1
|
Faucher L, Matsushita K, Kikuchi S, Tatarcheh T, Marchandot B, Granier A, Amissi S, Trimaille A, Jesel L, Ohlmann P, Hibi K, Schini‐Kerth V, Morel O. Mortality risk stratification for Takotsubo syndrome: Evaluating CRP measurement alongside the InterTAK prognostic score. ESC Heart Fail 2025; 12:1427-1436. [PMID: 39821701 PMCID: PMC11911587 DOI: 10.1002/ehf2.15161] [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: 06/19/2024] [Revised: 09/19/2024] [Accepted: 10/24/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Initially described as a benign acute cardiomyopathy, Takotsubo syndrome has been linked to elevated mortality rates. Emerging evidence suggests that unresolved myocardial inflammation may contribute to this adverse prognosis. This study aimed to evaluate the incremental prognostic utility of C-reactive protein (CRP) in conjunction with the InterTAK prognosis score for stratifying long-term mortality in Takotsubo syndrome. METHODS A retrospective analysis was conducted from a multicentre registry encompassing 307 patients diagnosed with Takotsubo syndrome between 2008 and 2020. Patients were stratified into quartiles based on the InterTAK prognosis score. The discriminatory potential of CRP in predicting long-term mortality was assessed. The primary endpoint was defined as all-cause mortality within 1 year. RESULTS A stepwise increase of CRP at discharge that corresponds to INTERTAK quartiles was observed: 9.5 mg/L (25th percentile) in the first quartile, 15.8 mg/L (median) in the second quartile, 25.3 mg/L (75th percentile) in the third quartile and 41.2 mg/L (maximum) in the fourth quartile. Receiver operating-characteristic curves analysis revealed that CRP value at discharge was predictive of 1 year mortality (area under the curve = 0.81; 95% confidence interval = 0.68-0.90) with an optimal threshold set at 33 mg/L (sensitivity: 65%; specificity: 87%). When considering the InterTAK score, the incorporation of CRP at discharge with a cut-off of 33 mg/L exhibited a significant enhancement in the prediction of 1 year mortality in 'intermediate' risk (25% vs. 1%; P = 0.008) or 'very high' risk (40% vs. 10%; P = 0.02) patients. CONCLUSIONS In Takotsubo syndrome, the persistence of inflammatory burden at hospital discharge emerged as an independent predictor of 1 year mortality, augmenting the predictive capacity of the conventional InterTAK prognosis score.
Collapse
Affiliation(s)
- Loïc Faucher
- Université de Strasbourg, Pôle d'Activité Médico‐Chirurgicale Cardio‐Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier UniversitaireStrasbourgFrance
| | - Kensuke Matsushita
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
- Division of Cardiology, Yokohama City University Medical CenterYokohama City University Graduate School of MedicineYokohamaJapan
| | - Shinnosuke Kikuchi
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
- Division of Cardiology, Yokohama City University Medical CenterYokohama City University Graduate School of MedicineYokohamaJapan
| | - Taraneh Tatarcheh
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Benjamin Marchandot
- Université de Strasbourg, Pôle d'Activité Médico‐Chirurgicale Cardio‐Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier UniversitaireStrasbourgFrance
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Amandine Granier
- Université de Strasbourg, Pôle d'Activité Médico‐Chirurgicale Cardio‐Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier UniversitaireStrasbourgFrance
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Said Amissi
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Antonin Trimaille
- Université de Strasbourg, Pôle d'Activité Médico‐Chirurgicale Cardio‐Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier UniversitaireStrasbourgFrance
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Laurence Jesel
- Université de Strasbourg, Pôle d'Activité Médico‐Chirurgicale Cardio‐Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier UniversitaireStrasbourgFrance
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Patrick Ohlmann
- Université de Strasbourg, Pôle d'Activité Médico‐Chirurgicale Cardio‐Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier UniversitaireStrasbourgFrance
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical CenterYokohama City University Graduate School of MedicineYokohamaJapan
| | - Valérie Schini‐Kerth
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
| | - Olivier Morel
- Université de Strasbourg, Pôle d'Activité Médico‐Chirurgicale Cardio‐Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier UniversitaireStrasbourgFrance
- Translational Cardiovascular MedicineCRBS, University of StrasbourgStrasbourgFrance
- Hanoï Medical UniversityHanoiVietnam
| |
Collapse
|
2
|
Maatz H, Lindberg EL, Adami E, López-Anguita N, Perdomo-Sabogal A, Cocera Ortega L, Patone G, Reichart D, Myronova A, Schmidt S, Elsanhoury A, Klein O, Kühl U, Wyler E, Landthaler M, Yousefian S, Haas S, Kurth F, Teichmann SA, Oudit GY, Milting H, Noseda M, Seidman JG, Seidman CE, Heidecker B, Sander LE, Sawitzki B, Klingel K, Doeblin P, Kelle S, Van Linthout S, Hubner N, Tschöpe C. The cellular and molecular cardiac tissue responses in human inflammatory cardiomyopathies after SARS-CoV-2 infection and COVID-19 vaccination. NATURE CARDIOVASCULAR RESEARCH 2025; 4:330-345. [PMID: 39994453 PMCID: PMC11913730 DOI: 10.1038/s44161-025-00612-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025]
Abstract
Myocarditis, characterized by inflammatory cell infiltration, can have multiple etiologies, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or, rarely, mRNA-based coronavirus disease 2019 (COVID-19) vaccination. The underlying cellular and molecular mechanisms remain poorly understood. In this study, we performed single-nucleus RNA sequencing on left ventricular endomyocardial biopsies from patients with myocarditis unrelated to COVID-19 (Non-COVID-19), after SARS-CoV-2 infection (Post-COVID-19) and after COVID-19 vaccination (Post-Vaccination). We identified distinct cytokine expression patterns, with interferon-γ playing a key role in Post-COVID-19, and upregulated IL16 and IL18 expression serving as a hallmark of Post-Vaccination myocarditis. Although myeloid responses were similar across all groups, the Post-Vaccination group showed a higher proportion of CD4+ T cells, and the Post-COVID-19 group exhibited an expansion of cytotoxic CD8+ T and natural killer cells. Endothelial cells showed gene expression changes indicative of vascular barrier dysfunction in the Post-COVID-19 group and ongoing angiogenesis across all groups. These findings highlight shared and distinct mechanisms driving myocarditis in patients with and without a history of SARS-CoV-2 infection or vaccination.
Collapse
Affiliation(s)
- Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Eleonora Adami
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Natalia López-Anguita
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alvaro Perdomo-Sabogal
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Lucía Cocera Ortega
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniel Reichart
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital Boston, Boston, MA, USA
| | - Anna Myronova
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sabine Schmidt
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ahmed Elsanhoury
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Oliver Klein
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Uwe Kühl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institut für Biologie, Humboldt Universität zu Berlin, Berlin, Germany
| | - Schayan Yousefian
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Simon Haas
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, DKFZ and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Sarah A Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, UK
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital Boston, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive Medicine CBF, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Leif E Sander
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Translational Immunology, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Patrick Doeblin
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Kelle
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Van Linthout
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany.
| | - Carsten Tschöpe
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany.
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Kuang J, Jia Z, Chong TK, Chen J, Liu K, Wang X, Li Z, Zhang J, Kong Y, Deng L, Cadieras M, Sun Y, Sun R, Lu Q, Liu Y. Sacubitril/valsartan attenuates inflammation and myocardial fibrosis in Takotsubo-like cardiomyopathy. J Mol Cell Cardiol 2025; 200:24-39. [PMID: 39832528 DOI: 10.1016/j.yjmcc.2025.01.003] [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: 10/30/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Takotsubo syndrome (TTS) primarily manifests as a cardiomyopathy induced by physical or emotional stress, remains a poorly understood condition with no established treatments. In this study, we investigated the potential of sacubitril/valsartan (sac/val) to increase the survival of TTS patients and reduce inflammation and myocardial fibrosis in experimental models. AIM This study aimed to evaluate whether sac/val could improve survival rates in TTS patients, mitigate cardiac remodeling in vivo, and explore its anti-inflammatory and antifibrotic mechanisms in vitro. METHODS Clinical cases from the Chinese Takotsubo syndrome (ChiTTS) registry were analyzed to assess patient survival rates. In addition, we used isoprenaline (ISO)-induced TTS-like animal models, pre-treated with sac/val, to evaluate cardiac function and inflammatory response. Additionally, the effects of isoprenaline on cardiomyocytes and myocardial fibroblasts, as well as protection from rhBNP, were thoroughly studied. RESULTS In TTS patients with a left ventricular ejection fraction (LVEF) ≤ 0.45, hyperglycemia, emotional stress, and inflammation were identified as independent risk factors. Moreover, the baseline characteristics of the TTS patients, heart rate, emotional triggers, female sex (%), WBC count, IL-6 concentration, PCT, ALT, AST and TG were significantly associated with decreasing left ventricular ejection fraction. In TTS patients, sac/val reduced inflammation, evidenced by lower levels of white blood cells and interleukin 6, compared to patients who did not receive sac/val by day 30. In animal models, Sac/val improved cardiac dysfunction in ISO-induced TTS-like cardiomyopathy and decreased myocardial inflammatory responses (IL-18 and Mac-3) by inhibiting the TLR4/NF-κB pathway and fibrosis through the inhibition of the TGFβ1/Smad pathway. CONCLUSIONS This study revealed that sac/val decreased inflammatory responses, myocardial edema, and fibrosis, resulting in an increased percentage of survivors in the TTS group. Similar to findings from in vivo and in vitro experiments, sac/val exerted cardioprotective effects by reducing the inflammatory response and reversing myocardial remodeling mediated by the TLR4/NF-κB and TGFβ1/Smad pathways. In conclusion, these findings highlight the anti-inflammatory and antifibrotic effects of sac/val in individuals with TTS.
