1
|
Sayour NV, Gergely TG, Váradi B, Tóth VÉ, Ágg B, Kovács T, Kucsera D, Kovácsházi C, Brenner GB, Giricz Z, Ferdinandy P, Varga ZV. Comparison of mouse models of heart failure with reduced ejection fraction. ESC Heart Fail 2025; 12:87-100. [PMID: 39243187 PMCID: PMC11769617 DOI: 10.1002/ehf2.15031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024] Open
Abstract
AIMS Heart failure with reduced ejection fraction (HFrEF) is a leading cause of death worldwide; thus, therapeutic improvements are needed. In vivo preclinical models are essential to identify molecular drug targets for future therapies. Transverse aortic constriction (TAC) is a well-established model of HFrEF; however, highly experienced personnel are needed for the surgery, and several weeks of follow-up are necessary to develop HFrEF. To this end, we aimed (i) to develop an easy-to-perform mouse model of HFrEF by treating Balb/c mice with angiotensin-II (Ang-II) for 2 weeks by minipump and (ii) to compare its cardiac phenotype and transcriptome to the well-established TAC model of HFrEF in C57BL/6J mice. METHODS Mortality and gross pathological data, cardiac structural and functional characteristics assessed by echocardiography and immunohistochemistry and differential gene expression obtained by RNA-sequencing and gene-ontology analyses were used to characterize and compare the two models. To achieve statistical comparability between the two models, changes in treatment groups related to the corresponding control were compared (ΔTAC vs. ΔAng-II). RESULTS Compared with the well-established TAC model, chronic Ang-II treatment of Balb/c mice shares similarities in cardiac systolic functional decline (left ventricular ejection fraction: -57.25 ± 7.17% vs. -43.68 ± 5.31% in ΔTAC vs. ΔAng-II; P = 0.1794) but shows a lesser degree of left ventricular dilation (left ventricular end-systolic volume: 190.81 ± 44.13 vs. 57.37 ± 10.18 mL in ΔTAC vs. ΔAng-II; P = 0.0252) and hypertrophy (cell surface area: 58.44 ± 6.1 vs. 10.24 ± 2.87 μm2 in ΔTAC vs. ΔAng-II; P < 0.001); nevertheless, transcriptomic changes in the two HFrEF models show strong correlation (Spearman's r = 0.727; P < 0.001). In return, Ang-II treatment in Balb/c mice needs significantly less procedural time [38 min, interquartile range (IQR): 31-46 min in TAC vs. 6 min, IQR: 6-7 min in Ang-II; P < 0.001] and surgical expertise, is less of an object for peri-procedural mortality (15.8% in TAC vs. 0% in Ang-II; P = 0.105) and needs significantly shorter follow-up for developing HFrEF. CONCLUSIONS Here, we demonstrate for the first time that chronic Ang-II treatment of Balb/c mice is also a relevant, reliable but significantly easier-to-perform preclinical model to identify novel pathomechanisms and targets in future HFrEF research.
Collapse
Affiliation(s)
- Nabil V. Sayour
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- HCEMM‐SU Cardiometabolic Immunology Research GroupBudapestHungary
- MTA‐SE Momentum Cardio‐Oncology and Cardioimmunology Research GroupBudapestHungary
| | - Tamás G. Gergely
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- HCEMM‐SU Cardiometabolic Immunology Research GroupBudapestHungary
- MTA‐SE Momentum Cardio‐Oncology and Cardioimmunology Research GroupBudapestHungary
| | - Barnabás Váradi
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- HCEMM‐SU Cardiometabolic Immunology Research GroupBudapestHungary
| | - Viktória É. Tóth
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- HCEMM‐SU Cardiometabolic Immunology Research GroupBudapestHungary
- MTA‐SE Momentum Cardio‐Oncology and Cardioimmunology Research GroupBudapestHungary
| | - Bence Ágg
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- Pharmahungary GroupSzegedHungary
- HUN‐REN–SU System Pharmacology Research Group, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Tamás Kovács
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- HCEMM‐SU Cardiometabolic Immunology Research GroupBudapestHungary
- MTA‐SE Momentum Cardio‐Oncology and Cardioimmunology Research GroupBudapestHungary
| | - Dániel Kucsera
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- HCEMM‐SU Cardiometabolic Immunology Research GroupBudapestHungary
- MTA‐SE Momentum Cardio‐Oncology and Cardioimmunology Research GroupBudapestHungary
| | - Csenger Kovácsházi
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Gábor B. Brenner
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Zoltán Giricz
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Péter Ferdinandy
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- Pharmahungary GroupSzegedHungary
- HUN‐REN–SU System Pharmacology Research Group, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Zoltán V. Varga
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- HCEMM‐SU Cardiometabolic Immunology Research GroupBudapestHungary
- MTA‐SE Momentum Cardio‐Oncology and Cardioimmunology Research GroupBudapestHungary
| |
Collapse
|
2
|
Fröhlich E. Animals in Respiratory Research. Int J Mol Sci 2024; 25:2903. [PMID: 38474149 DOI: 10.3390/ijms25052903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The respiratory barrier, a thin epithelial barrier that separates the interior of the human body from the environment, is easily damaged by toxicants, and chronic respiratory diseases are common. It also allows the permeation of drugs for topical treatment. Animal experimentation is used to train medical technicians, evaluate toxicants, and develop inhaled formulations. Species differences in the architecture of the respiratory tract explain why some species are better at predicting human toxicity than others. Some species are useful as disease models. This review describes the anatomical differences between the human and mammalian lungs and lists the characteristics of currently used mammalian models for the most relevant chronic respiratory diseases (asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary hypertension, pulmonary fibrosis, and tuberculosis). The generation of animal models is not easy because they do not develop these diseases spontaneously. Mouse models are common, but other species are more appropriate for some diseases. Zebrafish and fruit flies can help study immunological aspects. It is expected that combinations of in silico, in vitro, and in vivo (mammalian and invertebrate) models will be used in the future for drug development.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria
| |
Collapse
|
3
|
Park C, Singh M, Saeed MY, Nguyen CT, Roche ET. Biorobotic hybrid heart as a benchtop cardiac mitral valve simulator. DEVICE 2024; 2:100217. [PMID: 38312504 PMCID: PMC10836162 DOI: 10.1016/j.device.2023.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
In this work, we developed a high-fidelity beating heart simulator that provides accurate mitral valve pathophysiology. The benchtop platform is based on a biorobotic hybrid heart that combines preserved intracardiac tissue with soft robotic cardiac muscle providing dynamic left ventricular motion and precise anatomical features designed for testing intracardiac devices, particularly for mitral valve repair. The heart model is integrated into a mock circulatory loop, and the active myocardium drives fluid circulation producing physiological hemodynamics without an external pulsatile pump. Using biomimetic soft robotic technology, the heart can replicate both ventricular and septal wall motion, as well as intraventricular pressure-volume relationships. This enables the system to recreate the natural motion and function of the mitral valve, which allows us to demonstrate various surgical and interventional techniques. The biorobotic cardiovascular simulator allows for real-time hemodynamic data collection, direct visualization of the intracardiac procedure, and compatibility with clinical imaging modalities.
Collapse
Affiliation(s)
- Clara Park
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology; Cambridge, MA, USA 02139
- Department of Mechanical Engineering, Massachusetts Institute of Technology; Cambridge, MA, USA 02139
| | - Manisha Singh
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology; Cambridge, MA, USA 02139
| | - Mossab Y. Saeed
- Department of Cardiac Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA 02115
| | - Christopher T. Nguyen
- Cardiovascular Research Center, Massachusetts General Hospital; Charlestown, MA, USA 02114
- Cardiovascular Innovation Research Center, Heart Vascular Thoracic Institute, Cleveland Clinic; Cleveland, OH, USA 44195
- Imaging Sciences, Imaging Institute, Cleveland Clinic; Cleveland, OH, USA 44195
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic; Cleveland, OH, USA 44196
| | - Ellen T. Roche
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology; Cambridge, MA, USA 02139
- Department of Mechanical Engineering, Massachusetts Institute of Technology; Cambridge, MA, USA 02139
| |
Collapse
|
4
|
Singh M, Bonnemain J, Ozturk C, Ayers B, Saeed MY, Quevedo-Moreno D, Rowlett M, Park C, Fan Y, Nguyen CT, Roche ET. Robotic right ventricle is a biohybrid platform that simulates right ventricular function in (patho)physiological conditions and intervention. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1310-1326. [PMID: 39183977 PMCID: PMC11343235 DOI: 10.1038/s44161-023-00387-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/02/2023] [Indexed: 08/27/2024]
Abstract
The increasing recognition of the right ventricle (RV) necessitates the development of RV-focused interventions, devices and testbeds. In this study, we developed a soft robotic model of the right heart that accurately mimics RV biomechanics and hemodynamics, including free wall, septal and valve motion. This model uses a biohybrid approach, combining a chemically treated endocardial scaffold with a soft robotic synthetic myocardium. When connected to a circulatory flow loop, the robotic right ventricle (RRV) replicates real-time hemodynamic changes in healthy and pathological conditions, including volume overload, RV systolic failure and pressure overload. The RRV also mimics clinical markers of RV dysfunction and is validated using an in vivo porcine model. Additionally, the RRV recreates chordae tension, simulating papillary muscle motion, and shows the potential for tricuspid valve repair and replacement in vitro. This work aims to provide a platform for developing tools for research and treatment for RV pathophysiology.
Collapse
Affiliation(s)
- Manisha Singh
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jean Bonnemain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Caglar Ozturk
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brian Ayers
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Mossab Y. Saeed
- Department of Cardiac Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Diego Quevedo-Moreno
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Meagan Rowlett
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Clara Park
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yiling Fan
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Christopher T. Nguyen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
- Cardiovascular Innovation Research Center, Heart Vascular Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
- Imaging Sciences, Imaging Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ellen T. Roche
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
5
|
Farag A, Mandour AS, Hendawy H, Elhaieg A, Elfadadny A, Tanaka R. A review on experimental surgical models and anesthetic protocols of heart failure in rats. Front Vet Sci 2023; 10:1103229. [PMID: 37051509 PMCID: PMC10083377 DOI: 10.3389/fvets.2023.1103229] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Heart failure (HF) is a serious health and economic burden worldwide, and its prevalence is continuously increasing. Current medications effectively moderate the progression of symptoms, and there is a need for novel preventative and reparative treatments. The development of novel HF treatments requires the testing of potential therapeutic procedures in appropriate animal models of HF. During the past decades, murine models have been extensively used in fundamental and translational research studies to better understand the pathophysiological mechanisms of HF and develop more effective methods to prevent and control congestive HF. Proper surgical approaches and anesthetic protocols are the first steps in creating these models, and each successful approach requires a proper anesthetic protocol that maintains good recovery and high survival rates after surgery. However, each protocol may have shortcomings that limit the study's outcomes. In addition, the ethical regulations of animal welfare in certain countries prohibit the use of specific anesthetic agents, which are widely used to establish animal models. This review summarizes the most common and recent surgical models of HF and the anesthetic protocols used in rat models. We will highlight the surgical approach of each model, the use of anesthesia, and the limitations of the model in the study of the pathophysiology and therapeutic basis of common cardiovascular diseases.
