1
|
Silva J, Azevedo T, Ginja M, Oliveira PA, Duarte JA, Faustino-Rocha AI. Realistic Aspects of Cardiac Ultrasound in Rats: Practical Tips for Improved Examination. J Imaging 2024; 10:219. [PMID: 39330439 PMCID: PMC11433567 DOI: 10.3390/jimaging10090219] [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: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Echocardiography is a reliable and non-invasive method for assessing cardiac structure and function in both clinical and experimental settings, offering valuable insights into disease progression and treatment efficacy. The successful application of echocardiography in murine models of disease has enabled the evaluation of disease severity, drug testing, and continuous monitoring of cardiac function in these animals. However, there is insufficient standardization of echocardiographic measurements for smaller animals. This article aims to address this gap by providing a guide and practical tips for the appropriate acquisition and analysis of echocardiographic parameters in adult rats, which may also be applicable in other small rodents used for scientific purposes, like mice. With advancements in technology, such as ultrahigh-frequency ultrasonic transducers, echocardiography has become a highly sophisticated imaging modality, offering high temporal and spatial resolution imaging, thereby allowing for real-time monitoring of cardiac function throughout the lifespan of small animals. Moreover, it allows the assessment of cardiac complications associated with aging, cancer, diabetes, and obesity, as well as the monitoring of cardiotoxicity induced by therapeutic interventions in preclinical models, providing important information for translational research. Finally, this paper discusses the future directions of cardiac preclinical ultrasound, highlighting the need for continued standardization to advance research and improve clinical outcomes to facilitate early disease detection and the translation of findings into clinical practice.
Collapse
Affiliation(s)
- Jessica Silva
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
| | - Tiago Azevedo
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Centro de Investigação de Montanha (CIMO), Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Mário Ginja
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - José Alberto Duarte
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences (IUCS), Advanced Polytechnic and University Cooperative (CESPU), 4585-116 Gandra, Portugal;
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory (1H-TOXRUN), University Institute of Health Sciences (IUCS), Advanced Polytechnic and University Cooperative (CESPU), 4585-116 Gandra, Portugal
| | - Ana I. Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Department of Zootechnics, School of Sciences and Technology, University of Évora, 7004-516 Évora, Portugal
- Comprehensive Health Research Center (CHRC), University of Évora, 7004-516 Évora, Portugal
| |
Collapse
|
2
|
Phoon CK, Aristizábal O, Farhoud M, Turnbull DH, Wadghiri YZ. Mouse Cardiovascular Imaging. Curr Protoc 2024; 4:e1116. [PMID: 39222027 PMCID: PMC11371386 DOI: 10.1002/cpz1.1116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The mouse is the mammalian model of choice for investigating cardiovascular biology, given our ability to manipulate it by genetic, pharmacologic, mechanical, and environmental means. Imaging is an important approach to phenotyping both function and structure of cardiac and vascular components. This review details commonly used imaging approaches, with a focus on echocardiography and magnetic resonance imaging, with brief overviews of other imaging modalities. In this update, we also emphasize the importance of rigor and reproducibility in imaging approaches, experimental design, and documentation. Finally, we briefly outline emerging imaging approaches but caution that reliability and validity data may be lacking. © 2024 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Colin K.L. Phoon
- Division of Pediatric Cardiology, Department of Pediatrics, New York University Grossman School of Medicine, New York, NY
| | - Orlando Aristizábal
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, & Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY
- Preclinical Imaging, Division for Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY
| | | | - Daniel H. Turnbull
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, & Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Youssef Z. Wadghiri
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, & Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY
- Preclinical Imaging, Division for Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
3
|
Conley JM, Lambright CS, Evans N, Farraj AK, Smoot J, Grindstaff RD, Hill D, McCord J, Medlock-Kakaley E, Dixon A, Hines E, Gray LE. Dose additive maternal and offspring effects of oral maternal exposure to a mixture of three PFAS (HFPO-DA, NBP2, PFOS) during pregnancy in the Sprague-Dawley rat. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 892:164609. [PMID: 37271399 PMCID: PMC10681034 DOI: 10.1016/j.scitotenv.2023.164609] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Simultaneous exposure to multiple per- and polyfluoroalkyl substances (PFAS) is common in humans across the globe. Individual PFAS are associated with adverse health effects, yet the nature of mixture effects after exposure to two or more PFAS remains unclear. Previously we reported that oral administration of hexafluoropropylene oxide-dimer acid (HFPO-DA, or GenX), Nafion byproduct 2 (NBP2), or perfluorooctane sulfonate (PFOS) individually during pregnancy produced maternal and F1 effects. Here, we hypothesized that responses to the combined exposure to these three PFAS would be dose additive. Pregnant Sprague-Dawley rats were exposed to a fixed-ratio equipotent mixture where the top dose contained each PFAS at their ED50 for neonatal mortality (100 % dose = PFOS 3 mg/kg; NBP2 10 mg/kg; HFPO-DA 110 mg/kg), followed by a dilution series (33.3, 10, 3.3, and 1 %) and vehicle controls (0 % dose). Consistent with the single chemical studies, dams were exposed from gestation day (GD)14-18 or from GD8-postnatal day (PND2). Fetal and maternal livers on GD18 displayed multiple significantly upregulated genes associated with lipid and carbohydrate metabolism at all dose levels, while dams displayed significantly increased liver weight (≥3.3 % dose) and reduced serum thyroid hormones (≥33.3 % dose). Maternal exposure from GD8-PND2 significantly reduced pup bodyweights at birth (≥33.3 % dose) and PND2 (all doses), increased neonatal liver weights (≥3.3 % dose), increased pup mortality (≥3.3 % dose), and reduced maternal bodyweights and weight gain at the top dose. Echocardiography of adult F1 males and females identified significantly increased left ventricular anterior wall thickness (~10 % increase), whereas other cardiac morphological, functional, and transcriptomic measures were unaffected. Mixture effects in maternal and neonatal animals conformed to dose addition using a relative potency factor (RPF) analysis. Results support dose addition-based cumulative assessment approaches for estimating combined effects of PFAS co-exposure.
Collapse
Affiliation(s)
- Justin M Conley
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Christy S Lambright
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Nicola Evans
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Aimen K Farraj
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Jacob Smoot
- ORISE Participant, U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Rachel D Grindstaff
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA
| | - Donna Hill
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - James McCord
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Environmental Measurement and Modeling, Research Triangle Park, NC, USA.
| | - Elizabeth Medlock-Kakaley
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Aaron Dixon
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - Erin Hines
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| | - L Earl Gray
- U.S. Environmental Protection Agency, Office of Research & Development, Center for Public Health and Environmental Assessment, Research Triangle Park, NC, USA.
| |
Collapse
|
4
|
Farag A, Mandour AS, Kaneda M, Elfadadny A, Elhaieg A, Shimada K, Tanaka R. Effect of trehalose on heart functions in rats model after myocardial infarction: assessment of novel intraventricular pressure and heart rate variability. Front Cardiovasc Med 2023; 10:1182628. [PMID: 37469485 PMCID: PMC10353053 DOI: 10.3389/fcvm.2023.1182628] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/09/2023] [Indexed: 07/21/2023] Open
Abstract
Background Myocardial infarctions remain a leading cause of global deaths. Developing novel drugs to target cardiac remodeling after myocardial injury is challenging. There is an increasing interest in exploring natural cardioprotective agents and non-invasive tools like intraventricular pressure gradients (IVPG) and heart rate variability (HRV) analysis in myocardial infarctions. Trehalose (TRE), a natural disaccharide, shows promise in treating atherosclerosis, myocardial infarction, and neurodegenerative disorders. Objectives The objective of this study was to investigate the effectiveness of TRE in improving cardiac functions measured by IVPG and HRV and reducing myocardial remodeling following myocardial infarction in rat model. Methods Rats were divided into three groups: sham, myocardial infarction (MI), and trehalose-treated MI (TRE) groups. The animals in the MI and TRE groups underwent permanent ligation of the left anterior descending artery. The TRE group received 2% trehalose in their drinking water for four weeks after the surgery. At the end of the experiment, heart function was assessed using conventional echocardiography, novel color M-mode echocardiography for IVPG evaluation, and HRV analysis. After euthanasia, gross image scoring, histopathology, immunohistochemistry, and quantitative real-time PCR were performed to evaluate inflammatory reactions, oxidative stress, and apoptosis. Results The MI group exhibited significantly lower values in multiple IVPG parameters. In contrast, TRE administration showed an ameliorative effect on IVPG changes, with results comparable to the sham group. Additionally, TRE improved HRV parameters, mitigated morphological changes induced by myocardial infarction, reduced histological alterations in wall mass, and suppressed inflammatory reactions within the infarcted heart tissues. Furthermore, TRE demonstrated antioxidant, anti-apoptotic and anti-fibrotic properties. Conclusion The investigation into the effect of trehalose on a myocardial infarction rat model has yielded promising outcomes, as evidenced by improvements observed through conventional echocardiography, histological analysis, and immunohistochemical analysis. While minor trends were noticed in IVPG and HRV measurements. However, our findings offer valuable insights and demonstrate a correlation between IVPG, HRV, and other traditional markers of echo assessment in the myocardial infarction vs. sham groups. This alignment suggests the potential of IVPG and HRV as additional indicators for future research in this field.
Collapse
Affiliation(s)
- Ahmed Farag
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed S. Mandour
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ahmed Elfadadny
- Department of Animal Internal Medicine, Faculty of Veterinary Medicine, Damanhur University, Damanhur El-Beheira, Egypt
| | - Asmaa Elhaieg
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Kazumi Shimada
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Ryou Tanaka
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
5
|
Haustein R, Trogisch FA, Keles M, Hille S, Fuhrmann M, Weinzierl N, Hemanna S, Thackeray J, Dou Y, Zwadlo C, Froese N, Cordero J, Bengel F, Müller OJ, Bauersachs J, Dobreva G, Heineke J. C1q and Tumor Necrosis Factor Related Protein 9 Protects from Diabetic Cardiomyopathy by Alleviating Cardiac Insulin Resistance and Inflammation. Cells 2023; 12:cells12030443. [PMID: 36766785 PMCID: PMC9914367 DOI: 10.3390/cells12030443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
(1) Background: Diabetic cardiomyopathy is a major health problem worldwide. CTRP9, a secreted glycoprotein, is mainly expressed in cardiac endothelial cells and becomes downregulated in mouse models of diabetes mellitus; (2) Methods: In this study, we investigated the impact of CTRP9 on early stages of diabetic cardiomyopathy induced by 12 weeks of high-fat diet; (3) Results: While the lack of CTRP9 in knock-out mice aggravated insulin resistance and triggered diastolic left ventricular dysfunction, AAV9-mediated cardiac CTRP9 overexpression ameliorated cardiomyopathy under these conditions. At this early disease state upon high-fat diet, no fibrosis, no oxidative damage and no lipid deposition were identified in the myocardium of any of the experimental groups. Mechanistically, we found that CTRP9 is required for insulin-dependent signaling, cardiac glucose uptake in vivo and oxidative energy production in cardiomyocytes. Extensive RNA sequencing from myocardial tissue of CTRP9-overexpressing and knock-out as well as respective control mice revealed that CTRP9 acts as an anti-inflammatory mediator in the myocardium. Hence, CTRP9 knock-out exerted more, while CTRP9-overexpressing mice showed less leukocytes accumulation in the heart during high-fat diet; (4) Conclusions: In summary, endothelial-derived CTRP9 plays a prominent paracrine role to protect against diabetic cardiomyopathy and might constitute a therapeutic target.
Collapse
Affiliation(s)
- Ricarda Haustein
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Felix A. Trogisch
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Merve Keles
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Manuela Fuhrmann
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Nina Weinzierl
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Shruthi Hemanna
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - James Thackeray
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Yanliang Dou
- Cardiovascular Genomics and Epigenomics, ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Carolin Zwadlo
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Natali Froese
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Julio Cordero
- Cardiovascular Genomics and Epigenomics, ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Frank Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Oliver J. Müller
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Gergana Dobreva
- Cardiovascular Genomics and Epigenomics, ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- DZHK, Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Joerg Heineke
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- DZHK, Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
6
|
Hasbullah JS, Scott EN, Bhavsar AP, Gunaretnam EP, Miao F, Soliman H, Carleton BC, Ross CJD. All-trans retinoic acid (ATRA) regulates key genes in the RARG-TOP2B pathway and reduces anthracycline-induced cardiotoxicity. PLoS One 2022; 17:e0276541. [PMID: 36331922 PMCID: PMC9635745 DOI: 10.1371/journal.pone.0276541] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Abstract
The effectiveness of anthracycline chemotherapeutics (e.g., doxorubicin) is limited by anthracycline-induced cardiotoxicity (ACT). A nonsynonymous variant (S427L) in the retinoic acid receptor-γ (RARG) gene has been associated with ACT. This variant causes reduced RARG activity, which is hypothesized to lead to increased susceptibility to ACT through reduced activation of the retinoic acid pathway. This study explored the effects of activating the retinoic acid pathway using a RAR-agonist, all-trans retinoic acid (ATRA), in human cardiomyocytes and mice treated with doxorubicin. In human cardiomyocytes, ATRA induced the gene expression of RARs (RARG, RARB) and repressed the expression of topoisomerase II enzyme genes (TOP2A, TOP2B), which encode for the molecular targets of anthracyclines and repressed downstream ACT response genes. Importantly, ATRA enhanced cell survival of human cardiomyocytes exposed to doxorubicin. The protective effect of ATRA was also observed in a mouse model (B6C3F1/J) of ACT, in which ATRA treatment improved heart function compared to doxorubicin-only treated mice. Histological analyses of the heart also indicated that ATRA treatment reduced the pathology associated with ACT. These findings provide additional evidence for the retinoic acid pathway's role in ACT and suggest that the RAR activator ATRA can modulate this pathway to reduce ACT.