Collapse
Affiliation(s)
- Jiangying Kuang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Zhiyi Jia
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Tou Kun Chong
- Department of Cardiology, Kiang Wu Hospital, Macao Special Administrative Region of the People's Republic of China, PR China
| | - Jian Chen
- Department of Cardiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, PR China
| | - Kan Liu
- Division of Cardiology, Heart and Vascular Center, Washington University in St. Louis, Barnes-Jewish Hospital, St. Louis, MO, USA
| | - Xin Wang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Zhaohua Li
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Jing Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Yanru Kong
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Lin Deng
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, Guangdong, PR China
| | - Martin Cadieras
- Department of Internal Medicine, Cardiology, the University of California Davis, Sacramento, CA 95817, United States of America
| | - Yuanyuan Sun
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Rong Sun
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Qinghua Lu
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China
| | - Yusheng Liu
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250031, PR China.
| |
Collapse
|
4
|
Tian L, Liu B, Ren Y, Cui J, Pang Z. Proteomics of stress-induced cardiomyopathy: insights from differential expression, protein interaction networks, and functional pathway enrichment in an isoproterenol-induced TTC mouse model. PeerJ 2025; 13:e18984. [PMID: 39959819 PMCID: PMC11830371 DOI: 10.7717/peerj.18984] [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: 10/04/2024] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Backgrounds Takotsubo cardiomyopathy (TTC), also known as stress-induced cardiomyopathy, is a condition characterized by transient left ventricular dysfunction without coronary artery obstruction. Methods We utilized label-free quantitative proteomics to analyze protein expression in a murine model of TTC, induced by a high dose of isoproterenol (ISO) injection. Results We found that a single high dose of ISO injection in mice could induce stress-related cardiac dysfunction.The proteomic analysis revealed 81 differentially expressed proteins (DEPs) between the ISO and control groups-39 were upregulated, and 42 were downregulated. Key pathways enriched by Gene Ontology (GO) analysis included collagen fibril organization, cholesterol biosynthesis, and elastic fiber assembly. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment indicated significant changes in unsaturated fatty acid biosynthesis, glutathione metabolism, steroid biosynthesis, and ferroptosis. Key hub proteins identified by the protein-protein interaction (PPI) network included Ntrk2, Fdft1, Serpine1, and Cyp1a1. Gene set enrichment analysis (GSEA) showed upregulation in terpenoid backbone biosynthesis, oxidative phosphorylation, and ferroptosis, with downregulation in pathways such as systemic lupus erythematosus and Rap1 signaling. Conclusions This study employed high-throughput liquid chromatography-tandem mass spectrometry (LC-MS/MS) to identify key proteins associated with energy metabolism, oxidative stress, inflammation, and cell death in TTC. These findings provide new insights into the molecular mechanisms of stress-induced myocardial injury and may offer potential therapeutic targets for mitigating cardiovascular damage under stress conditions.
Collapse
Affiliation(s)
- Liuyang Tian
- Interventional Center of Valvular Heart Disease, Beijing Anzhen Hospital, Beijing, China
| | - Botao Liu
- China Medical University, Shenyang, China
| | - Ying Ren
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
- Department of Cardiology, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Jian Cui
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
- Department of Cardiology, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Zhihua Pang
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
- Department of Cardiology, The First Affiliated Hospital of Nankai University, Tianjin, China
| |
Collapse
|
5
|
Psarras S. The Macrophage-Fibroblast Dipole in the Context of Cardiac Repair and Fibrosis. Biomolecules 2024; 14:1403. [PMID: 39595580 PMCID: PMC11591949 DOI: 10.3390/biom14111403] [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: 09/24/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Stromal and immune cells and their interactions have gained the attention of cardiology researchers and clinicians in recent years as their contribution in cardiac repair is increasingly recognized. The repair process in the heart is a particularly critical constellation of complex molecular and cellular events and interactions that characteristically fail to ensure adequate recovery following injury, insult, or exposure to stress conditions in this regeneration-hostile organ. The tremendous consequence of this pronounced inability to maintain homeostatic states is being translated in numerous ways promoting progress into heart failure, a deadly, irreversible condition requiring organ transplantation. Fibrosis is in fact a repair response eventually promoting cardiac dysfunction and cardiac fibroblasts are the major cellular players in this process, overproducing collagens and other extracellular matrix components when activated. On the other hand, macrophages may differentially affect fibroblasts and cardiac repair depending on their status and subsets. The opposite interaction is also probable. We discuss here the multifaceted aspects and crosstalk of this cell dipole and the opportunities it may offer for beneficial manipulation approaches that will hopefully lead to progress in heart disease interventions.
Collapse
Affiliation(s)
- Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 115 27 Athens, Greece
| |
Collapse
|
6
|
Hong A, Vollett KDW, Cheng HLM. A Nitric Oxide-Sensing T1 Contrast Agent for In Vivo Molecular MR Imaging of Inflammatory Disease. ACS Sens 2024; 9:5374-5383. [PMID: 39377688 DOI: 10.1021/acssensors.4c01604] [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: 10/09/2024]
Abstract
Nitric oxide (NO) is a signaling molecule that not only appears in the very early stage of inflammatory disease but also persists in chronic conditions. Its detection in vivo can, therefore, potentially enable early disease detection and treatment monitoring. Due to its transient nature and low abundance, however, noninvasive and deep-tissue imaging of NO dynamics is challenging. In this study, we present a magnetic resonance imaging (MRI) contrast agent based on a manganese porphyrin for specific imaging of NO. This agent is activated by NO, binds to tissue protein, accumulates so long as NO is actively produced, and confers a substantial bright contrast on T1-weighted MRI. In vitro tests confirm the specificity of activation by NO over other reactive oxygen or nitrogen species, absence of inflammation induced by the contrast agent, and sensitivity to NO levels in the tens of micromolar. In vivo demonstration in a mouse model of stress-induced acute myocardial inflammation revealed an over 2.2-times increase in T1 reduction in the inflamed heart compared to a healthy heart. This new NO-activatable T1 contrast agent holds the potential to provide early diagnosis of inflammatory disease, characterize different stages of inflammation, and ultimately guide the design of novel anti-inflammation therapeutics.
Collapse
Affiliation(s)
- Anlan Hong
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3E2, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto M5G 1M1, Canada
| | - Kyle D W Vollett
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3E2, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto M5G 1M1, Canada
| | - Hai-Ling Margaret Cheng
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3E2, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto M5G 1M1, Canada
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
| |
Collapse
|
7
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Klein HG, Feng J, Lertora J, Parizi‐Torabi P, Danner RL, Solomon MA, Chen MY, Natanson C. In a Canine Model of Septic Shock, Cardiomyopathy Occurs Independent of Catecholamine Surges and Cardiac Microvascular Ischemia. J Am Heart Assoc 2024; 13:e034027. [PMID: 39101496 PMCID: PMC11964065 DOI: 10.1161/jaha.123.034027] [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: 02/27/2024] [Accepted: 06/20/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND High levels of catecholamines are cardiotoxic and associated with stress-induced cardiomyopathies. Using a septic shock model that reproduces the reversible cardiomyopathy seen over 10 days associated with human septic shock, we investigated the effects of catecholamines on microcirculatory perfusion and cardiac dysfunction. METHODS AND RESULTS Purpose-bred beagles received intrabronchial Staphylococcus aureus (n=30) or saline (n=6). The septic animals were than randomized to epinephrine (1 μg/kg per minute, n=15) or saline (n=15) infusions from 4 to 44 hours. Serial cardiac magnetic resonance imaging, catecholamine levels, and troponins were collected over 92 hours. Serial adenosine-stress perfusion cardiac magnetic resonance imaging was performed on septic animals randomized to receive saline (n=8 out of 15) or epinephrine (n=8 out of 15). High-dose sedation was given to suppress endogenous catecholamine release. Despite catecholamine levels largely remaining within the normal range throughout, by 48 hours, septic animals receiving saline versus nonseptic animals still developed significant worsening of left ventricular ejection fraction, circumferential strain, and ventricular-aortic coupling. In septic animals that received epinephrine versus saline infusions, plasma epinephrine levels increased 800-fold, but epinephrine produced no significant further worsening of left ventricular ejection fraction, circumferential strain, or ventricular-aortic coupling. Septic animals receiving saline had a significant increase in microcirculatory reserve without troponin elevations. Septic animals receiving epinephrine had decreased edema, blunted microcirculatory perfusion, and elevated troponin levels that persisted for hours after the epinephrine infusion stopped. CONCLUSIONS Cardiac dysfunction during sepsis is not primarily due to elevated endogenous or exogenous catecholamines nor due to decreased microvascular perfusion-induced ischemia. However, epinephrine itself has potentially harmful long-lasting ischemic effects during sepsis including impaired cardiac microvascular perfusion that persists after stopping the infusion.