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
- *Correspondence: Ahmed Farag
| | - Ahmed S. Mandour
- Department of Animal Medicine (Internal Medicine), Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
- Ahmed S. Mandour
| | - Hanan Hendawy
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Asmaa Elhaieg
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Ahmed Elfadadny
- Department of Animal Internal Medicine, Faculty of Veterinary Medicine, Damanhur University, Damanhur El-Beheira, Egypt
| | - Ryou Tanaka
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Ryou Tanaka
| |
Collapse
|
6
|
Gunata M, Parlakpinar H. Experimental heart failure models in small animals. Heart Fail Rev 2023; 28:533-554. [PMID: 36504404 DOI: 10.1007/s10741-022-10286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is one of the most critical health and economic burdens worldwide, and its prevalence is continuously increasing. HF is a disease that occurs due to a pathological change arising from the function or structure of the heart tissue and usually progresses. Numerous experimental HF models have been created to elucidate the pathophysiological mechanisms that cause HF. An understanding of the pathophysiology of HF is essential for the development of novel efficient therapies. During the past few decades, animal models have provided new insights into the complex pathogenesis of HF. Success in the pathophysiology and treatment of HF has been achieved by using animal models of HF. The development of new in vivo models is critical for evaluating treatments such as gene therapy, mechanical devices, and new surgical approaches. However, each animal model has advantages and limitations, and none of these models is suitable for studying all aspects of HF. Therefore, the researchers have to choose an appropriate experimental model that will fully reflect HF. Despite some limitations, these animal models provided a significant advance in the etiology and pathogenesis of HF. Also, experimental HF models have led to the development of new treatments. In this review, we discussed widely used experimental HF models that continue to provide critical information for HF patients and facilitate the development of new treatment strategies.
Collapse
Affiliation(s)
- Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Türkiye
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Türkiye.
| |
Collapse
|
7
|
Effect of injection of different doses of isoproterenol on the hearts of mice. BMC Cardiovasc Disord 2022; 22:409. [PMID: 36096747 PMCID: PMC9469628 DOI: 10.1186/s12872-022-02852-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022] Open
Abstract
Background Heart failure (HF) is one of the diseases that seriously threaten human health today and its mechanisms are very complex. Our study aims to confirm the optimal dose ISO-induced chronic heart failure mice model for better study of HF-related mechanisms and treatments in the future. Methods C57BL/6 mice were used to establish mice model of chronic heart failure. We injected isoproterenol subcutaneously in a dose gradient of 250 mg/kg, 200 mg/kg, 150 mg/kg, 100 mg/kg and 50 mg/kg. Echocardiography and ELISA were performed to figure out the occurrence of HF. We also supplemented the echocardiographic changes in mice over 30 days. Results Except group S and group E, echocardiographic abnormalities were found in other groups, suggesting a decrease in cardiac function. Except group S, myofibrolysis were found in the hearts of mice in other groups. Brain natriuretic peptide was significantly increased in groups B and D, and C-reactive protein was significantly increased in each group. Conclusion Our research finally found that the HFrEF mice model created by injection at a dose of 100 mg/kg for 7 days was the most suitable and a relatively stable chronic heart failure model could be obtained by placing it for 21 days. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02852-x.
Collapse
|
8
|
Chachques JC, Lila N, Soler-Botija C, Martinez-Ramos C, Valles A, Autret G, Perier MC, Mirochnik N, Monleon-Pradas M, Bayes-Genis A, Semino CE. Elastomeric cardiopatch scaffold for myocardial repair and ventricular support. Eur J Cardiothorac Surg 2021; 57:545-555. [PMID: 31539050 DOI: 10.1093/ejcts/ezz252] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/30/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Prevention of postischaemic ventricular dilatation progressing towards pathological remodelling is necessary to decrease ventricular wall deterioration. Myocardial tissue engineering may play a therapeutic role due to its capacity to replace the extracellular matrix, thereby creating niches for cell homing. In this experimental animal study, a biomimetic cardiopatch was created with elastomeric scaffolds and nanotechnologies. METHODS In an experimental animal study in 18 sheep, a cardiopatch was created with adipose tissue-derived progenitor cells seeded into an engineered bioimplant consisting of 3-dimensional bioabsorbable polycaprolactone scaffolds filled with a peptide hydrogel (PuraMatrix™). This patch was then transplanted to cover infarcted myocardium. Non-absorbable poly(ethyl) acrylate polymer scaffolds were used as controls. RESULTS Fifteen sheep were followed with ultrasound scans at 6 months, including echocardiography scans, tissue Doppler and spectral flow analysis and speckle-tracking imaging, which showed a reduction in longitudinal left ventricular deformation in the cardiopatch-treated group. Magnetic resonance imaging (late gadolinium enhancement) showed reduction of infarct size relative to left ventricular mass in the cardiopatch group versus the controls. Histopathological analysis at 6 months showed that the cardiopatch was fully anchored and integrated to the infarct area with minimal fibrosis interface, thereby promoting angiogenesis and migration of adipose tissue-derived progenitor cells to surrounding tissues. CONCLUSIONS This study shows the feasibility and effectiveness of a cardiopatch grafted onto myocardial infarction scars in an experimental animal model. This treatment decreased fibrosis, limited infarct scar expansion and reduced postischaemic ventricular deformity. A capillary network developed between our scaffold and the heart. The elastomeric cardiopatch seems to have a positive impact on ventricular remodelling and performance in patients with heart failure.
Collapse
Affiliation(s)
- Juan Carlos Chachques
- Laboratory Biosurgical Research, Alain Carpentier Foundation, Cardiac Surgery Pompidou Hospital, University Paris-Descartes, Paris, France
| | - Nermine Lila
- Laboratory Biosurgical Research, Alain Carpentier Foundation, Cardiac Surgery Pompidou Hospital, University Paris-Descartes, Paris, France
| | - Carolina Soler-Botija
- Research Cardiology Institute, Germans-Trias-Pujol Hospital, Badalona, Spain.,CIBER Cardiovascular, Carlos III Health Institute, Madrid, Spain
| | - Cristina Martinez-Ramos
- Center for Biomaterials and Tissue Engineering, Polytechnic University Valencia, Valencia, Spain
| | - Ana Valles
- Center for Biomaterials and Tissue Engineering, Polytechnic University Valencia, Valencia, Spain
| | - Gwennhael Autret
- Microcirculation Imaging Lab, Paris Cardiovascular Research Center (PARCC), University Paris, Paris, France
| | | | - Nicolas Mirochnik
- Cardiology Department, Pompidou Hospital, University Paris, Paris, France
| | - Manuel Monleon-Pradas
- Center for Biomaterials and Tissue Engineering, Polytechnic University Valencia, Valencia, Spain
| | - Antoni Bayes-Genis
- Research Cardiology Institute, Germans-Trias-Pujol Hospital, Badalona, Spain
| | - Carlos E Semino
- Bioengineering Department, IQS-School Engineering, Ramon-Llull University, Barcelona, Spain
| |
Collapse
|
9
|
Congestive heart failure model representing aortic banding induced hypertrophy: A study to analyse extent of pressure overload and alteration in myocardial structure and function. IJC HEART & VASCULATURE 2021; 34:100755. [PMID: 33816767 PMCID: PMC8008182 DOI: 10.1016/j.ijcha.2021.100755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 11/23/2022]
Abstract
Congestive Heart failure (CHF) is a severe pathology representing a major public health problem in industrialized nations which is increasing in prevalence and incidence. The aortic banding rat model provides steady progression of cardiac dysfunction under chronic pressure overload. Present study evaluated two abdominal aortic constriction techniques including constriction of aorta above renal arteries and between renal arteries. The extent of constriction was varied with 22 G and 24 G needles and the duration for evaluation of CHF was also varied by terminating the banded animals after 6 and 8 weeks of banding. Various hemodynamic, ECG and tissue parameters were evaluated after banding to see the progression of CHF. The findings revealed that the constriction of the aorta above both renal arteries with 24 G needle is a better technique amongst other tested banding techniques as the rate of progression of CHF was found to be maximum with it. On the basis of above study, it was concluded that, aortic banding above both renal arteries with 24 G needle is a better technique for induction of pressure overload and for further observation in transition of the cardiac compensatory to decompensatory phase, the duration of the model needs to be prolonged.
Collapse
|
10
|
Sabbah HN, Zhang K, Gupta RC, Xu J, Singh-Gupta V, Ma M, Stauber K, Nguyen N, Adams J. Intravenous Infusion of the β 3-Adrenergic Receptor Antagonist APD418 Improves Left Ventricular Systolic Function in Dogs With Systolic Heart Failure. J Card Fail 2020; 27:242-252. [PMID: 33352205 DOI: 10.1016/j.cardfail.2020.12.008] [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] [Received: 08/07/2020] [Revised: 11/08/2020] [Accepted: 12/08/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Unlike β1- and β2-adrenergic receptors (ARs), β3-AR stimulation inhibits cardiac contractility and relaxation. In the failing left ventricular (LV) myocardium, β3-ARs are upregulated, and can be maladaptive in the setting of decompensation by contributing to LV dysfunction. This study examined the effects of intravenous infusions of the β3-AR antagonist APD418 on cardiovascular function and safety in dogs with systolic heart failure (HF). METHODS AND RESULTS Three separate studies were performed in 21 dogs with coronary microembolization-induced HF (LV ejection fraction [LVEF] of approximately 35%). Studies 1 and 2 (n = 7 dogs each) were APD418 dose escalation studies (dosing range, 0.35-15.00 mg/kg/h) designed to identify an effective dose of APD418 to be used in study 3. Study 3, the sustained efficacy study, (n = 7 dogs) was a 6-hour constant intravenous infusion of APD418 at a dose of 4.224 mg/kg (0.70 mg/kg/h) measuring key hemodynamic endpoints (e.g., EF, cardiac output, the time velocity integral of the mitral inflow velocity waveform representing early filling to time-velocity integral representing left atrial contraction [Ei/Ai]). Studies 1 and 2 showed a dose-dependent increase of LVEF and Ei/Ai, the latter being an index of LV diastolic function. In study 3, infusion of APD418 over 6 hours increased LVEF from 31 ± 1% to 38 ± 1% (P < .05) and increased Ei/Ai from 3.4 ± 0.4 to 4.9 ± 0.5 (P < .05). Vehicle had no effect on the LVEF or Ei/Ai. In study 3, APD418 had no significant effects on the HR or the systemic blood pressure. CONCLUSIONS Intravenous infusions of APD418 in dogs with systolic HF elicit significant positive inotropic and lusitropic effects. These findings support the development of APD418 for the in-hospital treatment of patients with an acute exacerbation of chronic HF.