Collapse
Affiliation(s)
- Jafar S. Hasbullah
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Erika N. Scott
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Amit P. Bhavsar
- Department of Medical Microbiology and Immunology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Erandika P. Gunaretnam
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Fudan Miao
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Hesham Soliman
- School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce C. Carleton
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin J. D. Ross
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Villalba-Orero M, Garcia-Pavia P, Lara-Pezzi E. Non-invasive assessment of HFpEF in mouse models: current gaps and future directions. BMC Med 2022; 20:349. [PMID: 36229816 PMCID: PMC9563110 DOI: 10.1186/s12916-022-02546-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 09/01/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Heart failure (HF) with preserved ejection fraction (HFpEF) prevalence is increasing, and large clinical trials have failed to reduce mortality. A major reason for this outcome is the failure to translate results from basic research to the clinics. Evaluation of HFpEF in mouse models requires assessing three major key features defining this complex syndrome: the presence of a preserved left ventricular ejection fraction (LVEF), diastolic dysfunction, and the development of HF. In addition, HFpEF is associated with multiple comorbidities such as systemic arterial hypertension, chronic obstructive pulmonary disease, sleep apnea, diabetes, and obesity; thus, non-cardiac disorders assessment is crucial for a complete phenotype characterization. Non-invasive procedures present unquestionable advantages to maintain animal welfare and enable longitudinal analyses. However, unequivocally determining the presence of HFpEF using these methods remains challenging. MAIN TEXT Transthoracic echocardiography (TTE) represents an invaluable tool in HFpEF diagnosis, allowing evaluation of LVEF, diastolic dysfunction, and lung congestion in mice. Since conventional parameters used to evaluate an abnormal diastole like E/A ratio, isovolumic relaxation time, and E/e' may pose limitations in mice, including advanced TTE techniques to characterize cardiac motion, including an assessment under stress, will improve diagnosis. Patients with HFpEF also show electrical cardiac remodelling and therefore electrocardiography may add valuable information in mouse models to assess chronotropic incompetence and sinoatrial node dysfunction, which are major contributors to exercise intolerance. To complete the non-invasive diagnosis of HF, low aerobic exercise capacity and fatigue using exercise tests, impaired oxygen exchange using metabolic cages, and determination of blood biomarkers can be determined. Finally, since HFpEF patients commonly present non-cardiac pathological conditions, acquisition of systemic and pulmonary arterial pressures, blood glucose levels, and performing glucose tolerance and insulin resistance tests are required for a complete phenotyping. CONCLUSION Identification of reliable models of HFpEF in mice by using proper diagnosis tools is necessary to translate basic research results to the clinics. Determining the presence of several HFpEF indicators and a higher number of abnormal parameters will lead to more reliable evidence of HFpEF.
Collapse
Affiliation(s)
- María Villalba-Orero
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Av. Puerta de Hierro, s/n, 28040, Madrid, Spain. .,Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Melchor Fernández Almagro, 3, 28029, Madrid, Spain. .,Centro de investigación Biomédica en Red Cardiovascular (CIBERCV), Madrid, Spain.
| | - Pablo Garcia-Pavia
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,Centro de investigación Biomédica en Red Cardiovascular (CIBERCV), Madrid, Spain.,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, Madrid, Spain.,Universidad Francisco de Vitoria, Madrid, Spain
| | - Enrique Lara-Pezzi
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Melchor Fernández Almagro, 3, 28029, Madrid, Spain. .,Centro de investigación Biomédica en Red Cardiovascular (CIBERCV), Madrid, Spain.
| |
Collapse
|
8
|
Huang H, Chang WT, Huang CC. High-Spatiotemporal-Resolution Visualization of Myocardial Strains Through Vector Doppler Estimation: A Small-Animal Study. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:1859-1870. [PMID: 35108204 DOI: 10.1109/tuffc.2022.3148873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
High-frequency ultrasound (HFUS) imaging is extensively used for cardiac diseases in small animals due to its high spatial resolution. However, there is a lack of a system that can provide a 2-D high-spatiotemporal dynamic visualization of mouse myocardial strains. In this article, a dynamic HFUS (40 MHz) high-resolution strain imaging was developed through the vector Doppler imaging. Following in vitro tests using a rubber balloon phantom, in vivo experiments were performed on wild-type (WT) and myocardial infarction (MI) mice. High-resolution dynamic images of myocardial strains were obtained in the longitudinal, radial, and circumferential directions at a frame rate of 1 kHz. Global peak strain values for WT mice were -19.3% ± 1.3% (longitudinal), 31.4% ± 1.7% (radial in the long axis), -19.9% ±.8% (circumferential), and 34.4% ± 1.9% (radial in the short axis); those for the MI mice were -16.1% ±.9% (longitudinal), 26.8% ± 2.9% (radial in the long axis), -15.2% ± 2.7% (circumferential), and 21.6% ± 4.8% (radial in the short axis). These results indicate that the strains for MI mice are significantly lower than those for WT mice. Regional longitudinal strain curves in the epicardial, midcardial, and endocardial layers were measured and the peak strain values for WT mice were -22.% and -16.8% in the endocardial and epicardial layers, respectively. However, no significant difference in the layer-based values was noted for the MI mice. Regional analysis results revealed obvious myocardial strain variation in the apical anterior region in the MI mice. The experimental results demonstrate that the proposed dynamic cardiac strain imaging can be useful in high-performance imaging of small-animal cardiac diseases.
Collapse
|
9
|
Rajanathan R, Pedersen TM, Thomsen MB, Botker HE, Matchkov VV. Phenylephrine-Induced Cardiovascular Changes in the Anesthetized Mouse: An Integrated Assessment of in vivo Hemodynamics Under Conditions of Controlled Heart Rate. Front Physiol 2022; 13:831724. [PMID: 35250634 PMCID: PMC8891648 DOI: 10.3389/fphys.2022.831724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Investigating the cardiovascular system is challenging due to its complex regulation by humoral and neuronal factors. Despite this complexity, many existing research methods are limited to the assessment of a few parameters leading to an incomplete characterization of cardiovascular function. Thus, we aim to establish a murine in vivo model for integrated assessment of the cardiovascular system under conditions of controlled heart rate. Utilizing this model, we assessed blood pressure, cardiac output, stroke volume, total peripheral resistance, and electrocardiogram (ECG). Hypothesis We hypothesize that (i) our in vivo model can be utilized to investigate cardiac and vascular responses to pharmacological intervention with the α1-agonist phenylephrine, and (ii) we can study cardiovascular function during artificial pacing of the heart, modulating cardiac function without a direct vascular effect. Methods We included 12 mice that were randomly assigned to either vehicle or phenylephrine intervention through intraperitoneal administration. Mice were anesthetized with isoflurane and intubated endotracheally for mechanical ventilation. We measured blood pressure via a solid-state catheter in the aortic arch, blood flow via a probe on the ascending aorta, and ECG from needle electrodes on the extremities. Right atrium was electrically paced at a frequency ranging from 10 to 11.3 Hz before and after either vehicle or phenylephrine administration. Results Phenylephrine significantly increased blood pressure, stroke volume, and total peripheral resistance compared to the vehicle group. Moreover, heart rate was significantly decreased following phenylephrine administration. Pacing significantly decreased stroke volume and cardiac output both prior to and after drug administration. However, phenylephrine-induced changes in blood pressure and total peripheral resistance were maintained with increasing pacing frequencies compared to the vehicle group. Total peripheral resistance was not significantly altered with increasing pacing frequencies suggesting that the effect of phenylephrine is primarily of vascular origin. Conclusion In conclusion, this in vivo murine model is capable of distinguishing between changes in peripheral vascular and cardiac functions. This study underlines the primary effect of phenylephrine on vascular function with secondary changes to cardiac function. Hence, this in vivo model is useful for the integrated assessment of the cardiovascular system.
Collapse
Affiliation(s)
- Rajkumar Rajanathan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Rajkumar Rajanathan,
| | | | - Morten B. Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
10
|
Imaging techniques in Veterinary Medicine. Part I: Radiography and Ultrasonography. Eur J Radiol Open 2021; 8:100382. [PMID: 34712745 PMCID: PMC8529508 DOI: 10.1016/j.ejro.2021.100382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
In Veterinary Medicine all the Imaging techniques are used and described but, due to organizational, managerial and, mostly, economical reasons, Radiography and Ultrasonography are the most used. Veterinary Radiology teaching has a relatively small number of educational credits in the degree courses but, nowday, educational opportunities are remarkably increased thanks to a number of post-degree courses and masters, organized both by the universities and private veterinary associations. The relevance of Diagnostic Imaging is particularly true in Veterinary Medicine, in which Radiology could be considered “indispensable” for diagnosis, prognosis and follow up. Furthermore, it should not be forgotten that the diagnostic image represents a “document” and, has a relevant role in legal medical debats. In this first part, Radiography and Ultrasonography are described.
Collapse
|
11
|
Prospective analysis of myocardial strain through the evolution of Chagas disease in the hamster animal model. Int J Cardiovasc Imaging 2021; 38:117-129. [PMID: 34535853 PMCID: PMC8818632 DOI: 10.1007/s10554-021-02379-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Speckle tracking echocardiography (STE) enables early diagnosis of myocardial damage by evaluating myocardial strain. We aimed to study sequential changes in structural and ventricular functional parameters during Chagas disease (CD) natural history in an animal model. 37 Syrian hamsters were inoculated intraperitoneally with Trypanosoma cruzi (Chagas) and 20 with saline (Control). Echocardiography was performed before the infection (baseline), at 1 month (acute phase), 4, 6, and 8 months (chronic phase) using Vevo 2100 (Fujifilm Inc.) ultrasound system. Left ventricular end-diastolic diameter, Left ventricular end-systolic diameter (LVESD), Left ventricular ejection fraction (LVEF), Global longitudinal (GLS), circumferential (GCS) and radial (GRS) strain were evaluated. Tricuspid annular plane systolic excursion (TAPSE) was used to assess right ventricular function. At 8 months, animals were euthanized and LV myocardial samples were analyzed for quantitation of inflammation and fibrosis. LVEF decreased over time in Chagas group and a difference from Control was detected at 6 months (p-value of groups#time interaction = 0.005). There was a pronounced decrease in GLS, GCS and TAPSE in Chagas group (p-value of groups#time interaction = 0.003 for GLS, < 0.001 for GCS and < 0.009 for TAPSE vs Control) since the first month. LVESD, LVEF and GLS were significantly correlated to the number of inflammatory cells (r = 0.41, p = 0.046; r = − 0.42, p = 0.042; r = 0.41, p = 0.047) but not to fibrosis. In the Syrian hamster model of CD STE parameters (GLS and GCS) showed an early decrease. Changes in LVEF, LVESD, and GLS were correlated to myocardial inflammation but not to fibrosis.
Collapse
|
12
|
Vargas AL, Dias BDP, Moreira HT, Oliveira Filho ECD, Tanaka DM, Simões MV, Maciel BC, Schmidt A, Marin Neto JA, Romano MMD. Prospective study of ventricular function and myocardial deformation related to survival in acute Chagas disease: an experimental animal model. Rev Inst Med Trop Sao Paulo 2021; 63:e61. [PMID: 34378764 PMCID: PMC8357302 DOI: 10.1590/s1678-9946202163061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/23/2021] [Indexed: 11/22/2022] Open
Abstract
Chagas disease (CD) has been changing from an endemic Latino-American disease to a condition found outside endemic regions, due to migratory movements. Although often subclinical, its acute phase can be lethal. This study aimed to assess survival during the acute phase of CD and its relationship with ventricular function in an experimental model. To this end, 30 Syrian hamsters were inoculated with Trypanosoma cruzi (IG) and other 15 animals received saline solution (CG). Groups were monitored daily and submitted to echocardiography in two moments: before the challenge and 15 days post-infection. Left ventricular ejection fraction (LVEF) and global longitudinal myocardial strain (GLS) of the LV were measured. The IG was divided into groups of animals with and without clinical signs of disease. ANOVA for mixed models was used to compare ventricular function parameters. Survival analysis was studied using Kaplan-Meier curves and the log-rank test. The follow-up lasted 60 days. LVEF in IG was reduced through time (53.80 to 43.55%) compared to CG (57.86 to 59.73%) (p=0.002). There was also a reduction of GLS (-18.97% to -12.44%) in the IG compared to CG (p=0.012). Twelve animals from IG died compared to one animal from CG. Eleven out of the 12 animals from the IG group died before presenting with clinical signs of infection. Survival was reduced in the IG compared to CG over time (p=0.02). The reduced survival during the acute phase of this experimental model of Chagas disease was related to the significant reduction of LV function. The mortality rate in the IG was higher in the group presenting with clinical signs of infection.
Collapse
Affiliation(s)
- Arthur Lauand Vargas
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - Beatriz de Paula Dias
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - Henrique Turin Moreira
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - Edgard Camilo de Oliveira Filho
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - Denise Mayumi Tanaka
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - Marcus Vinicius Simões
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - Benedito Carlos Maciel
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - André Schmidt
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - José Antônio Marin Neto
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| | - Minna Moreira Dias Romano
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Centro de Cardiologia, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
13
|
Development of a semi-automated segmentation tool for high frequency ultrasound image analysis of mouse echocardiograms. Sci Rep 2021; 11:6559. [PMID: 33753835 PMCID: PMC7985193 DOI: 10.1038/s41598-021-85971-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 03/08/2021] [Indexed: 11/08/2022] Open
Abstract
Echocardiography is a widely used and clinically translatable imaging modality for the evaluation of cardiac structure and function in preclinical drug discovery and development. Echocardiograms are among the first in vivo diagnostic tools utilized to evaluate the heart due to its relatively low cost, high throughput acquisition, and non-invasive nature; however lengthy manual image analysis, intra- and inter-operator variability, and subjective image analysis presents a challenge for reproducible data generation in preclinical research. To combat the image-processing bottleneck and address both variability and reproducibly challenges, we developed a semi-automated analysis algorithm workflow to analyze long- and short-axis murine left ventricle (LV) ultrasound images. The long-axis B-mode algorithm executes a script protocol that is trained using a reference library of 322 manually segmented LV ultrasound images. The short-axis script was engineered to analyze M-mode ultrasound images in a semi-automated fashion using a pixel intensity evaluation approach, allowing analysts to place two seed-points to triangulate the local maxima of LV wall boundary annotations. Blinded operator evaluation of the semi-automated analysis tool was performed and compared to the current manual segmentation methodology for testing inter- and intra-operator reproducibility at baseline and after a pharmacologic challenge. Comparisons between manual and semi-automatic derivation of LV ejection fraction resulted in a relative difference of 1% for long-axis (B-mode) images and 2.7% for short-axis (M-mode) images. Our semi-automatic workflow approach reduces image analysis time and subjective bias, as well as decreases inter- and intra-operator variability, thereby enhancing throughput and improving data quality for pre-clinical in vivo studies that incorporate cardiac structure and function endpoints.
