1
|
Cortada E, Yao J, Xia Y, Dündar F, Zumbo P, Yang B, Rubio-Navarro A, Perder B, Qiu M, Pettinato AM, Homan EA, Stoll L, Betel D, Cao J, Lo JC. Cross-species single-cell RNA-seq analysis reveals disparate and conserved cardiac and extracardiac inflammatory responses upon heart injury. Commun Biol 2024; 7:1611. [PMID: 39627536 PMCID: PMC11615278 DOI: 10.1038/s42003-024-07315-x] [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: 08/12/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
The immune system coordinates the response to cardiac injury and controls regenerative and fibrotic scar outcomes in the heart and subsequent chronic low-grade inflammation associated with heart failure. Adult mice and humans lack the ability to fully recover while adult zebrafish spontaneously regenerate after heart injury. Here we profile the inflammatory response to heart cryoinjury in zebrafish and coronary artery ligation in mouse using single cell transcriptomics. We interrogate the extracardiac reaction to cardiomyocyte necrosis to assess the specific peripheral tissue and immune cell reaction to chronic stress. Cardiac macrophages play a critical role in determining tissue homeostasis by healing versus scarring. We identify distinct transcriptional clusters of monocytes/macrophages (mono/Mϕ) in each species and find analogous pairs in zebrafish and mice. However, the reaction to myocardial injury is largely disparate between mice and zebrafish. The dichotomous response to heart damage between the murine and zebrafish mono/Mϕ and/or the presence of distinct zebrafish mono/Mϕ subtypes may underlie the impaired regenerative process in adult mammals and humans. Our study furnishes a direct cross-species comparison of immune responses between regenerative and profibrotic myocardial injury models, providing a useful resource to the fields of regenerative biology and cardiovascular research.
Collapse
Affiliation(s)
- Eric Cortada
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Jun Yao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Friederike Dündar
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Boris Yang
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Björn Perder
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Anthony M Pettinato
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
| | - Edwin A Homan
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA.
- Institute for Computational Biomedicine, Division of Hematology and Medical, Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA.
| | - James C Lo
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA.
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Seropian IM, El-Diasty M, El-Sherbini AH, González GE, Rabinovich GA. Central role of Galectin-3 at the cross-roads of cardiac inflammation and fibrosis: Implications for heart failure and transplantation. Cytokine Growth Factor Rev 2024; 80:47-58. [PMID: 39482190 DOI: 10.1016/j.cytogfr.2024.10.002] [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: 09/02/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024]
Abstract
Cardiac inflammation and fibrosis are central pathogenic mechanisms leading to heart failure. Transplantation is still the treatment of choice for many patients undergoing end-stage heart failure who remain symptomatic despite optimal medical therapy. In spite of considerable progress, the molecular mechanisms linking inflammation, fibrosis and heart failure remain poorly understood. Galectin-3 (GAL3), a chimera-type member of the galectin family, has emerged as a critical mediator implicated in cardiac inflammatory, vascular and fibrotic processes through modulation of different cellular compartments including monocytes and macrophages, fibroblasts, endothelial cells and vascular smooth muscle cells via glycan-dependent or independent mechanisms. GAL3-driven circuits may hierarchically amplify cytokine production and function, immune cell activation and fibrosis cascades, influencing a wide range of cardiovascular disorders. Thus, GAL3 emerges as a potential therapeutic target to counteract aberrant inflammation and fibrosis during heart failure and a potential biomarker of heart failure and clinical outcome of heart transplantation.
Collapse
Affiliation(s)
- Ignacio M Seropian
- Servicio de Hemodinamia y Cardiología Intervencionista, Hospital Italiano de Buenos Aires, Ciudad de Buenos Aires C1199, Argentina; Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Medicina, Pontificia Universidad Católica Argentina, Ciudad de Buenos Aires & Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Ciudad de Buenos Aires, Argentina
| | - Mohammad El-Diasty
- Harrington Heart and Vascular Institute, University Hospital Cleveland Medical Center, Cleveland, OH 44106, USA; Faculty of Health Sciences, Queen's University, Kingston, ON K7L 2V7, Canada
| | | | - Germán E González
- Laboratorio de Patología Cardiovascular Experimental e Hipertensión Arterial, Instituto de Investigaciones Biomédicas (UCA-CONICET), Facultad de Medicina, Pontificia Universidad Católica Argentina, Ciudad de Buenos Aires & Departamento de Patología, Instituto de Salud Comunitaria, Universidad Nacional de Hurlingham, Ciudad de Buenos Aires, Argentina.
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires C1428, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, C1428, Argentina.
| |
Collapse
|
3
|
Ding Z, Liu X, Jiang H, Zhao J, Temme S, Bouvain P, Alter C, Rafii P, Scheller J, Flögel U, Zhu H, Ding Z. A refined TTC assay precisely detects cardiac injury and cellular viability in the infarcted mouse heart. Sci Rep 2024; 14:25214. [PMID: 39448689 PMCID: PMC11502796 DOI: 10.1038/s41598-024-76414-w] [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: 06/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Histological analysis with 2,3,5-triphenyltetrazolium chloride (TTC) staining is the most frequently used tool to detect myocardial ischemia/reperfusion injury. However, its practicality is often challenged by poor image quality in gross histology, leading to an equivocal infarct-boundary delineation and potentially compromised measurement accuracy. Here, we introduce several crucial refinements in staining protocol and sample processing, which enable TTC images to be analyzed with light microscopy. The refined protocol involves a two-step TTC staining process (perfusion and immersion) and subsequent Zamboni fixation to differentiate myocardial viability and necrosis, and use of Coomassie brilliant blue to label area-at-risk. After the duo-staining steps were completed, the heart sample was embedded and sliced transversally by a cryostat into a series of thin sections (50 µm) for microscopic analysis. The refined TTC (redTTC) assay yielded remarkably high-quality images with striking color intensity and sharply defined boundaries, permitting unambiguous and reliable delineation of the infarct and area-at-risk. In the same animals, the redTTC assay showed good agreement with the in-vivo gold standard measurements (LGE and MEMRI). Meanwhile, redTTC imaging allows tracking of viable cardiomyocytes at cellular resolution, and with this enhanced capability, we convincingly demonstrated the pro-survival action of stem cells based-therapy. Therefore, the redTTC assay represents a significant technical advance that permits precise detection of the true extent of cardiac injury and cardiomyocyte viability. This approach is cost-effective and may be adapted for use in diverse applications, making it highly appealing to many laboratories performing ischemia/reperfusion injury experiments.
Collapse
Affiliation(s)
- Zheheng Ding
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Xueqing Liu
- Department of Cardiology, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, West Xinmin Rd. 2, Danyang, 212300, China
| | - Hongyan Jiang
- Department of Cardiology, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, West Xinmin Rd. 2, Danyang, 212300, China
| | - Jianfeng Zhao
- Department of Cardiology, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, West Xinmin Rd. 2, Danyang, 212300, China
| | - Sebastian Temme
- Institute of Anesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Pascal Bouvain
- Institute of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Christina Alter
- Institute of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Ulrich Flögel
- Institute of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Hongtao Zhu
- Department of Cardiology, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, West Xinmin Rd. 2, Danyang, 212300, China.
| | - Zhaoping Ding
- Institute of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
4
|
Li YL, Chen BY, Feng ZH, Zhou LJ, Liu T, Lin WZ, Zhu H, Xu S, Bai XB, Meng XQ, Zhang J, Liu Y, Pu J, Jiang M, Duan SZ. Roles of oral and gut microbiota in acute myocardial infarction. J Adv Res 2024:S2090-1232(24)00463-6. [PMID: 39447641 DOI: 10.1016/j.jare.2024.10.009] [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: 12/10/2023] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
INTRODUCTION The significance of oral/gut microbiota in acute myocardial infarction (AMI) has been increasingly appreciated. However, correlations between oral/gut microbiota and AMI parameter, as well as the key microbiota that may have a crucial function in this process, remain unclear. OBJECTIVES To investigate the composition and structure of oral and gut microbiota associated with AMI and explore the roles of specific bacterial species in the progression of AMI. METHODS We conducted a case-control study with 37 AMI patients and 36 controls. Oral and gut sample were collected and sequenced. Using correlation analysis, we combined bioinformatics data with AMI clinical parameters and obtained heatmaps of correlation coefficients. Additionally, we used antibiotics to eliminate the gut microbiota of C57BL/6J mice, followed by the transplantation of selected bacteria to verify the gut colonization of oral bacteria and their impact on AMI. RESULTS The component of oral and gut microbiota of AMI group showed significant alterations when compared to the control group. 17 salivary genera, 21 subgingival genera, and 8 gut genera in AMI group substantially differed from those in control group. Additionally, 19 genera from saliva, 19 genera from subgingival plaque, and 11 genera from feces substantially correlated with AMI clinical parameters. Orally administrated S.o (Streptococcus oralis subsp. dentisani), S.p (Streptococcus parasanguinis), and S.s (Streptococcus salivarius) were able to colonize in the gut and exacerbate myocardial infarction. CONCLUSION There is a strong correlation between oral/gut microbiota and AMI. Streptococcus spp. is capable to transmit from oral to gut and exacerbate myocardial infarction in mice. Monitoring and control of specific oral microbiota may be an effective new strategy for improving the therapy of AMI.
Collapse
Affiliation(s)
- Yu-Lin Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Department of General Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ze-Hao Feng
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China; Department of Cardiology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu-Jun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Department of General Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wen-Zhen Lin
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Department of General Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Hong Zhu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shuo Xu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xue-Bing Bai
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Department of General Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Xiao-Qian Meng
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jun Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China.
| | - Meng Jiang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China.
| | - Sheng-Zhong Duan
- Stomatology Hospital, School of Stomatology, and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Köhler D, Leiss V, Beichert L, Killinger S, Grothe D, Kushwaha R, Schröter A, Roslan A, Eggstein C, Focken J, Granja T, Devanathan V, Schittek B, Lukowski R, Weigelin B, Rosenberger P, Nürnberg B, Beer-Hammer S. Targeting Gα i2 in neutrophils protects from myocardial ischemia reperfusion injury. Basic Res Cardiol 2024; 119:717-732. [PMID: 38811421 PMCID: PMC11461587 DOI: 10.1007/s00395-024-01057-x] [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: 10/09/2023] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
Neutrophils are not only involved in immune defense against infection but also contribute to the exacerbation of tissue damage after ischemia and reperfusion. We have previously shown that genetic ablation of regulatory Gαi proteins in mice has both protective and deleterious effects on myocardial ischemia reperfusion injury (mIRI), depending on which isoform is deleted. To deepen and analyze these findings in more detail the contribution of Gαi2 proteins in resident cardiac vs circulating blood cells for mIRI was first studied in bone marrow chimeras. In fact, the absence of Gαi2 in all blood cells reduced the extent of mIRI (22,9% infarct size of area at risk (AAR) Gnai2-/- → wt vs 44.0% wt → wt; p < 0.001) whereas the absence of Gαi2 in non-hematopoietic cells increased the infarct damage (66.5% wt → Gnai2-/- vs 44.0% wt → wt; p < 0.001). Previously we have reported the impact of platelet Gαi2 for mIRI. Here, we show that infarct size was substantially reduced when Gαi2 signaling was either genetically ablated in neutrophils/macrophages using LysM-driven Cre recombinase (AAR: 17.9% Gnai2fl/fl LysM-Cre+/tg vs 42.0% Gnai2fl/fl; p < 0.01) or selectively blocked with specific antibodies directed against Gαi2 (AAR: 19.0% (anti-Gαi2) vs 49.0% (IgG); p < 0.001). In addition, the number of platelet-neutrophil complexes (PNCs) in the infarcted area were reduced in both, genetically modified (PNCs: 18 (Gnai2fl/fl; LysM-Cre+/tg) vs 31 (Gnai2fl/fl); p < 0.001) and in anti-Gαi2 antibody-treated (PNCs: 9 (anti-Gαi2) vs 33 (IgG); p < 0.001) mice. Of note, significant infarct-limiting effects were achieved with a single anti-Gαi2 antibody challenge immediately prior to vessel reperfusion without affecting bleeding time, heart rate or cellular distribution of neutrophils. Finally, anti-Gαi2 antibody treatment also inhibited transendothelial migration of human neutrophils (25,885 (IgG) vs 13,225 (anti-Gαi2) neutrophils; p < 0.001), collectively suggesting that a therapeutic concept of functional Gαi2 inhibition during thrombolysis and reperfusion in patients with myocardial infarction should be further considered.
Collapse
Affiliation(s)
- David Köhler
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Veronika Leiss
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Lukas Beichert
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Simon Killinger
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Daniela Grothe
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Ragini Kushwaha
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Agnes Schröter
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Anna Roslan
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Claudia Eggstein
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Jule Focken
- Division of Dermatooncology, Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Tiago Granja
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Vasudharani Devanathan
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, 517507, India
| | - Birgit Schittek
- Division of Dermatooncology, Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Bettina Weigelin
- Department of Preclinical Imaging and Radiopharmacy, Multiscale Immunoimaging, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, Eberhard Karls University, Tübingen, Germany
| | - Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomic, Eberhard Karls University, and Interfaculty Center of Pharmacogenomic and Drug Research, Wilhelmstrasse 56, 72074, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
6
|
Kain V, Grilo GA, Upadhyay G, Nadler JL, Serhan CN, Halade GV. Macrophage-specific lipoxygenase deletion amplify cardiac repair activating Treg cells in chronic heart failure. J Leukoc Biol 2024; 116:864-875. [PMID: 38785336 PMCID: PMC11444306 DOI: 10.1093/jleuko/qiae113] [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: 12/06/2023] [Revised: 04/01/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Splenic leukocytes, particularly macrophage-expressed lipoxygenases, facilitate the biosynthesis of resolution mediators essential for cardiac repair. Next, we asked whether deletion of 12/15 lipoxygenase (12/15LOX) in macrophages impedes the resolution of inflammation following myocardial infarction (MI). Using 12/15flox/flox and LysMcre scheme, we generated macrophage-specific 12/15LOX (Mɸ-12/15LOX-/-) mice. Young C57BL/6J wild-type and Mɸ-12/15LOX-/- male mice were subjected to permanent coronary ligation microsurgery. Mice were monitored at day 1 (d1) to d5 (as acute heart failure [AHF]) and to d56 (chronic HF) post-MI, maintaining no MI as d0 naïve control animals. Post ligation, Mɸ-12/15LOX-/- mice showed increased survival (88% vs 56%) and limited heart dysfunction compared with wild-type. In AHF, Mɸ-12/15LOX-/- mice have increased biosynthesis of epoxyeicosatrienoic acid by 30%, with the decrease in D-series resolvins, protectin, and maresin by 70% in the infarcted heart. Overall, myeloid cell profiling from the heart and spleen indicated that Mɸ-12/15LOX-/- mice showed higher immune cells with reparative Ly6Clow macrophages during AHF. In addition, the detailed immune profiling revealed reparative macrophage phenotype (Ly6Clow) in Mɸ-12/15LOX-/- mice in a splenocardiac manner post-MI. Mɸ-12/15LOX-/- mice showed an increase in myeloid population that coordinated increase of T regulatory cells (CD4+/Foxp3+) in the spleen and injured heart at chronic HF compared with wild-type. Thus, macrophage-specific deletion of 12/15LOX directs reparative macrophage phenotype to facilitate cardiac repair. The presented study outlines the complex role of 12/15LOX in macrophage plasticity and T regulatory cell signaling that indicates that resolution mediators are viable targets to facilitate cardiac repair in HF post-MI.