Collapse
Affiliation(s)
- Hani N Sabbah
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan.
| | - Kefei Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Ramesh C Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Jiang Xu
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Vinita Singh-Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Michael Ma
- Arena Pharmaceuticals, Inc., San Diego, California
| | | | | | - John Adams
- Arena Pharmaceuticals, Inc., San Diego, California
| |
Collapse
|
11
|
Cobo AA, Margallo FMS, Díaz CB, Blázquez VB, Bueno IG, Crisóstomo V. Anesthesia Protocols used to Create Ischemia Reperfusion Myocardial Infarcts in Swine. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2020; 59:478-487. [PMID: 32709259 DOI: 10.30802/aalas-jaalas-19-000137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The porcine ischemia-reperfusion model is one of the most commonly used for cardiology research and for testing interventions for myocardial regeneration. In creating ischemic reperfusion injury, the anesthetic protocol is important for assuring hemodynamic stability of the animal during the induction of the experimental lesion and may affect its postoperative survival. This paper reviews the many drugs and anesthetic protocols used in recent studies involving porcine models of ischemiareperfusion injury. The paper also summarizes the most important characteristics of some commonly used anesthetic drugs. Literature was selected for inclusion in this review if the authors described the anesthetic protocol used and also reported the mortality rate attributed to the creation of the model. This information is an important consideration because the anesthetic protocol can influence hemodynamic stability during the experimental induction of an acute myocardial infarction, thereby impacting the survival rate and affecting the number of animals needed for each study.
Collapse
Affiliation(s)
- Ana Abad Cobo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain;,
| | | | - Claudia Báez Díaz
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain; CIBERCV, Madrid, Spain
| | | | | | - Verónica Crisóstomo
- Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain; CIBERCV, Madrid, Spain
| |
Collapse
|
12
|
Torregroza C, Sadat N, Gomez Hamacher CJR, Scheiber D, von der Beek JC, Westenfeld R, Knorr IJ, Akhyari P, Sager M, Lichtenberg A, Saeed D. Chronic stable heart failure model in ovine species. Artif Organs 2020; 44:947-954. [PMID: 32645761 DOI: 10.1111/aor.13772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/23/2020] [Accepted: 06/30/2020] [Indexed: 11/28/2022]
Abstract
Establishing a chronic heart failure (HF) model is challenging, particularly in the ovine model. The aim of this study was to establish a reproducible model of HF in an ovine model. Seventeen sheep were operated using the left thoracotomy approach. Chronic HF was induced through ligation of the diagonal and marginal branches only. Perioperative hemodynamic and echocardiographic parameters were compared. A total of (3 ± 1) coronary ligations were used. Thirteen animals survived the procedure and were followed up for (15 ± 5) days. The mean arterial pressure, heart rate (HR), mean pulmonary artery pressure (mPAP), central venous pressure, and cardiac output at baseline and prior to animal sacrifice was (75 ± 14 mmHg) and (68 ± 16 mmHg) P = .261; (72 ± 9 bpm), (100 ± 28 bpm) P = .01; (15 ± 4 mmHg) and (18 ± 5 mmHg) P = .034; (10 ± 6 mmHg) and (8 ± 4 mmHg) P = .326; (3.4 ± 1 L/min) and (3.9 ± 1 L/min) P = .286, respectively. The LVEF at baseline and prior to animal sacrifice was (63 ± 13%) and (43 ± 6%) P = .012. Twelve surviving animals were supported with LVAD in a follow-up procedure. Chronic stable HF in sheep was successively established. Clinical symptoms and drastic increase in the mPAP and HR as well as echo findings were the most sensitive parameters of HF. This reproducible ovine model has proven to be highly promising for research regarding HF.
Collapse
Affiliation(s)
- Carolin Torregroza
- Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Najla Sadat
- Department of Cardiovascular Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
| | | | - Daniel Scheiber
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Jil-Cathrin von der Beek
- Central Unit for Animal Research and Animal Welfare Affairs, Heinrich Heine University, Duesseldorf, Germany
| | - Ralf Westenfeld
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Ivonne Jeanette Knorr
- Central Unit for Animal Research and Animal Welfare Affairs, Heinrich Heine University, Duesseldorf, Germany
| | - Payam Akhyari
- Department of Cardiovascular Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Martin Sager
- Central Unit for Animal Research and Animal Welfare Affairs, Heinrich Heine University, Duesseldorf, Germany
| | - Artur Lichtenberg
- Department of Cardiovascular Surgery, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Diyar Saeed
- University Department for Cardiac Surgery, Leipzig Heart Center, Leipzig, Germany
| |
Collapse
|
13
|
Niu P, Li L, Yin Z, Du J, Tan W, Huo Y. Speckle tracking echocardiography could detect the difference of pressure overload-induced myocardial remodelling between young and adult rats. J R Soc Interface 2020; 17:20190808. [PMID: 32093537 DOI: 10.1098/rsif.2019.0808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The assessment by speckle tracking echocardiography (STE) provides useful information on regional and global left ventricular (LV) functions. The aim of the study is to investigate if STE-based strain analysis could detect the difference of pressure overload-induced myocardial remodelling between young and adult rats. Physiological, haemodynamic, histological measurements were performed post-operatively in young and adult rats with transverse aortic constriction (TAC) as well as the age-matched shams. Two-way ANOVA was used to detect the statistical difference of various measured parameters. Pressure overload decreased the ejection fraction, fractional shortening, dp/dtmax and |dp/dtmin|, but increased the LV end-diastolic (ED) pressure in adult rat hearts for nine weeks after TAC operation than those in young rat hearts. Pressure overload also resulted in different changes of peak strain and strain rate in the free wall, but similar changes in the interventricular septum of young and adult rat hearts. The changes in myocardial remodelling were confirmed by the histological analysis including the increased apoptosis rate of myocytes and collagen area ratio in the free wall of adult rat hearts of LV hypertrophy when compared with the young. Pressure overload alters myocardial components in different degrees between young and adult animals. STE-based strain analysis could detect the subtle difference of pressure overload-induced myocardial remodelling between young and adult rats.
Collapse
Affiliation(s)
- Pei Niu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, People's Republic of China
| | - Li Li
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, People's Republic of China
| | - Zhongjie Yin
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, People's Republic of China
| | - Jie Du
- Beijing Anzhen Hospital Capital Medical University, Beijing, People's Republic of China
| | - Wenchang Tan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, People's Republic of China.,PKU-HKUST Shenzhen-Hongkong Institution, Shenzhen, People's Republic of China.,Shenzhen Graduate School, Peking University, Shenzhen, Guangdong, People's Republic of China
| | - Yunlong Huo
- PKU-HKUST Shenzhen-Hongkong Institution, Shenzhen, People's Republic of China.,Institute of Mechanobiology and Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
14
|
Law ML, Cohen H, Martin AA, Angulski ABB, Metzger JM. Dysregulation of Calcium Handling in Duchenne Muscular Dystrophy-Associated Dilated Cardiomyopathy: Mechanisms and Experimental Therapeutic Strategies. J Clin Med 2020; 9:jcm9020520. [PMID: 32075145 PMCID: PMC7074327 DOI: 10.3390/jcm9020520] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
: Duchenne muscular dystrophy (DMD) is an X-linked recessive disease resulting in the loss of dystrophin, a key cytoskeletal protein in the dystrophin-glycoprotein complex. Dystrophin connects the extracellular matrix with the cytoskeleton and stabilizes the sarcolemma. Cardiomyopathy is prominent in adolescents and young adults with DMD, manifesting as dilated cardiomyopathy (DCM) in the later stages of disease. Sarcolemmal instability, leading to calcium mishandling and overload in the cardiac myocyte, is a key mechanistic contributor to muscle cell death, fibrosis, and diminished cardiac contractile function in DMD patients. Current therapies for DMD cardiomyopathy can slow disease progression, but they do not directly target aberrant calcium handling and calcium overload. Experimental therapeutic targets that address calcium mishandling and overload include membrane stabilization, inhibition of stretch-activated channels, ryanodine receptor stabilization, and augmentation of calcium cycling via modulation of the Serca2a/phospholamban (PLN) complex or cytosolic calcium buffering. This paper addresses what is known about the mechanistic basis of calcium mishandling in DCM, with a focus on DMD cardiomyopathy. Additionally, we discuss currently utilized therapies for DMD cardiomyopathy, and review experimental therapeutic strategies targeting the calcium handling defects in DCM and DMD cardiomyopathy.
Collapse
Affiliation(s)
- Michelle L. Law
- Department of Family and Consumer Sciences, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA;
| | - Houda Cohen
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Ashley A. Martin
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Addeli Bez Batti Angulski
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
- Correspondence: ; Tel.: +1-612-625-5902; Fax: +1-612-625-5149
| |
Collapse
|
15
|
Wireless, battery-free, fully implantable multimodal and multisite pacemakers for applications in small animal models. Nat Commun 2019; 10:5742. [PMID: 31848334 PMCID: PMC6917818 DOI: 10.1038/s41467-019-13637-w] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 11/12/2019] [Indexed: 11/09/2022] Open
Abstract
Small animals support a wide range of pathological phenotypes and genotypes as versatile, affordable models for pathogenesis of cardiovascular diseases and for exploration of strategies in electrotherapy, gene therapy, and optogenetics. Pacing tools in such contexts are currently limited to tethered embodiments that constrain animal behaviors and experimental designs. Here, we introduce a highly miniaturized wireless energy-harvesting and digital communication electronics for thin, miniaturized pacing platforms weighing 110 mg with capabilities for subdermal implantation and tolerance to over 200,000 multiaxial cycles of strain without degradation in electrical or optical performance. Multimodal and multisite pacing in ex vivo and in vivo studies over many days demonstrate chronic stability and excellent biocompatibility. Optogenetic stimulation of cardiac cycles with in-animal control and induction of heart failure through chronic pacing serve as examples of modes of operation relevant to fundamental and applied cardiovascular research and biomedical technology. Pacing tools that support small animals and can serve as models for pathogenesis of cardiovascular diseases are currently not available. Here, the authors report a miniaturized wireless battery-free implantable multimodal and multisite pacemaker that provides unlimited stimulation to test subjects.