Collapse
|
14
|
The Progress of Advanced Ultrasonography in Assessing Aortic Stiffness and the Application Discrepancy between Humans and Rodents. Diagnostics (Basel) 2021; 11:diagnostics11030454. [PMID: 33800855 PMCID: PMC8001300 DOI: 10.3390/diagnostics11030454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/26/2022] Open
Abstract
Aortic stiffening is a fundamental pathological alteration of atherosclerosis and other various aging-associated vascular diseases, and it is also an independent risk factor of cardiovascular morbidity and mortality. Ultrasonography is a critical non-invasive method widely used in assessing aortic structure, function, and hemodynamics in humans, playing a crucial role in predicting the pathogenesis and adverse outcomes of vascular diseases. However, its applications in rodent models remain relatively limited, hindering the progress of the research. Here, we summarized the progress of the advanced ultrasonographic techniques applied in evaluating aortic stiffness. With multiple illustrative images, we mainly characterized various ultrasound techniques in assessing aortic stiffness based on the alterations of aortic structure, hemodynamics, and tissue motion. We also discussed the discrepancy of their applications in humans and rodents and explored the potential optimized strategies in the experimental research with animal models. This updated information would help to better understand the nature of ultrasound techniques and provide a valuable prospect for their applications in assessing aortic stiffness in basic science research, particularly with small animals.
Collapse
|
15
|
Barros Filho ACL, Moreira HT, Dias BP, Ribeiro FFF, Tanaka DM, Schmidt A, Maciel BC, Simões MV, Marin-Neto JA, Romano MMD. Feasibility and reference intervals assessed by conventional and speckle-tracking echocardiography in normal hamsters. Physiol Rep 2021; 9:e14776. [PMID: 33650789 PMCID: PMC7923569 DOI: 10.14814/phy2.14776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/05/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES This study aimed to determine feasibility, reference intervals, and reproducibility of left ventricular ejection fraction (LVEF) and speckle-tracking echocardiography (STE) in adult Syrian hamsters. BACKGROUND Syrian hamster is an experimental model for several heart diseases. Echocardiography allows the evaluation of structure and function with bidimensional conventional techniques and STE. However, there is no data regarding reference values for bidimensional LVEF and myocardial strain in hamsters. METHODS A total of 135 female Syrian hamsters were anesthetized and studied with a small animal dedicated echocardiography system. Echocardiography measurements were obtained from M-mode and B-mode images. Feasibility and 95% reference intervals were obtained for LVEF using three different approaches: LVEF_Teichholz (from M-mode linear measurements), LVEF_BMode (from area-length method), and LVEF_ STE (from strain), and for global longitudinal (GLS), circumferential (GCS), and radial (GRS) endocardial strain. Reproducibility was assessed as intra-class correlation coefficients. RESULTS Feasibility of LVEF and endocardial strain was high (95% in FEVE_Teichholz, 93% in the LVEF_BMode, 84% in the LVEF_STE, 84% from PSLAX, and 80% from PSSAX). Values of LVEF_Teichholz were significantly higher than values of LVEF_BMode, and LVEF_STE-derived methods (59.0 ± 5.8, 53.8 ± 4.7, 46.3 ± 5.7, p < 0.0001). The 95% reference intervals for GLS, GCS, and GRS were respectively -13.6(-7.5;-20.4)%, -20.5 ± 3.1%, and + 34,7 ± 7.0%. Intra-class correlation coefficients were 0.49 - 0.91 for LVEF measurements, 0.73 - 0.92 for STE, with better results for LVEF_Teichholz and GLS. CONCLUSIONS Evaluation of LVEF by several methods and STE parameters is feasible in hamsters. Reference intervals for LVEF and STE obtained for this experimental animal model can be applied at future research.
Collapse
Affiliation(s)
- Antonio C L Barros Filho
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - Henrique T Moreira
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - Beatriz P Dias
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - Fernando F F Ribeiro
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - Denise M Tanaka
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - André Schmidt
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - Benedito C Maciel
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - Marcus V Simões
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - José A Marin-Neto
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| | - Minna M D Romano
- Cardiology Center of the Medical School of Ribeirao Preto, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Li H, Qu Y, Metze P, Sommerfeld F, Just S, Abaei A, Rasche V. Quantification of Biventricular Myocardial Strain Using CMR Feature Tracking: Reproducibility in Small Animals. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8492705. [PMID: 33553431 PMCID: PMC7847329 DOI: 10.1155/2021/8492705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/16/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
Myocardial strain is a well-validated parameter for evaluating myocardial contraction. Cardiovascular magnetic resonance myocardial feature tracking (CMR-FT) is a novel method for the quantitative measurements of myocardial strain from routine cine acquisitions. In this study, we investigated the influence of temporal resolution on tracking accuracy of CMR-FT and the intraobserver, interobserver, and interstudy reproducibilities for biventricular strain analysis in mice from self-gated CMR at 11.7 T. 12 constitutive nexilin knockout (Nexn-KO) mice, heterozygous (Het, N = 6) and wild-type (WT, N = 6), were measured with a well-established self-gating sequence twice within two weeks. CMR-FT measures of biventricular global and segmental strain parameters were derived. Interstudy, intraobserver, and interobserver reproducibilities were investigated. For the assessment of the impact of the temporal resolution for the outcome in CMR-FT, highly oversampled semi-4 chamber and midventricular short-axis data were acquired and reconstructed with 10 to 80 phases per cardiac cycle. A generally reduced biventricular myocardial strain was observed in Nexn-KO Het mice. Excellent intraobserver and interobserver reproducibility was achieved in all global strains (ICC range from 0.76 to 0.99), where global right ventricle circumferential strain (RCSSAX) showed an only good interobserver reproducibility (ICC 0.65, 0.11-0.89). For interstudy reproducibility, left ventricle longitudinal strain (LLSLAX) was the most reproducible measure of strain (ICC 0.90, 0.71-0.97). The left ventricle radial strain (LRSSAX) (ICC 0.50, 0.10-0.83) showed fair reproducibility and RCSSAX (ICC 0.36, 0.14-0.74) showed only poor reproducibility. In general, compared with global strains, the segmental strains showed relatively lower reproducibility. A minimal temporal resolution of 20 phases per cardiac cycle appeared sufficient for CMR-FT strain analysis. The analysis of myocardial strain from high-resolution self-gated cine images by CMR-FT provides a highly reproducible method for assessing myocardial contraction in small rodent animals. Especially, global LV longitudinal and circumferential strain revealed excellent reproducibility of intra- and interobserver and interstudy measurements.
Collapse
Affiliation(s)
- Hao Li
- Core Facility Small Animal Imaging, Ulm University, Ulm, Germany
| | - Yangyang Qu
- Department of Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| | - Patrick Metze
- Department of Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| | | | - Steffen Just
- Department of Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| | - Alireza Abaei
- Core Facility Small Animal Imaging, Ulm University, Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal Imaging, Ulm University, Ulm, Germany
- Department of Internal Medicine II, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
17
|
Sztechman D, Żera T, Czarzasta K, Wojciechowska M, Szczepańska-Sadowska E, Cudnoch-Jędrzejewska A. Transthoracic echocardiography: from guidelines for humans to cardiac ultrasound of the heart in rats. Physiol Meas 2020; 41:10TR02. [PMID: 33164918 DOI: 10.1088/1361-6579/abb3a3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ultrasound examination of the heart is a cornerstone of clinical evaluation of patients with established or suspected cardiovascular conditions. Advancements in ultrasound imaging technology have brought transthoracic echocardiography to preclinical murine models of cardiovascular diseases. The translational potential of cardiac ultrasound is critically important in rat models of myocardial infarction and ischemia-reperfusion injury, congestive heart failure, arterial hypertension, cardiac hypertrophy, pulmonary hypertension, right heart failure, Takotsubo cardiomyopathy, hypertrophic and dilated cardiomyopathies, developmental disorders, and metabolic syndrome. Modern echocardiographic machines capable of high-frame-rate image acquisition and fitted with high-frequency transducers allow for cardiac ultrasound in rats that yields most of the echocardiographic measurements and indices recommended by international guidelines for cardiac ultrasound in human patients. Among them are dimensions of cardiac chambers and walls, indices of systolic and diastolic cardiac function, and valvular function. In addition, measurements of cardiac dimensions and ejection fraction can be significantly improved by intravenous administration of ultrasound enhancing agents (UEAs). In this article we discuss echocardiography in rats, describe a technique for minimally invasive intravenous administration of UEAs via the saphenous vein and present a step-by-step approach to cardiac ultrasound in rats.
Collapse
Affiliation(s)
- Dorota Sztechman
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
18
|
Anti-apoptotic peptide for long term cardioprotection in a mouse model of myocardial ischemia-reperfusion injury. Sci Rep 2020; 10:18116. [PMID: 33093627 PMCID: PMC7582178 DOI: 10.1038/s41598-020-75154-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/05/2020] [Indexed: 01/19/2023] Open
Abstract
Reperfusion therapy during myocardial infarction (MI) leads to side effects called ischemia–reperfusion (IR) injury for which no treatment exists. While most studies have targeted the intrinsic apoptotic pathway to prevent IR injury with no successful clinical translation, we evidenced recently the potent cardioprotective effect of the anti-apoptotic Tat-DAXXp (TD) peptide targeting the FAS-dependent extrinsic pathway. The aim of the present study was to evaluate TD long term cardioprotective effects against IR injury in a MI mouse model. TD peptide (1 mg/kg) was administered in mice subjected to MI (TD; n = 21), 5 min prior to reperfusion, and were clinically followed-up during 6 months after surgery. Plasma cTnI concentration evaluated 24 h post-MI was 70%-decreased in TD (n = 16) versus Ctrl (n = 20) mice (p***). Strain echocardiography highlighted a 24%-increase (p****) in the ejection fraction mean value in TD-treated (n = 12) versus Ctrl mice (n = 17) during the 6 month-period. Improved cardiac performance was associated to a 54%-decrease (p**) in left ventricular fibrosis at 6 months in TD (n = 16) versus Ctrl (n = 20). In conclusion, targeting the extrinsic pathway with TD peptide at the onset of reperfusion provided long-term cardioprotection in a mouse model of myocardial IR injury by improving post-MI cardiac performance and preventing cardiac remodeling.
Collapse
|
19
|
Alonso-Herranz L, Sahún-Español Á, Paredes A, Gonzalo P, Gkontra P, Núñez V, Clemente C, Cedenilla M, Villalba-Orero M, Inserte J, García-Dorado D, Arroyo AG, Ricote M. Macrophages promote endothelial-to-mesenchymal transition via MT1-MMP/TGFβ1 after myocardial infarction. eLife 2020; 9:57920. [PMID: 33063665 PMCID: PMC7609061 DOI: 10.7554/elife.57920] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022] Open
Abstract
Macrophages (Mφs) produce factors that participate in cardiac repair and remodeling after myocardial infarction (MI); however, how these factors crosstalk with other cell types mediating repair is not fully understood. Here we demonstrated that cardiac Mφs increased the expression of Mmp14 (MT1-MMP) 7 days post-MI. We selectively inactivated the Mmp14 gene in Mφs using a genetic strategy (Mmp14f/f:Lyz2-Cre). This conditional KO (MAC-Mmp14 KO) resulted in attenuated post-MI cardiac dysfunction, reduced fibrosis, and preserved cardiac capillary network. Mechanistically, we showed that MT1-MMP activates latent TGFβ1 in Mφs, leading to paracrine SMAD2-mediated signaling in endothelial cells (ECs) and endothelial-to-mesenchymal transition (EndMT). Post-MI MAC-Mmp14 KO hearts contained fewer cells undergoing EndMT than their wild-type counterparts, and Mmp14-deficient Mφs showed a reduced ability to induce EndMT in co-cultures with ECs. Our results indicate the contribution of EndMT to cardiac fibrosis and adverse remodeling post-MI and identify Mφ MT1-MMP as a key regulator of this process.
Collapse
Affiliation(s)
- Laura Alonso-Herranz
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Álvaro Sahún-Español
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Paredes
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Pilar Gonzalo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Polyxeni Gkontra
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Vanessa Núñez
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina Clemente
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Marta Cedenilla
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María Villalba-Orero
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Vall d'Hebron University Hospital and Research Institute (VHIR), Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - David García-Dorado
- Cardiovascular Diseases Research Group, Vall d'Hebron University Hospital and Research Institute (VHIR), Barcelona, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Alicia G Arroyo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Mercedes Ricote
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
20
|
Costantino S, Akhmedov A, Melina G, Mohammed SA, Othman A, Ambrosini S, Wijnen WJ, Sada L, Ciavarella GM, Liberale L, Tanner FC, Matter CM, Hornemann T, Volpe M, Mechta-Grigoriou F, Camici GG, Sinatra R, Lüscher TF, Paneni F. Obesity-induced activation of JunD promotes myocardial lipid accumulation and metabolic cardiomyopathy. Eur Heart J 2020; 40:997-1008. [PMID: 30629164 DOI: 10.1093/eurheartj/ehy903] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/24/2018] [Accepted: 12/19/2018] [Indexed: 12/24/2022] Open
Abstract
AIMS Metabolic cardiomyopathy (MC)-characterized by intra-myocardial triglyceride (TG) accumulation and lipotoxic damage-is an emerging cause of heart failure in obese patients. Yet, its mechanisms remain poorly understood. The Activator Protein 1 (AP-1) member JunD was recently identified as a key modulator of hepatic lipid metabolism in obese mice. The present study investigates the role of JunD in obesity-induced MC. METHODS AND RESULTS JunD transcriptional activity was increased in hearts from diet-induced obese (DIO) mice and was associated with myocardial TG accumulation and left ventricular (LV) dysfunction. Obese mice lacking JunD were protected against MC. In DIO hearts, JunD directly binds PPARγ promoter thus enabling transcription of genes involved in TG synthesis, uptake, hydrolysis, and storage (i.e. Fas, Cd36, Lpl, Plin5). Cardiac-specific overexpression of JunD in lean mice led to PPARγ activation, cardiac steatosis, and dysfunction, thereby mimicking the MC phenotype. In DIO hearts as well as in neonatal rat ventricular myocytes exposed to palmitic acid, Ago2 immunoprecipitation, and luciferase assays revealed JunD as a direct target of miR-494-3p. Indeed, miR-494-3p was down-regulated in hearts from obese mice, while its overexpression prevented lipotoxic damage by suppressing JunD/PPARγ signalling. JunD and miR-494-3p were also dysregulated in myocardial specimens from obese patients as compared with non-obese controls, and correlated with myocardial TG content, expression of PPARγ-dependent genes, and echocardiographic indices of LV dysfunction. CONCLUSION miR-494-3p/JunD is a novel molecular axis involved in obesity-related MC. These results pave the way for approaches to prevent or treat LV dysfunction in obese patients.