Collapse
Affiliation(s)
- Vasundhara Kain
- Division of Cardiovascular Sciences, Heart Institute, Department of Medicine, University of South Florida, 560 Channelside Dr, Tampa, FL 33602, United States
| | - Gabriel Araujo Grilo
- Division of Cardiovascular Sciences, Heart Institute, Department of Medicine, University of South Florida, 560 Channelside Dr, Tampa, FL 33602, United States
| | - Gunjan Upadhyay
- Division of Cardiovascular Sciences, Heart Institute, Department of Medicine, University of South Florida, 560 Channelside Dr, Tampa, FL 33602, United States
| | - Jerry L Nadler
- Department of Medicine, New York Medical College, 40 Sunshine Cottage Rd, Valhalla, NY 10595, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, 60, Fenwood Road, Boston, MA 02115, USA
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Heart Institute, Department of Medicine, University of South Florida, 560 Channelside Dr, Tampa, FL 33602, United States
| |
Collapse
|
7
|
Standley A, Xie J, Lau AW, Grote L, Gifford AJ. Working with Miraculous Mice: Mus musculus as a Model Organism. Curr Protoc 2024; 4:e70021. [PMID: 39435766 DOI: 10.1002/cpz1.70021] [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: 10/23/2024]
Abstract
The laboratory mouse has been described as a "miracle" model organism, providing a window by which we may gain an understanding of ourselves. Since the first recorded mouse experiment in 1664, the mouse has become the most used animal model in biomedical research. Mice are ideally suited as a model organism because of their small size, short gestation period, large litter size, and genetic similarity to humans. This article provides a broad overview of the laboratory mouse as a model organism and is intended for undergraduates and those new to working with mice. We delve into the history of the laboratory mouse and outline important terminology to accurately describe research mice. The types of laboratory mice available to researchers are reviewed, including outbred stocks, inbred strains, immunocompromised mice, and genetically engineered mice. The critical role mice have played in advancing knowledge in the areas of oncology, immunology, and pharmacology is highlighted by examining the significant contribution of mice to Nobel Prize winning research. International mouse mutagenesis programs and accurate phenotyping of mouse models are outlined. We also explain important considerations for working with mice, including animal ethics; the welfare principles of replacement, refinement, and reduction; and the choice of mouse model in experimental design. Finally, we present practical advice for maintaining a mouse colony, which involves adequate training of staff, the logistics of mouse housing, monitoring colony health, and breeding strategies. Useful resources for working with mice are also listed. The aim of this overview is to equip the reader with a broad appreciation of the enormous potential and some of the complexities of working with the laboratory mouse in a quest to improve human health. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Anick Standley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Jinhan Xie
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Angelica Wy Lau
- Garvan Institute of Medical Research, St Vincent's Clinical School, Darlinghurst, NSW, Australia
| | - Lauren Grote
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- Anatomical Pathology, NSW Heath Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
8
|
Cousineau JP, Dawe AM, Alpaugh M. Investigating the Interplay between Cardiovascular and Neurodegenerative Disease. BIOLOGY 2024; 13:764. [PMID: 39452073 PMCID: PMC11505144 DOI: 10.3390/biology13100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024]
Abstract
Neurological diseases, including neurodegenerative diseases (NDDs), are the primary cause of disability worldwide and the second leading cause of death. The chronic nature of these conditions and the lack of disease-modifying therapies highlight the urgent need for developing effective therapies. To accomplish this, effective models of NDDs are required to increase our understanding of underlying pathophysiology and for evaluating treatment efficacy. Traditionally, models of NDDs have focused on the central nervous system (CNS). However, evidence points to a relationship between systemic factors and the development of NDDs. Cardiovascular disease and related risk factors have been shown to modify the cerebral vasculature and the risk of developing Alzheimer's disease. These findings, combined with reports of changes to vascular density and blood-brain barrier integrity in other NDDs, such as Huntington's disease and Parkinson's disease, suggest that cardiovascular health may be predictive of brain function. To evaluate this, we explore evidence for disruptions to the circulatory system in murine models of NDDs, evidence of disruptions to the CNS in cardiovascular disease models and summarize models combining cardiovascular disruption with models of NDDs. In this study, we aim to increase our understanding of cardiovascular disease and neurodegeneration interactions across multiple disease states and evaluate the utility of combining model systems.
Collapse
Affiliation(s)
| | | | - Melanie Alpaugh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.P.C.); (A.M.D.)
| |
Collapse
|
9
|
Guevara A, Smith CER, Wang L, Caldwell JL, Tapa S, Francis Stuart SD, Ma BW, Ng GA, Habecker BA, Wang Z, Ripplinger CM. Sympathetic structural and electrophysiological remodeling in a rabbit model of reperfused myocardial infarction. Am J Physiol Heart Circ Physiol 2024; 327:H631-H638. [PMID: 39028283 PMCID: PMC11442023 DOI: 10.1152/ajpheart.00398.2024] [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: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/20/2024]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) inhibit sympathetic reinnervation in rodent hearts post-myocardial infarction (MI), causing regional hypoinnervation that is associated with supersensitivity of β-adrenergic receptors and increased arrhythmia susceptibility. To investigate the role of CSPGs and hypoinnervation in the heart of larger mammals, we used a rabbit model of reperfused MI and tested electrophysiological responses to sympathetic nerve stimulation (SNS). Innervated hearts from MI and sham rabbits were optically mapped using voltage and Ca2+-sensitive dyes. SNS was performed with electrical stimulation of the spinal cord, and β-adrenergic responsiveness was tested using isoproterenol. Sympathetic nerve density and CSPG expression were evaluated using immunohistochemistry. CSPGs were robustly expressed in the infarct region of all MI hearts, and the presence of CSPGs was associated with reduced sympathetic nerve density in the infarct versus remote region. Action potential duration (APD) dispersion and tendency for induction of ventricular tachycardia/fibrillation (VT/VF) were increased with SNS in MI but not sham hearts. SNS decreased APD at 80% repolarization (APD80) in MI but not sham hearts, whereas isoproterenol decreased APD80 in both groups. Isoproterenol also shortened Ca2+ transient duration at 80% repolarization in both groups but to a greater extent in MI hearts. Our data suggest that sympathetic remodeling post-MI is similar between rodents and rabbits, with CSPGs associated with sympathetic hypoinnervation. Despite a reduction in sympathetic nerve density, the infarct region of MI hearts remained responsive to both physiological SNS and isoproterenol, potentially through preserved or elevated β-adrenergic responsiveness, which may underlie increased APD dispersion and tendency for VT/VF.NEW & NOTEWORTHY Here, we show that CSPGs are present in the infarcts of rabbit hearts with reperfused MI, where they are associated with reduced sympathetic nerve density. Despite hypoinnervation, sympathetic responsiveness is maintained or enhanced in MI rabbit hearts, which also demonstrate increased APD dispersion and tendency for arrhythmias following sympathetic modulation. Together, this study indicates that the mechanisms of sympathetic remodeling post-MI are similar between rodents and rabbits, with hypoinnervation likely associated with enhanced β-adrenergic sensitivity.
Collapse
Affiliation(s)
- Amanda Guevara
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Charlotte E R Smith
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Lianguo Wang
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Jessica L Caldwell
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Srinivas Tapa
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | | | - Betty W Ma
- Campus Veterinary Services, University of California Davis, Davis, California, United States
| | - G Andre Ng
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health & Care Research, Leicester Biomedical Research Centre, Leicester, United Kingdom
- Glenfield Hospital, Leicester, United Kingdom
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
- Department of Medicine and Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Zhen Wang
- Department of Pharmacology, University of California Davis, Davis, California, United States
- Shantou University Medical College, Shantou, China
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis, Davis, California, United States
| |
Collapse
|
10
|
Kruithof BPT, Mousavi Gourabi B, van de Merbel AF, DeRuiter MC, Goumans MJ. A New Ex Vivo Model to Study Cardiac Fibrosis in Whole Mouse Hearts. JACC Basic Transl Sci 2024; 9:1005-1022. [PMID: 39297130 PMCID: PMC11405901 DOI: 10.1016/j.jacbts.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 09/21/2024]
Abstract
Fibrosis is a characteristic of many cardiac diseases for which no effective treatment exists. We have developed an ex vivo flow system, which allows induction of cardiac fibrosis in intact adult mouse hearts. Lineage-tracing studies indicated that the collagen-producing myofibroblasts originated from the resident fibroblasts. The extent of fibrosis was flow rate dependent, and pharmacological inhibition of the transforming growth factor beta signaling pathway prevented fibrosis. Therefore, in this powerful system, the cellular and molecular mechanisms underlying cardiac fibrosis can be studied. In addition, new targets can be tested on organ level for their ability to inhibit fibrosis.
Collapse
Affiliation(s)
- Boudewijn P T Kruithof
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Babak Mousavi Gourabi
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Marco C DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
11
|
Black N, Bradley J, Schelbert EB, Bonnett LJ, Lewis GA, Lagan J, Orsborne C, Brown PF, Soltani F, Fröjdh F, Ugander M, Wong TC, Fukui M, Cavalcante JL, Naish JH, Williams SG, McDonagh T, Schmitt M, Miller CA. Remote myocardial fibrosis predicts adverse outcome in patients with myocardial infarction on clinical cardiovascular magnetic resonance imaging. J Cardiovasc Magn Reson 2024; 26:101064. [PMID: 39053856 PMCID: PMC11347049 DOI: 10.1016/j.jocmr.2024.101064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Heart failure (HF) most commonly occurs in patients who have had a myocardial infarction (MI), but factors other than MI size may be deterministic. Fibrosis of myocardium remote from the MI is associated with adverse remodeling. We aimed to 1) investigate the association between remote myocardial fibrosis, measured using cardiovascular magnetic resonance (CMR) extracellular volume fraction (ECV), and HF and death following MI, 2) identify predictors of remote myocardial fibrosis in patients with evidence of MI and determine the relationship with infarct size. METHODS Multicenter prospective cohort study of 1199 consecutive patients undergoing CMR with evidence of MI on late gadolinium enhancement. Median follow-up was 1133 (895-1442) days. Cox proportional hazards modeling was used to identify factors predictive of the primary outcome, a composite of first hospitalization for HF (HHF) or all-cause mortality, post-CMR. Linear regression modeling was used to identify determinants of remote ECV. RESULTS Remote myocardial fibrosis was a strong predictor of primary outcome (χ2: 15.6, hazard ratio [HR]: 1.07 per 1% increase in ECV, 95% confidence interval [CI]: 1.04-1.11, p < 0.001) and was separately predictive of both HHF and death. The strongest predictors of remote ECV were diabetes, sex, natriuretic peptides, and body mass index, but, despite extensive phenotyping, the adjusted model R2 was only 0.283. The relationship between infarct size and remote fibrosis was very weak. CONCLUSION Myocardial fibrosis, measured using CMR ECV, is a strong predictor of HHF and death in patients with evidence of MI. The mechanisms underlying remote myocardial fibrosis formation post-MI remain poorly understood, but factors other than infarct size appear to be important.
Collapse
Affiliation(s)
- Nicholas Black
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK
| | - Joshua Bradley
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK
| | - Erik B Schelbert
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, Pennsylvania, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Laura J Bonnett
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Gavin A Lewis
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK; Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| | - Jakub Lagan
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK; Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| | - Christopher Orsborne
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK
| | - Pamela F Brown
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK; South Tees NHS Foundation Trust, Middlesbrough, UK
| | - Fardad Soltani
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK
| | - Fredrika Fröjdh
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden
| | - Martin Ugander
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, Stockholm, Sweden; Kolling Institute, Royal North Shore Hospital, and University of Sydney, Sydney, Australia
| | - Timothy C Wong
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, Pennsylvania, USA
| | - Miho Fukui
- Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Joao L Cavalcante
- Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, Minnesota, USA
| | - Josephine H Naish
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK
| | - Simon G Williams
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Matthias Schmitt
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK
| | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Manchester University NHS Foundation Trust, Manchester, UK; Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
12
|
Warrington JP, Collins HE, Davidge ST, do Carmo JM, Goulopoulou S, Intapad S, Loria AS, Sones JL, Wold LE, Zinkhan EK, Alexander BT. Guidelines for in vivo models of developmental programming of cardiovascular disease risk. Am J Physiol Heart Circ Physiol 2024; 327:H221-H241. [PMID: 38819382 PMCID: PMC11380980 DOI: 10.1152/ajpheart.00060.2024] [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: 01/31/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
Research using animals depends on the generation of offspring for use in experiments or for the maintenance of animal colonies. Although not considered by all, several different factors preceding and during pregnancy, as well as during lactation, can program various characteristics in the offspring. Here, we present the most common models of developmental programming of cardiovascular outcomes, important considerations for study design, and provide guidelines for producing and reporting rigorous and reproducible cardiovascular studies in offspring exposed to normal conditions or developmental insult. These guidelines provide considerations for the selection of the appropriate animal model and factors that should be reported to increase rigor and reproducibility while ensuring transparent reporting of methods and results.