Collapse
|
16
|
Janssen PML, Elnakish MT. Modeling heart failure in animal models for novel drug discovery and development. Expert Opin Drug Discov 2019; 14:355-363. [PMID: 30861352 DOI: 10.1080/17460441.2019.1582636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION When investigating drugs that treat heart diseases, it is critical when choosing an animal model for the said model to produce data that is translatable to the human patient population, while keeping in mind the principles of reduction, refinement, and replacement of the animal model in the research. Areas covered: In this review, the authors focus on mammalian models developed to study the impact of drug treatments on human heart failure. Furthermore, the authors address human patient variability and animal model invariability as well as the considerations that need to be made regarding choice of species. Finally, the authors discuss some of the most common models for the two most prominent human heart failure etiologies; increased load on the heart and myocardial ischemia. Expert opinion: In the authors' opinion, the data generated by drug studies is often heavily impacted by the choice of species and the physiologically relevant conditions under which the data are collected. Approaches that use multiple models and are not restricted to small rodents but involve some verification on larger mammals or on human myocardium, are needed to advance drug discovery for the very large patient population that suffers from heart failure.
Collapse
Affiliation(s)
- Paul M L Janssen
- a Department of Physiology and Cell Biology , The Ohio State University Wexner Medical Center , Columbus, OH, USA.,b Dorothy M. Davis Heart and Lung Research Institute , The Ohio State University Wexner Medical Center , Columbus, OH, USA.,c Department of Internal Medicine , The Ohio State University Wexner Medical Center , Columbus, OH, USA
| | - Mohammad T Elnakish
- a Department of Physiology and Cell Biology , The Ohio State University Wexner Medical Center , Columbus, OH, USA.,b Dorothy M. Davis Heart and Lung Research Institute , The Ohio State University Wexner Medical Center , Columbus, OH, USA
| |
Collapse
|
17
|
Corremans R, Adão R, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Update on pathophysiology and preventive strategies of anthracycline-induced cardiotoxicity. Clin Exp Pharmacol Physiol 2018; 46:204-215. [DOI: 10.1111/1440-1681.13036] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/31/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Affiliation(s)
| | - Rui Adão
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | | | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
- Faculty of Nutrition and Food Sciences; University of Porto; Porto Portugal
| |
Collapse
|
18
|
Feng Y, Hemmeryckx B, Frederix L, Lox M, Wu J, Heggermont W, Lu HR, Gallacher D, Oyen R, Lijnen HR, Ni Y. Monitoring reperfused myocardial infarction with delayed left ventricular systolic dysfunction in rabbits by longitudinal imaging. Quant Imaging Med Surg 2018; 8:754-769. [PMID: 30306056 DOI: 10.21037/qims.2018.09.05] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background An experimental imaging platform for longitudinal monitoring and evaluation of cardiac morphology-function changes has been long desired. We sought to establish such a platform by using a rabbit model of reperfused myocardial infarction (MI) that develops chronic left ventricle systolic dysfunction (LVSD) within 7 weeks. Methods Fifty-five New Zeeland white (NZW) rabbits received sham-operated or 60-min left circumflex coronary artery (LCx) ligation followed by reperfusion. Cardiac magnetic resonance imaging (cMRI), transthoracic echocardiography (echo), and blood samples were collected at baseline, in acute (48 hours or 1 week) and chronic (7 weeks) stage subsequent to MI for in vivo assessment of infarct size, cardiac morphology, LV function, and myocardial enzymes. Seven weeks post MI, animals were sacrificed and heart tissues were processed for histopathological staining. Results The success rate of surgical operation was 87.27%. The animal mortality rates were 12.7% and 3.6% both in acute and chronic stage separately. Serum levels of the myocardial enzyme cardiac Troponin T (cTnT) were significantly increased in MI rabbits as compared with sham animals after 4 hours of operation (P<0.05). According to cardiac morphology and function changes, 4 groups could be distinguished: sham rabbits (n=12), and MI rabbits with no (MI_NO_LVSD; n=10), moderate (MI_M_LVSD; n=9) and severe (MI_S_LVSD; n=15) LVSD. No significant differences in cardiac function or wall thickening between sham and MI_NO_LVSD rabbits were observed at both stages using both cMRI and echo methods. cMRI data showed that MI_M_LVSD rabbits exhibited a reduction of ejection fraction (EF) and an increase in end-systolic volume (ESV) at the acute phase, while at the chronic stage these parameters did not change further. Moreover, in MI_S_LVSD animals, these observations were more striking at the acute stage followed by a further decline in EF and increase in ESV at the chronic stage. Lateral wall thickening determined by cMRI was significantly decreased in MI_M_LVSD versus MI_NO_LVSD animals at both stages (P<0.05). As for MI_S_LVSD versus MI_M_LVSD rabbits, the thickening of anterior, inferior and lateral walls was significantly more decreased at both stages (P<0.05). Echo confirmed the findings of cMRI. Furthermore, these in vivo outcomes including those from vivid cine cMRI could be supported by exactly matched ex vivo histomorphological evidences. Conclusions Our findings indicate that chronic LVSD developed over time after surgery-induced MI in rabbits can be longitudinally evaluated using non-invasive imaging techniques and confirmed by the entire-heart-slice histomorphology. This experimental LVSD platform in rabbits may interest researchers in the field of experimental cardiology and help strengthen drug development and translational research for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Yuanbo Feng
- Radiology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Bianca Hemmeryckx
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Liesbeth Frederix
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marleen Lox
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Jun Wu
- Ultrasound Diagnostic department, the second affiliated hospital of Dalian Medical University, Dalian 116000, China
| | - Ward Heggermont
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Hua Rong Lu
- Translational Sciences, Safety Pharmacology Research, Janssen Research & Development, Janssen Pharmaceutical NV, Beerse, Belgium
| | - David Gallacher
- Translational Sciences, Safety Pharmacology Research, Janssen Research & Development, Janssen Pharmaceutical NV, Beerse, Belgium
| | - Raymond Oyen
- Radiology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - H Roger Lijnen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Yicheng Ni
- Radiology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Li B, Cui Y, Zhang D, Luo X, Luo F, Li B, Tang Y. The characteristics of a porcine mitral regurgitation model. Exp Anim 2018; 67:463-477. [PMID: 29794373 PMCID: PMC6219876 DOI: 10.1538/expanim.18-0045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The porcine mitral regurgitation (MR) model is a common cardiovascular animal model.
Standardized manufacturing processes can improve the uniformity and success rate of the
model, and systematic research can evaluate its potential use. In this study, 17 pigs were
divided into an experimental group (n=11) and a control group (n=6). We used a homemade
retractor to cut the mitral chordae via the left atrial appendage to establish a model of
MR; the control group underwent a sham surgery. The model animals were followed for 30
months after the surgery. Enlargement and fibrosis of the left atrium were significant in
the experimental group compared with those in the control group, and left atrial systolic
function decreased significantly. In addition, model animals showed preserved left
ventricular systolic function. There were no differences in left atrial potential or left
ventricular myocardial fibrosis between the two groups. Atrial fibrillation susceptibility
in the experimental group was higher than that in the control group. Our method enables
the simple and effective production of a MR model with severe reflux that can be used for
pathophysiological studies of MR, as well as for the development of preclinical surgical
instruments and their evaluation. This model could also be used to study atrial
fibrillation and myocardial fibrosis but is not suitable for studies of heart failure.
Collapse
Affiliation(s)
- Bo Li
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North lishi Road, Xicheng District, Beijing 100037, China
| | - Yongchun Cui
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North lishi Road, Xicheng District, Beijing 100037, China
| | - Dong Zhang
- Department of Cardiovascular surgery, Beijing Jishuitan Hospital, No. 31 Xinjiekou East Street, Xicheng District, Beijing 100035, China
| | - Xiaokang Luo
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North lishi Road, Xicheng District, Beijing 100037, China
| | - Fuliang Luo
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North lishi Road, Xicheng District, Beijing 100037, China
| | - Bin Li
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North lishi Road, Xicheng District, Beijing 100037, China
| | - Yue Tang
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 North lishi Road, Xicheng District, Beijing 100037, China
| |
Collapse
|
20
|
Teneggi V, Sivakumar N, Chen D, Matter A. Drugs’ development in acute heart failure: what went wrong? Heart Fail Rev 2018; 23:667-691. [DOI: 10.1007/s10741-018-9707-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
21
|
Wu H, Li L, Niu P, Huang X, Liu J, Zhang F, Shen W, Tan W, Wu Y, Huo Y. The Structure-function remodeling in rabbit hearts of myocardial infarction. Physiol Rep 2018. [PMID: 28637704 PMCID: PMC5492201 DOI: 10.14814/phy2.13311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Animal models are of importance to investigate basic mechanisms for ischemic heart failure (HF). The objective of the study was to create a rabbit model through multiple coronary artery ligations to investigate the postoperative structure‐function remodeling of the left ventricle (LV) and coronary arterial trees. Here, we hypothesize that the interplay of the degenerated coronary vasculature and increased ventricle wall stress relevant to cardiac fibrosis in vicinity of myocardial infarction (MI) precipitates the incidence and progression of ischemic HF. Echocardiographic measurements showed an approximately monotonic drop of fractional shortening and ejection fraction from 40% and 73% down to 28% and 58% as well as persistent enlargement of LV cavity and slight mitral regurgitation at postoperative 12 weeks. Micro‐CT and histological measurements showed that coronary vascular rarefaction and cardiac fibrosis relevant to inflammation occurred concurrently in vicinity of MI at postoperative 12 weeks albeit there was compensatory vascular growth at postoperative 6 weeks. These findings validate the proposed rabbit model and prove the hypothesis. The post‐MI rabbit model can serve as a reference to test various drugs for treatment of ischemic HF.
