the many open questions that need to be answered to develop novel therapies that specifically address this unmet clinical need.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Gissler MC, Antiochos P, Ge Y, Heydari B, Gräni C, Kwong RY. Cardiac Magnetic Resonance Evaluation of LV Remodeling Post-Myocardial Infarction: Prognosis, Monitoring and Trial Endpoints. JACC Cardiovasc Imaging 2024; 17:1366-1380. [PMID: 38819335 DOI: 10.1016/j.jcmg.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/14/2024] [Indexed: 06/01/2024]
Abstract
Adverse left ventricular remodeling (ALVR) and subsequent heart failure after myocardial infarction (MI) remain a major cause of patient morbidity and mortality worldwide. Overt inflammation has been identified as the common pathway underlying myocardial fibrosis and development of ALVR post-MI. With its ability to simultaneously provide information about cardiac structure, function, perfusion, and tissue characteristics, cardiac magnetic resonance (CMR) is well poised to inform prognosis and guide early surveillance and therapeutics in high-risk cohorts. Further, established and evolving CMR-derived biomarkers may serve as clinical endpoints in prospective trials evaluating the efficacy of novel anti-inflammatory and antifibrotic therapies. This review provides an overview of post-MI ALVR and illustrates how CMR may help clinical adoption of novel therapies via mechanistic or prognostic imaging markers.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Panagiotis Antiochos
- Cardiology and Cardiac MR Centre, University Hospital Lausanne, Lausanne, Switzerland
| | - Yin Ge
- Division of Cardiology, St Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Bobak Heydari
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christoph Gräni
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raymond Y Kwong
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
19
|
Bonaventura A, Moroni F, Golino M, Del Buono MG, Vecchié A, Potere N, Abbate A. IL-1 blockade in cardiovascular disease: an appraisal of the evidence across different inflammatory paradigms. Minerva Cardiol Angiol 2024; 72:477-488. [PMID: 37705370 DOI: 10.23736/s2724-5683.23.06390-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Pre-clinical and clinical studies suggest a role for inflammation in the pathophysiology of cardiovascular (CV) diseases. The NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein 3) inflammasome is activated during tissue injury and releases interleukin-1β (IL-1β). We describe three paradigms in which the NLRP3 inflammasome and IL-1β contribute to CV diseases. During acute myocardial infarction (AMI), necrotic cell debris, including IL-1α, induce NLRP3 inflammasome activation and further damage the myocardium contributing to heart failure (HF) (acute injury paradigm). In chronic HF, IL-1β is induced by persistent myocardial overload and injury, neurohumoral activation and systemic comorbidities favoring infiltration and activation of immune cells into the myocardium, microvascular inflammation, and a pro-fibrotic response (chronic inflammation paradigm). In recurrent pericarditis, an autoinflammatory response triggered by cell injury and maintained by the NLRP3 inflammasome/IL-1β axis is present (autoinflammatory disease paradigm). Anakinra, recombinant IL-1 receptor antagonist, inhibits the acute inflammatory response in patients with ST elevation myocardial infarction (STEMI) and acute HF. Canakinumab, IL-1β antibody, blunts systemic inflammation and prevents complications of atherosclerosis in stable patients with prior AMI. In chronic HF, anakinra reduces systemic inflammation and improves cardiorespiratory fitness. In recurrent pericarditis, anakinra and rilonacept, a soluble IL-1 receptor chimeric fusion protein blocking IL-1α and IL-1β, treat and prevent acute flares. In conclusion, the NLRP3 inflammasome and IL-1 contribute to the pathophysiology of CV diseases, and IL-1 blockade is beneficial with different roles in the acute injury, chronic inflammation and autoinflammatory disease paradigms. Further research is needed to guide the optimal use of IL-1 blockers in clinical practice.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Division of Internal Medicine, Medical Center, Ospedale di Circolo & Fondazione Macchi, ASST Sette Laghi, Varese, Italy -
| | - Francesco Moroni
- Heart and Vascular Center, Division of Cardiology, Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Michele Golino
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
- VCU Health Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Marco G Del Buono
- Department of Cardiovascular and Thoracic Sciences, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Alessandra Vecchié
- Division of Internal Medicine, Medical Center, Ospedale di Circolo & Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Nicola Potere
- Department of Medicine and Ageing Sciences, "G. D'Annunzio" University, Chieti, Italy
| | - Antonio Abbate
- Heart and Vascular Center, Division of Cardiology, Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
20
|
Golino M, Morello M. Beyond Remodeling: Interleukin-1 Blockade as a Therapeutic Strategy to Prevent Heart Failure After Myocardial Infarction. JACC. HEART FAILURE 2024; 12:1800-1801. [PMID: 39384291 DOI: 10.1016/j.jchf.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 10/11/2024]
Affiliation(s)
- Michele Golino
- Virginia Commonwealth University, Richmond, Virginia, USA; University of Virginia, Charlottesville, Virginia, USA.
| | | |
Collapse
|
21
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, Dayer M, Alegría-Barrero E, De Sanctis JB, Parise Vasco JM, Riera Lizardo RJ, Nicola S, Martí-Amarista CE, Correa-Pérez A. Interleukin-receptor antagonist and tumour necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Cochrane Database Syst Rev 2024; 9:CD014741. [PMID: 39297531 PMCID: PMC11411914 DOI: 10.1002/14651858.cd014741.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ACVD) is worsened by chronic inflammatory diseases. Interleukin receptor antagonists (IL-RAs) and tumour necrosis factor-alpha (TNF) inhibitors have been studied to see if they can prevent cardiovascular events. OBJECTIVES The purpose of this study was to assess the clinical benefits and harms of IL-RAs and TNF inhibitors in the primary and secondary prevention of ACVD. SEARCH METHODS The Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, EBSCO CINAHL plus, and clinical trial registries for ongoing and unpublished studies were searched in February 2024. The reference lists of relevant studies, reviews, meta-analyses and health technology reports were searched to identify additional studies. No limitations on language, date of publication or study type were set. SELECTION CRITERIA RCTs that recruited people with and without pre-existing ACVD, comparing IL-RAs or TNF inhibitors versus placebo or usual care, were selected. The primary outcomes considered were all-cause mortality, myocardial infarction, unstable angina, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors, working independently at each step, selected studies, extracted data, assessed the risk of bias and used GRADE to judge the certainty of evidence. MAIN RESULTS We included 58 RCTs (22,053 participants; 21,308 analysed), comparing medication efficacy with placebo or usual care. Thirty-four trials focused on primary prevention and 24 on secondary prevention. The interventions included IL-1 RAs (anakinra, canakinumab), IL-6 RA (tocilizumab), TNF-inhibitors (etanercept, infliximab) compared with placebo or usual care. The certainty of evidence was low to very low due to biases and imprecision; all trials had a high risk of bias. Primary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality(RR 0.33, 95% CI 0.01 to 7.58, 1 trial), myocardial infarction (RR 0.71, 95% CI 0.04 to 12.48, I² = 39%, 2 trials), unstable angina (RR 0.24, 95% CI 0.03 to 2.11, I² = 0%, 2 trials), stroke (RR 2.42, 95% CI 0.12 to 50.15; 1 trial), adverse events (RR 0.85, 95% CI 0.59 to 1.22, I² = 54%, 3 trials), or infection (rate ratio 0.84, 95% 0.55 to 1.29, I² = 0%, 4 trials). Evidence is very uncertain about whether anakinra and cankinumab may reduce heart failure (RR 0.21, 95% CI 0.05 to 0.94, I² = 0%, 3 trials). Peripheral vascular disease (PVD) was not reported as an outcome. IL-6 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.68, 95% CI 0.12 to 3.74, I² = 30%, 3 trials), myocardial infarction (RR 0.27, 95% CI 0.04 to1.68, I² = 0%, 3 trials), heart failure (RR 1.02, 95% CI 0.11 to 9.63, I² = 0%, 2 trials), PVD (RR 2.94, 95% CI 0.12 to 71.47, 1 trial), stroke (RR 0.34, 95% CI 0.01 to 8.14, 1 trial), or any infection (rate ratio 1.10, 95% CI: 0.88 to 1.37, I2 = 18%, 5 trials). Adverse events may increase (RR 1.13, 95% CI 1.04 to 1.23, I² = 33%, 5 trials). No trial assessed unstable angina. TNF inhibitors The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.78, 95% CI 0.63 to 4.99, I² = 10%, 3 trials), myocardial infarction (RR 2.61, 95% CI 0.11 to 62.26, 1 trial), stroke (RR 0.46, 95% CI 0.08 to 2.80, I² = 0%; 3 trials), heart failure (RR 0.85, 95% CI 0.06 to 12.76, 1 trial). Adverse events may increase (RR 1.13, 95% CI 1.01 to 1.25, I² = 51%, 13 trials). No trial assessed unstable angina or PVD. Secondary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.94, 95% CI 0.84 to 1.06, I² = 0%, 8 trials), unstable angina (RR 0.88, 95% CI 0.65 to 1.19, I² = 0%, 3 trials), PVD (RR 0.85, 95% CI 0.19 to 3.73, I² = 38%, 3 trials), stroke (RR 0.94, 95% CI 0.74 to 1.2, I² = 0%; 7 trials), heart failure (RR 0.91, 95% 0.5 to 1.65, I² = 0%; 7 trials), or adverse events (RR 0.92, 95% CI 0.78 to 1.09, I² = 3%, 4 trials). There may be little to no difference between the groups in myocardial infarction (RR 0.88, 95% CI 0.0.75 to 1.04, I² = 0%, 6 trials). IL6-RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.09, 95% CI 0.61 to 1.96, I² = 0%, 2 trials), myocardial infarction (RR 0.46, 95% CI 0.07 to 3.04, I² = 45%, 3 trials), unstable angina (RR 0.33, 95% CI 0.01 to 8.02, 1 trial), stroke (RR 1.03, 95% CI 0.07 to 16.25, 1 trial), adverse events (RR 0.89, 95% CI 0.76 to 1.05, I² = 0%, 2 trials), or any infection (rate ratio 0.66, 95% CI 0.32 to 1.36, I² = 0%, 4 trials). No trial assessed PVD or heart failure. TNF inhibitors The evidence is very uncertain about the effect of the intervention on all-cause mortality (RR 1.16, 95% CI 0.69 to 1.95, I² = 47%, 5 trials), heart failure (RR 0.92, 95% 0.75 to 1.14, I² = 0%, 4 trials), or adverse events (RR 1.15, 95% CI 0.84 to 1.56, I² = 32%, 2 trials). No trial assessed myocardial infarction, unstable angina, PVD or stroke. Adverse events may be underestimated and benefits inflated due to inadequate reporting. AUTHORS' CONCLUSIONS This Cochrane review assessed the benefits and harms of using interleukin-receptor antagonists and tumour necrosis factor inhibitors for primary and secondary prevention of atherosclerotic diseases compared with placebo or usual care. However, the evidence for the predetermined outcomes was deemed low or very low certainty, so there is still a need to determine whether these interventions provide clinical benefits or cause harm from this perspective. In summary, the different biases and imprecision in the included studies limit their external validity and represent a limitation to determining the effectiveness of the intervention for both primary and secondary prevention of ACVD.