Collapse
Affiliation(s)
- Verity J. Ford
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
| | - Willard N. Applefeld
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
- Division of CardiologyDuke University Medical CenterDurhamNCUSA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
- Emory UniversityAtlantaGAUSA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
| | - Harvey G. Klein
- Department of Transfusion Medicine, Clinical CenterNational Institutes of HealthBethesdaMDUSA
| | - Jing Feng
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
| | - Juan Lertora
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLAUSA
| | | | - Robert L. Danner
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
| | - Michael A. Solomon
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
- National HeartLung and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Marcus Y. Chen
- National HeartLung and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical CenterNational Institutes of HealthBethesdaMDUSA
- National HeartLung and Blood Institute, National Institutes of HealthBethesdaMDUSA
| |
Collapse
|
8
|
La Vecchia G, Del Buono MG, Sanna T, Capecchi PL, Lazzerini PE, Golino M, Kron J, Rodriguez-Miguelez P, Pelargonio G, Abbate A. Life-Threatening Arrhythmias in Patients With Takotsubo Syndrome: Insights Into Pathophysiology and Treatment Innovations. JACC Clin Electrophysiol 2024; 10:1943-1952. [PMID: 38842970 DOI: 10.1016/j.jacep.2024.03.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/26/2024] [Accepted: 03/31/2024] [Indexed: 08/30/2024]
Abstract
Takotsubo syndrome (TTS) is a reversible form of acute myocardial injury due to a neurocardiogenic mechanism associated with a relevant risk for life-threatening ventricular arrhythmias, occurring in up to 25% of all patients and including both ventricular arrhythmias (especially) in the context of QT prolongation and atrial tachy- or bradyarrhythmias. The pathogenetic mechanisms of TTS-related arrhythmic complications are not completely understood, and there are no randomized clinical trials addressing the pharmacologic and nonpharmacologic management in this specific setting. In this narrative review, the authors provide an overview of the pathogenesis and the therapeutic management of arrhythmic complications in patients with TTS, along with the future perspectives and the remaining knowledge gaps in this field.
Collapse
Affiliation(s)
- Giulia La Vecchia
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy; Operative Unit of Diagnostic Interventional Cardiology, Isola Tiberina-Gemelli Isola, Rome, Italy
| | - Marco Giuseppe Del Buono
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Tommaso Sanna
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Division of Internal Medicine, Electroimmunology Unit, University Hospital "Le Scotte," Siena, Italy
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Division of Internal Medicine, Electroimmunology Unit, University Hospital "Le Scotte," Siena, Italy
| | - Michele Golino
- Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA; Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, Virginia, USA
| | - Jordana Kron
- Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Paula Rodriguez-Miguelez
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, USA; Division of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Gemma Pelargonio
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy; Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
9
|
Perez-Bonilla P, LaViolette B, Bhandary B, Ullas S, Chen X, Hirenallur-Shanthappa D. Isoproterenol induced cardiac hypertrophy: A comparison of three doses and two delivery methods in C57BL/6J mice. PLoS One 2024; 19:e0307467. [PMID: 39038017 PMCID: PMC11262646 DOI: 10.1371/journal.pone.0307467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Heart Failure (HF) continues to be a complex public health issue with increasing world population prevalence. Although overall mortality has decreased for HF and hypertrophic cardiomyopathy (HCM), a precursor for HF, their prevalence continues to increase annually. Because the etiology of HF and HCM is heterogeneous, it has been difficult to identify novel therapies to combat these diseases. Isoproterenol (ISP), a non-selective β-adrenoreceptor agonist, is commonly used to induce cardiotoxicity and cause acute and chronic HCM and HF in mice. However, the variability in dose and duration of ISP treatment used in studies has made it difficult to determine the optimal combination of ISP dose and delivery method to develop a reliable ISP-induced mouse model for disease. Here we examined cardiac effects induced by ISP via subcutaneous (SQ) and SQ-minipump (SMP) infusions across 3 doses (2, 4, and 10mg/kg/day) over 2 weeks to determine whether SQ and SMP ISP delivery induced comparable disease severity in C57BL/6J mice. To assess disease, we measured body and heart weight, surface electrocardiogram (ECG), and echocardiography recordings. We found all 3 ISP doses comparably increase heart weight, but these increases are more pronounced when ISP was administered via SMP. We also found that the combination of ISP treatment and delivery method induces contrasting heart rate, RR interval, and R and S amplitudes that may place SMP treated mice at higher risk for sustained disease burden. Mice treated via SMP also had increased heart wall thickness and LV Mass, but mice treated via SQ showed greater increase in gene markers for hypertrophy and fibrosis. Overall, these data suggest that at 2 weeks, mice treated with 2, 4, or 10mg/kg/day ISP via SQ and SMP routes cause similar pathological heart phenotypes but highlight the importance of drug delivery method to induce differing disease pathways.
Collapse
Affiliation(s)
- Patricia Perez-Bonilla
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Brianna LaViolette
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Bidur Bhandary
- Rare Diseases Research Unit, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Soumya Ullas
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Xian Chen
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| | - Dinesh Hirenallur-Shanthappa
- Global Discovery, Investigative & Translational Sciences–Animal Models and Imaging, Pfizer Inc, Cambridge, Massachusetts, United States of America
| |
Collapse
|
10
|
Li X, Yang JJ, Xu D. The role of inflammation in takotsubo syndrome: A new therapeutic target? J Cell Mol Med 2024; 28:e18503. [PMID: 38896112 PMCID: PMC11186299 DOI: 10.1111/jcmm.18503] [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: 11/14/2023] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Takotsubo syndrome (TTS) is a particular form of acute heart failure that can be challenging to distinguish from acute coronary syndrome at presentation. TTS was previously considered a benign self-limiting condition, but it is now known to be associated with substantial short- and long-term morbidity and mortality. Because of the poor understanding of its underlying pathophysiology, there are few evidence-based interventions to treat TTS. The hypotheses formulated so far can be grouped into endogenous adrenergic surge, psychological stress or preexisting psychiatric illness, coronary vasospasm with microvascular dysfunction, metabolic and energetic alterations, and inflammatory mechanisms. Current evidence demonstrates that the infiltration of immune cells such as macrophages and neutrophils play a pivotal role in TTS. At baseline, resident macrophages were the dominant subset in cardiac macrophages, however, it underwent a shift from resident macrophages to monocyte-derived infiltrating macrophages in TTS. Depletion of macrophages and monocytes in mice strongly protected them from isoprenaline-induced cardiac dysfunction. It is probable that immune cells, especially macrophages, may be new targets for the treatment of TTS.
Collapse
Affiliation(s)
- Xiao Li
- Department of Internal Cardiovascular MedicineSecond Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Jingmin Jing Yang
- Department of Internal Cardiovascular MedicineSecond Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Danyan Xu
- Department of Internal Cardiovascular MedicineSecond Xiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
11
|
Wang Y, Tang X, Cui J, Wang P, Yang Q, Chen Y, Zhang T. Ginsenoside Rb1 mitigates acute catecholamine surge-induced myocardial injuries in part by suppressing STING-mediated macrophage activation. Biomed Pharmacother 2024; 175:116794. [PMID: 38776673 DOI: 10.1016/j.biopha.2024.116794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Stress cardiomyopathy (SCM) is associated with cardiovascular mortality rates similar to acute coronary syndrome. Myocardial injuries driven by inflammatory mechanisms may in part account for the dismal prognosis of SCM. Currently, no inflammation-targeted therapies are available to mitigate SCM-associated myocardial injuries. In this study, acute catecholamine surge-induced SCM was modeled by stimulating the ovariectomized (OVX) mice with isoproterenol (ISO). The effects of ginsenoside Rb1 (Rb1) on SCM-associated myocardial injuries were assessed in the OVX-ISO compound mice. RAW 264.7 macrophages stimulated with calf thymus DNA (ctDNA) or STING agonist DMXAA were adopted to further understand the anti-inflammatory mechanisms of Rb1. The results show that estrogen deprivation increases the susceptibility to ISO-induced myocardial injuries. Rb1 mitigates myocardial injuries and attenuates cardiomyocyte necrosis as well as myocardial inflammation in the OVX-ISO mice. Bioinformatics analysis suggests that cytosolic DNA-sensing pathway is closely linked with ISO-triggered inflammatory responses and cell death in the heart. In macrophages, Rb1 lowers ctDNA-stimulated production of TNF-α, IL-6, CCL2 and IFN-β. RNA-seq analyses uncover that Rb1 offsets DNA-stimulated upregulation in multiple inflammatory response pathways and cytosolic DNA-sensing pathway. Furthermore, Rb1 directly mitigates DMXAA-stimulated STING activation and inflammatory responses in macrophages. In conclusion, the work here demonstrates for the first time that Rb1 protects against SCM-associated myocardial injuries in part by counteracting acute ISO stress-triggered cardiomyocyte necrosis and myocardial inflammation. Moreover, by evidencing that Rb1 downregulates cytosolic DNA-sensing machineries in macrophages, our findings warrant further investigation of therapeutic implications of the anti-inflammatory Rb1 in the treatment of SCM.
