1
|
Hatami M, Özbek A, Deán‐Ben XL, Gutierrez J, Schill A, Razansky D, Larin KV. Noninvasive Tracking of Embryonic Cardiac Dynamics and Development with Volumetric Optoacoustic Spectroscopy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400089. [PMID: 38526147 PMCID: PMC11165471 DOI: 10.1002/advs.202400089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/29/2024] [Indexed: 03/26/2024]
Abstract
Noninvasive monitoring of cardiac development can potentially prevent cardiac anomalies in adulthood. Mouse models provide unique opportunities to study cardiac development and disease in mammals. However, high-resolution noninvasive functional analyses of murine embryonic cardiac models are challenging because of the small size and fast volumetric motion of the embryonic heart, which is deeply embedded inside the uterus. In this study, a real time volumetric optoacoustic spectroscopy (VOS) platform for whole-heart visualization with high spatial (100 µm) and temporal (10 ms) resolutions is developed. Embryonic heart development on gestational days (GDs) 14.5-17.5 and quantify cardiac dynamics using time-lapse-4D image data of the heart is followed. Additionally, spectroscopic recordings enable the quantification of the blood oxygenation status in heart chambers in a label-free and noninvasive manner. This technology introduces new possibilities for high-resolution quantification of embryonic heart function at different gestational stages in mammalian models, offering an invaluable noninvasive method for developmental biology.
Collapse
Affiliation(s)
- Maryam Hatami
- Department of Biomedical EngineeringUniversity of HoustonHoustonTX77004USA
| | - Ali Özbek
- Institute for Biomedical Engineering and Institute of Pharmacology and ToxicologyFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8092Switzerland
| | - Xosé Luís Deán‐Ben
- Institute for Biomedical Engineering and Institute of Pharmacology and ToxicologyFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8092Switzerland
| | - Jessica Gutierrez
- Department of Biomedical EngineeringUniversity of HoustonHoustonTX77004USA
| | - Alexander Schill
- Department of Biomedical EngineeringUniversity of HoustonHoustonTX77004USA
| | - Daniel Razansky
- Institute for Biomedical Engineering and Institute of Pharmacology and ToxicologyFaculty of MedicineUniversity of ZurichZurich8057Switzerland
- Institute for Biomedical EngineeringDepartment of Information Technology and Electrical EngineeringETH ZurichZurich8092Switzerland
| | - Kirill V. Larin
- Department of Biomedical EngineeringUniversity of HoustonHoustonTX77004USA
- Department of Integrative PhysiologyBaylor College of MedicineHoustonTX77030USA
| |
Collapse
|
2
|
Hegemann N, Bintig W, Perret PL, Rees J, Viperino A, Eickholt B, Kuebler WM, Höpfner M, Nitzsche B, Grune J. In-ovo echocardiography for application in cardiovascular research. Basic Res Cardiol 2023; 118:19. [PMID: 37193927 PMCID: PMC10188421 DOI: 10.1007/s00395-023-00989-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/17/2023] [Accepted: 05/01/2023] [Indexed: 05/18/2023]
Abstract
Preclinical cardiovascular research relies heavily on non-invasive in-vivo echocardiography in mice and rats to assess cardiac function and morphology, since the complex interaction of heart, circulation, and peripheral organs are challenging to mimic ex-vivo. While n-numbers of annually used laboratory animals worldwide approach 200 million, increasing efforts are made by basic scientists aiming to reduce animal numbers in cardiovascular research according to the 3R's principle. The chicken egg is well-established as a physiological correlate and model for angiogenesis research but has barely been used to assess cardiac (patho-) physiology. Here, we tested whether the established in-ovo system of incubated chicken eggs interfaced with commercially available small animal echocardiography would be a suitable alternative test system in experimental cardiology. To this end, we defined a workflow to assess cardiac function in 8-13-day-old chicken embryos using a commercially available high resolution ultrasound system for small animals (Vevo 3100, Fujifilm Visualsonics Inc.) equipped with a high frequency probe (MX700; centre transmit: 50 MHz). We provide detailed standard operating procedures for sample preparation, image acquisition, data analysis, reference values for left and right ventricular function and dimensions, and inter-observer variabilities. Finally, we challenged incubated chicken eggs with two interventions well-known to affect cardiac physiology-metoprolol treatment and hypoxic exposure-to demonstrate the sensitivity of in-ovo echocardiography. In conclusion, in-ovo echocardiography is a feasible alternative tool for basic cardiovascular research, which can easily be implemented into the small animal research environment using existing infrastructure to replace mice and rat experiments, and thus, reduce use of laboratory animals according to the 3R principle.