Collapse
Key Words
- humans
- angina, unstable
- angina, unstable/mortality
- angina, unstable/prevention & control
- antibodies, monoclonal, humanized
- antibodies, monoclonal, humanized/administration & dosage
- antibodies, monoclonal, humanized/adverse effects
- atherosclerosis
- atherosclerosis/mortality
- atherosclerosis/prevention & control
- bias
- cause of death
- myocardial infarction
- myocardial infarction/mortality
- myocardial infarction/prevention & control
- primary prevention
- primary prevention/methods
- randomized controlled trials as topic
- receptors, interleukin-1
- receptors, interleukin-1/antagonists & inhibitors
- secondary prevention
- secondary prevention/methods
- tumor necrosis factor-alpha
- tumor necrosis factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
- Facultad de Medicina (Centro Cochrane Madrid), Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia , Venezuela
| | - Mario A Gemmato-Valecillos
- Icahn School of Medicine at Mount Sinai/ NYCHH Elmhurst Hospital Center, 79-01 Broadway, Elmhurst, New York 11373, USA
| | | | - Mark Dayer
- Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Palacky University, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | - Juan Marcos Parise Vasco
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Susana Nicola
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | | | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Hospital Pharmacy and Medical Devices Department, Hospital Central de la Defensa "Gómez Ulla" CSVE, Madrid, Spain
| |
Collapse
|
22
|
Zubirán R, Neufeld EB, Dasseux A, Remaley AT, Sorokin AV. Recent Advances in Targeted Management of Inflammation In Atherosclerosis: A Narrative Review. Cardiol Ther 2024; 13:465-491. [PMID: 39031302 PMCID: PMC11333429 DOI: 10.1007/s40119-024-00376-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/26/2024] [Indexed: 07/22/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of morbidity and mortality despite effective low-density lipoprotein cholesterol-targeted therapies. This review explores the crucial role of inflammation in the residual risk of ASCVD, emphasizing its impact on atherosclerosis progression and plaque stability. Evidence suggests that high-sensitivity C-reactive protein (hsCRP), and potentially other inflammatory biomarkers, can be used to identify the inflammatory residual ASCVD risk phenotype and may serve as future targets for the development of more efficacious therapeutic approaches. We review the biological basis for the association of inflammation with ASCVD, propose new therapeutic strategies for the use of inflammation-targeted treatments, and discuss current challenges in the implementation of this new treatment paradigm for ASCVD.
Collapse
Affiliation(s)
- Rafael Zubirán
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Edward B Neufeld
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amaury Dasseux
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V Sorokin
- Lipoprotein Metabolism Laboratory, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
- Section of Inflammation and Cardiometabolic Diseases, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
- Section of Lipoprotein Metabolism, Clinical Research Center, National Heart, Lung and Blood Institute, 9000 Rockville Pike, Bldg 10, Room 5-5150, Bethesda, MD, 20892, USA.
| |
Collapse
|
23
|
Muller R, Cauchois R, Lagarde M, Roffino S, Genovesio C, Fernandez S, Hache G, Guillet B, Kara Y, Marlinge M, Lenting P, Poullin P, Dignat-George F, Tellier E, Kaplanski G. Reduction of mortality, cardiac damage, and cerebral damage by IL-1 inhibition in a murine model of TTP. Blood 2024; 143:2791-2803. [PMID: 38598839 DOI: 10.1182/blood.2023021974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/09/2024] [Accepted: 03/09/2024] [Indexed: 04/12/2024] Open
Abstract
ABSTRACT Thrombotic thrombocytopenic purpura (TTP), a rare but fatal disease if untreated, is due to alteration in von Willebrand factor cleavage resulting in capillary microthrombus formation and ischemic organ damage. Interleukin-1 (IL-1) has been shown to drive sterile inflammation after ischemia and could play an essential contribution to postischemic organ damage in TTP. Our objectives were to evaluate IL-1 involvement during TTP and to test the efficacy of the recombinant IL-1 receptor antagonist, anakinra, in a murine TTP model. We retrospectively measured plasma IL-1 concentrations in patients with TTP and controls. Patients with TTP exhibited elevated plasma IL-1α and -1β concentrations, which correlated with disease course and survival. In a mouse model of TTP, we administered anakinra (IL-1 inhibitor) or placebo for 5 days and evaluated the efficacy of this treatment. Anakinra significantly reduced mortality of mice (P < .001). Anakinra significantly decreased TTP-induced cardiac damage as assessed by blood troponin concentrations, evaluation of left ventricular function by echocardiography, [18F]fluorodeoxyglucose positron emission tomography of myocardial glucose metabolism, and cardiac histology. Anakinra also significantly reduced brain TTP-induced damage evaluated through blood PS100b concentrations, nuclear imaging, and histology. We finally showed that IL-1α and -1β trigger endothelial degranulation in vitro, leading to the release of von Willebrand factor. In conclusion, anakinra significantly reduced TTP mortality in a preclinical model of the disease by inhibiting both endothelial degranulation and postischemic inflammation, supporting further evaluations in humans.
Collapse
Affiliation(s)
- Romain Muller
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Department of Clinical Immunology and Internal Medicine, CHU Conception, Marseille, France
| | - Raphaël Cauchois
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Department of Clinical Immunology and Internal Medicine, CHU Conception, Marseille, France
- French Reference Center for Thrombotic Microangiopathies, Paris, France
| | - Marie Lagarde
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
- French Reference Center for Thrombotic Microangiopathies, Paris, France
| | - Sandrine Roffino
- Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Aix-Marseille University, Marseille, France
| | - Cécile Genovesio
- Faculté de Pharmacie, Aix-Marseille University, Marseille, France
| | - Samantha Fernandez
- Assistance Publique des Hôpitaux de Marseille, INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Centre Européen de Recherche en Imagerie Médicale, CHU Timone, Aix-Marseille University, Marseille, France
| | - Guillaume Hache
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
- Biology Department, Assistance Publique des Hôpitaux de Marseille, INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Centre Européen de Recherche en Imagerie Médicale, CHU Timone, Aix-Marseille University, Marseille, France
| | - Benjamin Guillet
- Biology Department, Assistance Publique des Hôpitaux de Marseille, INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Centre Européen de Recherche en Imagerie Médicale, CHU Timone, Aix-Marseille University, Marseille, France
| | - Yéter Kara
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
| | - Marion Marlinge
- Biology Department, Assistance Publique des Hôpitaux de Marseille, INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Centre Européen de Recherche en Imagerie Médicale, CHU Timone, Aix-Marseille University, Marseille, France
| | - Peter Lenting
- INSERM, Hémostase Inflammation Thrombose HITh U1176, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Pascale Poullin
- French Reference Center for Thrombotic Microangiopathies, Paris, France
- Assistance Publique des Hôpitaux de Marseille, Service d'Hémaphérése, CHU Conception, Marseille, France
| | - Françoise Dignat-George
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
- Department of Hematology and Vascular Biology, Assistance Publique des Hôpitaux de Marseille, INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, CHU Conception, Aix-Marseille University, Marseille, France
| | - Edwige Tellier
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
- French Reference Center for Thrombotic Microangiopathies, Paris, France
| | - Gilles Kaplanski
- INSERM, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Centre de Recherche en CardioVasculaire et Nutrition, Aix Marseille University, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Department of Clinical Immunology and Internal Medicine, CHU Conception, Marseille, France
| |
Collapse
|
24
|
Hilgendorf I, Frantz S, Frangogiannis NG. Repair of the Infarcted Heart: Cellular Effectors, Molecular Mechanisms and Therapeutic Opportunities. Circ Res 2024; 134:1718-1751. [PMID: 38843294 PMCID: PMC11164543 DOI: 10.1161/circresaha.124.323658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through the activation of inflammatory and fibrogenic cascades that ultimately result in the formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of damage-associated molecular patterns that initiate both myocardial and systemic inflammatory responses. TLRs (toll-like receptors) and NLRs (NOD-like receptors) recognize damage-associated molecular patterns (DAMPs) and transduce downstream proinflammatory signals, leading to upregulation of cytokines (such as interleukin-1, TNF-α [tumor necrosis factor-α], and interleukin-6) and chemokines (such as CCL2 [CC chemokine ligand 2]) and recruitment of neutrophils, monocytes, and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart play a major role in the clearance of the infarct from dead cells and the subsequent stimulation of reparative pathways. Efferocytosis triggers the induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for the activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades, and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to the stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and inhibit arrhythmia generation in patients surviving myocardial infarction.