Collapse
Affiliation(s)
- Yujue Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Xinmiao Tang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Jingang Cui
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China; Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Peiwei Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China; Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Qinbo Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China; Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China
| | - Yu Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China; Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China; Laboratory of Clinical and Molecular Pharmacology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China.
| | - Teng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China; Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, 110 Ganhe Rd, Shanghai 200437, China.
| |
Collapse
|
12
|
Bruns B, Joos M, Elsous N, Katus HA, Schultz J, Frey N, Backs J, Meder B. Insulin resistance in Takotsubo syndrome. ESC Heart Fail 2024; 11:1515-1524. [PMID: 38123355 PMCID: PMC11098631 DOI: 10.1002/ehf2.14623] [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: 03/28/2023] [Revised: 11/07/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
AIMS Takotsubo syndrome (TTS) is an acute heart failure (AHF) syndrome mimicking the symptoms of acute myocardial infarction. Impaired outcome has been shown, making risk stratification and novel therapeutic concepts a necessity. We hypothesized insulin resistance with elevated plasma glucose and potentially myocardial glucose deprivation to contribute to the pathogenesis of TTS and investigated the therapeutic benefit of insulin in vivo. METHODS AND RESULTS First, we retrospectively analysed patient data of n = 265 TTS cases (85.7% female, mean age 71.1 ± 14.1 years) with documented initial plasma glucose from the Department of Cardiology of the University Hospital Heidelberg in Germany (May 2011 to May 2021). Median split of the study population according to glucose levels (≤123 mg/dL vs. >123 mg/dL) yielded significantly elevated mean heart rate (80.75 ± 18.96 vs. 90.01 ± 22.19 b.p.m., P < 0.001), left ventricular end-diastolic pressure (LVEDP, 18.51 ± 8.35 vs. 23.09 ± 7.97 mmHg, P < 0.001), C-reactive protein (26.14 ± 43.30 vs. 46.4 ± 68.6 mg/L, P = 0.006), leukocyte count (10.12 ± 4.29 vs. 15.05 ± 9.83/nL, P < 0.001), peak high-sensitive Troponin T (hs-TnT, 515.44 ± 672.15 vs. 711.40 ± 736.37 pg/mL, P = 0.005), reduced left ventricular ejection fraction (EF, 34.92 ± 8.94 vs. 31.35 ± 8.06%, P < 0.001), and elevated intrahospital mortality (2.3% vs. 12.1%, P = 0.002) in the high-glucose group (Student's t-test, Mann-Whitney U test, or chi-squared test). Linear regression indicated a significant association of glucose with HR (P < 0.001), LVEDP (P = 0.014), hs-TnT kinetics from admission to the next day (P < 0.001), hs-TnT the day after admission (P < 0.001), as well as peak hsTnT (P < 0.001). Logistic regression revealed significant association of glucose with a composite intrahospital outcome including catecholamine use, respiratory support, and resuscitation [OR 1.010 (1.004-1.015), P = 0.001]. To further investigate the potential role of glucose in TTS pathophysiology experimentally, we utilized an in vivo murine model of epinephrine (EPI)-driven reversible AHF. For this, male mice underwent therapeutic injection of insulin (INS, 1 IU/kg) or/and glucose (GLU, 0.5 g/kg) after EPI (2.5 mg/kg), both of which markedly improved mean EF (EPI 34.3% vs. EPI + INS + GLU 43.7%, P = 0.025) and significantly blunted mean hs-TnT (EPI 14 393 pg/mL vs. EPI + INS 6864 pg/mL at 24 h, P = 0.039). Particularly, insulin additionally ameliorated myocardial pro-inflammatory gene expression, suggesting an anti-inflammatory effect of acute insulin therapy. CONCLUSIONS Elevated initial plasma glucose was associated with adverse outcome-relevant parameters in TTS and may present a surrogate parameter of heightened catecholaminergic drive. In mice, insulin- and glucose injection both improved EPI-induced AHF and myocardial damage, indicating insulin resistance rather than detrimental effects of hyperglycaemia itself as the underlying cause. Future studies will investigate the role of HbA1c as a risk stratifier and of insulin-based therapy in TTS.
Collapse
Affiliation(s)
- Bastian Bruns
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- Institute of Experimental CardiologyHeidelberg University HospitalHeidelbergGermany
- Department of General Internal Medicine and PsychosomaticsHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Maximilian Joos
- Institute of Experimental CardiologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Nesrin Elsous
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Hugo A. Katus
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Jobst‐Hendrik Schultz
- Department of General Internal Medicine and PsychosomaticsHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Norbert Frey
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Johannes Backs
- Institute of Experimental CardiologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Benjamin Meder
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| |
Collapse
|
13
|
Bajic Z, Sobot T, Amidzic L, Vojinovic N, Jovicic S, Gajic Bojic M, Djuric DM, Stojiljkovic MP, Bolevich S, Skrbic R. Liraglutide Protects Cardiomyocytes against Isoprenaline-Induced Apoptosis in Experimental Takotsubo Syndrome. Biomedicines 2024; 12:1207. [PMID: 38927414 PMCID: PMC11200478 DOI: 10.3390/biomedicines12061207] [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: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 06/28/2024] Open
Abstract
Takotsubo syndrome (TTS) is a stress-induced cardiomyopathy, characterized by an increased concentration of catecholamines, free radicals, and inflammatory cytokines, endothelial dysfunction, and increased apoptotic activity. High doses of isoprenaline are used in animal models to induce Takotsubo (TT)-like myocardial injury. The aim of the study was to investigate the antiapoptotic effects of liraglutide in experimental TTS and its role in the NF-κB pathway. Wistar rats were pretreated with liraglutide for 10 days, and on days 9 and 10, TT-like myocardial injury was induced with isoprenaline. After the sacrifice on day 11, hearts were isolated for histopathological and immunohistochemical analysis. Liraglutide reduced isoprenaline-induced cardiomyocyte apoptosis by decreasing cleaved caspase-3 (CC3), BCL-2-associated X protein (BAX), and NF-κB and increasing B-cell lymphoma/leukemia-2 (BCL-2). An increase in NF-κB in isoprenaline-treated rats was in positive correlation with proapoptotic markers (BAX and CC3) and in negative correlation with antiapoptotic marker BCL-2. Liraglutide increased BCL-2 and decreased NF-κB, BAX, and CC3, preserving the same correlations of NF-κB to apoptotic markers. It is concluded that liraglutide protects cardiomyocytes against isoprenaline-induced apoptosis in experimental TT-like myocardial injury through downregulation of the NF-κB pathway.
Collapse
Affiliation(s)
- Zorislava Bajic
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina;
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
| | - Tanja Sobot
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina;
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
| | - Ljiljana Amidzic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Biology of Cell and Human Genetics, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Natasa Vojinovic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Biology of Cell and Human Genetics, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Sanja Jovicic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Histology and Embryology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Milica Gajic Bojic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Dragan M. Djuric
- Faculty of Medicine, Institute of Medical Physiology “Richard Burian”, University of Belgrade, 11 000 Belgrade, Serbia;
| | - Milos P. Stojiljkovic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
| | - Sergey Bolevich
- Department of Pathologic Physiology, First Moscow State Medical University I.M. Sechenov, 119435 Moscow, Russia;
| | - Ranko Skrbic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina; (L.A.); (N.V.); (S.J.); (M.G.B.); (M.P.S.); (R.S.)
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78 000 Banja Luka, Bosnia and Herzegovina
- Department of Pathologic Physiology, First Moscow State Medical University I.M. Sechenov, 119435 Moscow, Russia;
| |
Collapse
|
14
|
Zulfaj E, Nejat A, Haamid A, Elmahdy A, Espinosa A, Redfors B, Omerovic E. Animal models of Takotsubo syndrome: bridging the gap to the human condition. Front Cardiovasc Med 2024; 11:1351587. [PMID: 38841261 PMCID: PMC11152046 DOI: 10.3389/fcvm.2024.1351587] [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: 12/08/2023] [Accepted: 03/18/2024] [Indexed: 06/07/2024] Open
Abstract
Modelling human diseases serves as a crucial tool to unveil underlying mechanisms and pathophysiology. Takotsubo syndrome (TS), an acute form of heart failure resembling myocardial infarction, manifests with reversible regional wall motion abnormalities (RWMA) of the ventricles. Despite its mortality and clinical similarity to myocardial infarction, TS aetiology remains elusive, with stress and catecholamines playing central roles. This review delves into current animal models of TS, aiming to assess their ability to replicate key clinical traits and identifying limitations. An in-depth evaluation of published animal models reveals a variation in the definition of TS among studies. We notice a substantial prevalence of catecholamine-induced models, particularly in rodents. While these models shed light on TS, there remains potential for refinement. Translational success in TS research hinges on models that align with human TS features and exhibit the key features, including transient RWMA. Animal models should be comprehensively evaluated regarding the various systemic changes of the applied trigger(s) for a proper interpretation. This review acts as a guide for researchers, advocating for stringent TS model standards and enhancing translational validity.
Collapse
Affiliation(s)
- Ermir Zulfaj
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - AmirAli Nejat
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - Abdulhussain Haamid
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ahmed Elmahdy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - Aaron Espinosa
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
| | - Björn Redfors
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
- Core Facilities - Experimental Biomedicine, Sahlgrenska Academy, Gothenburg, Sweden
| | - Elmir Omerovic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
- Core Facilities - Experimental Biomedicine, Sahlgrenska Academy, Gothenburg, Sweden
| |
Collapse
|
15
|
Lin HB, Hong P, Yin MY, Yao ZJ, Zhang JY, Jiang YP, Huang XX, Xu SY, Li FX, Zhang HF. Monocyte-Derived Macrophages Aggravate Cardiac Dysfunction After Ischemic Stroke in Mice. J Am Heart Assoc 2024; 13:e034731. [PMID: 38700011 PMCID: PMC11179859 DOI: 10.1161/jaha.123.034731] [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: 02/01/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.