Collapse
Grants
- 20YVNR35490079 American Heart Association (AHA)
- R01HL139348 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL135158 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54GM115428 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01AG057046 HHS | NIH | National Institute on Aging (NIA)
- P20 GM104357 NIGMS NIH HHS
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P30 GM149404 NIGMS NIH HHS
- P20GM104357 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM135002 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01 HL163003 NHLBI NIH HHS
- R01HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01DK121411 HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
- R01HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Excellence Faculty Support Grant Jewish Heritage Fund
- P30GM149404 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P30GM14940 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- 23SFRNPCS1067044 American Heart Association (AHA)
- R01 HL146562 NHLBI NIH HHS
- R56HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U54 GM115428 NIGMS NIH HHS
- 1R01HL163076 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL51971 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- FS154313 CIHR
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Junie P Warrington
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Helen E Collins
- Division of Environmental Medicine, Department of Medicine, Center for Cardiometabolic Science, University of Louisville, Louisville, Kentucky, United States
| | - Sandra T Davidge
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jussara M do Carmo
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Styliani Goulopoulou
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, United States
- Department of Gynecology, and Obstetrics, Loma Linda University, Loma Linda, California, United States
| | - Suttira Intapad
- Department of Pharmacology, Tulane University, New Orleans, Louisiana, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Jenny L Sones
- Equine Reproduction Laboratory, Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, Colorado, United States
| | - Loren E Wold
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Erin K Zinkhan
- Department of Pediatrics, University of Utah and Intermountain Health, Salt Lake City, Utah, United States
- Intermountain Health, Salt Lake City, Utah, United States
| | - Barbara T Alexander
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
13
|
Omoto ACM, Earl CC, Richards AM, Annamalai K, Nelson B, Hall JE, Goergen CJ, da Silva AA. 4D ultrasound-based strain assessment of cardiac dysfunction in male rats with reperfused and nonreperfused myocardial infarction. Physiol Rep 2024; 12:e16159. [PMID: 39039717 PMCID: PMC11263132 DOI: 10.14814/phy2.16159] [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: 03/16/2024] [Revised: 06/04/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
Two-dimensional ultrasound (2DUS) echocardiography is the main noninvasive method used to evaluate cardiac function in animal models of myocardial infarction (MI). However, 2DUS echocardiography does not capture regional differences in cardiac contractility since it relies on planar images to estimate left ventricular (LV) geometry and global function. Thus, the current study was designed to evaluate the efficacy of a newly developed 4-dimensional ultrasound (4DUS) method in detecting cardiac functional differences between two models of MI, permanent ligation (PL), and ischemia/reperfusion (I/R) in rats. We found that only 4DUS was able to detect LV global functional differences between the two models and that 4DUS-derived surface area strain accurately detected infarcted regions within the myocardium that correlated well with histological infarct size analysis. We also found that 4DUS-derived strain, which includes circumferential, longitudinal, and surface area strain, correlated with the peak positive of the first derivative of left ventricular pressure (+dP/dtmax). In conclusion, 4DUS strain echocardiography effectively assesses myocardial mechanics following experimentally induced ischemia in rats and accurately estimates infarct size as early as 1 day after injury. 4DUS also correlates well with +dP/dtmax, a widely used marker of cardiac contractility.
Collapse
Affiliation(s)
- Ana C. M. Omoto
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Conner C. Earl
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
| | - Alyssa M. Richards
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
| | - Karthik Annamalai
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
| | - Benjamin Nelson
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - John E. Hall
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Craig J. Goergen
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
| | - Alexandre A. da Silva
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
14
|
Guevara A, Smith CER, Wang L, Caldwell JL, Tapa S, Stuart SDF, Ma BW, Ng GA, Habecker BA, Wang Z, Ripplinger CM. Sympathetic structural and electrophysiological remodeling in a rabbit model of reperfused myocardial infarction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597494. [PMID: 38895350 PMCID: PMC11185619 DOI: 10.1101/2024.06.05.597494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) inhibit sympathetic reinnervation in rodent hearts post myocardial infarction (MI), causing regional hypo-innervation that is associated with supersensitivity of β-adrenergic receptors and increased arrhythmia susceptibility. To investigate the role of CSPGs and hypo-innervation in the heart of larger mammals, we used a rabbit model of reperfused MI and tested electrophysiological responses to sympathetic nerve stimulation (SNS). Innervated hearts from MI and sham rabbits were optically mapped using voltage and Ca 2+ -sensitive dyes. SNS was performed with electrical stimulation of the spinal cord and β-adrenergic responsiveness was tested using isoproterenol. Sympathetic nerve density and CSPG expression were evaluated using immunohistochemistry. CSPGs were robustly expressed in the infarct and border zone of all MI hearts, and the presence of CSPGs was associated with reduced sympathetic nerve density in the infarct vs. remote region. Action potential duration (APD) dispersion and susceptibility to ventricular tachycardia/fibrillation (VT/VF) were increased with SNS in MI hearts but not in sham. SNS decreased APD 80 in MI but not sham hearts, while isoproterenol decreased APD 80 in both groups. Isoproterenol also shortened Ca 2+ transient duration (CaTD 80 ) in both groups but to a greater extent in MI hearts. Our data suggest sympathetic remodeling post-MI is similar between species, with CSPGs associated with sympathetic hypo-innervation. Despite a reduction in sympathetic nerve density, the infarct region of MI hearts remained responsive to both physiological SNS and isoproterenol, potentially through preserved or elevated β-adrenergic responsiveness, which may underly increased APD dispersion and susceptibility for VT/VF. NEW & NOTEWORTHY Here we show that CSPGs are present in the infarcts of rabbit hearts with reperfused MI, where they are associated with reduced sympathetic nerve density. Despite hypo-innervation, sympathetic responsiveness is maintained or enhanced in MI rabbit hearts, which also demonstrate increased APD dispersion and tendency for arrhythmias following sympathetic modulation. Together, this study indicates that the mechanisms of sympathetic remodeling post-MI are similar between species, with hypo-innervation likely associated with enhanced β-adrenergic sensitivity.
Collapse
|
15
|
Pędzińska-Betiuk A, Schlicker E, Weresa J, Malinowska B. Re-evaluation of the cardioprotective effects of cannabinoids against ischemia-reperfusion injury according to the IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) criteria. Front Pharmacol 2024; 15:1382995. [PMID: 38873412 PMCID: PMC11170160 DOI: 10.3389/fphar.2024.1382995] [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: 02/06/2024] [Accepted: 04/05/2024] [Indexed: 06/15/2024] Open
Abstract
Ischemic heart disease, associated with high morbidity and mortality, represents a major challenge for the development of drug-based strategies to improve its prognosis. Results of pre-clinical studies suggest that agonists of cannabinoid CB2 receptors and multitarget cannabidiol might be potential cardioprotective strategies against ischemia-reperfusion injury. The aim of our study was to re-evaluate the cardioprotective effects of cannabinoids against ischemia-reperfusion injury according to the IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) criteria published recently by the European Union (EU) CARDIOPROTECTION COST ACTION. To meet the minimum criteria of those guidelines, experiments should be performed (i) on healthy small animals subjected to ischemia with reperfusion lasting for at least 2 hours and (ii) confirmed in small animals with comorbidities and co-medications and (iii) in large animals. Our analysis revealed that the publications regarding cardioprotective effects of CB2 receptor agonists and cannabidiol did not meet all three strict steps of IMPACT. Thus, additional experiments are needed to confirm the cardioprotective activities of (endo)cannabinoids mainly on small animals with comorbidities and on large animals. Moreover, our publication underlines the significance of the IMPACT criteria for a proper planning of preclinical experiments regarding cardiac ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Anna Pędzińska-Betiuk
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, Bialystok, Poland
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Jolanta Weresa
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, Bialystok, Poland
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
16
|
Lindsey ML, Kirk JA, LeBlanc AJ, Brunt KR, Carter JR, Hansell Keehan K, Ripplinger CM, Kleinbongard P, Kassiri Z. Looking backward to plan forward. Am J Physiol Heart Circ Physiol 2024; 326:H1155-H1158. [PMID: 38551484 DOI: 10.1152/ajpheart.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/23/2024]
Affiliation(s)
- Merry L Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
- Research Service, Nashville Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois, United States
| | - Amanda J LeBlanc
- Department of Cardiovascular and Thoracic Surgery and Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| | - Jason R Carter
- Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, United States
| | - Kara Hansell Keehan
- American Journal of Physiology-Heart and Circulatory Physiology, American Physiological Society, Rockville, Maryland, United States
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California, United States
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
17
|
Wang H, Zhou T, Ma W, Zheng J, Cao Z, He C, Lemos PA, Luo J. Transcriptome analysis revealed the new mechanism of the intra-myocardial injectable alginate-hydrogel in the treatment of ventricular function degradation. J Thorac Dis 2024; 16:2443-2459. [PMID: 38738236 PMCID: PMC11087630 DOI: 10.21037/jtd-24-358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/10/2024] [Indexed: 05/14/2024]
Abstract
Background Myocardial infarction (MI) is one of the most lethal cardiovascular diseases. The loss of cardiomyocytes and the degradation of the extracellular matrix leads to high ventricular wall stress, which further drives the pathological thinning of the ventricular wall during MI. Injecting biomaterials to thicken the infarct ventricular wall provides mechanical support, thereby inhibiting the continued expansion of the heart. As an injectable biomaterial, alginate hydrogel has achieved exciting results in clinical trials, but further research needs to be conducted to determine whether it can improve cardiac function in addition to providing mechanical support. This study sought to explore these mechanisms in an animal model of MI. Methods A MI model was established in male C57BL/6J mice by ligation of the proximal left anterior descending (LAD) coronary artery. Intramyocardial injections (hydrogel or saline group) were performed in the proximal wall regions bordering the infarct area (with one 20-µL injection). Four weeks after MI, RNA sequencing revealed that 342 messenger RNAs (mRNAs) from the infarcted hearts were differentially expressed between the saline group and hydrogel group. We subsequently conducted a Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis to analyze the RNA sequencing data. In addition, we employed both western blotting and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) techniques to verify a number of genes that were differentially expressed and could potentially affect cardiac function after MI. Subsequently, we confirmed these findings through in vitro experiments. Results We found that compared with hydrogel treatment group, 250 mRNAs were upregulated and 92 mRNAs were downregulated in saline group (P<0.05). And by exploring the GO and KEGG signaling pathways as well as the protein-protein interaction (PPI) network, we found that administration of alginate hydrogel modulated cardiomyocyte inflammation-associated proteins as well as chemokine-related proteins during the inflammatory response phase after MI. In addition, our analysis at both the protein and RNA level revealed that B2M was effective in improving cardiac function after MI in the hydrogel treatment group, which was consistent in the myocardium oxygen and glucose deprivation (OGD) injury model. Conclusions We explored the transcriptome changes of infarcted hearts after alginate-hydrogel injection during the inflammatory response period. Our findings suggest that the injectable hydrogel directly alters the inflammatory response and the chemokine-mediated signaling pathway of cardiomyocytes, ultimately improving cardiac function.
Collapse
Affiliation(s)
- Huiyong Wang
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Zhou
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wentao Ma
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | | | | | - Caiyun He
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pedro A. Lemos
- Department of Cardiology, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
- Department of Cardiology, Heart Institute-InCor, University of Sao Paulo Medical School, Sao Paulo, SP, Brazil
| | - Jianfang Luo
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
18
|
Bryson TD, Bhat SY, Moore C, Taube D, Xu J, Peterson E, Harding P. Targeted Gene Deletion or Antagonism of the Prostaglandin E2 EP3 Receptor Protects Against Cardiac Injury Postmyocardial Infarction. Circ Heart Fail 2024; 17:e011089. [PMID: 38525608 PMCID: PMC11008451 DOI: 10.1161/circheartfailure.123.011089] [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: 07/28/2023] [Accepted: 02/06/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Prostaglandin E2 acts through 4 G-protein-coupled receptors (EP1-EP4). We previously reported that activation of the EP3 receptor reduces cardiac contractility, and its expression increases after a myocardial infarction (MI), mediating the reduction in cardiac function. In contrast, cardiac overexpression of the EP4 receptor in MI substantially improves cardiac function. Moreover, we recently reported that mice overexpressing EP3 have heart failure under basal conditions and worsened cardiac function after MI. Thus, the deleterious effects of the prostaglandin E2 EP receptors in the heart are mediated via its EP3 receptor. We, therefore, hypothesized that cardiomyocyte-specific knockout (CM-EP3 KO) or antagonism of the EP3 receptor protects the heart after MI. METHODS To test our hypothesis, we made the novel CM-EP3 KO mouse and subjected CM-EP3 KO or controls to sham or MI surgery for 2 weeks. In separate experiments, C57BL/6 mice were subjected to 2 weeks of MI and treated with either the EP3 antagonist L798 106 or vehicle starting 3 days post-MI. RESULTS CM-EP3 KO significantly prevented a decline in cardiac function after MI compared with WT animals and prevented an increase in hypertrophy and fibrosis. Excitingly, mice treated with L798 106 3 days after MI had significantly better cardiac function compared with vehicle-treated mice. CONCLUSIONS Altogether, these data suggest that EP3 may play a direct role in regulating cardiac function, and pharmaceutical targeting of the EP3 receptor may be a therapeutic option in the treatment of heart failure.