Collapse
Affiliation(s)
- Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine at the University of Freiburg, Freiburg, Germany
| | - Stefan Frantz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
25
|
Wang DD, Naumova AV, Isquith D, Sapp J, Huynh KA, Tucker I, Balu N, Voronyuk A, Chu B, Ordovas K, Maynard C, Tian R, Zhao XQ, Kim F. Dapagliflozin reduces systemic inflammation in patients with type 2 diabetes without known heart failure. Cardiovasc Diabetol 2024; 23:197. [PMID: 38849829 PMCID: PMC11161924 DOI: 10.1186/s12933-024-02294-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
OBJECTIVE Sodium glucose cotransporter 2 (SGLT2) inhibitors significantly improve cardiovascular outcomes in diabetic patients; however, the mechanism is unclear. We hypothesized that dapagliflozin improves cardiac outcomes via beneficial effects on systemic and cardiac inflammation and cardiac fibrosis. RESEARCH AND DESIGN METHODS This randomized placebo-controlled clinical trial enrolled 62 adult patients (mean age 62, 17% female) with type 2 diabetes (T2D) without known heart failure. Subjects were randomized to 12 months of daily 10 mg dapagliflozin or placebo. For all patients, blood/plasma samples and cardiac magnetic resonance imaging (CMRI) were obtained at time of randomization and at the end of 12 months. Systemic inflammation was assessed by plasma IL-1B, TNFα, IL-6 and ketone levels and PBMC mitochondrial respiration, an emerging marker of sterile inflammation. Global myocardial strain was assessed by feature tracking; cardiac fibrosis was assessed by T1 mapping to calculate extracellular volume fraction (ECV); and cardiac tissue inflammation was assessed by T2 mapping. RESULTS Between the baseline and 12-month time point, plasma IL-1B was reduced (- 1.8 pg/mL, P = 0.003) while ketones were increased (0.26 mM, P = 0.0001) in patients randomized to dapagliflozin. PBMC maximal oxygen consumption rate (OCR) decreased over the 12-month period in the placebo group but did not change in patients receiving dapagliflozin (- 158.9 pmole/min/106 cells, P = 0.0497 vs. - 5.2 pmole/min/106 cells, P = 0.41), a finding consistent with an anti-inflammatory effect of SGLT2i. Global myocardial strain, ECV and T2 relaxation time did not change in both study groups. CLINICAL TRIAL GOV REGISTRATION NCT03782259.
Collapse
Affiliation(s)
- Dennis D Wang
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Anna V Naumova
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Daniel Isquith
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jamie Sapp
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kim A Huynh
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Isabella Tucker
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Niranjan Balu
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Anna Voronyuk
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Baocheng Chu
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Karen Ordovas
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Charles Maynard
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Rong Tian
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Xue-Qiao Zhao
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Francis Kim
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA.
- University of Washington, 850 Republican St, Box 358055, Seattle, WA, 98104, USA.
| |
Collapse
|
26
|
Welt FGP, Batchelor W, Spears JR, Penna C, Pagliaro P, Ibanez B, Drakos SG, Dangas G, Kapur NK. Reperfusion Injury in Patients With Acute Myocardial Infarction: JACC Scientific Statement. J Am Coll Cardiol 2024; 83:2196-2213. [PMID: 38811097 DOI: 10.1016/j.jacc.2024.02.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 05/31/2024]
Abstract
Despite impressive improvements in the care of patients with ST-segment elevation myocardial infarction, mortality remains high. Reperfusion is necessary for myocardial salvage, but the abrupt return of flow sets off a cascade of injurious processes that can lead to further necrosis. This has been termed myocardial ischemia-reperfusion injury and is the subject of this review. The pathologic and molecular bases for myocardial ischemia-reperfusion injury are increasingly understood and include injury from reactive oxygen species, inflammation, calcium overload, endothelial dysfunction, and impaired microvascular flow. A variety of pharmacologic strategies have been developed that have worked well in preclinical models and some have shown promise in the clinical setting. In addition, there are newer mechanical approaches including mechanical unloading of the heart prior to reperfusion that are in current clinical trials.
Collapse
Affiliation(s)
- Frederick G P Welt
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA.
| | | | - J Richard Spears
- Department of Cardiovascular Medicine, Beaumont Systems, Royal Oak, Michigan, USA
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Madrid, Spain; Department of Cardiology, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Stavros G Drakos
- Department of Medicine, Division of Cardiovascular Medicine, University of Utah Hospital, Salt Lake City, Utah, USA; Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - George Dangas
- Division of Cardiology, Mount Sinai Health System, New York, New York, USA
| | - Navin K Kapur
- The CardioVascular Center and Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Jafree E, Del Buono MG, Canada JM, Carbone S, Kron J, Arena R, Van Tassell B, Abbate A, Trankle CR. Interleukin-1 Inhibition for the Prevention and Treatment of Heart Failure. J Cardiovasc Pharmacol 2024; 83:522-530. [PMID: 37815298 PMCID: PMC11004086 DOI: 10.1097/fjc.0000000000001497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
ABSTRACT Heart failure (HF) is a complex syndrome that remains a leading cause of morbidity and mortality worldwide. Abundant evidence suggests inflammation plays a key role in the development and perpetuation of HF, but there are currently no anti-inflammatory treatments approved for use in HF. Interleukin-1, the prototypical proinflammatory cytokine, has been implicated in adverse cardiac remodeling and left ventricular dysfunction. Multiple early phase clinical trials using interleukin-1 blockade in patients at risk for or diagnosed with HF have suggested favorable safety and efficacy in reducing inflammatory biomarkers, as well as positive signals in surrogate and clinical end points. Additional large scale clinical trials are urgently needed to confirm the safety and efficacy of this therapeutic approach specifically in HF. In this narrative review, we discuss current evidence regarding interleukin-1 blockade in the prevention and treatment of HF.
Collapse
Affiliation(s)
- Ehsan Jafree
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Justin M. Canada
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Salvatore Carbone
- Department of Kinesiology & Health Sciences, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA
| | - Jordana Kron
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Benjamin Van Tassell
- Department of Pharmacotherapy and Outcome Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- Division of Cardiology, University of Virginia, Charlottesville, VA
| | - Cory R. Trankle
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
28
|
Barbu E, Mihaila AC, Gan AM, Ciortan L, Macarie RD, Tucureanu MM, Filippi A, Stoenescu AI, Petrea SV, Simionescu M, Balanescu SM, Butoi E. The Elevated Inflammatory Status of Neutrophils Is Related to In-Hospital Complications in Patients with Acute Coronary Syndrome and Has Important Prognosis Value for Diabetic Patients. Int J Mol Sci 2024; 25:5107. [PMID: 38791147 PMCID: PMC11121518 DOI: 10.3390/ijms25105107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Despite neutrophil involvement in inflammation and tissue repair, little is understood about their inflammatory status in acute coronary syndrome (ACS) patients with poor outcomes. Hence, we investigated the potential correlation between neutrophil inflammatory markers and the prognosis of ACS patients with/without diabetes and explored whether neutrophils demonstrate a unique inflammatory phenotype in patients experiencing an adverse in-hospital outcome. The study enrolled 229 ACS patients with or without diabetes. Poor evolution was defined as either death, left ventricular ejection fraction (LVEF) <40%, Killip Class 3/4, ventricular arrhythmias, or mechanical complications. Univariate and multivariate analyses were employed to identify clinical and paraclinical factors associated with in-hospital outcomes. Neutrophils isolated from fresh blood were investigated using qPCR, Western blot, enzymatic assay, and immunofluorescence. Poor evolution post-myocardial infarction (MI) was associated with increased number, activity, and inflammatory status of neutrophils, as indicated by significant increase of Erythrocyte Sedimentation Rate (ESR), C-reactive protein (CRP), fibrinogen, interleukin-1β (IL-1β), and, interleukin-6 (IL-6). Among the patients with complicated evolution, neutrophil activity had an important prognosis value for diabetics. Neutrophils from patients with unfavorable evolution revealed a pro-inflammatory phenotype with increased expression of CCL3, IL-1β, interleukin-18 (IL-18), S100A9, intracellular cell adhesion molecule-1 (ICAM-1), matrix metalloprotease (MMP-9), of molecules essential in reactive oxygen species (ROS) production p22phox and Nox2, and increased capacity to form neutrophil extracellular traps. Inflammation is associated with adverse short-term prognosis in acute ACS, and inflammatory biomarkers exhibit greater specificity in predicting short-term outcomes in diabetics. Moreover, neutrophils from patients with unfavorable evolution exhibit distinct inflammatory patterns, suggesting that alterations in the innate immune response in this subgroup may exert detrimental effects on disease progression.