Collapse
Affiliation(s)
- Niklas Hegemann
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117, Berlin, Germany
| | - Willem Bintig
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany
| | - Paul-Lennard Perret
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117, Berlin, Germany
| | - Judith Rees
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Alessandra Viperino
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Britta Eickholt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biochemistry, Charitéplatz 1, 10117, Berlin, Germany
| | - Wolfgang M Kuebler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117, Berlin, Germany
| | - Michael Höpfner
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Bianca Nitzsche
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117, Berlin, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Institute of Physiology, Charitéplatz 1, 10117, Berlin, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117, Berlin, Germany.
| |
Collapse
|
3
|
Zhou YQ, Bonafiglia QA, Zhang H, Heximer SP, Bendeck MP. Comprehensive ultrasound imaging of right ventricular remodeling under surgically induced pressure overload in mice. Am J Physiol Heart Circ Physiol 2023; 324:H391-H410. [PMID: 36607797 DOI: 10.1152/ajpheart.00590.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This study reports a new methodology for right heart imaging by ultrasound in mice under right ventricular (RV) pressure overload. Pulmonary artery constriction (PAC) or sham surgeries were performed on C57BL/6 male mice at 8 wk of age. Ultrasound imaging was conducted at 2, 4, and 8 wk postsurgery using both classical and advanced ultrasound imaging modalities including electrocardiogram (ECG)-based kilohertz visualization, anatomical M-mode, and strain imaging. Based on pulsed Doppler, the PAC group demonstrated dramatically enhanced pressure gradient in the main pulmonary artery (MPA) as compared with the sham group. By the application of advanced imaging modalities in novel short-axis views of the ventricles, the PAC group demonstrated increased thickness of RV free wall, enlarged RV chamber, and reduced RV fractional shortening compared with the sham group. The PAC group also showed prolonged RV contraction, asynchronous interplay between RV and left ventricle (LV), and passive leftward motion of the interventricular septum (IVS) at early diastole. Consequently, the PAC group exhibited prolongation of LV isovolumic relaxation time, without change in LV wall thickness or systolic function. Significant correlations were found between the maximal pressure gradient in MPA measured by Doppler and the RV systolic pressure by catheterization, as well as the morphological and functional parameters of RV by ultrasound.NEW & NOTEWORTHY The established protocol overcomes the challenges in right heart imaging in mice, thoroughly elucidating the changes of RV, the dynamics of IVS, and the impact on LV and provides new insights into the pathophysiological mechanism of RV remodeling.
Collapse
Affiliation(s)
- Yu-Qing Zhou
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada.,Institute of Biomedical Engineering, Faculty of Applied Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Quinn A Bonafiglia
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hangjun Zhang
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Scott P Heximer
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michelle P Bendeck
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Karsenty C, Guilbeau-Frugier C, Genet G, Seguelas MH, Alzieu P, Cazorla O, Montagner A, Blum Y, Dubroca C, Maupoint J, Tramunt B, Cauquil M, Sulpice T, Richard S, Arcucci S, Flores-Flores R, Pataluch N, Montoriol R, Sicard P, Deney A, Couffinhal T, Senard JM, Galés C. Ephrin-B1 regulates the adult diastolic function through a late postnatal maturation of cardiomyocyte surface crests. eLife 2023; 12:e80904. [PMID: 36649053 PMCID: PMC9844986 DOI: 10.7554/elife.80904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
The rod-shaped adult cardiomyocyte (CM) harbors a unique architecture of its lateral surface with periodic crests, relying on the presence of subsarcolemmal mitochondria (SSM) with unknown role. Here, we investigated the development and functional role of CM crests during the postnatal period. We found in rodents that CM crest maturation occurs late between postnatal day 20 (P20) and P60 through both SSM biogenesis, swelling and crest-crest lateral interactions between adjacent CM, promoting tissue compaction. At the functional level, we showed that the P20-P60 period is dedicated to the improvement of relaxation. Interestingly, crest maturation specifically contributes to an atypical CM hypertrophy of its short axis, without myofibril addition, but relying on CM lateral stretching. Mechanistically, using constitutive and conditional CM-specific knock-out mice, we identified ephrin-B1, a lateral membrane stabilizer, as a molecular determinant of P20-P60 crest maturation, governing both the CM lateral stretch and the diastolic function, thus highly suggesting a link between crest maturity and diastole. Remarkably, while young adult CM-specific Efnb1 KO mice essentially exhibit an impairment of the ventricular diastole with preserved ejection fraction and exercise intolerance, they progressively switch toward systolic heart failure with 100% KO mice dying after 13 months, indicative of a critical role of CM-ephrin-B1 in the adult heart function. This study highlights the molecular determinants and the biological implication of a new late P20-P60 postnatal developmental stage of the heart in rodents during which, in part, ephrin-B1 specifically regulates the maturation of the CM surface crests and of the diastolic function.