Collapse
Affiliation(s)
- Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| | - Giovanni Melina
- Department of Cardiac Surgery, Sant'Andrea Hospital, "Sapienza" University, Via di Grottarossa, 1035, Rome, Italy
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| | - Alaa Othman
- Institute for Clinical Chemistry, University Hospital Zürich, Ramistrasse 100, Zurich, Switzerland
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| | - Winandus J Wijnen
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| | - Lidia Sada
- Department of Clinical and Molecular Medicine, Cardiology Unit, Faculty of Medicine and Psychology, "Sapienza" University, Via di Grottarossa, 1035, Rome, Italy
| | - Giuseppino M Ciavarella
- Department of Clinical and Molecular Medicine, Cardiology Unit, Faculty of Medicine and Psychology, "Sapienza" University, Via di Grottarossa, 1035, Rome, Italy
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland.,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, Genoa, Italy
| | - Felix C Tanner
- University Heart Center, Cardiology, University Hospital Zürich, Ramistrasse 100, Zurich, Switzerland
| | - Christian M Matter
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland.,University Heart Center, Cardiology, University Hospital Zürich, Ramistrasse 100, Zurich, Switzerland
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital Zürich, Ramistrasse 100, Zurich, Switzerland
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Cardiology Unit, Faculty of Medicine and Psychology, "Sapienza" University, Via di Grottarossa, 1035, Rome, Italy.,IRCCS Neuromed, Pozzilli, Via Atinense, 18, Pozzilli (IS), Italy
| | - Fatima Mechta-Grigoriou
- Institut Curie, Stress and Cancer Laboratory, Equipe Labelisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, Paris, France.,Inserm, U830, 26, rue d'Ulm, Paris, France
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland.,University Heart Center, Cardiology, University Hospital Zürich, Ramistrasse 100, Zurich, Switzerland
| | - Riccardo Sinatra
- Department of Cardiac Surgery, Sant'Andrea Hospital, "Sapienza" University, Via di Grottarossa, 1035, Rome, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland.,Cardiology, Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland.,University Heart Center, Cardiology, University Hospital Zürich, Ramistrasse 100, Zurich, Switzerland
| |
Collapse
|
21
|
Zacchigna S, Paldino A, Falcão-Pires I, Daskalopoulos EP, Dal Ferro M, Vodret S, Lesizza P, Cannatà A, Miranda-Silva D, Lourenço AP, Pinamonti B, Sinagra G, Weinberger F, Eschenhagen T, Carrier L, Kehat I, Tocchetti CG, Russo M, Ghigo A, Cimino J, Hirsch E, Dawson D, Ciccarelli M, Oliveti M, Linke WA, Cuijpers I, Heymans S, Hamdani N, de Boer M, Duncker DJ, Kuster D, van der Velden J, Beauloye C, Bertrand L, Mayr M, Giacca M, Leuschner F, Backs J, Thum T. Towards standardization of echocardiography for the evaluation of left ventricular function in adult rodents: a position paper of the ESC Working Group on Myocardial Function. Cardiovasc Res 2020; 117:43-59. [PMID: 32365197 DOI: 10.1093/cvr/cvaa110] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/28/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Echocardiography is a reliable and reproducible method to assess non-invasively cardiac function in clinical and experimental research. Significant progress in the development of echocardiographic equipment and transducers has led to the successful translation of this methodology from humans to rodents, allowing for the scoring of disease severity and progression, testing of new drugs, and monitoring cardiac function in genetically modified or pharmacologically treated animals. However, as yet, there is no standardization in the procedure to acquire echocardiographic measurements in small animals. This position paper focuses on the appropriate acquisition and analysis of echocardiographic parameters in adult mice and rats, and provides reference values, representative images, and videos for the accurate and reproducible quantification of left ventricular function in healthy and pathological conditions.
Collapse
Affiliation(s)
- Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy.,International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessia Paldino
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy
| | - Inês Falcão-Pires
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Evangelos P Daskalopoulos
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Belgium, Brussels
| | - Matteo Dal Ferro
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy
| | - Simone Vodret
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Pierluigi Lesizza
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy
| | - Antonio Cannatà
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy
| | - Daniela Miranda-Silva
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - André P Lourenço
- Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Bruno Pinamonti
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy
| | - Gianfranco Sinagra
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy
| | - Florian Weinberger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Izhak Kehat
- Department of Physiology, Biophysics and System Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Michele Russo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - James Cimino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | | | | | - Wolfgang A Linke
- Institute of Physiology 2, University of Muenster, Muenster, Germany
| | - Ilona Cuijpers
- Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands.,Center of Molecular and Vascular Biology (CMVB), KU Leuven, Leuven, Belgium
| | - Stephane Heymans
- Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands.,Center of Molecular and Vascular Biology (CMVB), KU Leuven, Leuven, Belgium
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Division Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Martine de Boer
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Diederik Kuster
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
| | - Christophe Beauloye
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Belgium, Brussels.,Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), Belgium, Brussels
| | - Manuel Mayr
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina, strada di Fiume 447, 34149 Trieste (TS), Italy.,International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Florian Leuschner
- Institute of Experimental Cardiology, Department of Cardiology, Angiology & Pulmology, Heidelberg University Hospital, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, Department of Cardiology, Angiology & Pulmology, Heidelberg University Hospital, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
Palmisano A, Piccoli M, Monti CB, Canu T, Cirillo F, Napolitano A, Perani L, Signorelli P, Vignale D, Anastasia L, Esposito A. Single-shot morpho-functional and structural characterization of the left-ventricle in a mouse model of acute ischemia-reperfusion injury with an optimized 3D IntraGate cine FLASH sequence at 7T MR. Magn Reson Imaging 2020; 68:127-135. [PMID: 32004712 DOI: 10.1016/j.mri.2020.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/08/2020] [Accepted: 01/26/2020] [Indexed: 11/16/2022]
Abstract
Preclinical cardiac MR is challenging and time-consuming. A fast and comprehensive acquisition protocol and standardized image post-processing may improve preclinical research, reducing acquisition time, costs and variability of results. In the present study, we evaluated the feasibility of a contrast-enhanced 3D IntraGate steady-state cine sequence (ce-3D-IG-cine) with short acquisition time (11 min) for a single-shot combined characterization of left ventricle (LV) remodeling and infarct size (IS) in a mouse model of acute ischemia-reperfusion injury. Sixteen male C57BL/6N mice underwent 7T cardiac MR (Bruker, BioSpec 70/30) including optimized ce-3D-IG-cine (total scan time 11 min) at day 1, 5 and 28 after surgery. LV end-diastolic volume (EDVMR) and ejection fraction (EFMR) extracted from MR were compared to ones from short-axis (SA-EDVecho, SA-EFecho) and parasternal long-axis (LA-EDVecho, LA-EFecho) echocardiography. IS was manually and semiautomatically segmented from ce-3D-IG-cine using different standard deviation (SD +2, +3, +4, +5, +6 in respect to a reference tissue). Mice were sacrificed at day 28, immediately after imaging. IS at day 28 was compared to injury burden at histology. MR and echocardiographic morpho-functional parameters were compared, as IS from MR and histology. Bland-Altman plots were used to assess the agreement in ischemic burden segmentation. Volumetric and functional parameters measured on ce-3D-IG-cine correlated to the correspondent echocardiographic parameter (EDVMR vs SA-EDVecho: ρ = 0.813; EDVMR vs LA-EDVecho: ρ = 0.845; EFMR vs SA-EFecho ρ = 0.612; EFMR vs LA-EFecho ρ = 0.791; p < 0.001 in all cases). Manually segmented IS strongly correlated with the scar at histology (ρ = 0.904, p < 0.001). A threshold of +3SD showed the highest performance for semiautomatic assessment of IS compared to manual segmentation (ρ = 0.965, p < 0.001), with an overall reproducibility of 73%, and a peak reproducibility of 80% at day 1. The ce-3D-IG-cine sequence, manually or semiautomatically segmented using 3SD threshold, allows fast and comprehensive LV morpho-functional and structural characterization in myocardial ischemia-reperfusion injury model.
Collapse
Affiliation(s)
- Anna Palmisano
- Vita-Salute San Raffaele University, Milan, Italy; Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Piccoli
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | | | - Tamara Canu
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Cirillo
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Angela Napolitano
- Vita-Salute San Raffaele University, Milan, Italy; Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Perani
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Signorelli
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Davide Vignale
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Anastasia
- Vita-Salute San Raffaele University, Milan, Italy; Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Antonio Esposito
- Vita-Salute San Raffaele University, Milan, Italy; Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
23
|
de Lucia C, Wallner M, Eaton DM, Zhao H, Houser SR, Koch WJ. Echocardiographic Strain Analysis for the Early Detection of Left Ventricular Systolic/Diastolic Dysfunction and Dyssynchrony in a Mouse Model of Physiological Aging. J Gerontol A Biol Sci Med Sci 2019; 74:455-461. [PMID: 29917053 PMCID: PMC6417453 DOI: 10.1093/gerona/gly139] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Indexed: 01/31/2023] Open
Abstract
Heart disease is the leading cause of hospitalization and death worldwide, severely affecting health care costs. Aging is a significant risk factor for heart disease, and the senescent heart is characterized by structural and functional changes including diastolic and systolic dysfunction as well as left ventricular (LV) dyssynchrony. Speckle tracking-based strain echocardiography (STE) has been shown as a noninvasive, reproducible, and highly sensitive methodology to evaluate LV function in both animal models and humans. Herein, we describe the efficiency of this technique as a comprehensive and sensitive method for the detection of age-related cardiac dysfunction in mice. Compared with conventional echocardiographic measurements, radial and longitudinal strain, and reverse longitudinal strain were able to detect subtle changes in systolic and diastolic cardiac function in mice at an earlier time point during aging. Additionally, the data show a gradual and consistent decrease with age in regional contractility throughout the entire LV, in both radial and longitudinal axes. Furthermore, we observed that LV segmental dyssynchrony in longitudinal axis reliably differentiated between aged and young mice. Therefore, we propose the use of echocardiographic strain as a highly sensitive and accurate technology enabling and evaluating the effect of new treatments to fight age-induced cardiac disease.
Collapse
Affiliation(s)
- Claudio de Lucia
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Markus Wallner
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Deborah M Eaton
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Huaqing Zhao
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Steven R Houser
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Walter J Koch
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Acuna A, Berman AG, Damen FW, Meyers BA, Adelsperger AR, Bayer KC, Brindise MC, Bungart B, Kiel AM, Morrison RA, Muskat JC, Wasilczuk KM, Wen Y, Zhang J, Zito P, Goergen CJ. Computational Fluid Dynamics of Vascular Disease in Animal Models. J Biomech Eng 2019; 140:2676341. [PMID: 29570754 DOI: 10.1115/1.4039678] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Indexed: 12/19/2022]
Abstract
Recent applications of computational fluid dynamics (CFD) applied to the cardiovascular system have demonstrated its power in investigating the impact of hemodynamics on disease initiation, progression, and treatment outcomes. Flow metrics such as pressure distributions, wall shear stresses (WSS), and blood velocity profiles can be quantified to provide insight into observed pathologies, assist with surgical planning, or even predict disease progression. While numerous studies have performed simulations on clinical human patient data, it often lacks prediagnosis information and can be subject to large intersubject variability, limiting the generalizability of findings. Thus, animal models are often used to identify and manipulate specific factors contributing to vascular disease because they provide a more controlled environment. In this review, we explore the use of CFD in animal models in recent studies to investigate the initiating mechanisms, progression, and intervention effects of various vascular diseases. The first section provides a brief overview of the CFD theory and tools that are commonly used to study blood flow. The following sections are separated by anatomical region, with the abdominal, thoracic, and cerebral areas specifically highlighted. We discuss the associated benefits and obstacles to performing CFD modeling in each location. Finally, we highlight animal CFD studies focusing on common surgical treatments, including arteriovenous fistulas (AVF) and pulmonary artery grafts. The studies included in this review demonstrate the value of combining CFD with animal imaging and should encourage further research to optimize and expand upon these techniques for the study of vascular disease.
Collapse
Affiliation(s)
- Andrea Acuna
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Alycia G Berman
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Frederick W Damen
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Brett A Meyers
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907 e-mail:
| | - Amelia R Adelsperger
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Kelsey C Bayer
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Melissa C Brindise
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907 e-mail:
| | - Brittani Bungart
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Alexander M Kiel
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Rachel A Morrison
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Joseph C Muskat
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Kelsey M Wasilczuk
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Yi Wen
- Department of Agricultural and Biological Engineering, Purdue University, 225 South University Street, West Lafayette, IN 47907 e-mail:
| | - Jiacheng Zhang
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907 e-mail:
| | - Patrick Zito
- Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| | - Craig J Goergen
- ASME Membership Bioengineering Division, Weldon School of Biomedical Engineering, Purdue University, 206 S. Martin Jischke Drive, West Lafayette, IN 47907 e-mail:
| |
Collapse
|
25
|
van Opbergen CJM, Noorman M, Pfenniger A, Copier JS, Vermij SH, Li Z, van der Nagel R, Zhang M, de Bakker JMT, Glass AM, Mohler PJ, Taffet SM, Vos MA, van Rijen HVM, Delmar M, van Veen TAB. Plakophilin-2 Haploinsufficiency Causes Calcium Handling Deficits and Modulates the Cardiac Response Towards Stress. Int J Mol Sci 2019; 20:E4076. [PMID: 31438494 PMCID: PMC6747156 DOI: 10.3390/ijms20174076] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 01/06/2023] Open
Abstract
Human variants in plakophilin-2 (PKP2) associate with most cases of familial arrhythmogenic cardiomyopathy (ACM). Recent studies show that PKP2 not only maintains intercellular coupling, but also regulates transcription of genes involved in Ca2+ cycling and cardiac rhythm. ACM penetrance is low and it remains uncertain, which genetic and environmental modifiers are crucial for developing the cardiomyopathy. In this study, heterozygous PKP2 knock-out mice (PKP2-Hz) were used to investigate the influence of exercise, pressure overload, and inflammation on a PKP2-related disease progression. In PKP2-Hz mice, protein levels of Ca2+-handling proteins were reduced compared to wildtype (WT). PKP2-Hz hearts exposed to voluntary exercise training showed right ventricular lateral connexin43 expression, right ventricular conduction slowing, and a higher susceptibility towards arrhythmias. Pressure overload increased levels of fibrosis in PKP2-Hz hearts, without affecting the susceptibility towards arrhythmias. Experimental autoimmune myocarditis caused more severe subepicardial fibrosis, cell death, and inflammatory infiltrates in PKP2-Hz hearts than in WT. To conclude, PKP2 haploinsufficiency in the murine heart modulates the cardiac response to environmental modifiers via different mechanisms. Exercise upon PKP2 deficiency induces a pro-arrhythmic cardiac remodeling, likely based on impaired Ca2+ cycling and electrical conduction, versus structural remodeling. Pathophysiological stimuli mainly exaggerate the fibrotic and inflammatory response.