Collapse
Affiliation(s)
- Elena Barbu
- Department of Cardiology, Elias Emergency Hospital, 011461, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (S.M.B.)
| | - Andreea Cristina Mihaila
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Ana-Maria Gan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Letitia Ciortan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Razvan Daniel Macarie
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Monica Madalina Tucureanu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Alexandru Filippi
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Andra Ioana Stoenescu
- Department of Cardiology, Elias Emergency Hospital, 011461, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (S.M.B.)
| | | | - Maya Simionescu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| | - Serban Mihai Balanescu
- Department of Cardiology, Elias Emergency Hospital, 011461, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (S.M.B.)
| | - Elena Butoi
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (A.-M.G.); (L.C.); (R.D.M.); (M.M.T.); (M.S.)
| |
Collapse
|
29
|
Sagris M, Apostolos A, Theofilis P, Ktenopoulos N, Katsaros O, Tsalamandris S, Tsioufis K, Toutouzas K, Tousoulis D. Myocardial Ischemia-Reperfusion Injury: Unraveling Pathophysiology, Clinical Manifestations, and Emerging Prevention Strategies. Biomedicines 2024; 12:802. [PMID: 38672157 PMCID: PMC11048318 DOI: 10.3390/biomedicines12040802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/02/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) remains a challenge in the context of reperfusion procedures for myocardial infarction (MI). While early revascularization stands as the gold standard for mitigating myocardial injury, recent insights have illuminated the paradoxical role of reperfusion, giving rise to the phenomenon known as ischemia-reperfusion injury. This comprehensive review delves into the intricate pathophysiological pathways involved in MIRI, placing a particular focus on the pivotal role of endothelium. Beyond elucidating the molecular intricacies, we explore the diverse clinical manifestations associated with MIRI, underscoring its potential to contribute substantially to the final infarct size, up to 50%. We further navigate through current preventive approaches and highlight promising emerging strategies designed to counteract the devastating effects of the phenomenon. By synthesizing current knowledge and offering a perspective on evolving preventive interventions, this review serves as a valuable resource for clinicians and researchers engaged in the dynamic field of MIRI.
Collapse
Affiliation(s)
- Marios Sagris
- Correspondence: ; Tel.: +30-213-2088099; Fax: +30-2132088676
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Toldo S, Abbate A. The role of the NLRP3 inflammasome and pyroptosis in cardiovascular diseases. Nat Rev Cardiol 2024; 21:219-237. [PMID: 37923829 PMCID: PMC11550901 DOI: 10.1038/s41569-023-00946-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2023] [Indexed: 11/06/2023]
Abstract
An intense, stereotyped inflammatory response occurs in response to ischaemic and non-ischaemic injury to the myocardium. The NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is a finely regulated macromolecular protein complex that senses the injury and triggers and amplifies the inflammatory response by activation of caspase 1; cleavage of pro-inflammatory cytokines, such as pro-IL-1β and pro-IL-18, to their mature forms; and induction of inflammatory cell death (pyroptosis). Inhibitors of the NLRP3 inflammasome and blockers of IL-1β and IL-18 activity have been shown to reduce injury to the myocardium and pericardium, favour resolution of the inflammation and preserve cardiac function. In this Review, we discuss the components of the NLRP3 inflammasome and how it is formed and activated in various ischaemic and non-ischaemic cardiac pathologies (acute myocardial infarction, cardiac dysfunction and remodelling, atherothrombosis, myocarditis and pericarditis, cardiotoxicity and cardiac sarcoidosis). We also summarize current preclinical and clinical evidence from studies of agents that target the NLRP3 inflammasome and related cytokines.
Collapse
Affiliation(s)
- Stefano Toldo
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
31
|
Dankar R, Wehbi J, Refaat MM. Tailoring Treatment in Cardiovascular Diseases: The Role of Targeted Therapies. Pharmaceutics 2024; 16:461. [PMID: 38675122 PMCID: PMC11054164 DOI: 10.3390/pharmaceutics16040461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/19/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality around the globe. To address this public health burden, innovative therapeutic agents are being developed to specifically target molecular and genetic markers. Various therapeutic modalities have been implemented, including vaccines, monoclonal or bispecific antibodies, and gene-based therapies. Such drugs precisely target the underlying disease pathophysiology, aiming at notable molecules such as lipid metabolism regulators, proinflammatory cytokines, and growth factors. This review focuses on the latest advancements in different targeted therapies. It provides an insightful overview of the current landscape of targeted cardiovascular therapies, highlighting promising strategies with potential to transform the treatment of CVDs into an era of precision medicine.
Collapse
Affiliation(s)
- Razan Dankar
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon; (R.D.); (J.W.)
- Department of Internal Medicine, Division of Cardiology, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon
| | - Jad Wehbi
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon; (R.D.); (J.W.)
- Department of Internal Medicine, Division of Cardiology, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan M. Refaat
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon; (R.D.); (J.W.)
- Department of Internal Medicine, Division of Cardiology, American University of Beirut Faculty of Medicine and Medical Center, Beirut P.O. Box 11-0236, Lebanon
| |
Collapse
|
32
|
Sattler S. Finding the right balance on the challenging path to clinical translation of anti-inflammatory therapies for ischemic heart disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:245-247. [PMID: 39196119 DOI: 10.1038/s44161-024-00440-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London, UK.
- Department of Pharmacology, Otto-Loewi Research Center, Medical University of Graz, Graz, Austria.
- Department of Cardiology, LKH Univ. Klinikum Graz, Medical University of Graz, Graz, Austria.
| |
Collapse
|
33
|
Moroni F, Corna G, Del Buono MG, Golino M, Talasaz AH, Decotto S, Markley R, Trankle C, Biondi-Zoccai G, Carbone S, Agatiello CR, Van Tassell B, Abbate A. Impact of C-reactive protein levels and role of anakinra in patients with ST-elevation myocardial infarction. Int J Cardiol 2024; 398:131610. [PMID: 38016623 PMCID: PMC10896664 DOI: 10.1016/j.ijcard.2023.131610] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/30/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Interleukin-1 blockade with anakinra reduces C-reactive protein (CRP) levels and prevents heart failure (HF) events after ST-segment myocardial infarction (STEMI). The effectiveness of anakinra according to the degree of systemic inflammation in STEMI has not been addressed. METHODS We analyzed 139 patients from three Virginia Commonwealth University Anakinra Response Trial randomized clinical trials to assess whether CRP levels predicted HF hospitalization or death in patients with STEMI, and if CRP levels influenced the effects of treatment with anakinra. RESULTS CRP cut-off levels for prediction of the composite of death or HF hospitalization for CRP at admission, 3 and 14 days were, respectively 6.45 mg/L (100% of sensitivity and 66.1% specificity), 26 mg/L (100% of sensitivity and 78% specificity) and 9.56 mg/L (100% of sensitivity and 80% specificity). More patients with elevated CRP levels died or had a HF hospitalization (5/47 [11%] vs 0/82 [0%], p = 0.004 for CRP at admission; 5/32 [15.6%] vs 0/92 [0%], p < 0.001 for day 3 and 5/26 [19%] vs 0/89 [0%], p < 0.001 for day 14). A greater number of patients treated with anakinra had low CRP levels at 3 and 14 days compared to placebo (Odds Ratio 0.11 [95% IC 0.04-0.28], p < 0.0001 and OR 0.35 [95% CI 0.14-0.86], p = 0.02, respectively). Anakinra significantly prevented death or HF hospitalization in patients with high inflammatory burden (p = 0.04 for admission, p = 0.24 for day 3, and p = 0.05 for day 14). CONCLUSION Patients with elevated CRP had higher incidence of HF hospitalization or death. Anakinra reduced the number of patients with elevated CRP levels and prevented death or HF hospitalization in patients with elevated CRP levels.