Collapse
Affiliation(s)
- Clement Karsenty
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Pediatric Cardiology, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Celine Guilbeau-Frugier
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Forensic Medicine, Centre Hospitalier Universitaire de Toulouse, Université de ToulouseToulouseFrance
| | - Gaël Genet
- Department of Cell Biology, University of Virginia School of MedicineCharlottesvilleUnited States
| | - Marie-Helene Seguelas
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Philippe Alzieu
- Université de Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
| | - Olivier Cazorla
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Alexandra Montagner
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Yuna Blum
- IGDR UMR 6290, CNRS, Université de Rennes 1RennesFrance
| | | | | | - Blandine Tramunt
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Diabetology, Metabolic Diseases & Nutrition, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Marie Cauquil
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | | | - Sylvain Richard
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Silvia Arcucci
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Remy Flores-Flores
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Nicolas Pataluch
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Romain Montoriol
- Department of Forensic Medicine, Centre Hospitalier Universitaire de Toulouse, Université de ToulouseToulouseFrance
| | - Pierre Sicard
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Antoine Deney
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Thierry Couffinhal
- Université de Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
- Service des Maladies Cardiaques et Vasculaires, CHU de BordeauxBordeauxFrance
| | - Jean-Michel Senard
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Clinical Pharmacology, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Celine Galés
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| |
Collapse
|
5
|
Valero-Muñoz M, Saw EL, Hekman RM, Blum BC, Hourani Z, Granzier H, Emili A, Sam F. Proteomic and phosphoproteomic profiling in heart failure with preserved ejection fraction (HFpEF). Front Cardiovasc Med 2022; 9:966968. [PMID: 36093146 PMCID: PMC9452734 DOI: 10.3389/fcvm.2022.966968] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Although the prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing, evidence-based therapies for HFpEF remain limited, likely due to an incomplete understanding of this disease. This study sought to identify the cardiac-specific features of protein and phosphoprotein changes in a murine model of HFpEF using mass spectrometry. HFpEF mice demonstrated moderate hypertension, left ventricle (LV) hypertrophy, lung congestion and diastolic dysfunction. Proteomics analysis of the LV tissue showed that 897 proteins were differentially expressed between HFpEF and Sham mice. We observed abundant changes in sarcomeric proteins, mitochondrial-related proteins, and NAD-dependent protein deacetylase sirtuin-3 (SIRT3). Upregulated pathways by GSEA analysis were related to immune modulation and muscle contraction, while downregulated pathways were predominantly related to mitochondrial metabolism. Western blot analysis validated SIRT3 downregulated cardiac expression in HFpEF vs. Sham (0.8 ± 0.0 vs. 1.0 ± 0.0; P < 0.001). Phosphoproteomics analysis showed that 72 phosphosites were differentially regulated between HFpEF and Sham LV. Aberrant phosphorylation patterns mostly occurred in sarcomere proteins and nuclear-localized proteins associated with contractile dysfunction and cardiac hypertrophy. Seven aberrant phosphosites were observed at the z-disk binding region of titin. Additional agarose gel analysis showed that while total titin cardiac expression remained unaltered, its stiffer N2B isoform was significantly increased in HFpEF vs. Sham (0.144 ± 0.01 vs. 0.127 ± 0.01; P < 0.05). In summary, this study demonstrates marked changes in proteins related to mitochondrial metabolism and the cardiac contractile apparatus in HFpEF. We propose that SIRT3 may play a role in perpetuating these changes and may be a target for drug development in HFpEF.