Collapse
Affiliation(s)
- Haotian Wu
- School of Basic Medical Sciences, Nanjing University of Traditional Chinese Medicine, Nanjing, China.,Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang, China
| | - Li Li
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Pei Niu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China.,College of Medicine, Hebei University, Baoding, China
| | - Xu Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Jinyi Liu
- College of Medicine, Hebei University, Baoding, China
| | | | - Wenzeng Shen
- College of Medicine, Hebei University, Baoding, China
| | - Wenchang Tan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China.,Shenzhen Graduate School, Peking University, Shenzhen, China.,PKU-HKUST Shenzhen-Hongkong Institution, Shenzhen, China
| | - Yiling Wu
- School of Basic Medical Sciences, Nanjing University of Traditional Chinese Medicine, Nanjing, China .,Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang, China.,Key Laboratory, State Administration of Traditional Chinese Medicine (Cardiovascular and cerebrovascular collateral diseases), Shijiazhuang, China.,Hebei Province Key Laboratory of Collateral Diseases, Shijiazhuang, China
| | - Yunlong Huo
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China .,PKU-HKUST Shenzhen-Hongkong Institution, Shenzhen, China
| |
Collapse
|
22
|
ANGPTL8 reverses established adriamycin cardiomyopathy by stimulating adult cardiac progenitor cells. Oncotarget 2018; 7:80391-80403. [PMID: 27823982 PMCID: PMC5348328 DOI: 10.18632/oncotarget.13061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 10/07/2016] [Indexed: 12/18/2022] Open
Abstract
Established adriamycin cardiomyopathy is a lethal disease. When congestive heart failure develops, mortality is approximately 50% in a year. It has been known that ANGPTLs has various functions in lipid metabolism, inflammation, cancer cell invasion, hematopoietic stem activity and diabetes. We hypothesized that ANGPTL8 is capable of maintaining heart function by stimulating adult cardiac progenitor cells to initiate myocardial regeneration. We employed UTMD to deliver piggybac transposon plasmids with the human ANGPTL8 gene to the liver of rats with adriamycin cardiomyopathy. After ANGPTL8 gene liver delivery, overexpression of transgenic human ANGPTL8 was found in rat liver cells and blood. UTMD- ANGPTL8 gene therapy restored LV mass, fractional shortening index, and LV posterior wall diameter to nearly normal. Our results also showed that ANGPTL8 reversed established ADM cardiomyopathy. This was associated with activation of ISL-1 positive cardiac progenitor cells in the epicardium. A time-course experiment shown that ISL-1 cardiac progenitor cells proliferated and formed a niche in the epicardial layer and then migrated into sub-epicardium. The observed myocardial regeneration accompanying reversal of adriamycin cardiomyopathy was associated with upregulation of PirB expression on the cell membrane of cardiac muscle cells or progenitor cells stimulated by ANGPTL8.
Collapse
|
23
|
Short-term and long-term models of doxorubicin-induced cardiomyopathy in rats: A comparison of functional and histopathological changes. ACTA ACUST UNITED AC 2017; 69:213-219. [PMID: 28153388 DOI: 10.1016/j.etp.2017.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 12/28/2016] [Accepted: 01/16/2017] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Doxorubicin (DXR), an anthracyclic antineoplastic agent, is one of the most commonly drug utilized to induce dilated cardiomyopathy (DCM) and heart failure (HF), but the well optimized protocol for cardiomyopathy induction leading to development of cardiac systolic dysfunction is unclear. This study aims to critically compare short-term and long-term DXR injection protocols for the induction of DCM in rats. METHODS Animals were allocated into 3 experimental groups: a ST (short-term DXR injection) group, in which animals received 6 intraperitoneal (i.p.) injections of DXR (2.5mg/kg per dose) over a period of 2 weeks (cumulative dose of 15mg/kg); a LT (long-term DXR injection) group in which animals received weekly i.p. injections of DXR (2mg/kg per dose) over a period of 9 weeks (cumulative dose of 18mg/kg); and a control group in which animals received an appropriate volume of 0.9% saline i.p. All animals were submitted to echocardiography analysis at baseline and after completion treatment. Afterwards, the hearts were collected for conventional light microscopy and collagen quantification. RESULTS Morphological myocardial analysis of both DXR-treated groups showed an identical pattern of swollen and vacuolated cardiomyocytes and disorganization of myofibrils. There was pronounced interstitial fibrosis in both groups of DXR-treated hearts as compared to controls, as assessed by the interstitial collagen volume fraction. There was no difference in interstitial fibrosis between the ST and LT groups. The echocardiography analysis of the LT group showed structural and functional findings compatible with DCM, including increased left ventricular systolic (5.02±0.96mm) and diastolic (7.68±0.96mm) dimensions and reduction of ejection fraction (69.40±8.51%) as compared to the ST group (4.10±0.89mm, 7.32±0.84, and 79.68±7.23%, respectively) and control group (4.07±0.72mm, 7.17±0.68mm and 80.08±4.71%, respectively), ANOVA p<0.01. CONCLUSIONS These results indicate that LT injection of DXR is more effective than ST injection in inducing left ventricular dysfunction and structural cardiac changes resembling those found in dilated cardiomyopathy.
Collapse
|
24
|
Giri SR, Bhoi B, Jain MR, Gatne MM. Cardioprotective role of peroxisome proliferator-activated receptor-γ agonist, rosiglitazone in a unique murine model of diabetic cardiopathy. Life Sci 2016; 162:1-13. [PMID: 27530514 DOI: 10.1016/j.lfs.2016.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/09/2016] [Accepted: 08/12/2016] [Indexed: 10/21/2022]
Abstract
AIMS Rosiglitazone (RSZ), a PPARγ agonist was potent efficacious insulin sensitizing blockbuster drug for treatment of Type 2 diabetes mellitus (T2DM) but the benefit of PPARγ activation in congestive heart failure (CHF) was controversial. The present work was planned to study the role of RSZ in diabetic cardiopathy. MAIN METHODS Zucker fa/fa rats, the genetic model of T2DM were subjected to constriction of suprarenal abdominal aorta so that they represent a combined model of diabetes and cardiopathy. The development cardiopathy was assessed biochemically (plasma BNP and aldosterone levels), using echocardiography and expression angiotensin II receptor type 1a gene in heart and Endothelin-1 gene in aorta. Rats were treated with RSZ and in combination with amiloride for four weeks and were assessed to evaluate the effect of RSZ or amiloride or its combination on antidiabetic activity, adverse or toxic effects and congestive heart failure status. KEY FINDINGS RSZ shows its anti-diabetic effect from 0.3mg/kg dose onwards and at 3mg/kg dose levels it caused beneficial effects (reduction of blood pressure) on cardiovascular system and at highest (30mg/kg) dose it starts showing adverse effects like body weight gain, edema, left ventricular hypertrophy. However, when highest dose of RSZ animals were treated with amiloride (ENaC inhibitor) at 2mg/kg the reversal of the adverse effects was evident, indicating the combination of RSZ and amiloride is beneficial in diabetic cardiopathy model. SIGNIFICANCE RSZ and amiloride combination appeared promising treatment in diabetic patients with cardiopathy without any side effect.
Collapse
Affiliation(s)
- Suresh R Giri
- Department of Pharmacology & Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad 382 213, Gujarat, India; Department of Pharmacology & Toxicology, Bombay Veterinary College, Parel, Mumbai 400012, India.
| | - Bibhuti Bhoi
- Department of Pharmacology & Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad 382 213, Gujarat, India
| | - Mukul R Jain
- Department of Pharmacology & Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad 382 213, Gujarat, India
| | - Madhumanjiri M Gatne
- Department of Pharmacology & Toxicology, Bombay Veterinary College, Parel, Mumbai 400012, India
| |
Collapse
|
25
|
Abstract
With the impressive advancement in high-throughput 'omics' technologies over the past two decades, epigenetic mechanisms have emerged as the regulatory interface between the genome and environmental factors. These mechanisms include DNA methylation, histone modifications, ATP-dependent chromatin remodeling and RNA-based mechanisms. Their highly interdependent and coordinated action modulates the chromatin structure controlling access of the transcription machinery and thereby regulating expression of target genes. Given the rather limited proliferative capability of human cardiomyocytes, epigenetic regulation appears to play a particularly important role in the myocardium. The highly dynamic nature of the epigenome allows the heart to adapt to environmental challenges and to respond quickly and properly to cardiac stress. It is now becoming evident that histone-modifying and chromatin-remodeling enzymes as well as numerous non-coding RNAs play critical roles in cardiac development and function, while their dysregulation contributes to the onset and development of pathological cardiac remodeling culminating in HF. This review focuses on up-to-date knowledge about the epigenetic mechanisms and highlights their emerging role in the healthy and failing heart. Uncovering the determinants of epigenetic regulation holds great promise to accelerate the development of successful new diagnostic and therapeutic strategies in human cardiac disease.
Collapse
Affiliation(s)
- José Marín-García
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Ave., Highland Park, NJ, 08904, USA,
| | | |
Collapse
|
26
|
Common swine models of cardiovascular disease for research and training. Lab Anim (NY) 2016; 45:67-74. [DOI: 10.1038/laban.935] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/28/2015] [Indexed: 12/14/2022]
|
27
|
Gong X, Qin S, Huang Z, Zhou N, Yang Z, Nie Z, Dai S, Yao R, Wei Z, Ge J, Su Y, Shu X. “Pacing Bigeminal”. Int Heart J 2016; 57:747-752. [DOI: 10.1536/ihj.16-043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Xue Gong
- Department of Echocardiography, Zhongshan Hospital, Fudan University
| | - Shengmei Qin
- Department of Cardiology, Zhongshan Hospital, Fudan University
| | - Zheyong Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University
| | - Nianwei Zhou
- Department of Echocardiography, Zhongshan Hospital, Fudan University
| | - Zhaohua Yang
- Department of Cardiology Surgical, Zhongshan Hospital, Fudan University
| | - Zhenning Nie
- Department of Cardiology, Zhongshan Hospital, Fudan University
| | - Shimo Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University
| | - Ruiming Yao
- Department of Cardiology, Zhongshan Hospital, Fudan University
| | | | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University
| | - Yangang Su
- Department of Cardiology, Zhongshan Hospital, Fudan University
| | - Xianhong Shu
- Department of Echocardiography, Zhongshan Hospital, Fudan University
- Department of Cardiology, Zhongshan Hospital, Fudan University
| |
Collapse
|
28
|
Akhmedov AT, Rybin V, Marín-García J. Mitochondrial oxidative metabolism and uncoupling proteins in the failing heart. Heart Fail Rev 2015; 20:227-49. [PMID: 25192828 DOI: 10.1007/s10741-014-9457-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite significant progress in cardiovascular medicine, myocardial ischemia and infarction, progressing eventually to the final end point heart failure (HF), remain the leading cause of morbidity and mortality in the USA. HF is a complex syndrome that results from any structural or functional impairment in ventricular filling or blood ejection. Ultimately, the heart's inability to supply the body's tissues with enough blood may lead to death. Mechanistically, the hallmarks of the failing heart include abnormal energy metabolism, increased production of reactive oxygen species (ROS) and defects in excitation-contraction coupling. HF is a highly dynamic pathological process, and observed alterations in cardiac metabolism and function depend on the disease progression. In the early stages, cardiac remodeling characterized by normal or slightly increased fatty acid (FA) oxidation plays a compensatory, cardioprotective role. However, upon progression of HF, FA oxidation and mitochondrial oxidative activity are decreased, resulting in a significant drop in cardiac ATP levels. In HF, as a compensatory response to decreased oxidative metabolism, glucose uptake and glycolysis are upregulated, but this upregulation is not sufficient to compensate for a drop in ATP production. Elevated mitochondrial ROS generation and ROS-mediated damage, when they overwhelm the cellular antioxidant defense system, induce heart injury and contribute to the progression of HF. Mitochondrial uncoupling proteins (UCPs), which promote proton leak across the inner mitochondrial membrane, have emerged as essential regulators of mitochondrial membrane potential, respiratory activity and ROS generation. Although the physiological role of UCP2 and UCP3, expressed in the heart, has not been clearly established, increasing evidence suggests that these proteins by promoting mild uncoupling could reduce mitochondrial ROS generation and cardiomyocyte apoptosis and ameliorate thereby myocardial function. Further investigation on the alterations in cardiac UCP activity and regulation will advance our understanding of their physiological roles in the healthy and diseased heart and also may facilitate the development of novel and more efficient therapies.