Collapse
Affiliation(s)
- Chantal J M van Opbergen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands
| | - Maartje Noorman
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands
| | - Anna Pfenniger
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
| | - Jaël S Copier
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands
| | - Sarah H Vermij
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern 3012, Switzerland
| | - Zhen Li
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
| | - Roel van der Nagel
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands
| | - Mingliang Zhang
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
| | - Jacques M T de Bakker
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands
- Department of Medical Biology, Academic Medical Center Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Aaron M Glass
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH 43210, USA
- Departments of Physiology & Cell Biology and Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University College of Medicine Wexner Medical Center, Columbus, OH 43210, USA
| | - Steven M Taffet
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Marc A Vos
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands
| | - Harold V M van Rijen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands
| | - Mario Delmar
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
| | - Toon A B van Veen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, Utrecht 3584CM, The Netherlands.
| |
Collapse
|
26
|
Li X, Sun Y, Zhang X, Wang J. Reductions in gut microbiota‑derived metabolite trimethylamine N‑oxide in the circulation may ameliorate myocardial infarction‑induced heart failure in rats, possibly by inhibiting interleukin‑8 secretion. Mol Med Rep 2019; 20:779-786. [PMID: 31180562 DOI: 10.3892/mmr.2019.10297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/25/2019] [Indexed: 11/06/2022] Open
Abstract
Myocardial infarction (MI) is a common cause of chronic heart failure (HF). Increasing evidence has revealed that trimethylamine N‑oxide (TMAO), a gut‑microbiota‑derived metabolite, contributes to the pathogenesis of cardiovascular disease by promoting inflammation. Elevated levels of circulating TMAO have been reported in patients following MI and were associated with unfavorable outcomes. The present study examined whether reductions in circulating TMAO could attenuate the progression of HF in rats following MI. Sprague‑Dawley rats underwent coronary ligation to induce MI or a sham operation. Echocardiography confirmed MI and cardiac dysfunction one day following coronary ligation. MI and sham rats were then treated with either vehicle (tap water) or 1.0% 3,3‑dimethyl‑1‑butanol (DMB, a trimethylamine formation inhibitor) in tap water, for 8 weeks. At the end of the experiment, TMAO plasma levels were markedly elevated in vehicle‑treated MI rats compared with vehicle‑treated sham rats; however, TMAO plasma levels were reduced in DMB‑treated MI rats compared with vehicle‑treated MI rats. Both MI groups exhibited cardiac hypertrophy, lung congestion, left ventricular remodeling and impaired cardiac function, according to the results of anatomical analysis, echocardiography and left ventricular hemodynamics; however, these manifestations of MI‑induced HF were significantly improved in DMB‑treated MI rats compared with vehicle‑treated MI rats. The plasma levels of the chemokine interleukin (IL)‑8, and cardiac expression of IL‑8 and its receptors were significantly increased in vehicle‑treated MI rats compared with vehicle‑treated sham rats; however, these were normalized in DMB‑treated MI rats. In addition, elevated TMAO plasma level was positively correlated with increased IL‑8 plasma level in MI groups. Notably, DMB treatment of sham rats also reduced plasma TMAO, but did not alter other parameters. These results indicated that reducing circulating TMAO may ameliorate the development of chronic HF following MI in rats, potentially by inhibiting IL‑8 secretion. The results from the present study suggested that inhibition of TMAO synthesis may be considered as a novel therapeutic approach for the prevention and treatment of patients with chronic MI‑induced HF.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Yongcun Sun
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Xinru Zhang
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Jing Wang
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
27
|
Ziegler KA, Ahles A, Wille T, Kerler J, Ramanujam D, Engelhardt S. Local sympathetic denervation attenuates myocardial inflammation and improves cardiac function after myocardial infarction in mice. Cardiovasc Res 2019; 114:291-299. [PMID: 29186414 PMCID: PMC5852629 DOI: 10.1093/cvr/cvx227] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/23/2017] [Indexed: 01/09/2023] Open
Abstract
Aims Cardiac inflammation has been suggested to be regulated by the sympathetic nervous system (SNS). However, due to the lack of methodology to surgically eliminate the myocardial SNS in mice, neuronal control of cardiac inflammation remains ill-defined. Here, we report a procedure for local cardiac sympathetic denervation in mice and tested its effect in a mouse model of heart failure post-myocardial infarction. Methods and results Upon preparation of the carotid bifurcation, the right and the left superior cervical ganglia were localized and their pre- and postganglionic branches dissected before removal of the ganglion. Ganglionectomy led to an almost entire loss of myocardial sympathetic innervation in the left ventricular anterior wall. When applied at the time of myocardial infarction (MI), cardiac sympathetic denervation did not affect acute myocardial damage and infarct size. In contrast, cardiac sympathetic denervation significantly attenuated chronic consequences of MI, including myocardial inflammation, myocyte hypertrophy, and overall cardiac dysfunction. Conclusion These data suggest a critical role for local sympathetic control of cardiac inflammation. Our model of myocardial sympathetic denervation in mice should prove useful to further dissect the molecular mechanisms underlying cardiac neural control.
Collapse
Affiliation(s)
- Karin A Ziegler
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany
| | - Andrea Ahles
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, Munich 80802, Germany
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, Munich 80937, Germany
| | - Julia Kerler
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, Munich 80802, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, Munich 80802, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, Munich 80802, Germany
| |
Collapse
|
28
|
Accurate assessment of LV function using the first automated 2D-border detection algorithm for small animals - evaluation and application to models of LV dysfunction. Cardiovasc Ultrasound 2019; 17:7. [PMID: 31010431 PMCID: PMC6477743 DOI: 10.1186/s12947-019-0156-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/26/2019] [Indexed: 01/01/2023] Open
Abstract
Echocardiography is the most commonly applied technique for non-invasive assessment of cardiac function in small animals. Manual tracing of endocardial borders is time consuming and varies with operator experience. Therefore, we aimed to evaluate a novel automated two-dimensional software algorithm (Auto2DE) for small animals and compare it to the standard use of manual 2D-echocardiographic assessment (2DE). We hypothesized that novel Auto2DE will provide rapid and robust data sets, which are in agreement with manually assessed data of animals.2DE and Auto2DE were carried out using a high-resolution imaging-system for small animals. First, validation cohorts of mouse and rat cine loops were used to compare Auto2DE against 2DE. These data were stratified for image quality by a blinded expert in small animal imaging. Second, we evaluated 2DE and Auto2DE in four mouse models and four rat models with different cardiac pathologies.Automated assessment of LV function by 2DE was faster than conventional 2DE analysis and independent of operator experience levels. The accuracy of Auto2DE-assessed data in healthy mice was dependent on cine loop quality, with excellent agreement between Auto2DE and 2DE in cine loops with adequate quality. Auto2DE allowed for valid detection of impaired cardiac function in animal models with pronounced cardiac phenotypes, but yielded poor performance in diabetic animal models independent of image quality.Auto2DE represents a novel automated analysis tool for rapid assessment of LV function, which is suitable for data acquisition in studies with good and very good echocardiographic image quality, but presents systematic problems in specific pathologies.
Collapse
|
29
|
Zhang M, Wu J, Sun R, Tao X, Wang X, Kang Q, Wang H, Zhang L, Liu P, Zhang J, Xia Y, Zhao Y, Yang Y, Xiong Y, Guan KL, Zou Y, Ye D. SIRT5 deficiency suppresses mitochondrial ATP production and promotes AMPK activation in response to energy stress. PLoS One 2019; 14:e0211796. [PMID: 30759120 PMCID: PMC6373945 DOI: 10.1371/journal.pone.0211796] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 01/21/2019] [Indexed: 01/30/2023] Open
Abstract
Sirtuin 5 (SIRT5) is a member of the NAD+-dependent sirtuin family of protein deacylase that catalyzes removal of post-translational modifications, such as succinylation, malonylation, and glutarylation on lysine residues. In light of the SIRT5's roles in regulating mitochondrion function, we show here that SIRT5 deficiency leads to suppression of mitochondrial NADH oxidation and inhibition of ATP synthase activity. As a result, SIRT5 deficiency decreases mitochondrial ATP production, increases AMP/ATP ratio, and subsequently activates AMP-activated protein kinase (AMPK) in cultured cells and mouse hearts under energy stress conditions. Moreover, Sirt5 knockout attenuates transverse aortic constriction (TAC)-induced cardiac hypertrophy and cardiac dysfunction in mice, which is associated with decreased ATP level, increased AMP/ATP ratio and enhanced AMPK activation. Our study thus uncovers an important role of SIRT5 in regulating cellular energy metabolism and AMPK activation in response to energy stress.
Collapse
Affiliation(s)
- Mengli Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Renqiang Sun
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoting Tao
- Department of Thoracic Surgery and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoxia Wang
- Waters corporation Shanghai Science & Technology Co Ltd, Shanghai, China
| | - Qi Kang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Wang
- Waters corporation Shanghai Science & Technology Co Ltd, Shanghai, China
| | - Lei Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Liu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinye Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yukun Xia
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuzheng Zhao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yi Yang
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yue Xiong
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
- Lineberger Comprehensive Cancer Center, Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Kun-Liang Guan
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, California, United States
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Dan Ye
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and The Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Oliveira AC, Melo MB, Motta-Santos D, Peluso AA, Souza-Neto F, da Silva RF, Almeida JFQ, Canta G, Reis AM, Goncalves G, Cerri G, Coutinho D, Guedes de Jesus IC, Guatimosim S, Linhares ND, Alenina N, Bader M, Campagnole-Santos MJ, Santos RAS. Genetic deletion of the alamandine receptor MRGD leads to dilated cardiomyopathy in mice. Am J Physiol Heart Circ Physiol 2019; 316:H123-H133. [DOI: 10.1152/ajpheart.00075.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have recently described a new peptide of the renin-angiotensin system, alamandine, a derivative of angiotensin-(1–7). Mas-related G protein-coupled receptor member D (MrgD) was identified as its receptor. Although similar cardioprotective effects of alamandine to those of angiotensin-(1–7) have been described, the significance of this peptide in heart function is still elusive. We aimed to evaluate the functional role of the alamandine receptor MrgD in the heart using MrgD-deficient mice. MrgD was localized in cardiomyocytes by immunofluorescence using confocal microscopy. High-resolution echocardiography was performed in wild-type and MrgD-deficient mice (2 and 12 wk old) under isoflurane anesthesia. Standard B-mode images were obtained in the right and left parasternal long and short axes for morphological and functional assessment and evaluation of cardiac deformation. Additional heart function evaluation was performed using Langendorff isolated heart preparations and inotropic measurements of isolated cardiomyocytes. Immunofluorescence indicated that the MrgD receptor is expressed in cardiomyocytes, mainly in the membrane and perinuclear and nuclear regions. Echocardiography showed left ventricular remodeling and severe dysfunction in MrgD-deficient mice. Strikingly, MrgD-deficient mice presented a pronounced dilated cardiomyopathy with a marked decrease in systolic function. Echocardiographic changes were supported by the data obtained in isolated hearts and inotropic measurements in cardiomyocytes. Our data add new evidence for a major role for alamandine/MrgD in the heart. Furthermore, our results indicate that we have identified a new gene implicated in dilated cardiomyopathy, unveiling a new target for translational approaches aimed to treat heart diseases. NEW & NOTEWORTHY The renin-angiotensin system is a key target for cardiovascular therapy. We have recently identified a new vasodepressor/cardioprotective angiotensin, alamandine. Here, we unmasked a key role for its receptor, Mas-related G protein-coupled receptor member D (MrgD), in heart function. The severe dilated cardiomyopathy observed in MrgD-deficient mice warrants clinical and preclinical studies to unveil its potential use in cardiovascular therapy. Listen to this article’s corresponding podcast at https://ajpheart.podbean.com/e/mrgd-deficiency-leads-to-dilated-cardiomyopathy/ .
Collapse
Affiliation(s)
- Aline Cristina Oliveira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Marcos Barrouin Melo
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Daisy Motta-Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - A. Augusto Peluso
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Fernando Souza-Neto
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Rafaela F. da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Jonathas F. Q. Almeida
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Giovanni Canta
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Adelina M. Reis
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Gleisy Goncalves
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela Cerri
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Danielle Coutinho
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Itamar Couto Guedes de Jesus
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Natalia D. Linhares
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- German Center for Cardiovascular Research, Berlin Partner Site, Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- German Center for Cardiovascular Research, Berlin Partner Site, Berlin, Germany
- Charite-University Medicine, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Maria José Campagnole-Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| | - Robson A. Souza Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute in Science and Technology NanoBioFar, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
31
|
Shah A, Cooke CLM, Kirschenman RD, Quon AL, Morton JS, Care AS, Davidge ST. Sex-specific effects of advanced maternal age on cardiovascular function in aged adult rat offspring. Am J Physiol Heart Circ Physiol 2018; 315:H1724-H1734. [DOI: 10.1152/ajpheart.00375.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pregnancy at an advanced maternal age has an increased risk of complications for both the mothers and their offspring. We have previously shown that advanced maternal age in a rat model leads to poor fetal outcomes, maternal vascular dysfunction, and hypertension, concordant with findings in humans. Moreover, offspring from aged dams had sex-specific cardiovascular dysfunction in young adulthood. However, the detrimental impact of aging on the cardiovascular system of the offspring in this model is unknown. We hypothesized that offspring born to aged dams (9.5–10 mo old) would have impaired cardiovascular function at 12 mo of age. Echocardiographic data revealed signs of mild left ventricular diastolic dysfunction in only male offspring from aged dams [isovolumetric relaxation time: 34.27 ± 2.04 in the young dam group vs. 27.61 ± 0.99 ms in the aged dam group, P < 0.01; mitral annular velocity ratio ( E′/ A′): 1.08 ± 0.04 in the young dam group vs. 0.96 ± 0.02 in the aged dam group, P < 0.05]. We have previously shown that in young adulthood (4 mo of age), male, but not female, offspring born to aged dams had impaired recovery from ischemia-reperfusion injury. Aging did not alter the susceptibility of female offspring to ischemia-reperfusion injury. Interestingly, wire myography data revealed that male offspring from aged dams had enhanced vascular sensitivity to methacholine (negative log of EC50: 7.4 ± 0.08 in young dams vs. 7.9 ± 0.11 in aged dams, P = 0.007) due, in part, to increased prostaglandin-mediated vasodilation. Despite intact endothelium-dependent relaxation, female offspring from aged dams had elevated systolic blood pressure (125.3 ± 4.2 mmHg in young dams vs. 144.0 ± 6.9 mmHg in aged dams, P = 0.03). These data highlight sex-specific mechanisms underlying cardiovascular programming in offspring born to dams of advanced age. NEW & NOTEWORTHY Our study demonstrated that adult male and female offspring (12 mo old) born to aged dams had impaired cardiac diastolic function and increased blood pressure, respectively, signifying sex-specific differential cardiovascular effects of advanced maternal age.