Collapse
MESH Headings
- Animals
- Macrophages/metabolism
- Disease Models, Animal
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- Ischemic Stroke/physiopathology
- Ischemic Stroke/metabolism
- Ischemic Stroke/pathology
- Mice, Inbred C57BL
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Male
- Mice, Knockout
- Mice
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/pathology
- Sympathetic Nervous System/physiopathology
- Myocardium/pathology
- Myocardium/metabolism
- Heart Diseases/etiology
- Heart Diseases/physiopathology
- Heart Diseases/pathology
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- CX3C Chemokine Receptor 1/deficiency
Collapse
Affiliation(s)
- Hong-Bin Lin
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Pu Hong
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Meng-Yu Yin
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Zhi-Jun Yao
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Jin-Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science Guangzhou Guangdong China
| | - Yan-Pin Jiang
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Xuan-Xuan Huang
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Shi-Yuan Xu
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Feng-Xian Li
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| |
Collapse
|
16
|
Wu Y, Ni T, Zhang M, Fu S, Ren D, Feng Y, Liang H, Zhang Z, Zhao Y, He Y, Yang Y, Tian Z, Yan T, Liu J. Treatment with β-Adrenoceptor Agonist Isoproterenol Reduces Non-parenchymal Cell Responses in LPS/D-GalN-Induced Liver Injury. Inflammation 2024; 47:733-752. [PMID: 38129360 PMCID: PMC11074027 DOI: 10.1007/s10753-023-01941-z] [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: 09/10/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
There is an increasing evidence indicating the involvement of the sympathetic nervous system (SNS) in liver disease development. To achieve an extensive comprehension of the obscure process by which the SNS alleviates inflammatory damage in non-parenchymal liver cells (NPCs) during acute liver failure (ALF), we employ isoproterenol (ISO), a beta-adrenoceptor agonist, to mimic SNS signaling. ISO was administered to C57BL/6J mice to establish an acute liver failure (ALF) model using LPS/D-GalN, which was defined as ISO + ALF. Non-parenchymal cells (NPCs) were isolated from liver tissues and digested for tandem mass tag (TMT) labeled proteomics to identify differentially expressed proteins (DEPs). The administration of ISO resulted in a decreased serum levels of pro-inflammatory cytokines, e.g., TNF-α, IL-1β, and IL-6 in ALF mice, which alleviated liver damage. By using TMT analysis, it was possible to identify 1587 differentially expressed proteins (DEPs) in isolated NPCs. Notably, over 60% of the DEPs in the ISO + ALF vs. ALF comparison were shared in the Con vs. ALF comparison. According to enrichment analysis, the DEPs influenced by ISO in ALF mice were linked to biological functions of heme and fatty acid metabolism, interferon gamma response, TNFA signaling pathway, and mitochondrial oxidation function. Protein-protein interaction network analysis indicated Mapk14 and Caspase3 may serve as potentially valuable indicators of ISO intervention. In addition, the markers on activated macrophages, such as Mapk14, Casp1, Casp8, and Mrc1, were identified downregulated after ISO initiation. ISO treatment increased the abundance of anti-inflammatory markers in mouse macrophages, as evidenced by the immunohistochemistry (IHC) slides showing an increase in Arg + staining and a reduction in iNOS + staining. Furthermore, pretreatment with ISO also resulted in a reduction of LPS-stimulated inflammation signaling markers, Mapk14 and NF-κB, in human THP-1 cells. Prior treatment with ISO may have the potential to modify the biological functions of NPCs and could serve as an innovative pharmacotherapy for delaying the pathogenesis and progression of ALF.
Collapse
Affiliation(s)
- Yuchao Wu
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Tianzhi Ni
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Mengmeng Zhang
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
- Honghui Hospital, Xi'an Jiaotong University, Xi'an City, China
| | - Shan Fu
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Danfeng Ren
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Yali Feng
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Huiping Liang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Ze Zhang
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Yingren Zhao
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Yingli He
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Yuan Yang
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China
| | - Zhen Tian
- Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, China.
| | - Taotao Yan
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China.
| | - Jinfeng Liu
- Department of Infectious Diseases and Hepatopathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, China.
| |
Collapse
|
17
|
Musigk N, Suwalski P, Golpour A, Fairweather D, Klingel K, Martin P, Frustaci A, Cooper LT, Lüscher TF, Landmesser U, Heidecker B. The inflammatory spectrum of cardiomyopathies. Front Cardiovasc Med 2024; 11:1251780. [PMID: 38464847 PMCID: PMC10921946 DOI: 10.3389/fcvm.2024.1251780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
Infiltration of the myocardium with various cell types, cytokines and chemokines plays a crucial role in the pathogenesis of cardiomyopathies including inflammatory cardiomyopathies and myocarditis. A more comprehensive understanding of the precise immune mechanisms involved in acute and chronic myocarditis is essential to develop novel therapeutic approaches. This review offers a comprehensive overview of the current knowledge of the immune landscape in cardiomyopathies based on etiology. It identifies gaps in our knowledge about cardiac inflammation and emphasizes the need for new translational approaches to improve our understanding thus enabling development of novel early detection methods and more effective treatments.
Collapse
Affiliation(s)
- Nicolas Musigk
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Phillip Suwalski
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Ainoosh Golpour
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
| | - Karin Klingel
- Cardiopathology Institute for Pathology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Pilar Martin
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Thomas F. Lüscher
- GZO-Zurich Regional Health Centre, Wetzikon & Cardioimmunology, Centre for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Royal Brompton & Harefield Hospitals and National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Bettina Heidecker
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| |
Collapse
|
18
|
Farjoud Kouhanjani M, Hosseini SA, Mousavi SM, Noroozi Z, Sadeghi P, Jokar-Derisi A, Jamshidi Mouselou MS, Ahmadi M, Attar A. Takotsubo Cardiomyopathy and Autoimmune Disorders: A Systematic Scoping Review of Published Cases. Int J Clin Pract 2024; 2024:7259200. [PMID: 38414580 PMCID: PMC10898954 DOI: 10.1155/2024/7259200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 02/29/2024] Open
Abstract
Introduction Takotsubo cardiomyopathy (TCM) features transient left ventricular apical dysfunction or ballooning. The underlying mechanism remains elusive; however, evidence suggests the role of different physical and psychological stressors. We systematically reviewed patients presenting with TCM and autoimmunity to explore the link between the two conditions. Methods We applied the Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) to report this review. Using keywords related to autoimmune/immune-mediated diseases and TCM, we searched PubMed, Scopus, and WOS in March 2022. The final results were added to a data extraction sheet. Data were analyzed by SPSS version 26.0. Results Our search yielded 121 studies, including 155 patients. Females were considerably predominant. Most patients had a history of autoimmune disease, and almost a third had a history of cardiovascular disease. Dyspnea and chest pain were the most common chief complaints. More than 70% of patients had experienced physical stress. Myasthenia gravis, systemic lupus erythematosus, and multiple sclerosis were the most frequently reported autoimmune diseases. Conclusion There were similarities in age and sex compared to classic TCM. TCM should be considered as a differential diagnosis for ACS, especially in patients with a positive background of autoimmunity. A precise reporting system is required for further studies.
Collapse
Affiliation(s)
- Mohsen Farjoud Kouhanjani
- Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Zahra Noroozi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paniz Sadeghi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armita Jokar-Derisi
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Meysam Ahmadi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armin Attar
- Department of Cardiology, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
19
|
Peng Y, Qin D, Wang Y, Gao W, Xu X. Pharmacological inhibition of ICOS attenuates the protective effect of exercise on cardiac fibrosis induced by isoproterenol. Eur J Pharmacol 2024; 965:176327. [PMID: 38224847 DOI: 10.1016/j.ejphar.2024.176327] [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: 08/27/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Abstract
AIMS To investigate the cardioprotective mechanism of exercise or exercise combined with inducible costimulatory molecules (ICOS) monoclonal antibody (mAb) therapy against isoproterenol (ISO)-induced cardiac remodeling. MAIN METHODS Totally 24 male C57BL/6J mice were randomly divided into four groups: the control group (normal saline treatment), ISO group (subcutaneous injection of isoproterenol, 10 mg/kg/day, once daily for 5 consecutive days), the exercise with subcutaneous ISO injection group (EPI), and the exercise with injected with ISO and ICOS mAb group (EPII). The mice in EPI and EPII group were trained on a small animal treadmill for 4 weeks (13 m/min, 0% grade, 60min/day). KEY FINDINGS Exercise significantly attenuated CD45+, Mac-2 inflammatory cell infiltration, cardiac fibrosis and inhibited the RIPK1/RIPK3/MLKL/CaMKII and cardiomyocyte pyroptosis pathways to counter ISO-induced severe cardiac injury. The administration of the ICOS mAb may inhibit the cardioprotection of exercise against ISO-induced heart damage. Compared to those in EPI, our data showed that the increasing levels of myocardial fibrosis, the leukocyte infiltration of cardiac tissue and proteins expression of cardiac myocyte necrosis and pyroptosis signaling pathways in the EPII group. SIGNIFICANCE Our results demonstrated that exercise decreased leukocyte infiltration in heart, inhibited the cardiomyocyte pyroptosis and necroptosis signaling pathways, and attenuated inflammatory responses to alleviate ISO-induced cardiac fibrosis. However, the antifibrotic effects of combined treatment with exercise and ICOS mAb intervention did not exhibit synergistic enhancement.