Collapse
Affiliation(s)
- María Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Eng Leng Saw
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Ryan M. Hekman
- Department of Biology, Boston University, Boston, MA, United States
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
| | - Benjamin C. Blum
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, United States
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, United States
| | - Andrew Emili
- Department of Biology, Boston University, Boston, MA, United States
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
6
|
Pitsch M, Kant S, Mytzka C, Leube RE, Krusche CA. Autophagy and Endoplasmic Reticulum Stress during Onset and Progression of Arrhythmogenic Cardiomyopathy. Cells 2021; 11:96. [PMID: 35011658 PMCID: PMC8750195 DOI: 10.3390/cells11010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a heritable, potentially lethal disease without a causal therapy. AC is characterized by focal cardiomyocyte death followed by inflammation and progressive formation of connective tissue. The pathomechanisms leading to structural disease onset and progression, however, are not fully elucidated. Recent studies revealed that dysregulation of autophagy and endoplasmic/sarcoplasmic reticulum (ER/SR) stress plays an important role in cardiac pathophysiology. We therefore examined the temporal and spatial expression patterns of autophagy and ER/SR stress indicators in murine AC models by qRT-PCR, immunohistochemistry, in situ hybridization and electron microscopy. Cardiomyocytes overexpressing the autophagy markers LC3 and SQSTM1/p62 and containing prominent autophagic vacuoles were detected next to regions of inflammation and fibrosis during onset and chronic disease progression. mRNAs of the ER stress markers Chop and sXbp1 were elevated in both ventricles at disease onset. During chronic disease progression Chop mRNA was upregulated in right ventricles. In addition, reduced Ryr2 mRNA expression together with often drastically enlarged ER/SR cisternae further indicated SR dysfunction during this disease phase. Our observations support the hypothesis that locally altered autophagy and enhanced ER/SR stress play a role in AC pathogenesis both at the onset and during chronic progression.
Collapse
Affiliation(s)
| | | | | | - Rudolf E. Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.P.); (S.K.); (C.M.)
| | - Claudia A. Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.P.); (S.K.); (C.M.)
| |
Collapse
|
7
|
Shekhar A, Aristizabal O, Fishman GI, Phoon CKL, Ketterling JA. Characterization of Vortex Flow in a Mouse Model of Ventricular Dyssynchrony by Plane-Wave Ultrasound Using Hexplex Processing. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:538-548. [PMID: 32763851 PMCID: PMC8054309 DOI: 10.1109/tuffc.2020.3014844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The rodent heart is frequently used to study human cardiovascular disease (CVD). Although advanced cardiovascular ultrasound imaging methods are available for human clinical practice, application of these techniques to small animals remains limited due to the temporal and spatial-resolution demands. Here, an ultrasound vector-flow workflow is demonstrated that enables visualization and quantification of the complex hemodynamics within the mouse heart. Wild type (WT) and fibroblast growth factor homologous factor 2 (FHF2)-deficient mice (Fhf2 KO/Y ), which present with hyperthermia-induced ECG abnormalities highly reminiscent of Brugada syndrome, were used as a mouse model of human CVD. An 18-MHz linear array was used to acquire high-speed (30 kHz), plane-wave data of the left ventricle (LV) while increasing core body temperature up to 41.5 °C. Hexplex (i.e., six output) processing of the raw data sets produced the output of vector-flow estimates (magnitude and phase); B-mode and color-Doppler images; Doppler spectrograms; and local time histories of vorticity and pericardium motion. Fhf2 WT/Y mice had repeatable beat-to-beat cardiac function, including vortex formation during diastole, at all temperatures. In contrast, Fhf2 KO/Y mice displayed dyssynchronous contractile motion that disrupted normal inflow vortex formation and impaired LV filling as temperature rose. The hexplex processing approach demonstrates the ability to visualize and quantify the interplay between hemodynamic and mechanical function in a mouse model of human CVD.