Collapse
Affiliation(s)
- Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, and Division of Cardiology, University of Virginia, Charlottesville, VA, United States; Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Cardiovascular Division, Medicine Department, Università Milano-Bicocca, Milan, Italy
| | - Giuliana Corna
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Marco Giuseppe Del Buono
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Michele Golino
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Azita H Talasaz
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Santiago Decotto
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States; Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Roshanak Markley
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Cory Trankle
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Roma, Italy; Mediterranea Cardiocentro, Via Orazio, 2, 80122 Napoli, NA, Italy
| | - Salvatore Carbone
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Carla R Agatiello
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Benjamin Van Tassell
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, and Division of Cardiology, University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
34
|
Jung JM, Gruber A, Heseltine P, Rajamani K, Ameriso SF, Fisher MJ. New Directions in Infection-Associated Ischemic Stroke. J Clin Neurol 2024; 20:140-152. [PMID: 38330416 PMCID: PMC10921058 DOI: 10.3988/jcn.2023.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/06/2023] [Accepted: 11/12/2023] [Indexed: 02/10/2024] Open
Abstract
The relationship between infections and stroke has not been fully characterized, probably delaying the development of specific treatments. This narrative review addresses mechanisms of stroke linked to infections, including hypercoagulability, endothelial dysfunction, vasculitis, and impaired thrombolysis. SARS-CoV-2, the virus that causes COVID-19, may promote the development of stroke, which may represent its most severe neurological complication. The development of specific therapies for infection-associated stroke remains a profound challenge. Perhaps the most important remaining issue is the distinction between infections that trigger a stroke versus infections that are truly incidental. This distinction likely requires the establishment of appropriate biomarkers, candidates of which are elevated levels of fibrin D-dimer and anticardiolipin/antiphospholipid antibodies. These candidate biomarkers might have potential use in identifying pathogenic infections preceding stroke, which is a precursor to establishing specific therapies for this syndrome.
Collapse
Affiliation(s)
- Jin-Man Jung
- Department of Neurology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea; Korea University Zebrafish, Translational Medical Research Center, Ansan, Korea
| | | | - Peter Heseltine
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kumar Rajamani
- Department of Neurology, Wayne State University-Detroit Medical Center, Detroit, MI, USA
| | - Sebastián F Ameriso
- Division of Vascular Neurology, Department of Neurology, Fleni, Autonomous City of Buenos Aires, Argentina
| | - Mark J Fisher
- Department of Neurology, University of California Irvine Medical Center, Orange, CA, USA.
| |
Collapse
|
35
|
Bolognese L, Reccia MR, Sabini A. Italian contributions to the history of acute myocardial infarction treatment. Minerva Cardiol Angiol 2024; 72:32-40. [PMID: 37310157 DOI: 10.23736/s2724-5683.23.06335-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The reduction in mortality from cardiovascular disease has been one of the crowning achievements of medicine over the past century. The evolution in management of acute myocardial infarction (AMI) has played a key role. Yet, the epidemiology of patients with STEMI continues to evolve. The Global Registry of Acute Coronary Events (GRACE) documented that STEMI accounted for ~36% of ACS cases. According to an analysis of a large USA database, the age-adjusted and sex-adjusted incidence of hospitalizations for STEMI significantly decreased from 133 per 100,000 person-years in 1999 to 50 per 100,000 person-years in 2008. Despite advances in both the early management and longer-term treatment of AMI, this condition still represents a leading cause of morbidity and mortality in western countries, making essential understanding its determinants. Early mortality gains noted in all AMI patients may not be sustained over the longer term and reciprocal trends of decreasing mortality after AMI accompanied by an increasing incidence of heart failure have been demonstrated in more recent years. Greater salvage of high-risk MI patients in recent periods may contribute to these trends. Over the past century, insights into the pathophysiology of AMI revolutionized approaches to management through different historical phases. This review provides a historic perspective on the underlying discoveries and pivotal trials that have been the foundation of the key changes of pharmacological and interventional treatment of AMI leading to the dramatic improvement of prognosis during the last tre decades, with special emphasis to the Italian contributions to the field.
Collapse
Affiliation(s)
- Leonardo Bolognese
- Department of Cardiovascular Surgery, San Donato Hospital, A.O. Toscana Sud-Est, Arezzo, Italy -
| | - Matteo R Reccia
- Department of Cardiovascular Surgery, San Donato Hospital, A.O. Toscana Sud-Est, Arezzo, Italy
| | - Alessandra Sabini
- Department of Cardiovascular Surgery, San Donato Hospital, A.O. Toscana Sud-Est, Arezzo, Italy
| |
Collapse
|
36
|
Del Buono MG, Bonaventura A, Vecchié A, Moroni F, Golino M, Bressi E, De Ponti R, Dentali F, Montone RA, Kron J, Lazzerini PE, Crea F, Abbate A. Pathogenic pathways and therapeutic targets of inflammation in heart diseases: A focus on Interleukin-1. Eur J Clin Invest 2024; 54:e14110. [PMID: 37837616 DOI: 10.1111/eci.14110] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND An exuberant and dysregulated inflammatory response contributes to the development and progression of cardiovascular diseases (CVDs). METHODS This narrative review includes original articles and reviews published over the past 20 years and found through PubMed. The following search terms (or combination of terms) were considered: "acute pericarditis," "recurrent pericarditis," "myocarditis," "cardiac sarcoidosis," "atherosclerosis," "acute myocardial infarction," "inflammation," "NLRP3 inflammasome," "Interleukin-1" and "treatment." RESULTS Recent evidence supports the role of inflammation across a wide spectrum of CVDs including myocarditis, pericarditis, inflammatory cardiomyopathies (i.e. cardiac sarcoidosis) as well as atherosclerotic CVD and heart failure. Interleukins (ILs) are the signalling mediators of the inflammatory response. The NACHT, leucine-rich repeat and pyrin-domain containing protein 3 (NLRP3) inflammasome play a key role in producing IL-1β, the prototypical pro-inflammatory cytokine involved in CVDs. Other pro-inflammatory cytokines (e.g. tumour necrosis factor) have been implicated in cardiac sarcoidosis. As a proof of this, IL-1 blockade has been proven efficacious in pericarditis and chronic coronary syndrome. CONCLUSION Tailored strategies aiming at quenching the inflammatory response have emerged as promising to treat CVDs. In this review article, we summarize recent evidence regarding the role of inflammation across a broad spectrum of CVDs. We also review novel evidence regarding targeted therapeutic strategies.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Aldo Bonaventura
- Department of Internal Medicine, Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Alessandra Vecchié
- Department of Internal Medicine, Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Michele Golino
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Edoardo Bressi
- Department of Cardiology, Policlinico Casilino, Rome, Italy
| | - Roberto De Ponti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Francesco Dentali
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Rocco Antonio Montone
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Jordana Kron
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
37
|
Corna G, Golino M, Talasaz AH, Moroni F, Del Buono MG, Damonte JI, Chiabrando JG, Mbualungu J, Trankle CR, Thomas GK, Markley R, Canada JM, Turlington J, Agatiello CR, VAN Tassell B, Abbate A. Response to interleukin-1 blockade with anakinra in women and men with ST-segment elevation myocardial infarction. Minerva Cardiol Angiol 2024; 72:67-75. [PMID: 37987681 DOI: 10.23736/s2724-5683.23.06439-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
BACKGROUND Interleukin-1 blockade with anakinra reduces high-sensitivity C-reactive protein (hsCRP) levels and prevents heart failure (HF) events after ST-segment myocardial infarction (STEMI). Sex-based differences in STEMI patients have been reported, but no data are available regarding response to anakinra. METHODS We analyzed the systemic inflammation and composite end-point of new-onset HF or death in women and men with STEMI treated with anakinra from three different Virginia Commonwealth University Anakinra Response Trial (VCUART) randomized clinical trials. RESULTS We analyzed 139 patients, 29 (21%) were women while 110 (79%) were men. Baseline hsCRP was higher in women compared to men (8.9 [5.2-13.5] vs. 4.2 [2.1-7.7] mg/L, P<0.001). Eighty-four patients were treated with anakinra (22 [75%] women and 62 [56%] men). The area under the curve of hsCRP (hsCRP-AUC) after 14 days was numerically lower in patients receiving anakinra versus placebo both in men (86 [37-130] vs. 223 [119-374] mg day/L) and in women (73 [46-313] vs. 242 [102-988] mg day/L) (P<0.001 for multiple groups, P for interaction 0.22). The incidence of the composite endpoint was also numerically lower in the anakinra group compared to placebo, both in men (4 [6.4%] vs. 14 [29.1%]) and in women (3 [13.6%] vs. 2 [28.5%]) (P=0.019 for multiple groups, P for interaction 0.44). There were no statistically significant differences between women and men in hsCRP-AUC and death or HF events when comparing separately the anakinra and placebo groups (all P>0.05). CONCLUSIONS Women were underrepresented in the VCUART trials, they appeared to have higher hsCRP levels at time of presentation, yet to benefit similar to men by treatment with anakinra in STEMI.