Collapse
Affiliation(s)
- Alexander T Akhmedov
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Avenue, Highland Park, NJ, 08904, USA
| | | | | |
Collapse
|
29
|
Laganà A, Veneziano D, Spata T, Tang R, Zhu H, Mohler PJ, Kilic A. Identification of General and Heart-Specific miRNAs in Sheep (Ovis aries). PLoS One 2015; 10:e0143313. [PMID: 26599010 PMCID: PMC4657999 DOI: 10.1371/journal.pone.0143313] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/03/2015] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) are small regulatory RNAs crucial for modulation of signaling pathways in multiple organs. While the link between miRNAs and heart disease has grown more readily apparent over the past three years, these data are primarily limited to small animal models or cell-based systems. Here, we performed a high-throughput RNA sequencing (RNAseq) analysis of left ventricle and other tissue from a pre-clinical ovine model. We identified 172 novel miRNA precursors encoding a total of 264 mature miRNAs. Notably, 84 precursors were detected in both the left ventricle and other tissues. However, 10 precursors, encoding 11 mature sequences, were specific to the left ventricle. Moreover, the total 168 novel miRNA precursors included 22 non-conserved ovine-specific sequences. Our data identify and characterize novel miRNAs in the left ventricle of sheep, providing fundamental new information for our understanding of protein regulation in heart and other tissues.
Collapse
Affiliation(s)
- Alessandro Laganà
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
- * E-mail: (AL); (AK)
| | - Dario Veneziano
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
- Department of Clinical and Molecular Biomedicine, University of Catania, Catania, Italy
| | - Tyler Spata
- Department of Surgery, The Ohio State University, Columbus, OH, United States of America
| | - Richard Tang
- Department of Surgery, The Ohio State University, Columbus, OH, United States of America
| | - Hua Zhu
- Department of Surgery, The Ohio State University, Columbus, OH, United States of America
| | - Peter J. Mohler
- The Davis Heart and Lung Research Institute, Departments of Physiology & Cell Biology and Internal Medicine, The Ohio State University Medical Center, Columbus, OH, United States of America
| | - Ahmet Kilic
- Department of Surgery, The Ohio State University, Columbus, OH, United States of America
- * E-mail: (AL); (AK)
| |
Collapse
|
30
|
Asif SM, Hansen J, Khan MS, Walden SD, Jensen MO, Braaten BD, Ewert DL. Design and In Vivo Test of a Batteryless and Fully Wireless Implantable Asynchronous Pacing System. IEEE Trans Biomed Eng 2015; 63:1070-1081. [PMID: 26357395 DOI: 10.1109/tbme.2015.2477403] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Goal: The aim of this study is to develop a novel fully wireless and batteryless technology for cardiac pacing. METHODS This technology uses radio frequency (RF) energy to power the implanted electrode in the heart. An implantable electrode antenna was designed for 1.2 GHz; then, it was tested in vitro and, subsequently, integrated with the rectifier and pacing circuit to make a complete electrode. The prototype implanted electrode was tested in vivo in an ovine subject, implanting it on the epicardial surface of the left ventricle. The RF energy, however, was transmitted to the implanted electrode using a horn antenna positioned 25 cm above the thorax of the sheep. RESULTS It was demonstrated that a small implanted electrode can capture and harvest enough safe recommended RF energy to achieve pacing. Electrocardiogram signals were recorded during the experiments, which demonstrated asynchronous pacing achieved at three different rates. CONCLUSION These results show that the proposed method has a great potential to be used for stimulating the heart and provides pacing, without requiring any leads or batteries. It hence has the advantage of potentially lasting indefinitely and may never require replacement during the life of the patient. SIGNIFICANCE The proposed method brings forward transformational possibilities in wireless cardiac pacing, and also in powering up the implantable devices.
Collapse
|
31
|
Thorn S, Sinusas AJ. Creation of clinically relevant model of chronic heart failure: Application of multi-modality imaging to define physiology. J Nucl Cardiol 2015; 22:673-6. [PMID: 25698482 DOI: 10.1007/s12350-015-0081-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 01/20/2015] [Indexed: 10/24/2022]
Affiliation(s)
- Stephanie Thorn
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, P.O. Box 208017, New Haven, CT, 06520-8017, USA
| | | |
Collapse
|
32
|
Crisostomo V, Casado JG, Baez-Diaz C, Blazquez R, Sanchez-Margallo FM. Allogeneic cardiac stem cell administration for acute myocardial infarction. Expert Rev Cardiovasc Ther 2015; 13:285-99. [DOI: 10.1586/14779072.2015.1011621] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
33
|
Martínez-Ramos C, Rodríguez-Pérez E, Garnes MP, Chachques JC, Moratal D, Vallés-Lluch A, Monleón Pradas M. Design and Assembly Procedures for Large-Sized Biohybrid Scaffolds as Patches for Myocardial Infarct. Tissue Eng Part C Methods 2014; 20:817-27. [DOI: 10.1089/ten.tec.2013.0489] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Eduard Rodríguez-Pérez
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Manuel Pérez Garnes
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Juan Carlos Chachques
- Laboratory of Biosurgical Research, Department of Cardiovascular Surgery, Georges Pompidou European Hospital, Paris, France
| | - David Moratal
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Ana Vallés-Lluch
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| |
Collapse
|
34
|
Gálvez-Montón C, Prat-Vidal C, Díaz-Güemes I, Crisóstomo V, Soler-Botija C, Roura S, Llucià-Valldeperas A, Perea-Gil I, Sánchez-Margallo FM, Bayes-Genis A. Comparison of two preclinical myocardial infarct models: coronary coil deployment versus surgical ligation. J Transl Med 2014; 12:137. [PMID: 24885652 PMCID: PMC4047266 DOI: 10.1186/1479-5876-12-137] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/13/2014] [Indexed: 11/16/2022] Open
Abstract
Background Despite recent advances, myocardial infarction (MI) remains the leading cause of death worldwide. Pre-clinical animal models that closely mimic human MI are pivotal for a quick translation of research and swine have similarities in anatomy and physiology. Here, we compared coronary surgical ligation versus coil embolization MI models in swine. Methods Fifteen animals were randomly distributed to undergo surgical ligation (n = 7) or coil embolization (n = 8). We evaluated infarct size, scar fibrosis, inflammation, myocardial vascularization, and cardiac function by magnetic resonance imaging (MRI). Results Thirty-five days after MI, there were no differences between the models in infarct size (P = 0.53), left ventricular (LV) ejection fraction (P = 0.19), LV end systolic volume (P = 0.22), LV end diastolic volume (P = 0.84), and cardiac output (P = 0.89). Histologically, cardiac scars did not differ and the collagen content, collagen type I (I), collagen type III (III), and the I/III ratio were similar in both groups. Inflammation was assessed using specific anti-CD3 and anti-CD25 antibodies. There was similar activation of inflammation throughout the heart after coil embolization (P = 0.78); while, there were more activated lymphocytes in the infarcted myocardium in the surgical occlusion model (P = 0.02). Less myocardial vascularization in the infarction areas compared with the border and remote zones only in coil embolization animals was observed (P = 0.004 and P = 0.014, respectively). Conclusions Our results support that surgical occlusion and coil embolization MI models generate similar infarct size, cardiac function impairment, and myocardial fibrosis; although, inflammation and myocardial vascularization levels were closer to those found in humans when coil embolization was performed.
Collapse
Affiliation(s)
- Carolina Gálvez-Montón
- ICREC (Heart Failure and Cardiac Regeneration) Research Program, IGTP, Cardiology Service, Hospital Universitari Germans Trias i Pujol, Crta, Can Ruti, Camí de les Escoles, s/n, 08916 Badalona, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Oliveira LFLD, Mejia J, Carvalho EEVD, Lataro RM, Frassetto SN, Fazan R, Salgado HC, Galvis-Alonso OY, Simões MV. Myocardial infarction area quantification using high-resolution SPECT images in rats. Arq Bras Cardiol 2014; 101:59-67. [PMID: 23917507 PMCID: PMC3998176 DOI: 10.5935/abc.20130110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 03/06/2013] [Indexed: 01/29/2023] Open
Abstract
Background Imaging techniques enable in vivo sequential assessment of the morphology and
function of animal organs in experimental models. We developed a device for
high-resolution single photon emission computed tomography (SPECT) imaging based
on an adapted pinhole collimator. Objective To determine the accuracy of this system for quantification of myocardial infarct
area in rats. Methods Thirteen male Wistar rats (250 g) underwent experimental myocardial infarction by
occlusion of the left coronary artery. After 4 weeks, SPECT images were acquired
1.5 hours after intravenous injection of 555 MBq of 99mTc-Sestamibi. The
tomographic reconstruction was performed by using specially developed software
based on the Maximum Likelihood algorithm. The analysis of the data included the
correlation between the area of perfusion defects detected by scintigraphy and
extent of myocardial fibrosis assessed by histology. Results The images showed a high target organ/background ratio with adequate visualization
of the left ventricular walls and cavity. All animals presenting infarction areas
were correctly identified by the perfusion images. There was no difference of the
infarct area as measured by SPECT (21.1 ± 21.2%) and by histology (21.7 ± 22.0%;
p=0.45). There was a strong correlation between individual values of the area of
infarction measured by these two methods. Conclusion The developed system presented adequate spatial resolution and high accuracy for
the detection and quantification of myocardial infarction areas, consisting in a
low cost and versatile option for high-resolution SPECT imaging of small
rodents.