Collapse
Affiliation(s)
- Amin Shah
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Christy-Lynn M. Cooke
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Lois Hole Hospital for Women, Edmonton, Alberta, Canada
| | - Raven D. Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Anita L. Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Jude S. Morton
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Alison S. Care
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Sandra T. Davidge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
- Women and Children’s Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
32
|
MacQueen LA, Sheehy SP, Chantre CO, Zimmerman JF, Pasqualini FS, Liu X, Goss JA, Campbell PH, Gonzalez GM, Park SJ, Capulli AK, Ferrier JP, Kosar TF, Mahadevan L, Pu WT, Parker KK. A tissue-engineered scale model of the heart ventricle. Nat Biomed Eng 2018; 2:930-941. [PMID: 31015723 PMCID: PMC6774355 DOI: 10.1038/s41551-018-0271-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 06/20/2018] [Indexed: 02/08/2023]
Abstract
Laboratory studies of the heart use cell and tissue cultures to dissect heart function yet rely on animal models to measure pressure and volume dynamics. Here, we report tissue-engineered scale models of the human left ventricle, made of nanofibrous scaffolds that promote native-like anisotropic myocardial tissue genesis and chamber-level contractile function. Incorporating neonatal rat ventricular myocytes or cardiomyocytes derived from human induced pluripotent stem cells, the tissue-engineered ventricles have a diastolic chamber volume of ~500 µl (comparable to that of the native rat ventricle and approximately 1/250 the size of the human ventricle), and ejection fractions and contractile work 50-250 times smaller and 104-108 times smaller than the corresponding values for rodent and human ventricles, respectively. We also measured tissue coverage and alignment, calcium-transient propagation and pressure-volume loops in the presence or absence of test compounds. Moreover, we describe an instrumented bioreactor with ventricular-assist capabilities, and provide a proof-of-concept disease model of structural arrhythmia. The model ventricles can be evaluated with the same assays used in animal models and in clinical settings.
Collapse
Affiliation(s)
- Luke A MacQueen
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Sean P Sheehy
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Christophe O Chantre
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - John F Zimmerman
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Francesco S Pasqualini
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Xujie Liu
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Josue A Goss
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Patrick H Campbell
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Grant M Gonzalez
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Sung-Jin Park
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Andrew K Capulli
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - John P Ferrier
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - T Fettah Kosar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - L Mahadevan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Kavli Institute for Bionano Science and Technology, Harvard University, Cambridge, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kevin Kit Parker
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
33
|
Santos E, Aptekmann K, Santos P, Reis A, Kuster M, Andrade Junior P. Echocardiographic parameters in dogs treated with acepromazine and fentanyl. ARQ BRAS MED VET ZOO 2018. [DOI: 10.1590/1678-4162-10002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Considering that the use of tranquillizers could optimize the performance of the echocardiogram, this study aimed to evaluate the effect of protocols with acepromazine and fentanyl on the echocardiographic parameters of healthy dogs, besides their effect in systolic blood pressure (SBP), respiratory rate (RR), heart rate (HR), time spent for examination and sedation scale. Ten adult dogs were submitted to different tranquilizing protocols 20 minutes before the echocardiographic examination, totalling five treatments for each pair, performed at seven-day intervals between evaluations. The treatments were CT (control treatment), IAT (intramuscular acepromazine), OAT (oral acepromazine), FT (fentanyl) and AFT (acepromazine associated with fentanyl). In addition to the echocardiographic evaluation, SBP, degree of reassurance, duration of the exam, HR and RR in the different protocols were evaluated. There was a significant decrease of SBP in OAT. There was a significant reduction in left ventricular diameter during systole and diastole and mitral annular movement in IAT, OAT and AFT, compared with CT. There was a decrease in tricuspid annular plane systolic excursion and increase in mitral E/mitral A ratio in IAT and OAT when compared with CT. All the tranquillizer protocols studied were found to significantly reduce HR, that facilitated the echocardiographic examination.
Collapse
Affiliation(s)
- E.A. Santos
- Universidade Federal do Espírito Santo, Brazil
| | | | | | - A.C. Reis
- Universidade Federal do Espírito Santo, Brazil
| | | | | |
Collapse
|
34
|
Stegmann H, Bäuerle T, Kienle K, Dittrich S, Alkassar M. 4D cardiac magnetic resonance imaging, 4D and 2D transthoracic echocardiography: a comparison of in-vivo assessment of ventricular function in rats. Lab Anim 2018; 53:169-179. [PMID: 30081741 DOI: 10.1177/0023677218789971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Preclinical cardiovascular research is the foundation of our understanding and broad knowledge of heart function and cardiovascular disease. Reliable cardiac imaging modalities are the basis for applicable results. Four-dimensional cardiac magnetic resonance (4D CMR) has been set as the gold standard for in-vivo assessment of ventricular function in rodents. However, technical improvements in echocardiography now allow us to image the whole heart, which makes four-dimensional echocardiography (4DE) a possible alternative to 4D CMR. To date, no study has systematically assessed 4DE in comparison with 4D CMR in rats. In total we studied 26 juvenile Sprague-Dawley rats (Crl: CD (SD) IGS). Twenty rats underwent echocardiographic imaging (2D and 4D) and 4D CMR. Five of those rats underwent a ligation of the superior and inferior vena cava to reduce the cardiac inflow as a disease model. Six additional rats were used to assess reproducibility of echocardiography and underwent three echocardiographic examinations. 4D CMR was performed on a 7T scanner; 2D and 4D echocardiography was conducted using a 40 MHz transducer. Correlation between 4D CMR, 4DE and 2DE for left-ventricular ejection fraction (LVEF) was assessed. An excellent correlation was observed between 4DE and 4D CMR ( r = 0.95, p < 0.001). Correlation of 2DE and 4D CMR was weak ( r = 0.57, p < 0.01). 4DE provides results that are equally precise as 4D CMR and highly reproducible with less technical effort than 4D CMR.
Collapse
Affiliation(s)
- Hedwig Stegmann
- 1 Department of Paediatric Cardiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Tobias Bäuerle
- 2 Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Katharina Kienle
- 1 Department of Paediatric Cardiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Sven Dittrich
- 1 Department of Paediatric Cardiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Muhannad Alkassar
- 1 Department of Paediatric Cardiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
35
|
Divorty N, Milligan G, Graham D, Nicklin SA. The Orphan Receptor GPR35 Contributes to Angiotensin II-Induced Hypertension and Cardiac Dysfunction in Mice. Am J Hypertens 2018; 31:1049-1058. [PMID: 29860395 PMCID: PMC6077831 DOI: 10.1093/ajh/hpy073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/23/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The orphan receptor G protein–coupled receptor 35 (GPR35) has been associated with a range of diseases, including cancer, inflammatory bowel disease, diabetes, hypertension, and heart failure. To assess the potential for GPR35 as a therapeutic target in cardiovascular disease, this study investigated the cardiovascular phenotype of a GPR35 knockout mouse under both basal conditions and following pathophysiological stimulation. METHODS Blood pressure was monitored in male wild-type and GPR35 knockout mice over 7–14 days using implantable telemetry. Cardiac function and dimensions were assessed using echocardiography, and cardiomyocyte morphology evaluated histologically. Two weeks of angiotensin II (Ang II) infusion was used to investigate the effects of GPR35 deficiency under pathophysiological conditions. Gpr35 messenger RNA expression in cardiovascular tissues was assessed using quantitative polymerase chain reaction. RESULTS There were no significant differences in blood pressure, cardiac function, or cardiomyocyte morphology in GPR35 knockout mice compared with wild-type mice. Following Ang II infusion, GPR35 knockout mice were protected from significant increases in systolic, diastolic, and mean arterial blood pressure or impaired left ventricular systolic function, in contrast to wild-type mice. There were no significant differences in Gpr35 messenger RNA expression in heart, kidney, and aorta following Ang II infusion in wild-type mice. CONCLUSIONS Although GPR35 does not appear to influence basal cardiovascular regulation, these findings demonstrate that it plays an important pathological role in the development of Ang II–induced hypertension and impaired cardiac function. This suggests that GPR35 is a potential novel drug target for therapeutic intervention in hypertension.
Collapse
Affiliation(s)
- Nina Divorty
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Stuart A Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
36
|
Grune J, Blumrich A, Brix S, Jeuthe S, Drescher C, Grune T, Foryst-Ludwig A, Messroghli D, Kuebler WM, Ott C, Kintscher U. Evaluation of a commercial multi-dimensional echocardiography technique for ventricular volumetry in small animals. Cardiovasc Ultrasound 2018; 16:10. [PMID: 29966517 PMCID: PMC6029342 DOI: 10.1186/s12947-018-0128-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/13/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The assessment of ventricular volumes using conventional echocardiography methods is limited with regards to the need of geometrical assumptions. In the present study, we aimed to evaluate a novel commercial system for three-dimensional echocardiography (3DE) in preclinical models by direct comparison with conventional 1D- and 2D-echocardiography (1DE; 2DE) and the gold-standard technique magnetic resonance imaging (MRI). Further, we provide a standard operating protocol for image acquisition and analysis with 3DE. METHODS 3DE was carried out using a 30 MHz center frequency transducer coupled to a Vevo®3100 Imaging System. We evaluated under different experimental conditions: 1) in vitro phantom measurements served as controlled setting in which boundaries were clearly delineated; 2) a validation cohort composed of healthy C57BL/6 J mice and New Zealand Obese (NZO) mice was used in order to validate 3DE against cardiac MRI; 3) a standard mouse model of pressure overload induced-heart failure was investigated to estimate the value of 3DE. RESULTS First, in vitro volumetry revealed good agreement between 3DE assessed volumes and the MRI-assessed volumes. Second, cardiac volume determination with 3DE showed smaller mean differences compared to cardiac MRI than conventional 1DE and 2DE. Third, 3DE was suitable to detect reduced ejection fractions in heart failure mice. Fourth, inter- and intra-observer variability of 3DE showed good to excellent agreement regarding absolute volumes in healthy mice, whereas agreement rates for the relative metrics ejection fraction and stroke volume demonstrated good to moderate observer variabilities. CONCLUSIONS 3DE provides a novel method for accurate volumetry in small animals without the need for spatial assumptions, demonstrating a technique for an improved analysis of ventricular function. Further validation work and highly standardized image analyses are required to increase reproducibility of this approach.
Collapse
Affiliation(s)
- Jana Grune
- Institute of Pharmacology, Center for Cardiovascular Research, Charité -Universitaetsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
- Institute of Physiology, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Annelie Blumrich
- Institute of Pharmacology, Center for Cardiovascular Research, Charité -Universitaetsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
| | - Sarah Brix
- Institute of Pharmacology, Center for Cardiovascular Research, Charité -Universitaetsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
| | - Sarah Jeuthe
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
- Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Cardiology, Charité University Medicine Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Cathleen Drescher
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany
| | - Tilman Grune
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany
- German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany
| | - Anna Foryst-Ludwig
- Institute of Pharmacology, Center for Cardiovascular Research, Charité -Universitaetsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
| | - Daniel Messroghli
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
- Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Cardiology, Charité University Medicine Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Wolfgang M Kuebler
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
- Institute of Physiology, Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christiane Ott
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany
| | - Ulrich Kintscher
- Institute of Pharmacology, Center for Cardiovascular Research, Charité -Universitaetsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany.
| |
Collapse
|
37
|
Heinen A, Raupach A, Behmenburg F, Hölscher N, Flögel U, Kelm M, Kaisers W, Nederlof R, Huhn R, Gödecke A. Echocardiographic Analysis of Cardiac Function after Infarction in Mice: Validation of Single-Plane Long-Axis View Measurements and the Bi-Plane Simpson Method. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:1544-1555. [PMID: 29706407 DOI: 10.1016/j.ultrasmedbio.2018.03.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/19/2018] [Accepted: 03/22/2018] [Indexed: 06/08/2023]
Abstract
Although echocardiography is commonly used to analyze cardiac function in small animal models of cardiac remodeling after myocardial infarction, the different echocardiographic methods are validated poorly. End-diastolic volume, end-systolic volume and ejection fraction were analyzed using either standard single-plane analysis from parasternal long-axis B-mode views (PSLAX) or the bi-plane Simpson method (using PSLAX and three short-axis views) and validated using magnetic resonance imaging as standard. Ejection fraction measured by PSLAX was moderately correlated with a coefficient of R2 = 0.49. The standard deviation of residuals was 9.91. Simpson analysis revealed an improved correlation coefficient of R2 = 0.77 and a reduction in standard deviation of residuals by 45% (5.45 vs. 9.92, p = 0.014). Subgroup analysis revealed that the high variation in PSLAX is due to changes in ventricular geometry after myocardial infarction. Our results indicate that the bi-plane Simpson method is advantageous for the assessment of cardiac function after myocardial infarction.