Collapse
MESH Headings
- Mice
- Animals
- Dinoprostone/metabolism
- Dinoprostone/pharmacology
- Receptors, Prostaglandin/genetics
- Receptors, Prostaglandin/metabolism
- Gene Deletion
- Heart Failure/drug therapy
- Heart Failure/genetics
- Heart Failure/prevention & control
- Mice, Inbred C57BL
- Myocardial Infarction
- Myocytes, Cardiac/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
Collapse
Affiliation(s)
- Timothy D. Bryson
- Hypertension & Vascular Research Division, Department of Internal Medicine (T.D.B., S.B., C.M., D.T., J.X., P.H.), Henry Ford Health, Detroit, MI
| | - Shaheen Y. Bhat
- Hypertension & Vascular Research Division, Department of Internal Medicine (T.D.B., S.B., C.M., D.T., J.X., P.H.), Henry Ford Health, Detroit, MI
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI (S.B., P.H.)
| | - Carlin Moore
- Hypertension & Vascular Research Division, Department of Internal Medicine (T.D.B., S.B., C.M., D.T., J.X., P.H.), Henry Ford Health, Detroit, MI
| | - David Taube
- Hypertension & Vascular Research Division, Department of Internal Medicine (T.D.B., S.B., C.M., D.T., J.X., P.H.), Henry Ford Health, Detroit, MI
| | - Jiang Xu
- Hypertension & Vascular Research Division, Department of Internal Medicine (T.D.B., S.B., C.M., D.T., J.X., P.H.), Henry Ford Health, Detroit, MI
- Department of Cardiovascular Research (J.X.), Henry Ford Health, Detroit, MI
| | - Edward Peterson
- Department of Public Health Sciences (E.P.), Henry Ford Health, Detroit, MI
| | - Pamela Harding
- Hypertension & Vascular Research Division, Department of Internal Medicine (T.D.B., S.B., C.M., D.T., J.X., P.H.), Henry Ford Health, Detroit, MI
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI (S.B., P.H.)
| |
Collapse
|
19
|
Livkisa D, Chang TH, Burnouf T, Czosseck A, Le NTN, Shamrin G, Yeh WT, Kamimura M, Lundy DJ. Extracellular vesicles purified from serum-converted human platelet lysates offer strong protection after cardiac ischaemia/reperfusion injury. Biomaterials 2024; 306:122502. [PMID: 38354518 DOI: 10.1016/j.biomaterials.2024.122502] [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: 09/27/2023] [Revised: 01/06/2024] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Extracellular vesicles (EVs) from cultured cells or bodily fluids have been demonstrated to show therapeutic value following myocardial infarction. However, challenges in donor variation, EV generation and isolation methods, and material availability have hindered their therapeutic use. Here, we show that human clinical-grade platelet concentrates from a blood establishment can be used to rapidly generate high concentrations of high purity EVs from sero-converted platelet lysate (SCPL-EVs) with minimal processing, using size-exclusion chromatography. Processing removed serum carrier proteins, coagulation factors and complement proteins from the original platelet lysate and the resultant SCPL-EVs carried a range of trophic factors and multiple recognised cardioprotective miRNAs. As such, SCPL-EVs protected rodent and human cardiomyocytes from hypoxia/re-oxygenation injury and stimulated angiogenesis of human cardiac microvessel endothelial cells. In a mouse model of myocardial infarction with reperfusion, SCPL-EV delivery using echo-guided intracavitary percutaneous injection produced large improvements in cardiac function, reduced scar formation and promoted angiogenesis. Since platelet-based biomaterials are already widely used clinically, we believe that this therapy could be rapidly suitable for a human clinical trial.
Collapse
Affiliation(s)
- Dora Livkisa
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Hsin Chang
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Thierry Burnouf
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; International Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Andreas Czosseck
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Nhi Thao Ngoc Le
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Gleb Shamrin
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Ting Yeh
- School of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Masao Kamimura
- Department of Medical and Robotic Engineering Design, Faculty of Advanced Engineering, Tokyo University of Science, Japan
| | - David J Lundy
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Center for Cell Therapy, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
20
|
Jimenez J, Amrute J, Ma P, Wang X, Dai R, Lavine KJ. CD40 is an immune checkpoint regulator that potentiates myocardial inflammation through activation and expansion of CCR2 + macrophages and CD8 T-cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.584418. [PMID: 38559055 PMCID: PMC10980053 DOI: 10.1101/2024.03.14.584418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Novel immune checkpoint therapeutics including CD40 agonists have tremendous promise to elicit antitumor responses in patients resistant to current therapies. Conventional immune checkpoint inhibitors (PD-1/PD-L1, CTLA-4 antagonists) are associated with serious adverse cardiac events including life-threatening myocarditis. However, little is known regarding the potential for CD40 agonists to trigger myocardial inflammation or myocarditis. Here, we leveraged genetic mouse models, single cell sequencing, and cell depletion studies to demonstrate that an anti-CD40 agonist antibody reshapes the cardiac immune landscape through activation of CCR2 + macrophages and subsequent recruitment of effector memory CD8 T-cells. We identify a positive feedback loop between CCR2 + macrophages and CD8 T-cells driven by IL12b, TNF, and IFN-γ signaling that promotes myocardial inflammation and show that prior exposure to CD40 agonists sensitizes the heart to secondary insults and accelerates LV remodeling. Collectively, these findings highlight the potential for CD40 agonists to promote myocardial inflammation and potentiate heart failure pathogenesis.
Collapse
|
21
|
Tjandra PM, Ripplinger CM, Christiansen BA. The heart-bone connection: relationships between myocardial infarction and osteoporotic fracture. Am J Physiol Heart Circ Physiol 2024; 326:H845-H856. [PMID: 38305753 PMCID: PMC11062618 DOI: 10.1152/ajpheart.00576.2023] [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: 09/18/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Myocardial infarction (MI) and osteoporotic fracture (Fx) are two of the leading causes of mortality and morbidity worldwide. Although these traumatic injuries are treated as if they are independent, there is epidemiological evidence linking the incidence of Fx and MI, thus raising the question of whether each of these events can actively influence the risk of the other. Atherosclerotic cardiovascular disease and osteoporosis, the chronic conditions leading to MI and Fx, are known to have shared pathoetiology. Furthermore, sustained systemic inflammation after traumas such as MI and Fx has been shown to exacerbate both underlying chronic conditions. However, the effects of MI and Fx outside their own system have not been well studied. The sympathetic nervous system (SNS) and the complement system initiate a systemic response after MI that could lead to subsequent changes in bone remodeling through osteoclasts. Similarly, SNS and complement system activation following fracture could lead to heart tissue damage and exacerbate atherosclerosis. To determine whether damaging bone-heart cross talk may be important comorbidity following Fx or MI, this review details the current understanding of bone loss after MI, cardiovascular damage after Fx, and possible shared underlying mechanisms of these processes.
Collapse
Affiliation(s)
- Priscilla M Tjandra
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, United States
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
| | - Crystal M Ripplinger
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
- Department of Pharmacology, University of California Davis Health, Davis, California, United States
| | - Blaine A Christiansen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, California, United States
| |
Collapse
|
22
|
Jung SE, Kim SW, Choi JW. Exploring Cardiac Exosomal RNAs of Acute Myocardial Infarction. Biomedicines 2024; 12:430. [PMID: 38398032 PMCID: PMC10886708 DOI: 10.3390/biomedicines12020430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI), often a frequent symptom of coronary artery disease (CAD), is a leading cause of death and disability worldwide. Acute myocardial infarction (AMI), a major form of cardiovascular disease, necessitates a deep understanding of its complex pathophysiology to develop innovative therapeutic strategies. Exosomal RNAs (exoRNA), particularly microRNAs (miRNAs) within cardiac tissues, play a critical role in intercellular communication and pathophysiological processes of AMI. METHODS This study aimed to delineate the exoRNA landscape, focusing especially on miRNAs in animal models using high-throughput sequencing. The approach included sequencing analysis to identify significant miRNAs in AMI, followed by validation of the functions of selected miRNAs through in vitro studies involving primary cardiomyocytes and fibroblasts. RESULTS Numerous differentially expressed miRNAs in AMI were identified using five mice per group. The functions of 20 selected miRNAs were validated through in vitro studies with primary cardiomyocytes and fibroblasts. CONCLUSIONS This research enhances understanding of post-AMI molecular changes in cardiac tissues and investigates the potential of exoRNAs as biomarkers or therapeutic targets. These findings offer new insights into the molecular mechanisms of AMIs, paving the way for RNA-based diagnostics and therapeutics and therapies and contributing to the advancement of cardiovascular medicine.
Collapse
Affiliation(s)
- Seung Eun Jung
- Medical Science Research Institute, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Sang Woo Kim
- International St. Mary's Hospital, Incheon 22711, Republic of Korea
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Jung-Won Choi
- Medical Science Research Institute, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| |
Collapse
|
23
|
Najjar RS, Roy RK, Stern JE, Feresin RG. Raspberry polyphenols target molecular pathways of heart failure. J Nutr Biochem 2024; 124:109535. [PMID: 37984734 DOI: 10.1016/j.jnutbio.2023.109535] [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: 06/01/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Approximately 650,000 new cases of heart failure (HF) are diagnosed annually with a 50% five-year mortality rate. HF is characterized by reduced left ventricular (LV) ejection fraction and hypertrophy of the LV wall. The pathophysiological remodeling of the heart is mediated by increased oxidative stress and inflammation. Raspberries are rich in polyphenols which may favorably impact enzymes involved in redox homeostasis while also targeting inflammatory signaling. Thus, the objective of this study was to investigate whether raspberry polyphenols could attenuate HF. Sprague Dawley rats consumed a 10% (w/w) raspberry diet for 7 weeks. At week 3, HF was surgically induced via coronary artery ligation. Hemodynamics and morphology of the heart were assessed. Expression of cardiac proteins involved in oxidative stress, inflammation, apoptosis, and remodeling were examined, and histological analysis was conducted. Additionally, human cardiomyocytes were treated with raspberry polyphenol extract (RBPE) followed by CoCl2 to chemically induce hypoxia. Redox status, apoptosis, and mitochondrial dysfunction were measured. Raspberries attenuated reductions in cardiac function and reduced morphological changes which coincided with reduced toll-like receptor (TLR)4 signaling. Reductions in oxidative stress, apoptosis, and remodeling occurred in vivo. Incubation of cardiomyocytes with RBPE attenuated CoCl2-induced oxidative stress and apoptosis despite pronounced hypoxia-inducible factor (HIF)-1α expression. These data indicate that consumption of raspberries can reduce the underlying molecular drivers of HF; thus, leading to the observed improvements in cardiac functional capacity and morphology. This dietary strategy may be an effective alternative strategy for treating HF. However, further investigation into alternative models of HF is warranted.
Collapse
Affiliation(s)
- Rami S Najjar
- Department of Nutrition, Georgia State University, Atlanta, Georgia, USA; Department of Chemistry, Georgia State University, Atlanta, Georgia, USA
| | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA
| | - Rafaela G Feresin
- Department of Nutrition, Georgia State University, Atlanta, Georgia, USA; Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
24
|
Barrett MS, Bauer TC, Li MH, Hegarty DM, Mota CMD, Amaefuna CJ, Ingram SL, Habecker BA, Aicher SA. Ischemia-reperfusion myocardial infarction induces remodeling of left cardiac-projecting stellate ganglia neurons. Am J Physiol Heart Circ Physiol 2024; 326:H166-H179. [PMID: 37947434 PMCID: PMC11213476 DOI: 10.1152/ajpheart.00582.2023] [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: 09/19/2023] [Revised: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023]
Abstract
Neurons in the stellate ganglion (SG) provide sympathetic innervation to the heart, brown adipose tissue (BAT), and other organs. Sympathetic innervation to the heart becomes hyperactive following myocardial infarction (MI). The impact of MI on the morphology of cardiac sympathetic neurons is not known, but we hypothesized that MI would stimulate increased cell and dendritic tree size in cardiac neurons. In this study, we examined the effects of ischemia-reperfusion MI on sympathetic neurons using dual retrograde tracing methods to allow detailed characterization of cardiac- and BAT-projecting neurons. Different fluorescently conjugated cholera toxin subunit B (CTb) tracers were injected into the pericardium and the interscapular BAT pads, respectively. Experimental animals received a 45-min occlusion of the left anterior descending coronary artery and controls received sham surgery. One week later, hearts were collected for assessment of MI infarct and SGs were collected for morphological or electrophysiological analysis. Cardiac-projecting SG neurons from MI mice had smaller cell bodies and shorter dendritic trees compared with sham animals, specifically on the left side ipsilateral to the MI. BAT-projecting neurons were not altered by MI, demonstrating the subpopulation specificity of the response. The normal size and distribution differences between BAT- and cardiac-projecting stellate ganglion neurons were not altered by MI. Patch-clamp recordings from cardiac-projecting left SG neurons revealed increased spontaneous excitatory postsynaptic currents despite the decrease in cell and dendritic tree size. Thus, increased dendritic tree size does not contribute to the enhanced sympathetic neural activity seen after MI.NEW & NOTEWORTHY Myocardial infarction (MI) causes structural and functional changes specifically in stellate ganglion neurons that project to the heart, but not in cells that project to brown adipose fat tissue.
Collapse
Affiliation(s)
- Madeleine S Barrett
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Temerity C Bauer
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Ming-Hua Li
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Deborah M Hegarty
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Clarissa M D Mota
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Chimezie J Amaefuna
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Susan L Ingram
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Sue A Aicher
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
25
|
Zhao X, Li D, Song Y, Xu J, Xiang FL. Drug Discovery for Adult Cardiomyocyte Regeneration: Opportunities and Challenges. Antioxid Redox Signal 2023; 39:1070-1087. [PMID: 37166381 DOI: 10.1089/ars.2023.0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Significance: Cardiovascular disease is a major contributor to human mortality and morbidity. The cardiac tissue undergoes fibrotic healing after injury because of the limited regenerative capacity of adult mammalian cardiomyocyte (CM). Extensive research has been performed to identify therapeutic targets for CM regeneration, as the success of promoting adult human CM regeneration to repair the injured heart is considered the Holy Grail in the field. Recent Advances: To date, more than 30 target genes have been shown to regulate adult mammalian CM proliferation. More than 20 targets have been validated in adult mouse myocardial infarction (MI) model in a therapeutic setting. In this review, the translational efficacy readouts from 17 selected pharmaceutical targets are summarized, among which the Hippo-yes-associated protein (Yap) pathway is the most extensively investigated and fits the criteria for a promising target for pro-CM-regeneration therapy development. Critical Issues and Future Directions: As the pro-CM-regeneration potential of current drug treatment for cardiovascular patients is limited, to help identify and fill the gap between basic research and drug discovery in this specific field, details regarding target identification, validation in mouse MI models, high-throughput screening assay development, and preclinical in vivo efficacy model optimization are discussed. Finally, suggestions and recommendations are also provided to help establish a common guideline for in vivo translational studies for drug discovery focusing on CM regeneration. Antioxid. Redox Signal. 39, 1070-1087.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Anesthesiology and the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Donghua Li
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yiyan Song
- Department of Anesthesiology and the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Xu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fu-Li Xiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
26
|
Venegas-Zamora L, Fiedler M, Perez W, Altamirano F. Bridging the Translational Gap in Heart Failure Research: Using Human iPSC-derived Cardiomyocytes to Accelerate Therapeutic Insights. Methodist Debakey Cardiovasc J 2023; 19:5-15. [PMID: 38028973 PMCID: PMC10655754 DOI: 10.14797/mdcvj.1295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/04/2023] [Indexed: 12/01/2023] Open
Abstract
Heart failure (HF) remains a leading cause of death worldwide, with increasing prevalence and burden. Despite extensive research, a cure for HF remains elusive. Traditionally, the study of HF's pathogenesis and therapies has relied heavily on animal experimentation. However, these models have limitations in recapitulating the full spectrum of human HF, resulting in challenges for clinical translation. To address this translational gap, research employing human cells, especially cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs), offers a promising solution. These cells facilitate the study of human genetic and molecular mechanisms driving cardiomyocyte dysfunction and pave the way for research tailored to individual patients. Further, engineered heart tissues combine hiPSC-CMs, other cell types, and scaffold-based approaches to improve cardiomyocyte maturation. Their tridimensional architecture, complemented with mechanical, chemical, and electrical cues, offers a more physiologically relevant environment. This review explores the advantages and limitations of conventional and innovative methods used to study HF pathogenesis, with a primary focus on ischemic HF due to its relative ease of modeling and clinical relevance. We emphasize the importance of a collaborative approach that integrates insights obtained in animal and hiPSC-CMs-based models, along with rigorous clinical research, to dissect the mechanistic underpinnings of human HF. Such an approach could improve our understanding of this disease and lead to more effective treatments.