Collapse
Affiliation(s)
- Yong Peng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China; Jiangsu Collaborative Innovation Center for Sports and Health Project, Nanjing Sport Institute, Nanjing, Jiangsu, China; Key Laboratory of Exercise Training and Rehabilitation for Jiangsu Province, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Di Qin
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Yudi Wang
- School of Physical Education and Nursing, Chengdu College of Arts and Sciences, Chengdu, Sichuan, China
| | - Wenyue Gao
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Xin Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
20
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Klein HG, Feng J, Lertora J, Parizi-Torabi P, Danner RL, Solomon MA, Chen MY, Natanson C. In a Canine Model of Septic Shock, Cardiomyopathy Occurs Independent of Catecholamine Surges and Cardiac Microvascular Ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578927. [PMID: 39803473 PMCID: PMC11722328 DOI: 10.1101/2024.02.05.578927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Background High levels of catecholamines are cardiotoxic and associated with stress-induced cardiomyopathies. Septic patients are routinely exposed to endogenously released and exogenously administered catecholamines, which may alter cardiac function and perfusion causing ischemia. Early during human septic shock, left ventricular ejection fraction (LVEF) decreases but normalizes in survivors over 7-10 days. Employing a septic shock model that reproduces these human septic cardiac findings, we investigated the effects of catecholamines on microcirculatory perfusion and cardiac function. Methods Purpose-bred beagles received intrabronchial Staphylococcus aureus (n=30) or saline (n=6) challenges and septic animals recieved either epinephrine (1mcg/kg/min, n=15) or saline (n=15) infusions from 4 to 44 hours. Serial cardiac magnetic resonance imaging (CMR), invasive hemodynamics and laboratory data including catecholamine levels and troponins were collected over 92 hours. Adenosine-stress perfusion CMR was performed on eight of the fifteen septic epinephrine, and eight of the fifteen septic saline animals. High-dose sedation was titrated for comfort and suppress endogenous catecholamine release. Results Catecholamine levels were largely within the normal range throughout the study in animals receiving an intrabronchial bacteria or saline challenge. However, septic versus non-septic animals developed significant worsening of LV; EF, strain, and -aortic coupling that was not explained by differences in afterload, preload, or heart rate. In septic animals that received epinephrine versus saline infusions, plasma epinephrine levels increased 800-fold, pulmonary and systemic pressures significantly increased, and cardiac edema decreased. Despite this, septic animals receiving epinephrine versus saline during and after infusions, had no significant further worsening of LV; EF, strain, or -aortic coupling. Animals receiving saline had a sepsis-induced increase in microcirculatory reserve without troponin elevations. In contrast, septic animals receiving epinephrine had blunted microcirculatory perfusion and elevated troponin levels that persisted for hours after the infusion stopped. During infusion, septic animals that received epinephrine versus saline had significantly greater lactate, creatinine, and alanine aminotransferase levels. Conclusions Cardiac dysfunction during sepsis is not primarily due to elevated endogenous or exogenous catecholamines nor is it principally due to decreased microvascular perfusion-induced ischemia. However, epinephrine itself has potentially harmful long lasting ischemic effects during sepsis including impaired microvascular perfusion that persists after stopping the infusion.
Collapse
Affiliation(s)
- Verity J. Ford
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Willard N. Applefeld
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- Emory, 100 Woodruff Circle, Atlanta, GA 30322
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Harvey G. Klein
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Juan Lertora
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808
| | - Parizad Parizi-Torabi
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Robert L. Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Michael A. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Marcus Y. Chen
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| |
Collapse
|
21
|
Wu Q, Yao Q, Hu T, Yu J, Jiang K, Wan Y, Tang Q. Dapagliflozin protects against chronic heart failure in mice by inhibiting macrophage-mediated inflammation, independent of SGLT2. Cell Rep Med 2023; 4:101334. [PMID: 38118414 PMCID: PMC10772464 DOI: 10.1016/j.xcrm.2023.101334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/06/2023] [Accepted: 11/20/2023] [Indexed: 12/22/2023]
Abstract
The specific mechanism of sodium-glucose cotransporter 2 (SGLT2) inhibitor in heart failure (HF) needs to be elucidated. In this study, we use SGLT2-global-knockout (KO) mice to assess the mechanism of SGLT2 inhibitor on HF. Dapagliflozin ameliorates both myocardial infarction (MI)- and transverse aortic constriction (TAC)-induced HF. Global SGLT2 deficiency does not exert protection against adverse remodeling in both MI- and TAC-induced HF models. Dapagliflozin blurs MI- and TAC-induced HF phenotypes in SGLT2-KO mice. Dapagliflozin causes major changes in cardiac fibrosis and inflammation. Based on single-cell RNA sequencing, dapagliflozin causes significant differences in the gene expression profile of macrophages and fibroblasts. Moreover, dapagliflozin directly inhibits macrophage inflammation, thereby suppressing cardiac fibroblasts activation. The cardio-protection of dapagliflozin is blurred in mice treated with a C-C chemokine receptor type 2 antagonist. Taken together, the protective effects of dapagliflozin against HF are independent of SGLT2, and macrophage inhibition is the main target of dapagliflozin against HF.
Collapse
Affiliation(s)
- Qingqing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, P.R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, P.R. China
| | - Tongtong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, P.R. China
| | - Jiabin Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, P.R. China
| | - Kebing Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, P.R. China
| | - Ying Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, P.R. China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, P.R. China.
| |
Collapse
|
22
|
Vendrov AE, Xiao H, Lozhkin A, Hayami T, Hu G, Brody MJ, Sadoshima J, Zhang YY, Runge MS, Madamanchi NR. Cardiomyocyte NOX4 regulates resident macrophage-mediated inflammation and diastolic dysfunction in stress cardiomyopathy. Redox Biol 2023; 67:102937. [PMID: 37871532 PMCID: PMC10598408 DOI: 10.1016/j.redox.2023.102937] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023] Open
Abstract
In acute sympathetic stress, catecholamine overload can lead to stress cardiomyopathy. We tested the hypothesis that cardiomyocyte NOX4 (NADPH oxidase 4)-dependent mitochondrial oxidative stress mediates inflammation and diastolic dysfunction in stress cardiomyopathy. Isoproterenol (ISO; 5 mg/kg) injection induced sympathetic stress in wild-type and cardiomyocyte (CM)-specific Nox4 knockout (Nox4CM-/-) mice. Wild-type mice treated with ISO showed higher CM NOX4 expression, H2O2 levels, inflammasome activation, and IL18, IL6, CCL2, and TNFα levels than Nox4CM-/- mice. Spectral flow cytometry and t-SNE analysis of cardiac cell suspensions showed significant increases in pro-inflammatory and pro-fibrotic embryonic-derived resident (CCR2-MHCIIhiCX3CR1hi) macrophages in wild-type mice 3 days after ISO treatment, whereas Nox4CM-/- mice had a higher proportion of embryonic-derived resident tissue-repair (CCR2-MHCIIloCX3CR1lo) macrophages. A significant increase in cardiac fibroblast activation and interstitial collagen deposition and a restrictive pattern of diastolic dysfunction with increased filling pressure was observed in wild-type hearts compared with Nox4CM-/- 7 days post-ISO. A selective NOX4 inhibitor, GKT137831, reduced myocardial mitochondrial ROS, macrophage infiltration, and fibrosis in ISO-injected wild-type mice, and preserved diastolic function. Our data suggest sympathetic overstimulation induces resident macrophage (CCR2-MHCII+) activation and myocardial inflammation, resulting in fibrosis and impaired diastolic function mediated by CM NOX4-dependent ROS.
Collapse
Affiliation(s)
- Aleksandr E Vendrov
- Frankel Cardiovascular Center, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Andrey Lozhkin
- Frankel Cardiovascular Center, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Takayuki Hayami
- Frankel Cardiovascular Center, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Guomin Hu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Matthew J Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Junichi Sadoshima
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, NJ, 07101, USA
| | - You-Yi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, 100191, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Marschall S Runge
- Frankel Cardiovascular Center, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nageswara R Madamanchi
- Frankel Cardiovascular Center, Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
23
|
Lim KRQ, Mann DL, Kenzaka T, Hayashi T. The Immunology of Takotsubo Syndrome. Front Immunol 2023; 14:1254011. [PMID: 37868970 PMCID: PMC10588665 DOI: 10.3389/fimmu.2023.1254011] [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: 07/06/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Takotsubo syndrome (TTS) is a disorder characterized by transient cardiac dysfunction with ventricular regional wall motion abnormalities, primarily thought to be caused by the effects of a sudden catecholamine surge on the heart. Although the majority of patients exhibit prompt recovery of their cardiac dysfunction, TTS remains associated with increased mortality rates acutely and at long-term, and there is currently no cure for TTS. Inflammation has been shown to play a key role in determining outcomes in TTS patients, as well as in the early pathogenesis of the disorder. There are also cases of TTS patients that have been successfully treated with anti-inflammatory therapies, supporting the importance of the inflammatory response in TTS. In this article, we provide a comprehensive review of the available clinical and pre-clinical literature on the immune response in TTS, in an effort to not only better understand the pathophysiology of TTS but also to generate insights on the treatment of patients with this disorder.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO, United States
| | - Douglas L. Mann
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO, United States
| | - Tsuneaki Kenzaka
- Division of Community Medicine and Career Development, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Internal Medicine, Hyogo Prefectural Tamba Medical Center, Tamba, Japan
| | - Tomohiro Hayashi
- Division of Community Medicine and Career Development, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Internal Medicine, Hyogo Prefectural Tamba Medical Center, Tamba, Japan
| |
Collapse
|
24
|
Yoganathan T, Perez-Liva M, Balvay D, Le Gall M, Lallemand A, Certain A, Autret G, Mokrani Y, Guillonneau F, Bruce J, Nguyen V, Gencer U, Schmitt A, Lager F, Guilbert T, Bruneval P, Vilar J, Maissa N, Mousseaux E, Viel T, Renault G, Kachenoura N, Tavitian B. Acute stress induces long-term metabolic, functional, and structural remodeling of the heart. Nat Commun 2023; 14:3835. [PMID: 37380648 DOI: 10.1038/s41467-023-39590-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Takotsubo cardiomyopathy is a stress-induced cardiovascular disease with symptoms comparable to those of an acute coronary syndrome but without coronary obstruction. Takotsubo was initially considered spontaneously reversible, but epidemiological studies revealed significant long-term morbidity and mortality, the reason for which is unknown. Here, we show in a female rodent model that a single pharmacological challenge creates a stress-induced cardiomyopathy similar to Takotsubo. The acute response involves changes in blood and tissue biomarkers and in cardiac in vivo imaging acquired with ultrasound, magnetic resonance and positron emission tomography. Longitudinal follow up using in vivo imaging, histochemistry, protein and proteomics analyses evidences a continued metabolic reprogramming of the heart towards metabolic malfunction, eventually leading to irreversible damage in cardiac function and structure. The results combat the supposed reversibility of Takotsubo, point to dysregulation of glucose metabolic pathways as a main cause of long-term cardiac disease and support early therapeutic management of Takotsubo.