Collapse
Affiliation(s)
- Giuliana Corna
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Michele Golino
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Azita H Talasaz
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Moroni
- Department of Internal Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Medicine, University of Milano-Bicocca, Milan, Italy
| | - Marco G Del Buono
- Department of Cardiovascular Medicine, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Juan I Damonte
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Juan G Chiabrando
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - James Mbualungu
- Division of Cardiology, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Cory R Trankle
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Georgia K Thomas
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Roshanak Markley
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Justin M Canada
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Jeremy Turlington
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Carla R Agatiello
- Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Benjamin VAN Tassell
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Antonio Abbate
- Division of Cardiology, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA -
| |
Collapse
|
38
|
Nelson AR, Christiansen SL, Naegle KM, Saucerman JJ. Logic-based mechanistic machine learning on high-content images reveals how drugs differentially regulate cardiac fibroblasts. Proc Natl Acad Sci U S A 2024; 121:e2303513121. [PMID: 38266046 PMCID: PMC10835125 DOI: 10.1073/pnas.2303513121] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024] Open
Abstract
Fibroblasts are essential regulators of extracellular matrix deposition following cardiac injury. These cells exhibit highly plastic responses in phenotype during fibrosis in response to environmental stimuli. Here, we test whether and how candidate anti-fibrotic drugs differentially regulate measures of cardiac fibroblast phenotype, which may help identify treatments for cardiac fibrosis. We conducted a high-content microscopy screen of human cardiac fibroblasts treated with 13 clinically relevant drugs in the context of TGFβ and/or IL-1β, measuring phenotype across 137 single-cell features. We used the phenotypic data from our high-content imaging to train a logic-based mechanistic machine learning model (LogiMML) for fibroblast signaling. The model predicted how pirfenidone and Src inhibitor WH-4-023 reduce actin filament assembly and actin-myosin stress fiber formation, respectively. Validating the LogiMML model prediction that PI3K partially mediates the effects of Src inhibition, we found that PI3K inhibition reduces actin-myosin stress fiber formation and procollagen I production in human cardiac fibroblasts. In this study, we establish a modeling approach combining the strengths of logic-based network models and regularized regression models. We apply this approach to predict mechanisms that mediate the differential effects of drugs on fibroblasts, revealing Src inhibition acting via PI3K as a potential therapy for cardiac fibrosis.
Collapse
Affiliation(s)
- Anders R. Nelson
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
| | - Steven L. Christiansen
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
- Department of Biochemistry, Brigham Young University, Provo, UT84602
| | - Kristen M. Naegle
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
| |
Collapse
|
39
|
Ter Mors B, Spieler V, Merino Asumendi E, Gantert B, Lühmann T, Meinel L. Bioresponsive Cytokine Delivery Responding to Matrix Metalloproteinases. ACS Biomater Sci Eng 2024; 10:29-37. [PMID: 37102329 DOI: 10.1021/acsbiomaterials.2c01320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Cytokines are regulated in acute and chronic inflammation, including rheumatoid arthritis (RA) and myocardial infarction (MI). However, the dynamic windows within which cytokine activity/inhibition is desirable in RA and MI change timely and locally during the disease. Therefore, traditional, static delivery regimens are unlikely to meet the idiosyncrasy of these highly dynamic pathophysiological and individual processes. Responsive delivery systems and biomaterials, sensing surrogate markers of inflammation (i.e., matrix metalloproteinases - MMPs) and answering with drug release, may present drug activity at the right time, manner, and place. This article discusses MMPs as surrogate markers for disease activity in RA and MI to clock drug discharge to MMP concentration profiles from MMP-responsive drug delivery systems and biomaterials.
Collapse
Affiliation(s)
- Björn Ter Mors
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Valerie Spieler
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Eduardo Merino Asumendi
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Benedikt Gantert
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| |
Collapse
|
40
|
Matter MA, Paneni F, Libby P, Frantz S, Stähli BE, Templin C, Mengozzi A, Wang YJ, Kündig TM, Räber L, Ruschitzka F, Matter CM. Inflammation in acute myocardial infarction: the good, the bad and the ugly. Eur Heart J 2024; 45:89-103. [PMID: 37587550 PMCID: PMC10771378 DOI: 10.1093/eurheartj/ehad486] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023] Open
Abstract
Convergent experimental and clinical evidence have established the pathophysiological importance of pro-inflammatory pathways in coronary artery disease. Notably, the interest in treating inflammation in patients suffering acute myocardial infarction (AMI) is now expanding from its chronic aspects to the acute setting. Few large outcome trials have proven the benefits of anti-inflammatory therapies on cardiovascular outcomes by targeting the residual inflammatory risk (RIR), i.e. the smouldering ember of low-grade inflammation persisting in the late phase after AMI. However, these studies have also taught us about potential risks of anti-inflammatory therapy after AMI, particularly related to impaired host defence. Recently, numerous smaller-scale trials have addressed the concept of targeting a deleterious flare of excessive inflammation in the early phase after AMI. Targeting different pathways and implementing various treatment regimens, those trials have met with varied degrees of success. Promising results have come from those studies intervening early on the interleukin-1 and -6 pathways. Taking lessons from such past research may inform an optimized approach to target post-AMI inflammation, tailored to spare 'The Good' (repair and defence) while treating 'The Bad' (smouldering RIR) and capturing 'The Ugly' (flaming early burst of excess inflammation in the acute phase). Key constituents of such a strategy may read as follows: select patients with large pro-inflammatory burden (i.e. large AMI); initiate treatment early (e.g. ≤12 h post-AMI); implement a precisely targeted anti-inflammatory agent; follow through with a tapering treatment regimen. This approach warrants testing in rigorous clinical trials.
Collapse
Affiliation(s)
- Michael A Matter
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Francesco Paneni
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Oberdürrbacher Strasse 6, 97080 Würzburg, Germany
| | - Barbara E Stähli
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Christian Templin
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy
| | - Yu-Jen Wang
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Lorenz Räber
- Department of Cardiology, Bern University Hospital, Inselspital, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Christian M Matter
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
41
|
Cheng T, You Y, Jia B, Wang H, Lv M, Zhu X, Hu Y. Knowledge mapping of B cell and atherosclerosis over the past 20 years: A bibliometric analysis. Hum Vaccin Immunother 2023; 19:2277567. [PMID: 37953301 PMCID: PMC10760366 DOI: 10.1080/21645515.2023.2277567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023] Open
Abstract
Atherosclerosis (AS) is the main underlying cause of cardiovascular disease, and B cells are considered a key immune cell type to regulate AS. So far, there is no bibliometric study on B cell and AS. This study aims to comprehensively analyze the scientific output about B cell and AS, summarize the literature characteristics, explore research hotspots, and point out emerging trends. We searched the literature from 2003 to 2022 from the Web of Science Core Collection (WoSCC) database. CiteSpace, VOSviewer, and the R package "Bibliometrix" were used for literature analysis and visualization. A total of 1,062 articles and reviews were identified. The number of annual publications generally showed an upward trend. The United States and China were the most productive countries. Medical University of Vienna was the most productive research institution, and Binder Christoph J. was the most productive author, who was also from Medical University of Vienna. "Arteriosclerosis Thrombosis and Vascular Biology" was the most published journal and the most frequently cited journal. The most cited reference was written by Caligiuri G (2002) in "Journal of Clinical Investigation." The most frequent keywords were "inflammation," "macrophages," "cardiovascular disease," "T cells," "apoptosis," "immunity," "cytokines," "lymphocytes," etc. The trend topics were mainly focused on "immune infiltration," "immunoglobulins," and "biomarkers." The complex role of B cell subtypes and a variety of B cell mediators is the main research direction at present. In-depth analysis of B cell-specific targets can provide new ideas and methods for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yaping You
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Bochao Jia
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Huan Wang
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Meng Lv
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Xueping Zhu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
42
|
Corna G, Golino M, Moroni F, Del Buono MG, Talasaz AH, Decotto S, Mbualungu J, Trankle CR, Thomas GK, Markley R, Canada JM, Turlington J, Agatiello CR, Keen L, Van Tassell B, Abbate A. Response to Interleukin-1 Blockade With Anakinra in Black and White Americans With ST-Segment Elevation Myocardial Infarction. Am J Cardiol 2023; 207:336-338. [PMID: 37774475 PMCID: PMC10840676 DOI: 10.1016/j.amjcard.2023.08.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 08/27/2023] [Indexed: 10/01/2023]
Affiliation(s)
- Giuliana Corna
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia; Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Michele Golino
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia; Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Francesco Moroni
- Department of Internal Medicine, University of Virginia, Charlottesville, Virginia; Department of Medicine, University of Milano-Bicocca, Milan, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Azita H Talasaz
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Santiago Decotto
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia; Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - James Mbualungu
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Cory R Trankle
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Georgia K Thomas
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Roshanak Markley
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Justin M Canada
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jeremy Turlington
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Carla R Agatiello
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Larry Keen
- Department of Psychology, Virginia State University, Petersburg, Virginia
| | - Benjamin Van Tassell
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, Virginia.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia School of Medicine, Charlottesville, Virginia.