Collapse
Affiliation(s)
- Leslye Venegas-Zamora
- Houston Methodist Research Institute, Houston, Texas, US
- Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Matthew Fiedler
- Houston Methodist Research Institute, Houston, Texas, US
- Weill Cornell Graduate School of Medical Sciences, New York, New York, US
| | - William Perez
- Houston Methodist Research Institute, Houston, Texas, US
| | - Francisco Altamirano
- Houston Methodist Research Institute, Houston, Texas, US
- Weill Cornell Medical College, New York, New York, US
| |
Collapse
|
27
|
Francisco J, Del Re DP. Inflammation in Myocardial Ischemia/Reperfusion Injury: Underlying Mechanisms and Therapeutic Potential. Antioxidants (Basel) 2023; 12:1944. [PMID: 38001797 PMCID: PMC10669026 DOI: 10.3390/antiox12111944] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Acute myocardial infarction (MI) occurs when blood flow to the myocardium is restricted, leading to cardiac damage and massive loss of viable cardiomyocytes. Timely restoration of coronary flow is considered the gold standard treatment for MI patients and limits infarct size; however, this intervention, known as reperfusion, initiates a complex pathological process that somewhat paradoxically also contributes to cardiac injury. Despite being a sterile environment, ischemia/reperfusion (I/R) injury triggers inflammation, which contributes to infarct expansion and subsequent cardiac remodeling and wound healing. The immune response is comprised of subsets of both myeloid and lymphoid-derived cells that act in concert to modulate the pathogenesis and resolution of I/R injury. Multiple mechanisms, including altered metabolic status, regulate immune cell activation and function in the setting of acute MI, yet our understanding remains incomplete. While numerous studies demonstrated cardiac benefit following strategies that target inflammation in preclinical models, therapeutic attempts to mitigate I/R injury in patients were less successful. Therefore, further investigation leveraging emerging technologies is needed to better characterize this intricate inflammatory response and elucidate its influence on cardiac injury and the progression to heart failure.
Collapse
Affiliation(s)
| | - Dominic P. Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
28
|
Schunke KJ, Rodriguez J, Dyavanapalli J, Schloen J, Wang X, Escobar J, Kowalik G, Cheung EC, Ribeiro C, Russo R, Alber BR, Dergacheva O, Chen SW, Murillo-Berlioz AE, Lee KB, Trachiotis G, Entcheva E, Brantner CA, Mendelowitz D, Kay MW. Outcomes of hypothalamic oxytocin neuron-driven cardioprotection after acute myocardial infarction. Basic Res Cardiol 2023; 118:43. [PMID: 37801130 PMCID: PMC10558415 DOI: 10.1007/s00395-023-01013-1] [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: 06/15/2022] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023]
Abstract
Altered autonomic balance is a hallmark of numerous cardiovascular diseases, including myocardial infarction (MI). Although device-based vagal stimulation is cardioprotective during chronic disease, a non-invasive approach to selectively stimulate the cardiac parasympathetic system immediately after an infarction does not exist and is desperately needed. Cardiac vagal neurons (CVNs) in the brainstem receive powerful excitation from a population of neurons in the paraventricular nucleus (PVN) of the hypothalamus that co-release oxytocin (OXT) and glutamate to excite CVNs. We tested if chemogenetic activation of PVN-OXT neurons following MI would be cardioprotective. The PVN of neonatal rats was transfected with vectors to selectively express DREADDs within OXT neurons. At 6 weeks of age, an MI was induced and DREADDs were activated with clozapine-N-oxide. Seven days following MI, patch-clamp electrophysiology confirmed the augmented excitatory neurotransmission from PVN-OXT neurons to downstream nuclei critical for parasympathetic activity with treatment (43.7 ± 10 vs 86.9 ± 9 pA; MI vs. treatment), resulting in stark improvements in survival (85% vs. 95%; MI vs. treatment), inflammation, fibrosis assessed by trichrome blue staining, mitochondrial function assessed by Seahorse assays, and reduced incidence of arrhythmias (50% vs. 10% cumulative incidence of ventricular fibrillation; MI vs. treatment). Myocardial transcriptomic analysis provided molecular insight into potential cardioprotective mechanisms, which revealed the preservation of beneficial signaling pathways, including muscarinic receptor activation, in treated animals. These comprehensive results demonstrate that the PVN-OXT network could be a promising therapeutic target to quickly activate beneficial parasympathetic-mediated cellular pathways within the heart during the early stages of infarction.
Collapse
Affiliation(s)
- Kathryn J Schunke
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA.
- Department of Anatomy, Biochemistry and Physiology, University of Hawaii, 651 Ilalo St, Honolulu, HI, BSB 211 96813, USA.
| | - Jeannette Rodriguez
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - John Schloen
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Xin Wang
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Joan Escobar
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Grant Kowalik
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Emily C Cheung
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Caitlin Ribeiro
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Rebekah Russo
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Bridget R Alber
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Sheena W Chen
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Alejandro E Murillo-Berlioz
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Kyongjune B Lee
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Gregory Trachiotis
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Christine A Brantner
- The GWU Nanofabrication and Imaging Center, 800 22nd Street NW, Washington, DC, 20052, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA.
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA.
| |
Collapse
|
29
|
Wang Y, Chen W, Wang Q. Segmental and transmural motion of the rat myocardium estimated using quantitative ultrasound with new strategies for infarct detection. Front Bioeng Biotechnol 2023; 11:1236108. [PMID: 37744251 PMCID: PMC10512837 DOI: 10.3389/fbioe.2023.1236108] [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: 06/07/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction: The estimation of myocardial motion abnormalities has great potential for the early diagnosis of myocardial infarction (MI). This study aims to quantitatively analyze the segmental and transmural myocardial motion in MI rats by incorporating two novel strategies of algorithm parameter optimization and transmural motion index (TMI) calculation. Methods: Twenty-one rats were randomly divided into three groups (n = 7 per group): sham, MI, and ischemia-reperfusion (IR) groups. Ultrasound radio-frequency (RF) signals were acquired from each rat heart at 1 day and 28 days after animal model establishment; thus, a total of six datasets were represented as Sham1, Sham28, MI1, MI28, IR1, and IR28. The systolic cumulative displacement was calculated using our previously proposed vectorized normalized cross-correlation (VNCC) method. A semiautomatic regional and layer-specific myocardium segmentation framework was proposed for transmural and segmental myocardial motion estimation. Two novel strategies were proposed: the displacement-compensated cross-correlation coefficient (DCCCC) for algorithm parameter optimization and the transmural motion index (TMI) for quantitative estimation of the cross-wall transmural motion gradient. Results: The results showed that an overlap value of 80% used in VNCC guaranteed a more accurate displacement calculation. Compared to the Sham1 group, the systolic myocardial motion reductions were significantly detected (p < 0.05) in the middle anteroseptal (M-ANT-SEP), basal anteroseptal (B-ANT-SEP), apical lateral (A-LAT), middle inferolateral (M-INF-LAT), and basal inferolateral (B-INF-LAT) walls as well as a significant TMI drop (p < 0.05) in the M-ANT-SEP wall in the MI1 rats; significant motion reductions (p < 0.05) were also detected in the B-ANT-SEP and A-LAT walls in the IR1 group. The motion improvements (p < 0.05) were detected in the M-INF-LAT wall in the MI28 group and the apical septal (A-SEP) wall in the IR28 group compared to the MI1 and IR1 groups, respectively. Discussion: Our results show that the MI-induced reductions and reperfusion-induced recovery in systolic myocardial contractility could be successfully evaluated using our method, and most post-MI myocardial segments could recover systolic function to various extents in the remodeling phase. In conclusion, the ultrasound-based quantitative estimation framework for estimating segmental and transmural motion of the myocardium proposed in our study has great potential for non-invasive, novel, and early MI detection.
Collapse
Affiliation(s)
- Yinong Wang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, China
| | - Wufan Chen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, China
| | - Qing Wang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Zhuang L, Zong X, Yang Q, Fan Q, Tao R. Interleukin-34-NF-κB signaling aggravates myocardial ischemic/reperfusion injury by facilitating macrophage recruitment and polarization. EBioMedicine 2023; 95:104744. [PMID: 37556943 PMCID: PMC10433018 DOI: 10.1016/j.ebiom.2023.104744] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Macrophage infiltration and polarization are integral to the progression of heart failure and cardiac fibrosis after ischemia/reperfusion (IR). Interleukin 34 (IL-34) is an inflammatory regulator related to a series of autoimmune diseases. Whether IL-34 mediates inflammatory responses and contributes to cardiac remodeling and heart failure post-IR remains unclear. METHODS IL-34 knock-out mice were used to determine the role of IL-34 on cardiac remodeling after IR surgery. Then, immunofluorescence, flow cytometry assays, and RNA-seq analysis were performed to explore the underlying mechanisms of IL-34-induced macrophage recruitment and polarization, and further heart failure after IR. FINDINGS By re-analyzing single-cell RNA-seq and single-nucleus RNA-seq data of murine and human ischemic hearts, we showed that IL-34 expression was upregulated after IR. IL-34 knockout mitigated cardiac remodeling, cardiac dysfunction, and fibrosis after IR and vice versa. RNA-seq analysis revealed that IL-34 deletion correlated negatively with immune responses and chemotaxis after IR injury. Consistently, immunofluorescence and flow cytometry assays demonstrated that IL-34 deletion attenuated macrophage recruitment and CCR2+ macrophage polarization. Mechanistically, IL-34 deficiency repressed both the canonical and noncanonical NF-κB signaling pathway, leading to marked reduction of P-IKKβ and P-IκBα kinase levels; downregulation of NF-κB p65, RelB, and p52 expression, which drove the decline in chemokine CCL2 expression. Finally, IL-34 and CCL2 levels were increased in the serum of acute coronary syndrome patients, with a positive correlation between circulating IL-34 and CCL2 levels in clinical patients. INTERPRETATION In conclusion, IL-34 sustains NF-κB pathway activation to elicit increased CCL2 expression, which contributes to macrophage recruitment and polarization, and subsequently exacerbates cardiac remodeling and heart failure post-IR. Strategies targeting IL-34-centered immunomodulation may provide new therapeutic approaches to prevent and reverse cardiac remodeling and heart failure in clinical MI patients after percutaneous coronary intervention. FUNDING This study was supported by the National Nature Science Foundation of China (81670352 and 81970327 to R T, 82000368 to Q F).
Collapse
Affiliation(s)
- Lingfang Zhuang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xiao Zong
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qian Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qin Fan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Rong Tao
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
31
|
De Rosa L, L’Abbate S, Kusmic C, Faita F. Applications of Deep Learning Algorithms to Ultrasound Imaging Analysis in Preclinical Studies on In Vivo Animals. Life (Basel) 2023; 13:1759. [PMID: 37629616 PMCID: PMC10455134 DOI: 10.3390/life13081759] [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: 06/13/2023] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND AND AIM Ultrasound (US) imaging is increasingly preferred over other more invasive modalities in preclinical studies using animal models. However, this technique has some limitations, mainly related to operator dependence. To overcome some of the current drawbacks, sophisticated data processing models are proposed, in particular artificial intelligence models based on deep learning (DL) networks. This systematic review aims to overview the application of DL algorithms in assisting US analysis of images acquired in in vivo preclinical studies on animal models. METHODS A literature search was conducted using the Scopus and PubMed databases. Studies published from January 2012 to November 2022 that developed DL models on US images acquired in preclinical/animal experimental scenarios were eligible for inclusion. This review was conducted according to PRISMA guidelines. RESULTS Fifty-six studies were enrolled and classified into five groups based on the anatomical district in which the DL models were used. Sixteen studies focused on the cardiovascular system and fourteen on the abdominal organs. Five studies applied DL networks to images of the musculoskeletal system and eight investigations involved the brain. Thirteen papers, grouped under a miscellaneous category, proposed heterogeneous applications adopting DL systems. Our analysis also highlighted that murine models were the most common animals used in in vivo studies applying DL to US imaging. CONCLUSION DL techniques show great potential in terms of US images acquired in preclinical studies using animal models. However, in this scenario, these techniques are still in their early stages, and there is room for improvement, such as sample sizes, data preprocessing, and model interpretability.
Collapse
Affiliation(s)
- Laura De Rosa
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (L.D.R.); (F.F.)
- Department of Information Engineering and Computer Science, University of Trento, 38123 Trento, Italy
| | - Serena L’Abbate
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy;
| | - Claudia Kusmic
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (L.D.R.); (F.F.)
| | - Francesco Faita
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (L.D.R.); (F.F.)
| |
Collapse
|
32
|
Lindsey ML, Kassiri Z, LeBlanc AJ, Ripplinger CM, Kirk JA, Carter JR, Kleinbongard P, Brunt KR. Spring cleaning: freshening up the portfolio. Am J Physiol Heart Circ Physiol 2023; 324:H840-H842. [PMID: 37115630 PMCID: PMC10190828 DOI: 10.1152/ajpheart.00219.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 04/29/2023]
Affiliation(s)
- Merry L Lindsey
- School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
- Research Service, Nashville Veterans Affairs Medical Center, Nashville, Tennessee, United States
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| | - Amanda J LeBlanc
- Department of Cardiovascular and Thoracic Surgery and Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States
| | - Crystal M Ripplinger
- Department of Pharmacology, UC Davis School of Medicine, Davis, California, United States
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois, United States
| | - Jason R Carter
- Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, United States
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Keith R Brunt
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| |
Collapse
|
33
|
Grilo GA, Cakir SN, Shaver PR, Iyer RP, Whitehead K, McClung JM, Vahdati A, de Castro Brás LE. Collagen matricryptin promotes cardiac function by mediating scar formation. Life Sci 2023; 321:121598. [PMID: 36963720 PMCID: PMC10120348 DOI: 10.1016/j.lfs.2023.121598] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023]
Abstract
AIMS A peptide mimetic of a collagen-derived matricryptin (p1159) was shown to reduce left ventricular (LV) dilation and fibrosis after 7 days delivery in a mouse model of myocardial infarction (MI). This suggested p1159 long-term treatment post-MI could have beneficial effects and reduce/prevent adverse LV remodeling. This study aimed to test the potential of p1159 to reduce adverse cardiac remodeling in a chronic MI model and to elucidate p1159 mode-of-action. MATERIALS AND METHODS Using a permanent occlusion MI rodent model, animals received p1159 or vehicle solution up to 28 days. We assessed peptide treatment effects on scar composition and structure and on systolic function. To assess peptide effects on scar vascularization, a cohort of mice were injected with Griffonia simplicifolia isolectin-B4. To investigate p1159 mode-of-action, LV fibroblasts from naïve animals were treated with increasing doses of p1159. KEY FINDINGS Matricryptin p1159 significantly improved systolic function post-MI (2-fold greater EF compared to controls) by reducing left ventricular dilation and inducing the formation of a compliant and organized infarct scar, which promoted LV contractility and preserved the structural integrity of the heart. Specifically, infarcted scars from p1159-treated animals displayed collagen fibers aligned parallel to the epicardium, to resist circumferential stretching, with reduced levels of cross-linking, and improved tissue perfusion. In addition, we found that p1159 increases cardiac fibroblast migration by activating RhoA pathways via the membrane receptor integrin α4. SIGNIFICANCE Our data indicate p1159 treatment reduced adverse LV remodeling post-MI by modulating the deposition, arrangement, and perfusion of the fibrotic scar.