Collapse
Affiliation(s)
| | | | - Daniel Balvay
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Université Paris Cité, Plateforme d'Imageries du Vivant, PARCC, F-75015, Paris, France
| | - Morgane Le Gall
- Université Paris Cité, P53 proteom'IC facility, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Alice Lallemand
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Anais Certain
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Gwennhael Autret
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Université Paris Cité, Plateforme d'Imageries du Vivant, PARCC, F-75015, Paris, France
| | - Yasmine Mokrani
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - François Guillonneau
- Institut de Cancérologie de l'Ouest, CNRS UMR6075 INSERM U1307, 15 rue André Boquel, F-49055, Angers, France
| | - Johanna Bruce
- Université Paris Cité, P53 proteom'IC facility, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Vincent Nguyen
- Sorbonne Université, Laboratoire d'Imagerie Biomédicale, Inserm, CNRS, F-75006, Paris, France
| | - Umit Gencer
- Service de Radiologie, AP-HP, hôpital européen Georges Pompidou, F-75015, Paris, France
| | - Alain Schmitt
- Université Paris Cité, Cochin Imaging, Electron microscopy, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Franck Lager
- Université Paris Cité, Plateforme d'Imageries du Vivant, Institut Cochin, Inserm-CNRS, F-75014, Paris, France
| | - Thomas Guilbert
- Université Paris Cité, Cochin Imaging Photonic, IMAG'IC, Institut Cochin, Inserm, CNRS, F-75014, Paris, France
| | | | - Jose Vilar
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Nawal Maissa
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Elie Mousseaux
- Service de Radiologie, AP-HP, hôpital européen Georges Pompidou, F-75015, Paris, France
| | - Thomas Viel
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Université Paris Cité, Plateforme d'Imageries du Vivant, PARCC, F-75015, Paris, France
| | - Gilles Renault
- Université Paris Cité, Plateforme d'Imageries du Vivant, Institut Cochin, Inserm-CNRS, F-75014, Paris, France
| | - Nadjia Kachenoura
- Sorbonne Université, Laboratoire d'Imagerie Biomédicale, Inserm, CNRS, F-75006, Paris, France
| | - Bertrand Tavitian
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France.
- Université Paris Cité, Plateforme d'Imageries du Vivant, PARCC, F-75015, Paris, France.
- Service de Radiologie, AP-HP, hôpital européen Georges Pompidou, F-75015, Paris, France.
| |
Collapse
|
25
|
Salerno N, Scalise M, Marino F, Filardo A, Chiefalo A, Panuccio G, Torella M, De Angelis A, De Rosa S, Ellison-Hughes GM, Urbanek K, Viglietto G, Torella D, Cianflone E. A Mouse Model of Dilated Cardiomyopathy Produced by Isoproterenol Acute Exposure Followed by 5-Fluorouracil Administration. J Cardiovasc Dev Dis 2023; 10:225. [PMID: 37367390 DOI: 10.3390/jcdd10060225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023] Open
Abstract
Appropriate dilated cardiomyopathy (DCM) animal models are highly desirable considering the pathophysiological and clinical heterogeneity of DCM. Genetically modified mice are the most widely and intensively utilized research animals for DCM. However, to translate discoveries from basic science into new and personalized medical applications, research in non-genetically based DCM models remains a key issue. Here, we characterized a mouse model of non-ischemic DCM induced by a stepwise pharmacologic regime of Isoproterenol (ISO) high dose bolus followed by a low dose systemic injection of the chemotherapy agent, 5-Fluorouracil (5-FU). C57BL/6J mice were injected with ISO and, 3 days after, were randomly assigned to saline or 5-FU. Echocardiography and a strain analysis show that ISO + 5FU in mice induces progressive left ventricular (LV) dilation and reduced systolic function, along with diastolic dysfunction and a persistent global cardiac contractility depression through 56 days. While mice treated with ISO alone recover anatomically and functionally, ISO + 5-FU causes persistent cardiomyocyte death, ensuing in cardiomyocyte hypertrophy through 56 days. ISO + 5-FU-dependent damage was accompanied by significant myocardial disarray and fibrosis along with exaggerated oxidative stress, tissue inflammation and premature cell senescence accumulation. In conclusions, a combination of ISO + 5FU produces anatomical, histological and functional cardiac alterations typical of DCM, representing a widely available, affordable, and reproducible mouse model of this cardiomyopathy.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Andrea Filardo
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Michele Torella
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Georgina M Ellison-Hughes
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 88121 Naples, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
26
|
Wu H, Su H, Zhu C, Wu S, Cui S, Zhou M. Establishment and effect evaluation of a stress cardiomyopathy mouse model induced by different doses of isoprenaline. Exp Ther Med 2023; 25:166. [PMID: 36936708 PMCID: PMC10015318 DOI: 10.3892/etm.2023.11865] [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: 09/03/2022] [Accepted: 02/01/2023] [Indexed: 03/04/2023] Open
Abstract
The optimum dose of isoprenaline (ISO) required to induce stress cardiomyopathy (SC) in mice is not known. The present study aimed to investigate the dose-response association and determine the optimum dose of ISO to establish a high-morbidity/low-mortality SC mouse model to simulate the clinical symptoms of SC. A total of 72 6-week-old wild-type female mice (C57BL/6) were randomly divided into control mice administered normal saline and mice treated with increasing ISO concentrations (5, 10, 25, 50 and 100 mg/kg ISO intraperitoneal injections daily for 14 consecutive days). All mice were analysed by body weight assessment, open field test (OFT), echocardiography (Echo), electrocardiogram (ECG), assessment of myocardial pathology and quantification of cortisol, brain natriuretic peptide (BNP), cardiac troponin T (cTnT), catecholamine (CA) and C-reactive protein (CRP). Compared with the control group, the 25 and 50 mg/kg ISO groups exhibited the most prominent weight changes and lower mortality. The open-field test showed a significant decrease in autonomous activity behaviour in the 25 and 50 mg/kg ISO groups compared with the control group (P<0.05). Echo revealed that the apex of the heart was balloon-like in the 25 and 50 mg/kg ISO groups, along with prominent left ventricular dyskinesia. ECG showed a significant increase in ST segment amplitude, QT interval and Q amplitude (P<0.05) in the 25 and 50 mg/kg ISO group compared with the control group. Haematoxylin and eosin staining of heart tissue showed a disordered arrangement of myocardial cells, dissolution of myocardial fibres and cytoplasm, notable widening of myocardial cell space, oedema and hyperaemia of the interstitium, whereas heart tissue of the control group was structurally intact. Compared with the control group, the 25 and 50 mg/kg ISO groups exhibited significantly higher levels of cortisol, BNP, cTNT, CA and CRP (P<0.05). A high-incidence low-mortality SC model was successfully and stably developed by administration of 25 and 50 mg/kg ISO. Such models may provide a basis for the development of other animal models of SC.
Collapse
Affiliation(s)
- Haosheng Wu
- Graduate School, Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230000, P.R. China
| | - Hang Su
- Graduate School, Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230000, P.R. China
| | - Chao Zhu
- Graduate School, Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230000, P.R. China
| | - Shengbing Wu
- Research Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, Anhui 230000, P.R. China
- College of Acupuncture and Massage, Anhui University of Chinese Medicine, Hefei, Anhui 230000, P.R. China
| | - Shuai Cui
- Research Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, Anhui 230000, P.R. China
- College of Acupuncture and Massage, Anhui University of Chinese Medicine, Hefei, Anhui 230000, P.R. China
| | - Meiqi Zhou
- Research Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, Anhui 230000, P.R. China
| |
Collapse
|
27
|
Hayashi T, Tiwary SK, Lim KRQ, Rocha-Resende C, Kovacs A, Weinheimer C, Mann DL. Refining the reproducibility of a murine model of stress-induced reversible cardiomyopathy. Am J Physiol Heart Circ Physiol 2023; 324:H229-H240. [PMID: 36563015 PMCID: PMC9886343 DOI: 10.1152/ajpheart.00684.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Despite the many advantages of isoproterenol (Iso)-induced models of cardiomyopathy, the extant literature suggests that the reproducibility of the Iso-induced stress cardiomyopathy phenotype varies considerably depending on the dose of Iso used, the mode of administration of Iso (subcutaneous vs. intraperitoneal), and the species of the animal that is being studied. Recently, we have shown that a single injection of Iso into female C57BL/6J mice provokes transient myocardial injury that is characterized by a brisk release of troponin I within 1 h, as well as a self-limited myocardial inflammatory response that is associated with increased myocardial tissue edema, inferoapical regional left ventricular (LV) wall motion abnormalities, and a transient decrease in global LV function, which were completely recovered by day 7 after the Iso injection (i.e., stress-induced reversible cardiomyopathy). Here we expand upon this initial report in this model by demonstrating important sexually dimorphic differences in the response to Iso-induced tissue injury, the ensuing myocardial inflammatory response, and changes in LV structure and function. We also provide information with respect to enhancing the reproducibility in this model by optimizing animal welfare during the procedure. The acute Iso-induced myocardial injury model provides a low-cost, relatively high-throughput small-animal model that mimics human disease (e.g., Takotsubo cardiomyopathy). Given that the model can be performed in different genetic backgrounds, as well as different experimental conditions, the acute Iso injury model should provide the cardiovascular community with a valuable nonsurgical animal model for understanding the myocardial response to tissue injury.NEW & NOTEWORTHY The present study highlights the importance of sexual dimorphism with respect to isoproterenol injury, as well as the importance of animal handling and welfare to obtain reproducible results from investigator to investigator. Based on serial observations of animal recovery (locomotor activity and grooming behavior), troponin I release, and inflammation, we identified that the method used to restrain the mice for the intraperitoneal injection was the single greatest source of variability in this model.