| |
Collapse
|
43
|
He F, Xie T, Ni D, Tang T, Cheng X. Efficacy and safety of inhibiting the NLRP3/IL-1β/IL-6 pathway in patients with ST-elevation myocardial infarction: A meta-analysis. Eur J Clin Invest 2023; 53:e14062. [PMID: 37427709 DOI: 10.1111/eci.14062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND The NLRP3/IL-1β/IL-6 pathway plays a key role in mediating inflammatory responses after ST-elevation myocardial infarction (STEMI). However, the clinical benefits of inhibiting this pathway in STEMI are uncertain. We aimed to evaluate the efficacy and safety of inhibiting the NLRP3/IL-1β/IL-6 pathway in STEMI patients. METHODS This study followed PRISMA guidelines. PubMed, Embase, CENTRAL and ClinicalTrials.gov databases were searched for randomized controlled trials (RCTs) of inhibiting the NLRP3/IL-1β/IL-6 pathway in STEMI patients within 7 days of symptom onset. The efficacy outcomes included all-cause death, cardiovascular death, recurrent MI, new-onset or worsening heart failure (HF) and stroke. The safety outcomes were serious infection, gastrointestinal adverse events and injection site reactions. RESULTS Of 316 screened records, nine trials with 1211 patients were included in the meta-analysis. Colchicine reduced the risk of recurrent MI (RR 0.28, 95% CI 0.10-0.74; I2 = 0.0%). Anakinra was associated with reduced risk of new-onset or worsening HF (RR 0.32, 95% CI 0.13-0.77; I2 = 0.0%) and decreased C-reactive protein levels (SMD -1.34, 95% CI -2.04 to -0.65; I2 = 0.0%). Colchicine and anakinra increased the risk of gastrointestinal adverse events (RR 4.43, 95% CI 2.75-7.13; I2 = 38.1%) and injection site reactions (RR 4.52, 95% CI 1.32-15.49; I2 = 0.8%), respectively. None of the three medications affected the risks of all-cause death, cardiovascular death, stroke and serious infection. CONCLUSIONS There is still no large-scale RCT evidence on the efficacy and safety of inhibiting the NLRP3/IL-1β/IL-6 pathway for the treatment of STEMI. Preliminary results from the available RCTs suggest colchicine and anakinra may respectively reduce the risks of recurrent MI and new-onset or worsening HF. The available RCTs in this meta-analysis lack power to determine any differences on mortality.
Collapse
Affiliation(s)
- Fang He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Ni
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Francisco J, Del Re DP. Inflammation in Myocardial Ischemia/Reperfusion Injury: Underlying Mechanisms and Therapeutic Potential. Antioxidants (Basel) 2023; 12:1944. [PMID: 38001797 PMCID: PMC10669026 DOI: 10.3390/antiox12111944] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Acute myocardial infarction (MI) occurs when blood flow to the myocardium is restricted, leading to cardiac damage and massive loss of viable cardiomyocytes. Timely restoration of coronary flow is considered the gold standard treatment for MI patients and limits infarct size; however, this intervention, known as reperfusion, initiates a complex pathological process that somewhat paradoxically also contributes to cardiac injury. Despite being a sterile environment, ischemia/reperfusion (I/R) injury triggers inflammation, which contributes to infarct expansion and subsequent cardiac remodeling and wound healing. The immune response is comprised of subsets of both myeloid and lymphoid-derived cells that act in concert to modulate the pathogenesis and resolution of I/R injury. Multiple mechanisms, including altered metabolic status, regulate immune cell activation and function in the setting of acute MI, yet our understanding remains incomplete. While numerous studies demonstrated cardiac benefit following strategies that target inflammation in preclinical models, therapeutic attempts to mitigate I/R injury in patients were less successful. Therefore, further investigation leveraging emerging technologies is needed to better characterize this intricate inflammatory response and elucidate its influence on cardiac injury and the progression to heart failure.
Collapse
Affiliation(s)
| | - Dominic P. Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
45
|
Nelson AR, Christiansen SL, Naegle KM, Saucerman JJ. Logic-based mechanistic machine learning on high-content images reveals how drugs differentially regulate cardiac fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530599. [PMID: 36909540 PMCID: PMC10002757 DOI: 10.1101/2023.03.01.530599] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Fibroblasts are essential regulators of extracellular matrix deposition following cardiac injury. These cells exhibit highly plastic responses in phenotype during fibrosis in response to environmental stimuli. Here, we test whether and how candidate anti-fibrotic drugs differentially regulate measures of cardiac fibroblast phenotype, which may help identify treatments for cardiac fibrosis. We conducted a high content microscopy screen of human cardiac fibroblasts treated with 13 clinically relevant drugs in the context of TGFβ and/or IL-1β, measuring phenotype across 137 single-cell features. We used the phenotypic data from our high content imaging to train a logic-based mechanistic machine learning model (LogiMML) for fibroblast signaling. The model predicted how pirfenidone and Src inhibitor WH-4-023 reduce actin filament assembly and actin-myosin stress fiber formation, respectively. Validating the LogiMML model prediction that PI3K partially mediates the effects of Src inhibition, we found that PI3K inhibition reduces actin-myosin stress fiber formation and procollagen I production in human cardiac fibroblasts. In this study, we establish a modeling approach combining the strengths of logic-based network models and regularized regression models, apply this approach to predict mechanisms that mediate the differential effects of drugs on fibroblasts, revealing Src inhibition acting via PI3K as a potential therapy for cardiac fibrosis.
Collapse
Affiliation(s)
- Anders R. Nelson
- University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Steven L. Christiansen
- University of Virginia School of Medicine, Charlottesville, VA 22903
- Brigham Young University Department of Biochemistry, Provo, UT 84602
| | - Kristen M. Naegle
- University of Virginia School of Medicine, Charlottesville, VA 22903
| | | |
Collapse
|
46
|
Talasaz AH, Sculthorpe R, Pak M, Lipinski M, Roberts C, Markley R, Trankle CR, Canada JM, Wohlford GF, Golino M, Dixon D, Van Tassell BW, Abbate A. Comparison of Safety and Biological Efficacy of Anakinra (Kineret) Dispensed in Polycarbonate Plastic versus Borosilicate Glass Syringes: A Patient-Level Analysis of VCUART2 and VCUART3 Clinical Trials. J Pharmacol Exp Ther 2023; 386:138-142. [PMID: 36868827 PMCID: PMC10353132 DOI: 10.1124/jpet.122.001404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/09/2023] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Anakinra is a recombinant human interleukin-1 receptor antagonist approved for the treatment of inflammatory diseases. Kineret is available as a solution prepared in a borosilicate glass syringe. For implementing a placebo-controlled double-blind randomized clinical trial, anakinra is commonly transferred into plastic syringes. However, there is limited data on anakinra's stability in polycarbonate syringes. We described the results of our previous studies on the use of anakinra in glass (VCUART3) versus plastic syringes (VCUART2) compared with placebo. These studies were conducted in patients with ST-segment elevation myocardial infarction (STEMI), and we assessed the anti-inflammatory effects of anakinra versus placebo by comparing the area under the curve for high-sensitivity cardiac reactive protein (AUC-CRP) levels during the first 14 days of STEMI, its clinical effects on heart failure (HF) hospitalization, cardiovascular death, or new diagnosis of HF as well as adverse events profile between groups. The levels of AUC-CRP were 75 (50-255 mg·day/l) for anakinra in plastic syringes versus 255 (116-592 mg·day/l) in placebo and 60 (24-139 mg·day/l) and 86 (43-123 mg·day/l) for anakinra once and twice daily in glass syringes, respectively, compared with placebo 214 (131-394 mg·day/l). The rate of adverse events was also comparable between groups. There were no differences in the rate of HF hospitalization or cardiovascular death in patients who received anakinra in plastic or glass syringes. Fewer cases of new-onset heart failure occurred in patients receiving anakinra in plastic or glass syringes compared with placebo. Anakinra stored in plastic (polycarbonate) syringes provides comparable biologic and clinical effect to glass (borosilicate) syringes. SIGNIFICANCE STATEMENT: Anakinra (Kineret) 100 mg administered subcutaneously in patients with ST-segment elevation myocardial infarction (STEMI) for a duration of up to 14 days appears to have comparable safety and biological efficacy signals when delivered in prefilled glass or transferred into plastic polycarbonate syringes. This may have important implications for the feasibility of designing clinical trials in STEMI and other clinical conditions.
Collapse
Affiliation(s)
- Azita H Talasaz
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Robin Sculthorpe
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Mary Pak
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Michael Lipinski
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Charlotte Roberts
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Roshanak Markley
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Cory R Trankle
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Justin M Canada
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - George F Wohlford
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Michele Golino
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Dave Dixon
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Benjamin W Van Tassell
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| | - Antonio Abbate
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy (A.H.T.), Investigation Drug Pharmacy Department (R.S., M.P., G.F.W., D.D., B.W.V.T.), and Pauley Heart Center (M.L., C.R., R.M., C.R.T., J.M.C., M.G., D.D., B.W.V.T., A.A.), Virginia Commonwealth University, Richmond, Virginia; and Berne Cardiovascular Research Center and Division of Cardiology, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia (A.A.)