Collapse
Affiliation(s)
- Andre Heinen
- Institute of Cardiovascular Physiology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| | - Annika Raupach
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Nina Hölscher
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ulrich Flögel
- Department of Molecular Cardiology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Wolfgang Kaisers
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Rianne Nederlof
- Institute of Cardiovascular Physiology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Ragnar Huhn
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Axel Gödecke
- Institute of Cardiovascular Physiology, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
38
|
Villalba-Orero M, López-Olañeta MM, González-López E, Padrón-Barthe L, Gómez-Salinero JM, García-Prieto J, Wai T, García-Pavía P, Ibáñez B, Jiménez-Borreguero LJ, Lara-Pezzi E. Lung ultrasound as a translational approach for non-invasive assessment of heart failure with reduced or preserved ejection fraction in mice. Cardiovasc Res 2018; 113:1113-1123. [PMID: 28472392 DOI: 10.1093/cvr/cvx090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/03/2017] [Indexed: 12/28/2022] Open
Abstract
Aims Heart failure (HF) has become an epidemic and constitutes a major medical, social, and economic problem worldwide. Despite advances in medical treatment, HF prognosis remains poor. The development of efficient therapies is hampered by the lack of appropriate animal models in which HF can be reliably determined, particularly in mice. The development of HF in mice is often assumed based on the presence of cardiac dysfunction, but HF itself is seldom proved. Lung ultrasound (LUS) has become a helpful tool for lung congestion assessment in patients at all stages of HF. We aimed to apply this non-invasive imaging tool to evaluate HF in mouse models of both systolic and diastolic dysfunction. Methods and results We used LUS to study HF in a mouse model of systolic dysfunction, dilated cardiomyopathy, and in a mouse model of diastolic dysfunction, diabetic cardiomyopathy. LUS proved to be a reliable and reproducible tool to detect pulmonary congestion in mice. The combination of LUS and echocardiography allowed discriminating those mice that develop HF from those that do not, even in the presence of evident cardiac dysfunction. The study showed that LUS can be used to identify the onset of HF decompensation and to evaluate the efficacy of therapies for this syndrome. Conclusions This novel approach in mouse models of cardiac disease enables for the first time to adequately diagnose HF non-invasively in mice with preserved or reduced ejection fraction, and will pave the way to a better understanding of HF and to the development of new therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Esther González-López
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Laura Padrón-Barthe
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | | - Jaime García-Prieto
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Timothy Wai
- Institut Necker-Enfants Malades, Université Paris Descartes, Paris, France
| | - Pablo García-Pavía
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain.,Centro de Investigación Biomédica en Red Para Cardiología (CIBERCV), Madrid, Spain.,Universidad Francisco de Vitoria, Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red Para Cardiología (CIBERCV), Madrid, Spain.,Department of Cardiology, Instituto de Investigación Sanitaria (IIS), Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Luis J Jiménez-Borreguero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Hospital de la Princesa, Madrid, Spain
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red Para Cardiología (CIBERCV), Madrid, Spain.,National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
39
|
Zhou Z, Wu S, Lin MY, Fang J, Liu HL, Tsui PH. Three-dimensional Visualization of Ultrasound Backscatter Statistics by Window-modulated Compounding Nakagami Imaging. ULTRASONIC IMAGING 2018; 40:171-189. [PMID: 29506441 DOI: 10.1177/0161734618756101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this study, the window-modulated compounding (WMC) technique was integrated into three-dimensional (3D) ultrasound Nakagami imaging for improving the spatial visualization of backscatter statistics. A 3D WMC Nakagami image was produced by summing and averaging a number of 3D Nakagami images (number of frames denoted as N) formed using sliding cubes with varying side lengths ranging from 1 to N times the transducer pulse. To evaluate the performance of the proposed 3D WMC Nakagami imaging method, agar phantoms with scatterer concentrations ranging from 2 to 64 scatterers/mm3 were made, and six stages of fatty liver (zero, one, two, four, six, and eight weeks) were induced in rats by methionine-choline-deficient diets (three rats for each stage, total n = 18). A mechanical scanning system with a 5-MHz focused single-element transducer was used for ultrasound radiofrequency data acquisition. The experimental results showed that 3D WMC Nakagami imaging was able to characterize different scatterer concentrations. Backscatter statistics were visualized with various numbers of frames; N = 5 reduced the estimation error of 3D WMC Nakagami imaging in visualizing the backscatter statistics. Compared with conventional 3D Nakagami imaging, 3D WMC Nakagami imaging improved the image smoothness without significant image resolution degradation, and it can thus be used for describing different stages of fatty liver in rats.
Collapse
Affiliation(s)
- Zhuhuang Zhou
- 1 College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
- 2 Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Shuicai Wu
- 1 College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Man-Yen Lin
- 3 Department of Electrical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Jui Fang
- 4 PhD Program in Biomedical Engineering, College of Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Li Liu
- 3 Department of Electrical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Po-Hsiang Tsui
- 5 Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- 6 Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University and Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- 7 Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| |
Collapse
|
40
|
Acin-Perez R, Lechuga-Vieco AV, del Mar Muñoz M, Nieto-Arellano R, Torroja C, Sánchez-Cabo F, Jiménez C, González-Guerra A, Carrascoso I, Benincá C, Quiros PM, López-Otín C, Castellano JM, Ruíz-Cabello J, Jiménez-Borreguero LJ, Enríquez JA. Ablation of the stress protease OMA1 protects against heart failure in mice. Sci Transl Med 2018; 10:10/434/eaan4935. [DOI: 10.1126/scitranslmed.aan4935] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/14/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
|
41
|
Chandramouli C, Reichelt ME, Curl CL, Varma U, Bienvenu LA, Koutsifeli P, Raaijmakers AJA, De Blasio MJ, Qin CX, Jenkins AJ, Ritchie RH, Mellor KM, Delbridge LMD. Diastolic dysfunction is more apparent in STZ-induced diabetic female mice, despite less pronounced hyperglycemia. Sci Rep 2018; 8:2346. [PMID: 29402990 PMCID: PMC5799292 DOI: 10.1038/s41598-018-20703-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/23/2018] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy is a distinct pathology characterized by early emergence of diastolic dysfunction. Increased cardiovascular risk associated with diabetes is more marked for women, but an understanding of the role of diastolic dysfunction in female susceptibility to diabetic cardiomyopathy is lacking. To investigate the sex-specific relationship between systemic diabetic status and in vivo occurrence of diastolic dysfunction, diabetes was induced in male and female mice by streptozotocin (5x daily i.p. 55 mg/kg). Echocardiography was performed at 7 weeks post-diabetes induction, cardiac collagen content assessed by picrosirius red staining, and gene expression measured using qPCR. The extent of diabetes-associated hyperglycemia was more marked in males than females (males: 25.8 ± 1.2 vs 9.1 ± 0.4 mM; females: 13.5 ± 1.5 vs 8.4 ± 0.4 mM, p < 0.05) yet in vivo diastolic dysfunction was evident in female (E/E' 54% increase, p < 0.05) but not male diabetic mice. Cardiac structural abnormalities (left ventricular wall thinning, collagen deposition) were similar in male and female diabetic mice. Female-specific gene expression changes in glucose metabolic and autophagy-related genes were evident. This study demonstrates that STZ-induced diabetic female mice exhibit a heightened susceptibility to diastolic dysfunction, despite exhibiting a lower extent of hyperglycemia than male mice. These findings highlight the importance of early echocardiographic screening of asymptomatic prediabetic at-risk patients.
Collapse
Affiliation(s)
- Chanchal Chandramouli
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
- National Heart Centre, Singapore, Singapore
| | - Melissa E Reichelt
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Claire L Curl
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Upasna Varma
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Laura A Bienvenu
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Parisa Koutsifeli
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Miles J De Blasio
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- School of Biosciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Cheng Xue Qin
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Alicia J Jenkins
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Kimberley M Mellor
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Lea M D Delbridge
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
42
|
Lindsey ML, Kassiri Z, Virag JAI, de Castro Brás LE, Scherrer-Crosbie M. Guidelines for measuring cardiac physiology in mice. Am J Physiol Heart Circ Physiol 2018; 314:H733-H752. [PMID: 29351456 PMCID: PMC5966769 DOI: 10.1152/ajpheart.00339.2017] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cardiovascular disease is a leading cause of death, and translational research is needed to understand better mechanisms whereby the left ventricle responds to injury. Mouse models of heart disease have provided valuable insights into mechanisms that occur during cardiac aging and in response to a variety of pathologies. The assessment of cardiovascular physiological responses to injury or insult is an important and necessary component of this research. With increasing consideration for rigor and reproducibility, the goal of this guidelines review is to provide best-practice information regarding how to measure accurately cardiac physiology in animal models. In this article, we define guidelines for the measurement of cardiac physiology in mice, as the most commonly used animal model in cardiovascular research. Listen to this article’s corresponding podcast at http://ajpheart.podbean.com/e/guidelines-for-measuring-cardiac-physiology-in-mice/.
Collapse
Affiliation(s)
- Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center , Jackson, Mississippi.,Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta , Edmonton, Alberta , Canada
| | - Jitka A I Virag
- Department of Physiology, Brody School of Medicine, East Carolina University , Greenville, North Carolina
| | - Lisandra E de Castro Brás
- Department of Physiology, Brody School of Medicine, East Carolina University , Greenville, North Carolina
| | | |
Collapse
|
43
|
Rivaud MR, Agullo-Pascual E, Lin X, Leo-Macias A, Zhang M, Rothenberg E, Bezzina CR, Delmar M, Remme CA. Sodium Channel Remodeling in Subcellular Microdomains of Murine Failing Cardiomyocytes. J Am Heart Assoc 2017; 6:e007622. [PMID: 29222390 PMCID: PMC5779058 DOI: 10.1161/jaha.117.007622] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/13/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Cardiac sodium channel (NaV1.5) dysfunction contributes to arrhythmogenesis during pathophysiological conditions. Nav1.5 localizes to distinct subcellular microdomains within the cardiomyocyte, where it associates with region-specific proteins, yielding complexes whose function is location specific. We herein investigated sodium channel remodeling within distinct cardiomyocyte microdomains during heart failure. METHODS AND RESULTS Mice were subjected to 6 weeks of transverse aortic constriction (TAC; n=32) to induce heart failure. Sham-operated on mice were used as controls (n=20). TAC led to reduced left ventricular ejection fraction, QRS prolongation, increased heart mass, and upregulation of prohypertrophic genes. Whole-cell sodium current (INa) density was decreased by 30% in TAC versus sham-operated on cardiomyocytes. On macropatch analysis, INa in TAC cardiomyocytes was reduced by 50% at the lateral membrane (LM) and by 40% at the intercalated disc. Electron microscopy and scanning ion conductance microscopy revealed remodeling of the intercalated disc (replacement of [inter-]plicate regions by large foldings) and LM (less identifiable T tubules and reduced Z-groove ratios). Using scanning ion conductance microscopy, cell-attached recordings in LM subdomains revealed decreased INa and increased late openings specifically at the crest of TAC cardiomyocytes, but not in groove/T tubules. Failing cardiomyocytes displayed a denser, but more stable, microtubule network (demonstrated by increased α-tubulin and Glu-tubulin expression). Superresolution microscopy showed reduced average NaV1.5 cluster size at the LM of TAC cells, in line with reduced INa. CONCLUSIONS Heart failure induces structural remodeling of the intercalated disc, LM, and microtubule network in cardiomyocytes. These adaptations are accompanied by alterations in NaV1.5 clustering and INa within distinct subcellular microdomains of failing cardiomyocytes.
Collapse
Affiliation(s)
- Mathilde R Rivaud
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, the Netherlands
- Division of Cardiology, New York University Medical Center, New York, NY
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Xianming Lin
- Division of Cardiology, New York University Medical Center, New York, NY
| | | | - Mingliang Zhang
- Division of Cardiology, New York University Medical Center, New York, NY
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, NYU-School of Medicine, New York, NY
| | - Connie R Bezzina
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, the Netherlands
| | - Mario Delmar
- Division of Cardiology, New York University Medical Center, New York, NY
| | - Carol Ann Remme
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
44
|
Shah A, Quon A, Morton JS, Davidge ST. Postnatal resveratrol supplementation improves cardiovascular function in male and female intrauterine growth restricted offspring. Physiol Rep 2017; 5:5/2/e13109. [PMID: 28108646 PMCID: PMC5269411 DOI: 10.14814/phy2.13109] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/18/2022] Open
Abstract
Intrauterine growth restriction (IUGR) may predispose offspring to an increased susceptibility of developing cardiovascular disease (CVD) in adult life. The window of opportunity to treat later life CVD programmed in fetal life is critical. The aim of this study was to identify the effect of resveratrol treatment of IUGR offspring at a time of known CV dysfunction. Sprague–Dawley male and female rat offspring who experienced normoxia (21% O2; control) or hypoxia (11% O2; IUGR) in utero were fed a high‐fat (HF) diet (3–21 weeks of age) or a HF diet (3–21 weeks of age) supplemented with resveratrol from 13 to 21 weeks of age. At 21 weeks of age, echocardiographic data showed that male IUGR offspring had mild in vivo diastolic dysfunction, whereas female IUGR offspring had early signs of cardiac diastolic dysfunction that was not altered by resveratrol treatment. Notably, male and female IUGR offspring demonstrated equal susceptibility to ex vivo cardiac dysfunction recovery after ischemia/reperfusion (I/R) injury and this was improved by resveratrol treatment, independent of sex. Resveratrol increased cardiac phospho‐adenosine monophosphate kinase (p‐AMPK) levels in only female IUGR offspring. IUGR or resveratrol did not alter cardiac superoxide levels. However, in male offspring, an overall effect of IUGR in reducing cardiac catalase levels was observed that was not altered by resveratrol. Interestingly, in only female IUGR offspring, resveratrol significantly increased cardiac superoxide dismutase (SOD) 2 levels. In conclusion, resveratrol treatment of adult IUGR offspring, at the time of known CV dysfunction, improved cardiac function recovery in both sexes and the mechanisms involved were partially sex‐specific.
Collapse
Affiliation(s)
- Amin Shah
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jude S Morton
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada .,Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
45
|
Jones S, Mann A, Worley MC, Fulford L, Hall D, Karani R, Jiang M, Robbins N, Rubinstein J, Koch SE. The role of transient receptor potential vanilloid 2 channel in cardiac aging. Aging Clin Exp Res 2017; 29:863-873. [PMID: 27804106 DOI: 10.1007/s40520-016-0663-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND The aging heart is characterized by cellular and molecular changes leading to a decline in physiologic function and cardiac remodeling, specifically the development of myocyte hypertrophy and fibrosis. Transient receptor potential vanilloid 2 (TRPV2), a stretch-mediated channel and regulator of calcium homeostasis, plays a key role in the function and structure of the heart. TRPV2 also plays an important role in the adaptive and maladaptive compensatory mechanisms of the heart in response to pathologic and exercise-induced stress. Our current study seeks to elucidate the potential role of TRPV2 channels in the regulation of cardiac function in aging. METHODS Wild-type (WT) and TRPV2 functional knockout (FKO) mice were aged out to various time points, and their cardiac function was measured using advanced echocardiography. Furthermore, we histologically analyzed the heart morphology to determine myocyte hypertrophy, the development of fibrosis and the relative expression of TRPV2. RESULTS Our results demonstrate that even though TRPV2-FKO mice have impaired function at baseline, their cardiac function as measured via standard and advanced echocardiographic parameters (ejection fraction, cardiac output and circumferential strain) decreased less with aging in comparison with the WT group. Furthermore, there was less fibrosis and hypertrophy in the TRPV2-FKO group with aging in comparison with the WT. The expression of TRPV2 in the WT group did not significantly change with aging. CONCLUSIONS TRPV2 functional deletion is compatible with aging and associated with a decreased development of myocyte hypertrophy and fibrosis. It may be an important target for prevention of age-induced cardiac remodeling.