Collapse
Affiliation(s)
- Tomohiro Hayashi
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Sajal K Tiwary
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Kenji Rowel Q Lim
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Cibele Rocha-Resende
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Attila Kovacs
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Carla Weinheimer
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Douglas L Mann
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
28
|
Sethi Y, Murli H, Kaiwan O, Vora V, Agarwal P, Chopra H, Padda I, Kanithi M, Popoviciu MS, Cavalu S. Broken Heart Syndrome: Evolving Molecular Mechanisms and Principles of Management. J Clin Med 2022; 12:jcm12010125. [PMID: 36614928 PMCID: PMC9821117 DOI: 10.3390/jcm12010125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Broken Heart Syndrome, also known as Takotsubo Syndrome (TS), is sudden and transient dysfunction of the left and/or right ventricle which often mimics Acute Coronary Syndrome (ACS). Japan was the first country to describe this syndrome in the 1990s, and since then it has received a lot of attention from researchers all around the world. Although TS was once thought to be a harmless condition, recent evidence suggests that it may be linked to serious complications and mortality on par with Acute Coronary Syndrome (ACS). The understanding of TS has evolved over the past few years. However, its exact etiology is still poorly understood. It can be classified into two main types: Primary and Secondary TS. Primary TS occurs when the symptoms of myocardial damage, which is typically preceded by emotional stress, are the reason for hospitalization. Secondary TS is seen in patients hospitalized for some other medical, surgical, obstetric, anesthetic, or psychiatric conditions, and the dysfunction develops as a secondary complication due to the activation of the sympathetic nervous system and the release of catecholamines. The etiopathogenesis is now proposed to include adrenergic hormones/stress, decreased estrogen levels, altered microcirculation, endothelial dysfunction, altered inflammatory response via cardiac macrophages, and disturbances in the brain-heart axis. The role of genetics in disease progression is becoming the focus of several upcoming studies. This review focuses on potential pathophysiological mechanisms for reversible myocardial dysfunction observed in TS, and comprehensively describes its epidemiology, clinical presentation, novel diagnostic biomarkers, and evolving principles of management. We advocate for more research into molecular mechanisms and promote the application of current evidence for precise individualized treatment.
Collapse
Affiliation(s)
- Yashendra Sethi
- PearResearch, Dehradun 248001, India
- Department of Medicine, Government Doon Medical College, Dehradun 248001, India
- Correspondence: (Y.S.); (M.S.P.)
| | - Hamsa Murli
- PearResearch, Dehradun 248001, India
- Department of Medicine, Lokmanya Tilak Municipal Medical College, Mumbai 400022, India
| | - Oroshay Kaiwan
- PearResearch, Dehradun 248001, India
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Vidhi Vora
- PearResearch, Dehradun 248001, India
- Department of Medicine, Lokmanya Tilak Municipal Medical College, Mumbai 400022, India
| | - Pratik Agarwal
- PearResearch, Dehradun 248001, India
- Department of Medicine, Lokmanya Tilak Municipal Medical College, Mumbai 400022, India
| | - Hitesh Chopra
- College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Inderbir Padda
- Richmond University Medical Center, Staten Island, NY 10310, USA
| | - Manasa Kanithi
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Mihaela Simona Popoviciu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
- Correspondence: (Y.S.); (M.S.P.)
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
29
|
Hayashi T, Tiwary SK, Lavine KJ, Acharya S, Brent M, Adamo L, Kovacs A, Mann DL. The Programmed Death-1 Signaling Axis Modulates Inflammation and LV Structure/Function in a Stress-Induced Cardiomyopathy Model. JACC Basic Transl Sci 2022; 7:1120-1139. [PMID: 36687266 PMCID: PMC9849278 DOI: 10.1016/j.jacbts.2022.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022]
Abstract
The role of immune checkpoints in the setting of tissue injury remains unknown. Using an experimental model of isoproterenol (ISO)-induced stress cardiomyopathy, we show that ISO-induced myocardial injury provokes tissue-autonomous up-regulation of the programmed death-1 (PD-1):programmed death ligand (PD-L) axis in cardiac resident innate immune cells and T cells. PD-1 signaling was responsible for modulating the acute inflammatory response, as well as normalization of impaired left ventricular structure and function after ISO injection. Necrotic cardiac extracts were sufficient to increase the expression of PD-1 in macrophages and T cells in vitro. Viewed together these studies suggest that the PD-1:PD-L signaling axis regulates immune responses to cardiac tissue injury and is important for restoring myocardial homeostasis.
Collapse
Affiliation(s)
- Tomohiro Hayashi
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Sajal K. Tiwary
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Kory J. Lavine
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Sandeep Acharya
- Department of Computer Science, Washington University, St Louis, Missouri, USA
| | - Michael Brent
- Department of Computer Science, Washington University, St Louis, Missouri, USA
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Luigi Adamo
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Attila Kovacs
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Douglas L. Mann
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
30
|
Resident cardiac macrophages: Heterogeneity and function in health and disease. Immunity 2022; 55:1549-1563. [PMID: 36103852 DOI: 10.1016/j.immuni.2022.08.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/20/2022]
Abstract
Understanding tissue macrophage biology has become challenging in recent years due the ever-increasing complexity in macrophage-subset identification and functional characterization. This is particularly important within the myocardium, as we have come to understand that macrophages play multifaceted roles in cardiac health and disease, and heart disease remains the leading cause of death worldwide. Here, we review recent progress in the field, focusing on resident cardiac macrophage heterogeneity, origins, and functions at steady state and after injury. We stratify resident cardiac macrophage functions by the ability of macrophages to either directly influence cardiac physiology or indirectly influence cardiac physiology through orchestrating multi-cellular communication with cardiomyocytes and stromal and immune populations.
Collapse
|
31
|
Gudenkauf B, Goetsch MR, Vakil RM, Cingolani O, Adamo L. Case Report: Steroid-Responsive Takotsubo Cardiomyopathy Associated With Cytokine Storm and Obstructive Shock. Front Cardiovasc Med 2022; 9:931070. [PMID: 35898274 PMCID: PMC9309717 DOI: 10.3389/fcvm.2022.931070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/23/2022] [Indexed: 12/01/2022] Open
Abstract
A growing body of evidence suggests that inflammation may play a key role in the development of Takotsubo stress cardiomyopathy. Here, we report the case of a 63-year-old woman who presented with chest pain and was diagnosed with this cardiomyopathy. After an initial improvement, the patient experienced a systemic inflammatory response of unclear origin and deteriorated rapidly into obstructive shock. Her presentation was considered consistent with cytokine storm. She was, therefore, treated with steroids with rapid improvement in her clinical picture. She relapsed after the taper. Endomyocardial biopsy soon after initiation of pulse dose steroids showed macrophage and lymphocytic infiltration. This case highlights the potential intimate connection between systemic inflammatory response and Takotsubo stress cardiomyopathy and contributes to the evolving understanding of inflammation in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Brent Gudenkauf
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael R. Goetsch
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rachit M. Vakil
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Oscar Cingolani
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Luigi Adamo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Luigi Adamo
| |
Collapse
|
32
|
Shi HT, Huang ZH, Xu TZ, Sun AJ, Ge JB. New diagnostic and therapeutic strategies for myocardial infarction via nanomaterials. EBioMedicine 2022; 78:103968. [PMID: 35367772 PMCID: PMC8983382 DOI: 10.1016/j.ebiom.2022.103968] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction is lethal to patients because of insufficient blood perfusion to vital organs. Several attempts have been made to improve its prognosis, among which nanomaterial research offers an opportunity to address this problem at the molecular level and has the potential to improve disease prevention, diagnosis, and treatment significantly. Up to now, nanomaterial-based technology has played a crucial role in broad novel diagnostic and therapeutic strategies for cardiac repair. This review summarizes various nanomaterial applications in myocardial infarction from multiple aspects, including high precision detection, pro-angiogenesis, regulating immune homeostasis, and miRNA and stem cell delivery vehicles. We also propose promising research hotspots that have not been reported much yet, such as conjugating pro-angiogenetic elements with nanoparticles to construct drug carriers, developing nanodrugs targeting other immune cells except for macrophages in the infarcted myocardium or the remote region. Though most of those strategies are preclinical and lack clinical trials, there is tremendous potential for their further applications in the future.
Collapse
Affiliation(s)
- Hong-Tao Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, China
| | - Zi-Hang Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Tian-Zhao Xu
- School of Life Science, Shanghai University, Shanghai, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China.
| | - Jun-Bo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China.
| |
Collapse
|