| |
Collapse
|
47
|
Del Buono MG, Damonte JI, Moroni F, Chiabrando JG, Markley R, Turlington J, Trankle CR, Kang L, Biondi-Zoccai G, Kontos MC, Roberts CS, Van Tassell BW, Abbate A. Clinical and Pharmacological Implications of Time to Treatment with Interleukin-1 Blockade in ST-Segment Elevation Myocardial Infarction. J Pharmacol Exp Ther 2023; 386:156-163. [PMID: 37037651 PMCID: PMC10353076 DOI: 10.1124/jpet.123.001601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
Interleukin-1 (IL-1) blockade with anakinra given within 12 hours from reperfusion has been shown to reduce the inflammatory response as well as prevent heart failure (HF) events in patients with STEMI. We sought to determine whether time-to-treatment influences the efficacy of anakinra on systemic inflammation and incidence of HF events in patients with STEMI. We divided the cohort in two groups base6d on the median time from percutaneous coronary intervention (PCI) to investigational drug, and analyzed the effects of anakinra on the area-under-the-curve for C reactive protein (AUC-CRP) and on incidence of the composite endpoint of death or new onset HF. We analyzed data from 139 patients: 84 (60%) treated with anakinra and 55 (40%) with placebo. The median time from PCI to investigational treatment was 271 (182-391) minutes. The AUC-CRP was significantly higher in patients receiving placebo versus anakinra both in those with time from PCI to treatment <271 minutes (222.6 [103.9-325.2] vs. 78.4 [44.3-131.2], P < 0.001) and those with time from PCI to treatment ≥271 minute (235.2 [131.4-603.4] vs. 75.5 [38.9-171.9], P < 0.001) (P > 0.05 for interaction). Anakinra significantly reduced the combined endpoint of death or new onset HF in patients with time from PCI to treatment <271 minutes (5 [11%] vs. 9n[36%], log-rank χ 2 5.985, P = 0.014) as well as in patients with time from PCI to drug ≥271 minutes (2n[5%] vs. 7 [23%], log-rank χ 2 3.995, P = 0.046) (P > 0.05 for interaction). IL-1 blockade with anakinra blunts the acute systemic inflammatory response and prevents HF events independent of time-to-treatment. SIGNIFICANCE STATEMENT: In patients with ST segment elevation presenting within 12 hours of pain onset and treated within 12 hours of reperfusion, interleukin-1 blockade with anakinra blunts the acute systemic inflammatory response, a surrogate of interleukin-1 activity, and prevents heart failure events independent of time-to-treatment.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Juan Ignacio Damonte
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Francesco Moroni
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Juan Guido Chiabrando
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Roshanak Markley
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Jeremy Turlington
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Cory R Trankle
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Le Kang
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Giuseppe Biondi-Zoccai
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Michael C Kontos
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Charlotte S Roberts
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| | - Antonio Abbate
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, West Hospital, Richmond, Virginia (M.G.D.B., J.I.D., F.M., J.G.C., R.M., J.T., C.R.T., M.C.K., C.S.R., B.W.V.T., A.A.); Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.G.D.B.); Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy (M.G.D.B.); Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina (J.I.D., J.G.C.); Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia (A.A.); Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia (L.K.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (G.B.-Z.); Mediterranea Cardiocentro, Napoli, Italy (G.B.-Z.); and Department of Pharmacotherapy and Outcomes Science, MedStar Washington Hospital Center, Washington, DC (B.W.V.T.)
| |
Collapse
|
48
|
Pagliaro P, Penna C. Inhibitors of NLRP3 Inflammasome in Ischemic Heart Disease: Focus on Functional and Redox Aspects. Antioxidants (Basel) 2023; 12:1396. [PMID: 37507935 PMCID: PMC10376505 DOI: 10.3390/antiox12071396] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is caused by several mechanisms, including the production of reactive oxygen species (ROS), altered cellular osmolarity, and inflammatory response. Calcium overload, altered oxygen levels, and mitochondrial ROS are also involved in these MIRI processes, resulting in the irreversible opening of the mitochondrial permeability transition pore (mPTP). These mechanisms and processes are associated with NLRP3 inflammasome priming and activation, which can also induce cell death by pyroptosis through the up-regulation of the caspase-1 pathway and IL-18 release. In addition, endothelial dysfunction, both in the presence and absence of MIRI, is also accompanied by altered oxygen levels, decreased nitric oxide production, and ROS overproduction, resulting in the expression of adhesion molecules and leukocyte infiltration in which the NLRP3 inflammasome plays a central role, thus contributing, through endothelial dysfunction, to the alteration of coronary flow, typical of ischemic heart disease. Given the intricate interrelationship between ROS and NLRP3, ROS inhibitors can reduce NLRP3 inflammasome activation, while NLRP3 inhibitors can reduce oxidative stress and inflammation. NLRP3 inhibitors have been intensively studied as anti-inflammatory agents in basic cardiovascular sciences. In this review, we analyze the interrelation between ROS and NLRP3 in ischemic heart disease and the effects of some NLRP3 inhibitors as possible therapeutic agents in this disease condition. All compounds considered in this review need larger studies to confirm their appropriate use in clinical scenarios as anti-ischemic drugs.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
49
|
Peng Z, Chen H, Wang M. Identification of the biological processes, immune cell landscape, and hub genes shared by acute anaphylaxis and ST-segment elevation myocardial infarction. Front Pharmacol 2023; 14:1211332. [PMID: 37469874 PMCID: PMC10353022 DOI: 10.3389/fphar.2023.1211332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/22/2023] [Indexed: 07/21/2023] Open
Abstract
Background: Patients with anaphylaxis are at risk for ST-segment elevation myocardial infarction (STEMI). However, the pathological links between anaphylaxis and STEMI remain unclear. Here, we aimed to explore shared biological processes, immune effector cells, and hub genes of anaphylaxis and STEMI. Methods: Gene expression data for anaphylactic (GSE69063) and STEMI (GSE60993) patients with corresponding healthy controls were pooled from the Gene Expression Omnibus database. Differential expression analysis, enrichment analysis, and CIBERSORT were used to reveal transcriptomic signatures and immune infiltration profiles of anaphylaxis and STEMI, respectively. Based on common differentially expressed genes (DEGs), Gene Ontology analysis, cytoHubba algorithms, and correlation analyses were performed to identify biological processes, hub genes, and hub gene-related immune cells shared by anaphylaxis and STEMI. The robustness of hub genes was assessed in external anaphylactic (GSE47655) and STEMI (GSE61144) datasets. Furthermore, a murine model of anaphylaxis complicated STEMI was established to verify hub gene expressions. The logistic regression analysis was used to evaluate the diagnostic efficiency of hub genes. Results: 265 anaphylaxis-related DEGs were identified, which were associated with immune-inflammatory responses. 237 STEMI-related DEGs were screened, which were involved in innate immune response and myeloid leukocyte activation. M0 macrophages and dendritic cells were markedly higher in both anaphylactic and STEMI samples compared with healthy controls, while CD4+ naïve T cells and CD8+ T cells were significantly lower. Enrichment analysis of 33 common DEGs illustrated shared biological processes of anaphylaxis and STEMI, including cytokine-mediated signaling pathway, response to reactive oxygen species, and positive regulation of defense response. Six hub genes were identified, and their expression levels were positively correlated with M0 macrophage abundance and negatively correlated with CD4+ naïve T cell abundance. In external anaphylactic and STEMI samples, five hub genes (IL1R2, FOS, MMP9, DUSP1, CLEC4D) were confirmed to be markedly upregulated. Moreover, experimentally induced anaphylactic mice developed impaired heart function featuring STEMI and significantly increased expression of the five hub genes. DUSP1 and CLEC4D were screened as blood diagnostic biomarkers of anaphylaxis and STEMI based on the logistic regression analysis. Conclusion: Anaphylaxis and STEMI share the biological processes of inflammation and defense responses. Macrophages, dendritic cells, CD8+ T cells, and CD4+ naïve T cells constitute an immune cell population that acts in both anaphylaxis and STEMI. Hub genes (DUSP1 and CLEC4D) identified here provide candidate genes for diagnosis, prognosis, and therapeutic targeting of STEMI in anaphylactic patients.
Collapse
Affiliation(s)
- Zekun Peng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
50
|
Soni SS, D'Elia AM, Rodell CB. Control of the post-infarct immune microenvironment through biotherapeutic and biomaterial-based approaches. Drug Deliv Transl Res 2023; 13:1983-2014. [PMID: 36763330 PMCID: PMC9913034 DOI: 10.1007/s13346-023-01290-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 02/11/2023]
Abstract
Ischemic heart failure (IHF) is a leading cause of morbidity and mortality worldwide, for which heart transplantation remains the only definitive treatment. IHF manifests from myocardial infarction (MI) that initiates tissue remodeling processes, mediated by mechanical changes in the tissue (loss of contractility, softening of the myocardium) that are interdependent with cellular mechanisms (cardiomyocyte death, inflammatory response). The early remodeling phase is characterized by robust inflammation that is necessary for tissue debridement and the initiation of repair processes. While later transition toward an immunoregenerative function is desirable, functional reorientation from an inflammatory to reparatory environment is often lacking, trapping the heart in a chronically inflamed state that perpetuates cardiomyocyte death, ventricular dilatation, excess fibrosis, and progressive IHF. Therapies can redirect the immune microenvironment, including biotherapeutic and biomaterial-based approaches. In this review, we outline these existing approaches, with a particular focus on the immunomodulatory effects of therapeutics (small molecule drugs, biomolecules, and cell or cell-derived products). Cardioprotective strategies, often focusing on immunosuppression, have shown promise in pre-clinical and clinical trials. However, immunoregenerative therapies are emerging that often benefit from exacerbating early inflammation. Biomaterials can be used to enhance these therapies as a result of their intrinsic immunomodulatory properties, parallel mechanisms of action (e.g., mechanical restraint), or by enabling cell or tissue-targeted delivery. We further discuss translatability and the continued progress of technologies and procedures that contribute to the bench-to-bedside development of these critically needed treatments.
Collapse
Affiliation(s)
- Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA.
| |
Collapse
|