Collapse
Affiliation(s)
- Shannon Jones
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA
| | - Adrien Mann
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA
| | - Mariah C Worley
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA
| | - Logan Fulford
- Department of Pathobiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - David Hall
- Department of Nutritional Sciences College of Allied Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Rajiv Karani
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA
| | - Min Jiang
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA
| | - Nathan Robbins
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA
| | - Sheryl E Koch
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0542, Cincinnati, OH, 45267, USA.
| |
Collapse
|
46
|
Travers JG, Kamal FA, Valiente-Alandi I, Nieman ML, Sargent MA, Lorenz JN, Molkentin JD, Blaxall BC. Pharmacological and Activated Fibroblast Targeting of Gβγ-GRK2 After Myocardial Ischemia Attenuates Heart Failure Progression. J Am Coll Cardiol 2017; 70:958-971. [PMID: 28818206 DOI: 10.1016/j.jacc.2017.06.049] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/26/2017] [Accepted: 06/15/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND Cardiac fibroblasts are a critical cell population responsible for myocardial extracellular matrix homeostasis. Upon injury or pathological stimulation, these cells transform to an activated myofibroblast state and play a fundamental role in myocardial fibrosis and remodeling. Chronic sympathetic overstimulation, a hallmark of heart failure (HF), induces pathological signaling through G protein βγ (Gβγ) subunits and their interaction with G protein-coupled receptor kinase 2 (GRK2). OBJECTIVES This study investigated the hypothesis that Gβγ-GRK2 inhibition and/or ablation after myocardial injury would attenuate pathological myofibroblast activation and cardiac remodeling. METHODS The therapeutic potential of small molecule Gβγ-GRK2 inhibition, alone or in combination with activated fibroblast- or myocyte-specific GRK2 ablation-each initiated after myocardial ischemia-reperfusion (I/R) injury-was investigated to evaluate the possible salutary effects on post-I/R fibroblast activation, pathological remodeling, and cardiac dysfunction. RESULTS Small molecule Gβγ-GRK2 inhibition initiated 1 week post-injury was cardioprotective in the I/R model of chronic HF, including preservation of cardiac contractility and a reduction in cardiac fibrotic remodeling. Systemic small molecule Gβγ-GRK2 inhibition initiated 1 week post-I/R in cardiomyocyte-restricted GRK2 ablated mice (also post-I/R) still demonstrated significant cardioprotection, which suggested a potential protective role beyond the cardiomyocyte. Inducible ablation of GRK2 in activated fibroblasts (i.e., myofibroblasts) post-I/R injury demonstrated significant functional cardioprotection with reduced myofibroblast transformation and fibrosis. Systemic small molecule Gβγ-GRK2 inhibition initiated 1 week post-I/R provided little to no further protection in mice with ablation of GRK2 in activated fibroblasts alone. Finally, Gβγ-GRK2 inhibition significantly attenuated activation characteristics of failing human cardiac fibroblasts isolated from end-stage HF patients. CONCLUSIONS These findings suggested consideration of a paradigm shift in the understanding of the therapeutic role of Gβγ-GRK2 inhibition in treating HF and the potential therapeutic role for Gβγ-GRK2 inhibition in limiting pathological myofibroblast activation, interstitial fibrosis, and HF progression.
Collapse
Affiliation(s)
- Joshua G Travers
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Fadia A Kamal
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, New York
| | - Iñigo Valiente-Alandi
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michelle L Nieman
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michelle A Sargent
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - John N Lorenz
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jeffery D Molkentin
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Burns C Blaxall
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
47
|
Oliveira LFLD, O'Connell JL, Carvalho EEVD, Pulici ÉCC, Romano MMD, Maciel BC, Simões MV. Comparison between Radionuclide Ventriculography and Echocardiography for Quantification of Left Ventricular Systolic Function in Rats Exposed to Doxorubicin. Arq Bras Cardiol 2017; 108:12-20. [PMID: 28146205 PMCID: PMC5245843 DOI: 10.5935/abc.20160194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022] Open
Abstract
Background Radionuclide ventriculography (RV) is a validated method to evaluate the left
ventricular systolic function (LVSF) in small rodents. However, no prior
study has compared the results of RV with those obtained by other imaging
methods in this context. Objectives To compare the results of LVSF obtained by RV and echocardiography (ECHO) in
an experimental model of cardiotoxicity due to doxorubicin (DXR) in rats.
Methods Adult male Wistar rats serving as controls (n = 7) or receiving DXR (n = 22)
in accumulated doses of 8, 12, and 16 mg/kg were evaluated with ECHO
performed with a Sonos 5500 Philips equipment (12-MHz transducer) and RV
obtained with an Orbiter-Siemens gamma camera using a pinhole collimator
with a 4-mm aperture. Histopathological quantification of myocardial
fibrosis was performed after euthanasia. Results The control animals showed comparable results in the LVSF analysis obtained
with ECHO and RV (83.5 ± 5% and 82.8 ± 2.8%, respectively, p
> 0.05). The animals that received DXR presented lower LVSF values when
compared with controls (p < 0.05); however, the LVSF values obtained by
RV (60.6 ± 12.5%) were lower than those obtained by ECHO (71.8
± 10.1%, p = 0.0004) in this group. An analysis of the correlation
between the LVSF and myocardial fibrosis showed a moderate correlation when
the LVSF was assessed by ECHO (r = -0.69, p = 0.0002) and a stronger
correlation when it was assessed by RV (r = -0.79, p < 0.0001). On
multiple regression analysis, only RV correlated independently with
myocardial fibrosis. Conclusion RV is an alternative method to assess the left ventricular function in small
rodents in vivo. When compared with ECHO, RV showed a better correlation
with the degree of myocardial injury in a model of DXR-induced
cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | - Benedito Carlos Maciel
- Centro de Cardiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Marcus Vinicius Simões
- Centro de Cardiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil
| |
Collapse
|
48
|
Cardiovascular susceptibility to in vivo ischemic myocardial injury in male and female rat offspring exposed to prenatal hypoxia. Clin Sci (Lond) 2017; 131:2303-2317. [DOI: 10.1042/cs20171122] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/20/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023]
Abstract
Intrauterine growth restriction (IUGR) following prenatal hypoxia exposure leads to a higher risk of developing cardiovascular disease (CVD) in later life. Our aim was to evaluate cardiac susceptibility and its pathophysiological mechanisms following acute myocardial infarction (MI) in adult rat offspring exposed to prenatal hypoxia. Male and female rat offspring, which experienced normoxia (21% O2) or hypoxia (11% O2) in utero underwent sham or MI surgery at 12 weeks of age. Echocardiographic data revealed that both sexes had systolic dysfunction following MI surgery, independent of prenatal hypoxia. Male offspring exposed to prenatal hypoxia, however, had left ventricular dilatation, global dysfunction, and signs of diastolic dysfunction following MI surgery as evident by increased left ventricular internal diameter (LVID) during diastole (MI effect, P<0.01), Tei index (MI effect, P<0.001), and E/E′ ratio (prenatal hypoxia or MI effect, P<0.01). In contrast, diastolic dysfunction in female offspring was not as evident. Cardiac superoxide levels increased only in prenatal hypoxia exposed male offspring. Cardiac sarcoendoplasmic reticulum Ca2+-ATPase2a (SERCA2a) levels, a marker of cardiac injury and dysfunction, decreased in both male and female MI groups independent of prenatal hypoxia. Prenatal hypoxia increased cardiac ryanodine receptor 2 (RYR2) protein levels, while MI reduced RYR2 in only male offspring. In conclusion, male offspring exposed to prenatal hypoxia had an increased susceptibility to ischemic myocardial injury involving cardiac phenotypes similar to heart failure involving diastolic dysfunction in adult life compared with both offspring from healthy pregnancies and their female counterparts.
Collapse
|
49
|
Cerrone M, Montnach J, Lin X, Zhao YT, Zhang M, Agullo-Pascual E, Leo-Macias A, Alvarado FJ, Dolgalev I, Karathanos TV, Malkani K, Van Opbergen CJM, van Bavel JJA, Yang HQ, Vasquez C, Tester D, Fowler S, Liang F, Rothenberg E, Heguy A, Morley GE, Coetzee WA, Trayanova NA, Ackerman MJ, van Veen TAB, Valdivia HH, Delmar M. Plakophilin-2 is required for transcription of genes that control calcium cycling and cardiac rhythm. Nat Commun 2017; 8:106. [PMID: 28740174 PMCID: PMC5524637 DOI: 10.1038/s41467-017-00127-0] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/02/2017] [Indexed: 12/19/2022] Open
Abstract
Plakophilin-2 (PKP2) is a component of the desmosome and known for its role in cell-cell adhesion. Mutations in human PKP2 associate with a life-threatening arrhythmogenic cardiomyopathy, often of right ventricular predominance. Here, we use a range of state-of-the-art methods and a cardiomyocyte-specific, tamoxifen-activated, PKP2 knockout mouse to demonstrate that in addition to its role in cell adhesion, PKP2 is necessary to maintain transcription of genes that control intracellular calcium cycling. Lack of PKP2 reduces expression of Ryr2 (coding for Ryanodine Receptor 2), Ank2 (coding for Ankyrin-B), Cacna1c (coding for CaV1.2) and Trdn (coding for triadin), and protein levels of calsequestrin-2 (Casq2). These factors combined lead to disruption of intracellular calcium homeostasis and isoproterenol-induced arrhythmias that are prevented by flecainide treatment. We propose a previously unrecognized arrhythmogenic mechanism related to PKP2 expression and suggest that mutations in PKP2 in humans may cause life-threatening arrhythmias even in the absence of structural disease.It is believed that mutations in desmosomal adhesion complex protein plakophilin 2 (PKP2) cause arrhythmia due to loss of cell-cell communication. Here the authors show that PKP2 controls the expression of proteins involved in calcium cycling in adult mouse hearts, and that lack of PKP2 can cause arrhythmia in a structurally normal heart.
Collapse
Affiliation(s)
- Marina Cerrone
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Jerome Montnach
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Xianming Lin
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Yan-Ting Zhao
- Center for Arrhythmia Research, Division of Cardiology, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Mingliang Zhang
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Esperanza Agullo-Pascual
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Alejandra Leo-Macias
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Francisco J Alvarado
- Department of Molecular and Integrative Physiology, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Igor Dolgalev
- Genome Technology Center, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Thomas V Karathanos
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, 3400N Charles St., Baltimore, MD, 21218, USA
| | - Kabir Malkani
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Chantal J M Van Opbergen
- Department of Medical Physiology Division of Heart & Lungs University Medical Centre Utrecht, Yalelaan 50, 3584CM, Utrecht, The Netherlands
| | - Joanne J A van Bavel
- Department of Medical Physiology Division of Heart & Lungs University Medical Centre Utrecht, Yalelaan 50, 3584CM, Utrecht, The Netherlands
| | - Hua-Qian Yang
- Department of Pediatrics, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Carolina Vasquez
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - David Tester
- Departments of Cardiovascular Diseases/Division of Heart Rhythm Services, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Pediatric and Adolescent Medicine/Division of Pediatric Cardiology, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Steven Fowler
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Fengxia Liang
- Department of Cell Biology and Microscopy Core, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Adriana Heguy
- Genome Technology Center, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Gregory E Morley
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - William A Coetzee
- Departments of Pediatrics, Physiology & Neuroscience and Biochemistry and Molecular Pharmacology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA
| | - Natalia A Trayanova
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, 3400N Charles St., Baltimore, MD, 21218, USA
| | - Michael J Ackerman
- Departments of Cardiovascular Diseases/Division of Heart Rhythm Services, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Pediatric and Adolescent Medicine/Division of Pediatric Cardiology, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Toon A B van Veen
- Department of Medical Physiology Division of Heart & Lungs University Medical Centre Utrecht, Yalelaan 50, 3584CM, Utrecht, The Netherlands
| | - Hector H Valdivia
- Center for Arrhythmia Research, Division of Cardiology, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Mario Delmar
- Leon H Charney Division of Cardiology, NYU School of Medicine, 520 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
50
|
Huang J, Wu J, Wang S, You J, Ye Y, Ding Z, Yang F, Wang X, Guo J, Ma L, Yuan J, Shen Y, Yang X, Sun A, Jiang H, Bu L, Backx PH, Ge J, Zou Y. Ultrasound biomicroscopy validation of a murine model of cardiac hypertrophic preconditioning: comparison with a hemodynamic assessment. Am J Physiol Heart Circ Physiol 2017; 313:H138-H148. [PMID: 28455286 DOI: 10.1152/ajpheart.00004.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/23/2017] [Accepted: 04/06/2017] [Indexed: 01/09/2023]
Abstract
In mice, myocardial hypertrophic preconditioning (HP), which is produced by the removal of short-term transverse aortic constriction (TAC), was recently reported to render the heart resistant to hypertrophic responses induced by subsequent reconstriction (Re-TAC). However, there is no efficient noninvasive method for ensuring that the repeated aortic manipulations were successfully performed. We previously demonstrated that ultrasound biomicroscopy (UBM) is a noninvasive and effective approach for predicting TAC success. Here, we investigated the value of UBM for serial predictions of load conditions in establishing a murine HP model. C57BL/6J mice were subjected to a sham operation, TAC, or Re-TAC, and the peak flow velocity at the aortic banding site (PVb) was measured by UBM. Left ventricular end-systolic pressure (LVESP) was examined by micromanometric catheterization. The PVb was positively associated with LVESP (R2 = 0.8204, P < 0.001, for TAC at 3 days and R2 = 0.7746, P < 0.001, for Re-TAC at 4 wk). PVb and LVESP values were markedly elevated after aortic banding, became attenuated to the sham-operated level after debanding, and increased after aortic rebanding. The cardiac hypertrophic responses were examined by UBM, histology, RT-PCR, and Western blot analysis. Four weeks after the last operation, with PVb ≥ 3.5 m/s as an indicator of successful aortic constriction, Re-TAC mice showed less cardiac hypertrophy, fetal gene expression, and ERK1/2 activation than TAC mice. Therefore, we successfully established a UBM protocol for the serial assessment of aortic flow and the prediction of LVESP during repeated aortic manipulations in mice, which might be useful for noninvasive evaluations of the murine HP model.NEW & NOTEWORTHY We successfully developed an ultrasound biomicroscopy protocol for the serial assessment of aortic bandings and the relevant left ventricular pressure in a murine model of cardiac hypertrophic preconditioning. The protocol may be of great importance in the successful establishment of the hypertrophic preconditioning model for further mechanistic and pharmacological studies.
Collapse
Affiliation(s)
- Jiayuan Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Jieyun You
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yong Ye
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Zhiwen Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Fenghua Yang
- Guangdong Laboratory Animal Monitoring Institute, Guangzhou, People's Republic of China
| | - Xingxu Wang
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Junjie Guo
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Leilei Ma
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Jie Yuan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Yunli Shen
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiangdong Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Aijun Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Hong Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Liping Bu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Peter H Backx
- Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario; and.,Division of Cardiology, University Health Network, Toronto, Ontario, Canada
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China;
| |
Collapse
|