Collapse
Affiliation(s)
- Gabriel A Grilo
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America
| | - Sirin N Cakir
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America
| | - Patti R Shaver
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America
| | - Rugmani P Iyer
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America
| | - Kaitlin Whitehead
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America
| | - Joseph M McClung
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America; Department of Cardiovascular Sciences, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America; East Carolina Diabetes and Obesity Institute, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America
| | - Ali Vahdati
- Department of Engineering, East Carolina University, Greenville, NC 27858, United States of America
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America; Department of Cardiovascular Sciences, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States of America.
| |
Collapse
|
34
|
Pistritu DV, Vasiliniuc AC, Vasiliu A, Visinescu EF, Visoiu IE, Vizdei S, Martínez Anghel P, Tanca A, Bucur O, Liehn EA. Phospholipids, the Masters in the Shadows during Healing after Acute Myocardial Infarction. Int J Mol Sci 2023; 24:8360. [PMID: 37176067 PMCID: PMC10178977 DOI: 10.3390/ijms24098360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Phospholipids are major components of cell membranes with complex structures, high heterogeneity and critical biological functions and have been used since ancient times to treat cardiovascular disease. Their importance and role were shadowed by the difficulty or incomplete available research methodology to study their biological presence and functionality. This review focuses on the current knowledge about the roles of phospholipids in the pathophysiology and therapy of cardiovascular diseases, which have been increasingly recognized. Used in singular formulation or in inclusive combinations with current drugs, phospholipids proved their positive and valuable effects not only in the protection of myocardial tissue, inflammation and fibrosis but also in angiogenesis, coagulation or cardiac regeneration more frequently in animal models as well as in human pathology. Thus, while mainly neglected by the scientific community, phospholipids present negligible side effects and could represent an ideal target for future therapeutic strategies in healing myocardial infarction. Acknowledging and understanding their mechanisms of action could offer a new perspective into novel therapeutic strategies for patients suffering an acute myocardial infarction, reducing the burden and improving the general social and economic outcome.
Collapse
Affiliation(s)
- Dan-Valentin Pistritu
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
| | | | - Anda Vasiliu
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
| | - Elena-Florentina Visinescu
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Ioana-Elena Visoiu
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Smaranda Vizdei
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Paula Martínez Anghel
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Business Academy Aarhus, 30 Sønderhøj, 8260 Viby J, Denmark
| | - Antoanela Tanca
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Octavian Bucur
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Viron Molecular Medicine Institute, 201 Washington Street, Boston, MA 02108, USA
| | - Elisa Anamaria Liehn
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Institute for Molecular Medicine, University of Southern Denmark, 25 J.B Winsløws Vej, 5230 Odense, Denmark
- National Heart Center Singapore, 5 Hospital Dr., Singapore 169609, Singapore
| |
Collapse
|
35
|
Mouton AJ, Aitken NM, Moak SP, do Carmo JM, da Silva AA, Omoto ACM, Li X, Wang Z, Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA, Hall JE. Temporal changes in glucose metabolism reflect polarization in resident and monocyte-derived macrophages after myocardial infarction. Front Cardiovasc Med 2023; 10:1136252. [PMID: 37215542 PMCID: PMC10196495 DOI: 10.3389/fcvm.2023.1136252] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/28/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction Metabolic reprogramming from glycolysis to the mitochondrial tricarboxylic acid (TCA) cycle and oxidative phosphorylation may mediate macrophage polarization from the pro-inflammatory M1 to the anti-inflammatory M2 phenotype. We hypothesized that changes in cardiac macrophage glucose metabolism would reflect polarization status after myocardial infarction (MI), ranging from the early inflammatory phase to the later wound healing phase. Methods MI was induced by permanent ligation of the left coronary artery in adult male C57BL/6J mice for 1 (D1), 3 (D3), or 7 (D7) days. Infarct macrophages were subjected to metabolic flux analysis or gene expression analysis. Monocyte versus resident cardiac macrophage metabolism was assessed using mice lacking the Ccr2 gene (CCR2 KO). Results By flow cytometry and RT-PCR, D1 macrophages exhibited an M1 phenotype while D7 macrophages exhibited an M2 phenotype. Macrophage glycolysis (extracellular acidification rate) was increased at D1 and D3, returning to basal levels at D7. Glucose oxidation (oxygen consumption rate) was decreased at D3, returning to basal levels at D7. At D1, glycolytic genes were elevated (Gapdh, Ldha, Pkm2), while TCA cycle genes were elevated at D3 (Idh1 and Idh2) and D7 (Pdha1, Idh1/2, Sdha/b). Surprisingly, Slc2a1 and Hk1/2 were increased at D7, as well as pentose phosphate pathway (PPP) genes (G6pdx, G6pd2, Pgd, Rpia, Taldo1), indicating increased PPP activity. Macrophages from CCR2 KO mice showed decreased glycolysis and increased glucose oxidation at D3, and decreases in Ldha and Pkm2 expression. Administration of dichloroacetate, a pyruvate dehydrogenase kinase inhibitor, robustly decreased pyruvate dehydrogenase phosphorylation in the non-infarcted remote zone, but did not affect macrophage phenotype or metabolism in the infarct zone. Discussion Our results indicate that changes in glucose metabolism and the PPP underlie macrophage polarization following MI, and that metabolic reprogramming is a key feature of monocyte-derived but not resident macrophages.
Collapse
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Nikaela M. Aitken
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
| | - Sydney P. Moak
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
| | - Jussara M. do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Alexandre A. da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Ana C. M. Omoto
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | - Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| | | | - Simona G. Codreanu
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States
| | - Stacy D. Sherrod
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States
| | - John A. McLean
- Department of Chemistry and Center for Innovative Technology, Vanderbilt University, Nashville, TN, United States
| | - John E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, United States
| |
Collapse
|
36
|
Halade GV, Mat Y, Gowda SGB, Jain S, Hui S, Yadav H, Kain V. Sleep deprivation in obesogenic setting alters lipidome and microbiome toward suboptimal inflammation in acute heart failure. FASEB J 2023; 37:e22899. [PMID: 37002889 DOI: 10.1096/fj.202300184r] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
Sleep is a fundamental medicine for cardiac homeostasis, and sleep-deprived individuals are prone to higher incidences of heart attack. The lipid-dense diet (obesogenic diet-OBD) is a cumulative risk factor for chronic inflammation in cardiovascular disease; thus, understanding how sleep fragmentation (SF) in an obesity setting impacts immune and cardiac health is an unmet medical need. We hypothesized whether the co-existence of SF with OBD dysregulates gut homeostasis and leukocyte-derived reparative/resolution mediators, thereby impairing cardiac repair. Two-month-old male C57BL/6J mice were randomized first into two groups, then four groups; Control, control + SF, OBD, and OBD + SF mice subjected to myocardial infarction (MI). OBD mice had higher levels of plasma linolenic acid with a decrease in eicosapentaenoic and docosahexaenoic acid. The OBD mice had lower Lactobacillus johnsonii indicating a loss of probiotic microbiota. SF in OBD mice increased Firmicutes/Bacteroidetes ratio indicative of a detrimental change in SF-directed microbiome. OBD + SF group increased in the neutrophil: lymphocyte ratio suggestive of suboptimal inflammation. As a result of SF, resolution mediators (RvD2, RvD3, RvD5, LXA4 , PD1, and MaR1) decreased and inflammatory mediators (PGD2 , PGE2 , PGF2a , 6k-PGF1a ) were increased in OBD mice post-MI. At the site of infarction, the proinflammatory cytokines Ccl2, IL1β, and IL-6 were amplified in OBD + SF indicating a robust proinflammatory milieu post-MI. Also, brain circadian genes (Bmal1, Clock) were downregulated in SF-subjected control mice, but remained elevated in OBD mice post-MI. SF superimposed on obesity dysregulated physiological inflammation and disrupted resolving response thereby impaired cardiac repair and signs of pathological inflammation.
Collapse
Affiliation(s)
- Ganesh V. Halade
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine University of South Florida Tampa Florida USA
| | - Yusuf Mat
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine University of South Florida Tampa Florida USA
| | | | - Shalini Jain
- USF Center for Microbiome Research Microbiomes Institute Tampa Florida USA
- Center for Aging and Brain Repair University of South Florida Tampa Florida USA
| | - Shu‐Ping Hui
- Faculty of Health Sciences Hokkaido University Sapporo Japan
| | - Hariom Yadav
- USF Center for Microbiome Research Microbiomes Institute Tampa Florida USA
- Center for Aging and Brain Repair University of South Florida Tampa Florida USA
| | - Vasundhara Kain
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine University of South Florida Tampa Florida USA
| |
Collapse
|
37
|
Rodrigues EA, Lima ARR, Gomes MJ, Souza LM, Pontes THD, Pagan LU, Murata GM, Damatto FC, Carvalho Depra I, Rego ABGC, Reyes DRA, Zornoff LAM, Okoshi K, Okoshi MP. Influence of Isolated Resistance Exercise on Cardiac Remodeling, Myocardial Oxidative Stress, and Metabolism in Infarcted Rats. Antioxidants (Basel) 2023; 12:896. [PMID: 37107271 PMCID: PMC10135620 DOI: 10.3390/antiox12040896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/29/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
INTRODUCTION Exercise is an important therapeutic strategy for preventing and treating myocardial infarction (MI)-induced cardiac remodeling and heart failure. However, the myocardial effects of resistance exercise on infarcted hearts are not completely established. In this study, we investigated the effects of resistance exercise on structural, functional, and molecular cardiac alterations in infarcted rats. METHODS Three months after MI induction or simulated surgery, Wistar rats were assigned into three groups: Sham (n = 14); MI (n = 9); and exercised MI (MI-Ex, n = 13). Exercised rats performed, 3 times a week for 12 weeks, four climbs on a ladder with progressive loads. Cardiac structure and left ventricle (LV) function were analyzed by echocardiogram. Myocyte diameters were evaluated in hematoxylin- and eosin-stained histological sections as the smallest distance between borders drawn across the nucleus. Myocardial energy metabolism, lipid hydroperoxide, malondialdehyde, protein carbonylation, and antioxidant enzyme activities were evaluated by spectrophotometry. Gene expressions of NADPH oxidase subunits were evaluated by RT-PCR. Statistical analyses were performed using ANOVA and Tukey or Kruskal-Wallis and Dunn's test. RESULTS Mortality did not differ between the MI-Ex and MI groups. MI had dilated left atrium and LV, with LV systolic dysfunction. Exercise increased the maximum load-carrying capacity, with no changes in cardiac structure or LV function. Myocyte diameters were lower in MI than in Sham and MI-Ex. Lactate dehydrogenase and creatine kinase activity were lower in MI than in Sham. Citrate synthase and catalase activity were lower in MI and MI-Ex than in Sham. Lipid hydroperoxide concentration was lower in MI-Ex than in MI. Nox2 and p22phox gene expressions were higher in MI-Ex than in Sham. Gene expression of Nox4 was higher in MI and MI-Ex than in Sham, and p47phox was lower in MI than in Sham. CONCLUSION Late resistance exercise was safe in infarcted rats. Resistance exercise improved maximum load-carrying capacity, reduced myocardial oxidative stress, and preserved myocardial metabolism, with no changes in cardiac structure or left ventricle function in infarcted rats.
Collapse
Affiliation(s)
- Eder Anderson Rodrigues
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Aline Regina Ruiz Lima
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Mariana Janini Gomes
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX 77845, USA
| | - Lidiane Moreira Souza
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Thierres Hernani Dias Pontes
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Luana Urbano Pagan
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Gilson Masahiro Murata
- LIM29, Division of Nephrology, University of Sao Paulo Medical School, Sao Paulo 01246-903, SP, Brazil
| | - Felipe Cesar Damatto
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Igor Carvalho Depra
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | | | - David Rafael Abreu Reyes
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Leonardo Antonio Mamede Zornoff
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Katashi Okoshi
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Marina Politi Okoshi
- Department of Internal Medicine, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| |
Collapse
|
38
|
Clasen L, Angendohr S, Becher S, Bartsch B, Enkel S, Meyer C, Kelm M, Makimoto H, Klöcker N. Cardiac ischemia and reperfusion in mice: a comprehensive hemodynamic, electrocardiographic and electrophysiological characterization. Sci Rep 2023; 13:5693. [PMID: 37029160 PMCID: PMC10082073 DOI: 10.1038/s41598-023-32346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/26/2023] [Indexed: 04/09/2023] Open
Abstract
Malignant ventricular arrhythmias (VA) after acute myocardial infarction remain a major threat. Aim of this study was to characterize the electrophysiological and autonomic sequelae of cardiac ischemia and reperfusion (I/R) in mice during the first week post incident. Left ventricular function was serially assessed using transthoracic echocardiography. VA were quantified by telemetric electrocardiogram (ECG) recordings and electrophysiological studies on the 2nd and 7th day after I/R. Cardiac autonomic function was evaluated by heart rate variability (HRV) and heart rate turbulence (HRT). Infarct size was quantified by planimetric measures. I/R caused significant myocardial scarring and diminished left ventricular ejection fraction. The ECG intervals QRS, QT, QTc, and JTc were prolonged in I/R mice. Both spontaneous VA scored higher and the inducibility of VA was raised in I/R mice. An analysis of HRV and HRT indicated a relative reduction in parasympathetic activity and disturbed baroreflex sensitivity up to 7 days after I/R. In summary, during the first week after I/R, the murine heart reflects essential features of the human heart after myocardial infarction, including a greater vulnerability for VA and a decreased parasympathetic tone accompanied by decelerated depolarization and repolarization parameters.