Collapse
Affiliation(s)
- Luciano Fonseca Lemos de Oliveira
- Divisão de Cardiologia, Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Nanjundappa A, Raza JA, Dieter RS, Mandapaka S, Cascio WE. Cell transplantation for treatment of left-ventricular dysfunction due to ischemic heart failure: from bench to bedside. Expert Rev Cardiovasc Ther 2014; 5:125-31. [PMID: 17187464 DOI: 10.1586/14779072.5.1.125] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cell transplantation is an innovative technology that involves the implantation of a variety of myogenic and angiogenic cell types. The transplanted cells proliferate and augment left ventricular performance and therein ameliorate the heart failure symptoms. The concept of cell transplantation has followed the footsteps of angiogenesis starting as bench side research. The latter half of the decade saw the transformation of this potential mechanism to a promising therapy for ischemic heart failure. More than 150 patients have been treated with cellular transplantation worldwide. This novel application has the potential to revolutionize alternative therapeutic approaches to management of heart failure.
Collapse
Affiliation(s)
- Aravinda Nanjundappa
- East Carolina University, Department of Internal Medicine, Division of Cardiology, 600 Moye Blvd, TA 378, Greenville, NC 27834, USA.
| | | | | | | | | |
Collapse
|
37
|
Fiedler LR, Maifoshie E, Schneider MD. Mouse models of heart failure: cell signaling and cell survival. Curr Top Dev Biol 2014; 109:171-247. [PMID: 24947238 DOI: 10.1016/b978-0-12-397920-9.00002-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Heart failure is one of the paramount global causes of morbidity and mortality. Despite this pandemic need, the available clinical counter-measures have not altered substantially in recent decades, most notably in the context of pharmacological interventions. Cell death plays a causal role in heart failure, and its inhibition poses a promising approach that has not been thoroughly explored. In previous approaches to target discovery, clinical failures have reflected a deficiency in mechanistic understanding, and in some instances, failure to systematically translate laboratory findings toward the clinic. Here, we review diverse mouse models of heart failure, with an emphasis on those that identify potential targets for pharmacological inhibition of cell death, and on how their translation into effective therapies might be improved in the future.
Collapse
Affiliation(s)
- Lorna R Fiedler
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK.
| | - Evie Maifoshie
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael D Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
38
|
A nonthoracotomy myocardial infarction model in an ovine using autologous platelets. BIOMED RESEARCH INTERNATIONAL 2013; 2013:938047. [PMID: 24367790 PMCID: PMC3866830 DOI: 10.1155/2013/938047] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/15/2013] [Indexed: 11/18/2022]
Abstract
Objective. There is a paucity of a biological large animal model of myocardial infarction (MI). We hypothesized that, using autologous-aggregated platelets, we could create an ovine model that was reproducible and more closely mimicked the pathophysiology of MI. Methods. Mepacrine stained autologous platelets from male sheep (n = 7) were used to create a myocardial infarction via catheter injection into the mid-left anterior descending (LAD) coronary artery. Serial daily serum troponin measurements were taken and tissue harvested on post-embolization day three. Immunofluorescence microscopy was used to detect the mepacrine-stained platelet-induced thrombus, and histology performed to identify three distinct myocardial (infarct, peri-ischemic “border zone,” and remote) zones. Results. Serial serum troponin levels (μg/mL) measured 0.0 ± 0.0 at baseline and peaked at 297.4 ± 58.0 on post-embolization day 1, followed by 153.0 ± 38.8 on day 2 and 76.7 ± 19.8 on day 3. Staining confirmed distinct myocardial regions of inflammation and fibrosis as well as mepacrine-stained platelets as the cause of intravascular thrombosis. Conclusion. We report a reproducible, unique model of a biological myocardial infarction in a large animal model. This technique can be used to study acute, regional myocardial changes following a thrombotic injury.
Collapse
|
39
|
Development of a closed chest model of chronic myocardial infarction in Swine: magnetic resonance imaging and pathological evaluation. ISRN CARDIOLOGY 2013; 2013:781762. [PMID: 24282645 PMCID: PMC3825272 DOI: 10.1155/2013/781762] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 09/12/2013] [Indexed: 12/21/2022]
Abstract
Our aim was to develop an easy-to-induce, reproducible, and low mortality clinically relevant closed-chest model of chronic myocardial infarction in swine using intracoronary ethanol
and characterize its evolution using MRI and pathology. We injected 3-4 mL of 100% ethanol into the mid-LAD of anesthetized swine. Heart function and infarct size were assessed serially using MRI.
Pigs were euthanized on days 7, 30, and 90 (n = 5 at each timepoint). Postoperative MRI revealed compromised contractility and decreased ejection fraction, from 53.8% ± 6.32% to 43.79% ±
7.72%
(P = 0.001). These values remained lower than baseline thorough the followup (46.54% ± 11.12%, 44.48% ± 7.77%, and 40.48% ± 6.40%, resp., P < 0.05). Progressive remodeling was seen in all animals.
Infarcted myocardium decreased on the first 30 days (from 18.09% ± 7.26% to 9.9% ± 5.68%) and then stabilized (10.2% ± 4.21%). Pathology revealed increasing collagen content and fibrous organization
over time, with a rim of preserved endocardial cells. In conclusion, intracoronary ethanol administration in swine consistently results in infarction. The sustained compromise in heart function and myocardial
thinning over time indicate that the model may be useful for the preclinical evaluation of and training in therapeutic approaches to heart failure.
Collapse
|
40
|
Laughner JI, Marrus SB, Zellmer ER, Weinheimer CJ, MacEwan MR, Cui SX, Nerbonne JM, Efimov IR. A fully implantable pacemaker for the mouse: from battery to wireless power. PLoS One 2013; 8:e76291. [PMID: 24194832 PMCID: PMC3806780 DOI: 10.1371/journal.pone.0076291] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/22/2013] [Indexed: 11/19/2022] Open
Abstract
Animal models have become a popular platform for the investigation of the molecular and systemic mechanisms of pathological cardiovascular physiology. Chronic pacing studies with implantable pacemakers in large animals have led to useful models of heart failure and atrial fibrillation. Unfortunately, molecular and genetic studies in these large animal models are often prohibitively expensive or not available. Conversely, the mouse is an excellent species for studying molecular mechanisms of cardiovascular disease through genetic engineering. However, the large size of available pacemakers does not lend itself to chronic pacing in mice. Here, we present the design for a novel, fully implantable wireless-powered pacemaker for mice capable of long-term (>30 days) pacing. This design is compared to a traditional battery-powered pacemaker to demonstrate critical advantages achieved through wireless inductive power transfer and control. Battery-powered and wireless-powered pacemakers were fabricated from standard electronic components in our laboratory. Mice (n = 24) were implanted with endocardial, battery-powered devices (n = 14) and epicardial, wireless-powered devices (n = 10). Wireless-powered devices were associated with reduced implant mortality and more reliable device function compared to battery-powered devices. Eight of 14 (57.1%) mice implanted with battery-powered pacemakers died following device implantation compared to 1 of 10 (10%) mice implanted with wireless-powered pacemakers. Moreover, device function was achieved for 30 days with the wireless-powered device compared to 6 days with the battery-powered device. The wireless-powered pacemaker system presented herein will allow electrophysiology studies in numerous genetically engineered mouse models as well as rapid pacing-induced heart failure and atrial arrhythmia in mice.
Collapse
Affiliation(s)
- Jacob I. Laughner
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Scott B. Marrus
- Department of Internal Medicine, Division of Cardiovascular Sciences, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Erik R. Zellmer
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Carla J. Weinheimer
- Department of Internal Medicine, Division of Cardiovascular Sciences, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Matthew R. MacEwan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Sophia X. Cui
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Jeanne M. Nerbonne
- Department of Developmental Biology, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
| | - Igor R. Efimov
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
41
|
Gomes AC, Falcão-Pires I, Pires AL, Brás-Silva C, Leite-Moreira AF. Rodent models of heart failure: an updated review. Heart Fail Rev 2013; 18:219-49. [PMID: 22446984 DOI: 10.1007/s10741-012-9305-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Heart failure (HF) is one of the major health and economic burdens worldwide, and its prevalence is continuously increasing. The study of HF requires reliable animal models to study the chronic changes and pharmacologic interventions in myocardial structure and function and to follow its progression toward HF. Indeed, during the past 40 years, basic and translational scientists have used small animal models to understand the pathophysiology of HF and find more efficient ways of preventing and managing patients suffering from congestive HF (CHF). Each species and each animal model has advantages and disadvantages, and the choice of one model over another should take them into account for a good experimental design. The aim of this review is to describe and highlight the advantages and drawbacks of some commonly used HF rodents models, including both non-genetically and genetically engineered models, with a specific subchapter concerning diastolic HF models.
Collapse
Affiliation(s)
- A C Gomes
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal
| | | | | | | | | |
Collapse
|
42
|
Targeting mitochondrial oxidative metabolism as an approach to treat heart failure. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:857-65. [DOI: 10.1016/j.bbamcr.2012.08.014] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 08/21/2012] [Accepted: 08/23/2012] [Indexed: 01/24/2023]
|
43
|
Hagen MK, Ludke A, Araujo AS, Mendes RH, Fernandes TG, Mandarino JMG, Llesuy S, Vogt de Jong E, Belló-Klein A. Antioxidant characterization of soy derived products in vitro and the effect of a soy diet on peripheral markers of oxidative stress in a heart disease model. Can J Physiol Pharmacol 2012; 90:1095-1103. [PMID: 22808939 DOI: 10.1139/y2012-028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
This study analyzed and compared the content of isoflavones in 2 soy products, the effectiveness of isoflavones as antioxidants, in vitro, and demonstrated the antioxidant effect of a soy diet in rats with myocardial infarction (MI). Isoflavone content was analyzed in soybean hypocotyl (SH) and isolated soy protein (ISP). The quality (TAR) and quantity (TRAP) of antioxidants present in the samples was quantified. The amount of daidzin was higher in SH (9 times) and genistein in ISP (5 times). SH presented a 3-fold increase in TAR, while both products exhibited same TRAP. The rats were fed an ISP diet for 9 weeks. Animals were distributed among 6 treatment groups: (i) Sham Casein; (ii) Infarct Casein < 25%; (iii) Infarct Casein > 25%; (iv) Sham Soy; (v) Infarct Soy < 25%; and (vi) Infarct Soy > 25%. MI was induced 5 weeks after the commencement of the diets. Lipid peroxidation (LPO), antioxidant enzyme activity, and levels of nitrites/nitrates were determined in blood. Rats receiving the ISP diet demonstrated increased activity of antioxidant enzyme activity and nitrite/nitrate content. In addition, the increase in LPO seen in rats subjected to MI was significantly mitigated when the ISP diet was given. These findings suggest a nutritional approach of using a soy-based diet for the prevention of oxidative-stress-related diseases such as heart failure.