Collapse
Affiliation(s)
- Lukas Clasen
- Department of Cardiology, Pulmonary and Vascular Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Cardiology, Rhythmology and Angiology, Josephs-Hospital Warendorf, Academic Teaching Hospital, University of Münster, Warendorf, Germany
| | - Stephan Angendohr
- Department of Cardiology, Pulmonary and Vascular Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stefanie Becher
- Department of Cardiology, Pulmonary and Vascular Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Benedikt Bartsch
- Department of Cardiology, Pulmonary and Vascular Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Stephan Enkel
- Department of Cardiology, Pulmonary and Vascular Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christian Meyer
- Division of Cardiology, Angiology, Intensive Care Medicine, EVK Düsseldorf, cNEP, Cardiac Neuro- and Electrophysiology Research Consortium, Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonary and Vascular Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hisaki Makimoto
- Department of Cardiology, Pulmonary and Vascular Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Nikolaj Klöcker
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
39
|
Cortada E, Yao J, Xia Y, Dündar F, Zumbo P, Yang B, Rubio-Navarro A, Perder B, Qiu M, Pettinato AM, Homan EA, Stoll L, Betel D, Cao J, Lo JC. Cross-species single-cell comparison of systemic and cardiac inflammatory responses after cardiac injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532865. [PMID: 36993713 PMCID: PMC10055080 DOI: 10.1101/2023.03.15.532865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
The immune system coordinates the response to cardiac injury and is known to control regenerative and fibrotic scar outcomes in the heart and subsequent chronic low-grade inflammation associated with heart failure. Here we profiled the inflammatory response to heart injury using single cell transcriptomics to compare and contrast two experimental models with disparate outcomes. We used adult mice, which like humans lack the ability to fully recover and zebrafish which spontaneously regenerate after heart injury. The extracardiac reaction to cardiomyocyte necrosis was also interrogated to assess the specific peripheral tissue and immune cell reaction to chronic stress. Cardiac macrophages are known to play a critical role in determining tissue homeostasis by healing versus scarring. We identified distinct transcriptional clusters of monocytes/macrophages in each species and found analogous pairs in zebrafish and mice. However, the reaction to myocardial injury was largely disparate between mice and zebrafish. The dichotomous response to heart damage between the mammalian and zebrafish monocytes/macrophages may underlie the impaired regenerative process in mice, representing a future therapeutic target.
Collapse
|
40
|
Lugrin J, Parapanov R, Milano G, Cavin S, Debonneville A, Krueger T, Liaudet L. The systemic deletion of interleukin-1α reduces myocardial inflammation and attenuates ventricular remodeling in murine myocardial infarction. Sci Rep 2023; 13:4006. [PMID: 36899010 PMCID: PMC10006084 DOI: 10.1038/s41598-023-30662-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Myocardial inflammation following myocardial infarction (MI) is crucial for proper myocardial healing, yet, dysregulated inflammation may promote adverse ventricular remodeling and heart failure. IL-1 signaling contributes to these processes, as shown by dampened inflammation by inhibition of IL-1β or the IL-1 receptor. In contrast, the potential role of IL-1α in these mechanisms has received much less attention. Previously described as a myocardial-derived alarmin, IL-1α may also act as a systemically released inflammatory cytokine. We therefore investigated the effect of IL-1α deficiency on post-MI inflammation and ventricular remodeling in a murine model of permanent coronary occlusion. In the first week post-MI, global IL-1α deficiency (IL-1α KO mice) led to decreased myocardial expression of IL-6, MCP-1, VCAM-1, hypertrophic and pro-fibrotic genes, and reduced infiltration with inflammatory monocytes. These early changes were associated with an attenuation of delayed left ventricle (LV) remodeling and systolic dysfunction after extensive MI. In contrast to systemic Il1a-KO, conditional cardiomyocyte deletion of Il1a (CmIl1a-KO) did not reduce delayed LV remodeling and systolic dysfunction. In conclusion, systemic Il1a-KO, but not Cml1a-KO, protects against adverse cardiac remodeling after MI due to permanent coronary occlusion. Hence, anti-IL-1α therapies could be useful to attenuate the detrimental consequences of post-MI myocardial inflammation.
Collapse
Affiliation(s)
- J Lugrin
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Laboratoire de Chirurgie Thoracique, Centre des Laboratoires d'Epalinges, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| | - R Parapanov
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - G Milano
- Department Coeur-Vaisseaux, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - S Cavin
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - A Debonneville
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - T Krueger
- Service of Thoracic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - L Liaudet
- Service of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
41
|
Siatra P, Vatsellas G, Chatzianastasiou A, Balafas E, Manolakou T, Papapetropoulos A, Agapaki A, Mouchtouri ET, Ruchaya PJ, Korovesi AG, Mavroidis M, Thanos D, Beis D, Kokkinopoulos I. Return of the Tbx5; lineage-tracing reveals ventricular cardiomyocyte-like precursors in the injured adult mammalian heart. NPJ Regen Med 2023; 8:13. [PMID: 36869039 PMCID: PMC9984483 DOI: 10.1038/s41536-023-00280-9] [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: 03/03/2022] [Accepted: 01/25/2023] [Indexed: 03/05/2023] Open
Abstract
The single curative measure for heart failure patients is a heart transplantation, which is limited due to a shortage of donors, the need for immunosuppression and economic costs. Therefore, there is an urgent unmet need for identifying cell populations capable of cardiac regeneration that we will be able to trace and monitor. Injury to the adult mammalian cardiac muscle, often leads to a heart attack through the irreversible loss of a large number of cardiomyocytes, due to an idle regenerative capability. Recent reports in zebrafish indicate that Tbx5a is a vital transcription factor for cardiomyocyte regeneration. Preclinical data underscore the cardioprotective role of Tbx5 upon heart failure. Data from our earlier murine developmental studies have identified a prominent unipotent Tbx5-expressing embryonic cardiac precursor cell population able to form cardiomyocytes, in vivo, in vitro and ex vivo. Using a developmental approach to an adult heart injury model and by employing a lineage-tracing mouse model as well as the use of single-cell RNA-seq technology, we identify a Tbx5-expressing ventricular cardiomyocyte-like precursor population, in the injured adult mammalian heart. The transcriptional profile of that precursor cell population is closer to that of neonatal than embryonic cardiomyocyte precursors. Tbx5, a cardinal cardiac development transcription factor, lies in the center of a ventricular adult precursor cell population, which seems to be affected by neurohormonal spatiotemporal cues. The identification of a Tbx5-specific cardiomyocyte precursor-like cell population, which is capable of dedifferentiating and potentially deploying a cardiomyocyte regenerative program, provides a clear target cell population for translationally-relevant heart interventional studies.
Collapse
Affiliation(s)
- Panagiota Siatra
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Giannis Vatsellas
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Greek Genome Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Athanasia Chatzianastasiou
- Department of Pharmacy, Laboratory of Pharmacology, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Balafas
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Theodora Manolakou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Department of Pharmacy, Laboratory of Pharmacology, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Agapaki
- Histochemistry Facility, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Prashant J Ruchaya
- School of Health, Sport and Biosciences, University of East London, London, UK
| | - Artemis G Korovesi
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Greek Genome Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Manolis Mavroidis
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Dimitrios Thanos
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Greek Genome Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Dimitris Beis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Ioannis Kokkinopoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.
| |
Collapse
|
42
|
Doudin A, Riebeling T, Staab J, Menon PR, Lühder F, Wirths O, Vinkemeier U, Ivetic A, Meyer T. Lack of STAT1 co-operative DNA binding protects against adverse cardiac remodelling in acute myocardial infarction. Front Cardiovasc Med 2023; 10:975012. [PMID: 36923955 PMCID: PMC10008942 DOI: 10.3389/fcvm.2023.975012] [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: 06/21/2022] [Accepted: 02/02/2023] [Indexed: 03/03/2023] Open
Abstract
In this study, we addressed the functional significance of co-operative DNA binding of the cytokine-driven transcription factor STAT1 (signal transducer and activator of transcription 1) in an experimental murine model of acute myocardial infarction (MI). STAT1 knock-in mice expressing a phenylalanine-to-alanine substitution at position 77 in the STAT1 amino-terminal domain were examined for the early clinical effects produced by ligation of the left anterior descending coronary artery (LAD), an established model for MI. The F77A mutation has been previously reported to disrupt amino-terminal interactions between adjacent STAT1 dimers resulting in impaired tetramerization and defective co-operative binding on DNA, while leaving other protein functions unaffected. Our results demonstrate that a loss of STAT1 tetramer stabilization improves survival of adult male mice and ameliorates left ventricular dysfunction in female mice, as determined echocardiographically by an increased ejection fraction and a reduced left intra-ventricular diameter. We found that the ratio of STAT3 to STAT1 protein level was higher in the infarcted tissue in knock-in mice as compared to wild-type (WT) mice, which was accompanied by an enhanced infiltration of immune cells in the infarcted area, as determined by histology. Additionally, RNA sequencing of the infarcted tissue 24 h after LAD ligation revealed an upregulation of inflammatory genes in the knock-in mice, as compared to their WT littermates. Concomitantly, genes involved in oxidative phosphorylation and other metabolic pathways showed a significantly more pronounced downregulation in the infarcted tissue from STAT1F77A/F77A mice than in WT animals. Based on these results, we propose that dysfunctional STAT1 signalling owing to a lack of oligomerisation results in a compensatory increase in STAT3 expression and promotes early infiltration of immune cells in the infarcted area, which has beneficial effects on left ventricular remodelling in early MI following LAD ligation.
Collapse
Affiliation(s)
- Asmma Doudin
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Centre Göttingen, and German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Theresa Riebeling
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Centre Göttingen, and German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.,Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Julia Staab
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Centre Göttingen, and German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Priyanka Rajeev Menon
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Centre Göttingen, and German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Göttingen, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
| | - Uwe Vinkemeier
- Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Aleksandar Ivetic
- British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom
| | - Thomas Meyer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Centre Göttingen, and German Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
43
|
Chalise U, Becirovic‐Agic M, Lindsey ML. The cardiac wound healing response to myocardial infarction. WIREs Mech Dis 2023; 15:e1584. [PMID: 36634913 PMCID: PMC10077990 DOI: 10.1002/wsbm.1584] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 01/14/2023]
Abstract
Myocardial infarction (MI) is defined as evidence of myocardial necrosis consistent with prolonged ischemia. In response to MI, the myocardium undergoes a series of wound healing events that initiate inflammation and shift to anti-inflammation before transitioning to tissue repair that culminates in scar formation to replace the region of the necrotic myocardium. The overall response to MI is determined by two major steps, the first of which is the secretion of proteases by infiltrating leukocytes to breakdown extracellular matrix (ECM) components, a necessary step to remove necrotic cardiomyocytes. The second step is the generation of new ECM that comprises the scar; and this step is governed by the cardiac fibroblasts as the major source of new ECM synthesis. The leukocyte component resides in the middle of the two-step process, contributing to both sides as the leukocytes transition from pro-inflammatory to anti-inflammatory and reparative cell phenotypes. The balance between the two steps determines the final quantity and quality of scar formed, which in turn contributes to chronic outcomes following MI, including the progression to heart failure. This review will summarize our current knowledge regarding the cardiac wound healing response to MI, primarily focused on experimental models of MI in mice. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Mediha Becirovic‐Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| |
Collapse
|
44
|
Novitskaya T, Nishat S, Covarrubias R, Wheeler DG, Chepurko E, Bermeo-Blanco O, Xu Z, Baer B, He H, Moore SN, Dwyer KM, Cowan PJ, Su YR, Absi TS, Schoenecker J, Bellan LM, Koch WJ, Bansal S, Feoktistov I, Robson SC, Gao E, Gumina RJ. Ectonucleoside triphosphate diphosphohydrolase-1 (CD39) impacts TGF-β1 responses: insights into cardiac fibrosis and function following myocardial infarction. Am J Physiol Heart Circ Physiol 2022; 323:H1244-H1261. [PMID: 36240436 PMCID: PMC9722260 DOI: 10.1152/ajpheart.00138.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
Extracellular purine nucleotides and nucleosides released from activated or injured cells influence multiple aspects of cardiac physiology and pathophysiology. Ectonucleoside triphosphate diphosphohydrolase-1 (ENTPD1; CD39) hydrolyzes released nucleotides and thereby regulates the magnitude and duration of purinergic signaling. However, the impact of CD39 activity on post-myocardial infarction (MI) remodeling is incompletely understood. We measured the levels and activity of ectonucleotidases in human left ventricular samples from control and ischemic cardiomyopathy (ICM) hearts and examined the impact of ablation of Cd39 expression on post-myocardial infarction remodeling in mice. We found that human CD39 levels and activity are significantly decreased in ICM hearts (n = 5) compared with control hearts (n = 5). In mice null for Cd39, cardiac function and remodeling are significantly compromised in Cd39-/- mice following myocardial infarction. Fibrotic markers including plasminogen activator inhibitor-1 (PAI-1) expression, fibrin deposition, α-smooth muscle actin (αSMA), and collagen expression are increased in Cd39-/- hearts. Importantly, we found that transforming growth factor β1 (TGF-β1) stimulates ATP release and induces Cd39 expression and activity on cardiac fibroblasts, constituting an autocrine regulatory pathway not previously appreciated. Absence of CD39 activity on cardiac fibroblasts exacerbates TGF-β1 profibrotic responses. Treatment with exogenous ectonucleotidase rescues this profibrotic response in Cd39-/- fibroblasts. Together, these data demonstrate that CD39 has important interactions with TGF-β1-stimulated autocrine purinergic signaling in cardiac fibroblasts and dictates outcomes of cardiac remodeling following myocardial infarction. Our results reveal that ENTPD1 (CD39) regulates TGF-β1-mediated fibroblast activation and limits adverse cardiac remodeling following myocardial infarction.NEW & NOTEWORTHY We show that CD39 is a critical modulator of TGF-β1-mediated fibroblast activation and cardiac remodeling following myocardial infarction via modulation of nucleotide signaling. TGF-β1-induced CD39 expression generates a negative feedback loop that attenuates cardiac fibroblast activation. In the absence of CD39 activity, collagen deposition is increased, elastin expression is decreased, and diastolic dysfunction is worsened. Treatment with ecto-apyrase attenuates the TGF-β1-induced profibrotic cardiac fibroblast phenotype, revealing a novel approach to combat post-myocardial infarction cardiac fibrosis.