Collapse
Affiliation(s)
- Martine Kienzle Hagen
- Department of Internal Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Houser SR, Margulies KB, Murphy AM, Spinale FG, Francis GS, Prabhu SD, Rockman HA, Kass DA, Molkentin JD, Sussman MA, Koch WJ. Animal models of heart failure: a scientific statement from the American Heart Association. Circ Res 2012; 111:131-50. [PMID: 22595296 DOI: 10.1161/res.0b013e3182582523] [Citation(s) in RCA: 342] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
45
|
Trumble DR, McGregor WE, Kerckhoffs RCP, Waldman LK. Cardiac assist with a twist: apical torsion as a means to improve failing heart function. J Biomech Eng 2012; 133:101003. [PMID: 22070328 DOI: 10.1115/1.4005169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Changes in muscle fiber orientation across the wall of the left ventricle (LV) cause the apex of the heart to turn 10-15 deg in opposition to its base during systole and are believed to increase stroke volume and lower wall stress in healthy hearts. Studies show that cardiac torsion is sensitive to various disease states, which suggests that it may be an important aspect of cardiac function. Modern imaging techniques have sparked renewed interest in cardiac torsion dynamics, but no work has been done to determine whether mechanically augmented apical torsion can be used to restore function to failing hearts. In this report, we discuss the potential advantages of this approach and present evidence that turning the cardiac apex by mechanical means can displace a clinically significant volume of blood from failing hearts. Computational models of normal and reduced-function LVs were created to predict the effects of applied apical torsion on ventricular stroke work and wall stress. These same conditions were reproduced in anesthetized pigs with drug-induced heart failure using a custom apical torsion device programmed to rotate over various angles during cardiac systole. Simulations of applied 90 deg torsion in a prolate spheroidal computational model of a reduced-function pig heart produced significant increases in stroke work (25%) and stroke volume with reduced fiber stress in the epicardial region. These calculations were in substantial agreement with corresponding in vivo measurements. Specifically, the computer model predicted torsion-induced stroke volume increases from 13.1 to 14.4 mL (9.9%) while actual stroke volume in a pig heart of similar size and degree of dysfunction increased from 11.1 to 13.0 mL (17.1%). Likewise, peak LV pressures in the computer model rose from 85 to 95 mm Hg (11.7%) with torsion while maximum ventricular pressures in vivo increased in similar proportion, from 55 to 61 mm Hg (10.9%). These data suggest that: (a) the computer model of apical torsion developed for this work is a fair and accurate predictor of experimental outcomes, and (b) supra-physiologic apical torsion may be a viable means to boost cardiac output while avoiding blood contact that occurs with other assist methods.
Collapse
Affiliation(s)
- Dennnis R Trumble
- Allegheny-Singer Research and the McGinnis Cardiovascular Institutes, Allegheny General Hospital, West Penn Allegheny Health System, Pittsburgh, PA 15212, USA.
| | | | | | | |
Collapse
|
46
|
Katz MG, Fargnoli AS, Tomasulo CE, Pritchette LA, Bridges CR. Model-specific selection of molecular targets for heart failure gene therapy. J Gene Med 2012; 13:573-86. [PMID: 21954055 DOI: 10.1002/jgm.1610] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Heart failure (HF) is a complex multifaceted problem of abnormal ventricular function and structure. In recent years, new information has been accumulated allowing for a more detailed understanding of the cellular and molecular alterations that are the underpinnings of diverse causes of HF, including myocardial ischemia, pressure-overload, volume-overload or intrinsic cardiomyopathy. Modern pharmacological approaches to treat HF have had a significant impact on the course of the disease, although they do not reverse the underlying pathological state of the heart. Therefore gene-based therapy holds a great potential as a targeted treatment for cardiovascular diseases. Here, we survey the relative therapeutic efficacy of genetic modulation of β-adrenergic receptor signaling, Ca(2+) handling proteins and angiogenesis in the most common extrinsic models of HF.
Collapse
Affiliation(s)
- Michael G Katz
- Department of Surgery, Division of Cardiovascular Surgery, The University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
47
|
Effective ventricular unloading by left ventricular assist device varies with stage of heart failure: cardiac simulator study. ASAIO J 2012; 57:407-13. [PMID: 21817896 DOI: 10.1097/mat.0b013e318229ca8d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Although the use of left ventricular assist devices (LVADs) as a bridge-to-recovery (BTR) has shown promise, clinical success has been limited due to the lack of understanding the timing of implantation, acute/chronic device setting, and explantation. This study investigated the effective ventricular unloading at different heart conditions by using a mock circulatory system (MCS) to provide a tool for pump parameter adjustments. We tested the hypothesis that effective unloading by LVAD at a given speed varies with the stage of heart failure. By using a MCS, systematic depression of cardiac performance was obtained. Five different stages of heart failure from control were achieved by adjusting the pneumatic systolic/diastolic pressure, filling pressure, and systemic resistance. The Heart Mate II® (Thoratec Corp., Pleasanton, CA) was used for volumetric and pressure unloading at different heart conditions over a given LVAD speed. The effective unloading at a given LVAD speed was greater in more depressed heart condition. The rate of unloading over LVAD speed was also greater in more depressed heart condition. In conclusion, to get continuous and optimal cardiac recovery, timely increase in LVAD speed over a period of support is needed while avoiding the akinesis of aortic valve.
Collapse
|
48
|
Kim KH, Kim YJ, Lee SP, Kim HK, Seo JW, Sohn DW, Oh BH, Park YB. Survival, exercise capacity, and left ventricular remodeling in a rat model of chronic mitral regurgitation: serial echocardiography and pressure-volume analysis. Korean Circ J 2011; 41:603-11. [PMID: 22125560 PMCID: PMC3221903 DOI: 10.4070/kcj.2011.41.10.603] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 06/24/2011] [Accepted: 06/27/2011] [Indexed: 01/19/2023] Open
Abstract
Background and Objectives The aims of this study were to establish a reliable model of chronic mitral regurgitation (MR) in rats and verify the pathophysiological features of this model by evaluating cardiac function using serial echocardiography and a pressure-volume analysis. Materials and Methods MR was created in 37 Sprague-Dawley rats by making a hole with a 23 gauge needle on the mitral leaflet through the left ventricular (LV) apex under the guidance of transesophageal echocardiography. Results Serial echocardiograms revealed that the LV began to dilate immediately after the MR operation and showed progressive dilation until the 14th week (LV end-systolic dimension at 14 weeks, 4.71±0.25 mm vs. 6.81±0.50 mm for sham vs. MR, p<0.01; LV end-diastolic dimension, 8.32±0.42 mm vs. 11.01±0.47 mm, p<0.01). The LV ejection fraction tended to increase immediately after the MR operation but started to decrease thereafter and showed a significant difference with the sham group from the 14th week (70.0±2.2% vs. 62.1±3.1% for sham vs. MR). In a pressure-volume analysis performed at the 14th week, the LV end-systolic pressure-volume relationship and +dp/dt decreased significantly in the MR group. A serial treadmill test revealed that exercise capacity remained in the normal range until the 14th week when it began to decrease (exercise duration, 406±45 seconds vs. 330±27 seconds, p<0.01). A pathological analysis showed no significance difference in interstitial fibrosis between the two groups. Conclusion We established a small animal model of chronic MR and verified its pathophysiological features. This model may provide a useful tool for future research on MR and volume overload heart failure.
Collapse
Affiliation(s)
- Kyung-Hee Kim
- Department of Internal Medicine, Cardiovascular Center, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Hollander JM, Baseler WA, Dabkowski ER. Proteomic remodeling of mitochondria in heart failure. ACTA ACUST UNITED AC 2011; 17:262-8. [PMID: 22103917 DOI: 10.1111/j.1751-7133.2011.00254.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Heart failure (HF) is a common disease that has been attributed, in part, to deprivation of cardiac energy. As a result, the interplay between metabolism and adenosine triphosphate production is fundamental in determining the mechanisms driving the disease progression. Due to its central role in energy production, metabolism, calcium homeostasis, and oxidative stress, the mitochondrion has been suggested to play a pivotal role in the progression of the heart to failure. Nevertheless, the mitochondrion's specific role(s) and the proteins contributing to the development and progression of HF are not entirely clear. Thus, changes in mitochondrial proteomic make-up during HF have garnered great interest. With the continued development of advanced tools for assessing proteomic make-up, characterization of mitochondrial proteomic changes during disease states such as HF are being realized. These studies have begun to identify potential biomarkers of disease progression as well as protein targets that may provide an avenue for therapeutic intervention. The goal of this review is to highlight some of the changes in mitochondrial proteomic make-up that are associated with the development of HF in an effort to identify target axes and candidate proteins contributing to disease development. Results from a number of different HF models will be evaluated to gain insight into some of the similarities and differences in mitochondrial proteomic alterations associated with morphological and functional changes that result from the disease. Congest Heart Fail.
Collapse
Affiliation(s)
- John M Hollander
- Division of Exercise Physiology and Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, USA.
| | | | | |
Collapse
|
50
|
Carll AP, Willis MS, Lust RM, Costa DL, Farraj AK. Merits of non-invasive rat models of left ventricular heart failure. Cardiovasc Toxicol 2011; 11:91-112. [PMID: 21279739 DOI: 10.1007/s12012-011-9103-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Heart failure (HF) is characterized as a limitation to cardiac output that prevents the heart from supplying tissues with adequate oxygen and predisposes individuals to pulmonary edema. Impaired cardiac function is secondary to either decreased contractility reducing ejection (systolic failure), diminished ventricular compliance preventing filling (diastolic failure), or both. To study HF etiology, many different techniques have been developed to elicit this condition in experimental animals, with varying degrees of success. Among rats, surgically induced HF models are the most prevalent, but they bear several shortcomings, including high mortality rates and limited recapitulation of the pathophysiology, etiology, and progression of human HF. Alternatively, a number of non-invasive HF induction methods avoid many of these pitfalls, and their merits in technical simplicity, reliability, survivability, and comparability to the pathophysiologic and pathogenic characteristics of HF are reviewed herein. In particular, this review focuses on the primary pathogenic mechanisms common to genetic strains (spontaneously hypertensive and spontaneously hypertensive heart failure), pharmacological models of toxic cardiomyopathy (doxorubicin and isoproterenol), and dietary salt models, all of which have been shown to induce left ventricular HF in the rat. Additional non-invasive techniques that may potentially enable the development of new HF models are also discussed.
Collapse
Affiliation(s)
- Alex P Carll
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, 27599 USA.
| | | | | | | | | |
Collapse
|