Collapse
Affiliation(s)
- Tatiana Novitskaya
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shamama Nishat
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Roman Covarrubias
- Division of Cardiac Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Debra G Wheeler
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Elena Chepurko
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Oscar Bermeo-Blanco
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Zhaobin Xu
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Bradly Baer
- Department of Mechanical Engineering, Vanderbilt University School of Engineering, Nashville, Tennessee
| | - Heng He
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Stephanie N Moore
- Division of Orthopedic Surgery, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Karen M Dwyer
- Immunology Research Center, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter J Cowan
- Immunology Research Center, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Yan Ru Su
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tarek S Absi
- Division of Cardiac Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan Schoenecker
- Division of Orthopedic Surgery, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leon M Bellan
- Department of Mechanical Engineering, Vanderbilt University School of Engineering, Nashville, Tennessee
| | | | - Shyam Bansal
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Igor Feoktistov
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Simon C Robson
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Erhe Gao
- Temple University, Philadelphia, Pennsylvania
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
45
|
Ricketts SN, Qian L. The heart of cardiac reprogramming: The cardiac fibroblasts. J Mol Cell Cardiol 2022; 172:90-99. [PMID: 36007393 DOI: 10.1016/j.yjmcc.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/29/2022] [Accepted: 08/13/2022] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide, outpacing pulmonary disease, infectious disease, and all forms of cancer. Myocardial infarction (MI) dominates cardiovascular disease, contributing to four out of five cardiovascular related deaths. Following MI, patients suffer adverse and irreversible myocardial remodeling associated with cardiomyocyte loss and infiltration of fibrotic scar tissue. Current therapies following MI only mitigate the cardiac physiological decline rather than restore damaged myocardium function. Direct cardiac reprogramming is one strategy that has promise in repairing injured cardiac tissue by generating new, functional cardiomyocytes from cardiac fibroblasts (CFs). With the ectopic expression of transcription factors, microRNAs, and small molecules, CFs can be reprogrammed into cardiomyocyte-like cells (iCMs) that display molecular signatures, structures, and contraction abilities similar to endogenous cardiomyocytes. The in vivo induction of iCMs following MI leads to significant reduction in fibrotic cardiac remodeling and improved heart function, indicating reprogramming is a viable option for repairing damaged heart tissue. Recent work has illustrated different methods to understand the mechanisms driving reprogramming, in an effort to improve the efficiency of iCM generation and create an approach translational into clinic. This review will provide an overview of CFs and describe different in vivo reprogramming methods.
Collapse
Affiliation(s)
- Shea N Ricketts
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
46
|
Abstract
Metabolic changes frequently occur in patients with chronic heart failure (HF). Therefore, detailed identification of these metabolic changes, and complementing them, will provide new therapeutic approaches. Here, using a mouse model, we demonstrated that succinyl-CoA levels are reduced in the myocardial mitochondria of hearts undergoing chronic HF, and this reduction impairs mitochondrial oxidative phosphorylation capacity. We identified increased heme synthesis as a cause of this succinyl-CoA reduction and demonstrated a method that can compensate substantially for the increased succinyl-CoA consumption. Reduction in succinyl-CoA levels has also been reported in HF patients. Our results provide an academic basis for the development of new treatment methodologies against HF, which target the altered metabolic activities that occur in HF by nutritional interventions. Heart failure (HF) is a leading cause of death and repeated hospitalizations and often involves cardiac mitochondrial dysfunction. However, the underlying mechanisms largely remain elusive. Here, using a mouse model in which myocardial infarction (MI) was induced by coronary artery ligation, we show the metabolic basis of mitochondrial dysfunction in chronic HF. Four weeks after ligation, MI mice showed a significant decrease in myocardial succinyl-CoA levels, and this decrease impaired the mitochondrial oxidative phosphorylation (OXPHOS) capacity. Heme synthesis and ketolysis, and protein levels of several enzymes consuming succinyl-CoA in these events, were increased in MI mice, while enzymes synthesizing succinyl-CoA from α-ketoglutarate and glutamate were also increased. Furthermore, the ADP-specific subunit of succinyl-CoA synthase was reduced, while its GDP-specific subunit was almost unchanged. Administration of 5-aminolevulinic acid, an intermediate in the pathway from succinyl-CoA to heme synthesis, appreciably restored succinyl-CoA levels and OXPHOS capacity and prevented HF progression in MI mice. Previous reports also suggested the presence of succinyl-CoA metabolism abnormalities in cardiac muscles of HF patients. Our results identified that changes in succinyl-CoA usage in different metabolisms of the mitochondrial energy production system is characteristic to chronic HF, and although similar alterations are known to occur in healthy conditions, such as during strenuous exercise, they may often occur irreversibly in chronic HF leading to a decrease in succinyl-CoA. Consequently, nutritional interventions compensating the succinyl-CoA consumption are expected to be promising strategies to treat HF.
Collapse
|
47
|
Halade GV, Kain V, Hossain S, Parcha V, Limdi NA, Arora P. Arachidonate 5-lipoxygenase is essential for biosynthesis of specialized pro-resolving mediators and cardiac repair in heart failure. Am J Physiol Heart Circ Physiol 2022; 323:H721-H737. [PMID: 36018758 PMCID: PMC9529265 DOI: 10.1152/ajpheart.00115.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022]
Abstract
Arachidonate 5-lipoxygenase (ALOX5)-derived leukotrienes are primary signals of leukocyte activation and inflammation in response to ischemic cardiac injury (MI; myocardial infarction). Using risk-free male C57BL/6J and ALOX5-null mice (8-12 wk), we quantitated leukocytes and ALOX5-derived bioactive lipids of the infarcted left ventricle (LV) and spleen to measure the physiological inflammation and cardiac repair. Our results showed that ALOX5 endogenously generates specialized pro-resolving mediators (SPMs) that facilitate cardiac repair post-MI. Deficiency of ALOX5 leads to increase in cyclooxygenase gene expression, 6-keto prostaglandin F1α, and delayed neutrophil clearance with signs of unresolved inflammation post-MI. Consequently, ALOX5 deficiency impaired the resolution of inflammation and cardiac repair, including increased myocardium rupture post-MI in acute heart failure. On-time ALOX5 activation is critical for leukocyte clearance from the infarcted heart, indicating an essential role of ALOX5 in the resolution of inflammation. In addition, to balance the inflammatory responses, ALOX5 is also necessary for fibroblast signaling, as the ALOX5-deficient fibroblast are prone to fibroblast-to-myofibroblast differentiation leading to defective scar formation in post-MI cardiac repair. Consistent with these findings, ALOX5-null mice showed an overly inflammatory response, defective fibrotic signaling, and unresolved inflammation. These findings are indicative of a critical role of ALOX5 in myocardium healing, inflammation-resolution signaling, cardiac repair, and fibroblast pathophysiology.NEW & NOTEWORTHY Arachidonate 5-lipoxygenase (ALOX5) is critical in synthesizing specialized pro-resolving mediators that facilitate cardiac repair after cardiac injury. Thus, ALOX5 orchestrates the overlapping phases of inflammation and resolution to facilitate myocardium healing in cardiac repair postmyocardial infarction.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Shahriare Hossain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, Florida
| | - Vibhu Parcha
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Nita A Limdi
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Pankaj Arora
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
48
|
McAndrew DJ, Lake HA, Zervou S, Schwedhelm E, Schneider JE, Neubauer S, Lygate CA. Homoarginine and creatine deficiency do not exacerbate murine ischaemic heart failure. ESC Heart Fail 2022; 10:189-199. [PMID: 36178450 PMCID: PMC9871656 DOI: 10.1002/ehf2.14183] [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: 07/07/2022] [Revised: 08/23/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
AIMS Low levels of homoarginine and creatine are associated with heart failure severity in humans, but it is unclear to what extent they contribute to pathophysiology. Both are synthesized via L-arginine:glycine amidinotransferase (AGAT), such that AGAT-/- mice have a combined creatine and homoarginine deficiency. We hypothesized that this would be detrimental in the setting of chronic heart failure. METHODS AND RESULTS Study 1: homoarginine deficiency-female AGAT-/- and wild-type mice were given creatine-supplemented diet so that both had normal myocardial creatine levels, but only AGAT-/- had low plasma homoarginine. Myocardial infarction (MI) was surgically induced and left ventricular (LV) structure and function assessed at 6-7 weeks by in vivo imaging and haemodynamics. Study 2: homoarginine and creatine-deficiency-as before, but AGAT-/- mice were given creatine-supplemented diet until 1 week post-MI, when 50% were changed to a creatine-free diet. Both groups therefore had low homoarginine levels, but one group also developed lower myocardial creatine levels. In both studies, all groups had LV remodelling and dysfunction commensurate with the development of chronic heart failure, for example, LV dilatation and mean ejection fraction <20%. However, neither homoarginine deficiency alone or in combination with creatine deficiency had a significant effect on mortality, LV remodelling, or on any indices of contractile and lusitropic function. CONCLUSIONS Low levels of homoarginine and creatine do not worsen chronic heart failure arguing against a major causative role in disease progression. This suggests that it is unnecessary to correct hArg deficiency in patients with heart failure, although supra-physiological levels may still be beneficial.
Collapse
Affiliation(s)
- Debra J. McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| | - Hannah A. Lake
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| | - Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jurgen E. Schneider
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,Experimental and Preclinical Imaging Centre (ePIC), Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK
| | - Craig A. Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| |
Collapse
|
49
|
Broughton P, Troncoso M, Corker A, Williams A, Bolus D, Munoz G, McWhorter C, Roerden H, Huebsch P, DeLeon-Pennell KY. Riding the wave: a quantitative report of electrocardiogram utilization for myocardial infarction confirmation. Am J Physiol Heart Circ Physiol 2022; 323:H378-H387. [PMID: 35802516 PMCID: PMC9359650 DOI: 10.1152/ajpheart.00201.2022] [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: 04/25/2022] [Revised: 06/22/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to generate a quantitative profile of electrocardiograms (ECGs) for confirming surgical success of permanent coronary artery ligation. An ECG was recorded at baseline, and 0, 1, and 5 min after ligation and analyzed using iWorkx LabScribe software. Cohort 1 (C57Bl6/J, n = 8/sex) was enrolled to determine ECG characteristics that were confirmed in cohort 2 (C57Bl6/J, n = 6/sex; CD8-/-n = 6 males/4 females). Of the 16 mice in cohort 1, 12 (6/sex) had an infarct ≥35% and four mice (2/sex) had <35% based on 2,3,5-triphenyltetrazolium chloride staining. After ligation, the QRS complex and R-S amplitude were significantly different compared with baseline. No differences were observed in the R-S amplitude between mice with infarcts ≥35% versus <35% at any time point, whereas the QRS complex was significant 1 min after ligation. Receiver operating characteristic (ROC) curve linked changes in the QRS complex but not the R-S amplitude at 1 and 5 min with surgical success. Data were normalized to baseline values to calculate fold change. ROC analysis of the normalized QRS data indicated strong sensitivity and specificity for infarcts ≥35%; normalized R-S amplitude remained nonsignificant. With a cutoff generated by ROC analysis of cohort 1 (>80% sensitivity; >90% specificity), the non-normalized QRS complex of cohort 2 had an 86% success rate (2 false positives; 1 false negative). The normalized data had a 77% success rate (2 false positives; 3 false negatives). Neither sex nor genotype was associated with false predictions (P = 0.18). Our data indicate that the area under the QRS complex 1 min after ligation can improve reproducibility in MI surgeries.NEW & NOTEWORTHY Our study describes a quantitative method for using an electrocardiogram (ECG) to determine which animals have infarcts that reflect coronary artery ligation. Using a quantitative ECG, investigators will have the benefit of having real-time feedback during the procedure, which will ultimately decrease the amount of time investigators spend performing experiments. This overall increase in efficiency will help investigators decrease animal numbers used due to better surgical outcomes.
Collapse
Affiliation(s)
- Philip Broughton
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Miguel Troncoso
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Alexa Corker
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alexus Williams
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dawson Bolus
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Gualberto Munoz
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline McWhorter
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Hallie Roerden
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Penny Huebsch
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
50
|
Li Y, Gao Y, Li G. Preclinical multi-target strategies for myocardial ischemia-reperfusion injury. Front Cardiovasc Med 2022; 9:967115. [PMID: 36072870 PMCID: PMC9444048 DOI: 10.3389/fcvm.2022.967115] [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: 06/12/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Despite promising breakthroughs in diagnosing and treating acute coronary syndromes, cardiovascular disease’s high global mortality rate remains indisputable. Nearly half of these patients died of ischemic heart disease. Primary percutaneous coronary intervention (PCI) and coronary artery bypass grafting can rapidly restore interrupted blood flow and become the most effective method for salvaging viable myocardium. However, restoring blood flow could increase the risk of other complications and myocardial cell death attributed to myocardial ischemia-reperfusion injury (IRI). How to reduce the damage of blood reperfusion to ischemic myocardium has become an urgent problem to be solved. In preclinical experiments, many treatments have substantial cardioprotective effects against myocardial IRI. However, the transition from these cardioprotective therapies to clinically beneficial therapies for patients with acute myocardial infarction remains elusive. The reasons for the failure of the clinical translation may be multi-faceted, and three points are summarized here: (1) Our understanding of the complex pathophysiological mechanisms of myocardial IRI is far from enough, and the classification of specific therapeutic targets is not rigorous, and not clear enough; (2) Most of the clinical patients have comorbidities, and single cardioprotective strategies including ischemia regulation strategies cannot exert their due cardioprotective effects under conditions of hyperglycemia, hypertension, hyperlipidemia, and aging; (3) Most preclinical experimental results are based on adult, healthy animal models. However, most clinical patients had comorbidities and received multiple drug treatments before reperfusion therapy. In 2019, COST Action proposed a multi-target drug combination initiative for prospective myocardial IRI; the optimal cardioprotective strategy may be a combination of additive or synergistic multi-target therapy, which we support. By establishing more reasonable preclinical models, screening multi-target drug combinations more in line with clinical practice will benefit the translation of clinical treatment strategies.
Collapse
|