Collapse
|
8
|
Keller BB, Kowalski WJ, Tinney JP, Tobita K, Hu N. Validating the Paradigm That Biomechanical Forces Regulate Embryonic Cardiovascular Morphogenesis and Are Fundamental in the Etiology of Congenital Heart Disease. J Cardiovasc Dev Dis 2020; 7:E23. [PMID: 32545681 PMCID: PMC7344498 DOI: 10.3390/jcdd7020023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/31/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
The goal of this review is to provide a broad overview of the biomechanical maturation and regulation of vertebrate cardiovascular (CV) morphogenesis and the evidence for mechanistic relationships between function and form relevant to the origins of congenital heart disease (CHD). The embryonic heart has been investigated for over a century, initially focusing on the chick embryo due to the opportunity to isolate and investigate myocardial electromechanical maturation, the ability to directly instrument and measure normal cardiac function, intervene to alter ventricular loading conditions, and then investigate changes in functional and structural maturation to deduce mechanism. The paradigm of "Develop and validate quantitative techniques, describe normal, perturb the system, describe abnormal, then deduce mechanisms" was taught to many young investigators by Dr. Edward B. Clark and then validated by a rapidly expanding number of teams dedicated to investigate CV morphogenesis, structure-function relationships, and pathogenic mechanisms of CHD. Pioneering studies using the chick embryo model rapidly expanded into a broad range of model systems, particularly the mouse and zebrafish, to investigate the interdependent genetic and biomechanical regulation of CV morphogenesis. Several central morphogenic themes have emerged. First, CV morphogenesis is inherently dependent upon the biomechanical forces that influence cell and tissue growth and remodeling. Second, embryonic CV systems dynamically adapt to changes in biomechanical loading conditions similar to mature systems. Third, biomechanical loading conditions dynamically impact and are regulated by genetic morphogenic systems. Fourth, advanced imaging techniques coupled with computational modeling provide novel insights to validate regulatory mechanisms. Finally, insights regarding the genetic and biomechanical regulation of CV morphogenesis and adaptation are relevant to current regenerative strategies for patients with CHD.
Collapse
Affiliation(s)
- Bradley B. Keller
- Cincinnati Children’s Heart Institute, Greater Louisville and Western Kentucky Practice, Louisville, KY 40202, USA
| | - William J. Kowalski
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA;
| | - Joseph P. Tinney
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY 40202, USA;
| | - Kimimasa Tobita
- Department of Medical Affairs, Abiomed Japan K.K., Muromachi Higashi Mitsui Bldg, Tokyo 103-0022, Japan;
| | - Norman Hu
- Department of Pediatrics, University of Utah, Salt Lake City, UT 84108, USA;
| |
Collapse
|
9
|
Fayaz MA, Awang-Junaidi AH, Singh J, Honaramooz A. Validation of ultrasound biomicroscopy for the assessment of xenogeneic testis tissue grafts and cell implants in recipient mice. Andrology 2020; 8:1332-1346. [PMID: 32030908 DOI: 10.1111/andr.12771] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Subcutaneous grafting/implantation of neonatal testis tissue/cells from diverse donor species into recipient mice can be used as an in vivo model to study testis development, spermatogenesis, and steroidogenesis. Ultrasound biomicroscopy (UBM) allows obtaining high definition cross-sectional images of tissues at microscopic resolutions. OBJECTIVES The present study was designed to (a) validate the use of UBM for non-invasive monitoring of grafts/implants overtime and to (b) correlate UBM findings with the morphological attributes of recovered grafts/implants. MATERIALS AND METHODS Testis tissue fragments (~14 mm3 , each) and cell aggregates (100 × 106 cells, each) obtained from 1-week-old donor piglets (n = 30) were grafted/implanted under the back skin of immunodeficient mice (n = 6) in eight analogous sites per mouse. Three-dimensional transcutaneous Doppler UBM was performed, and a randomly selected graft and its corresponding implant were recovered at 2, 4, 6, and 8 weeks. RESULTS Graft/implant weight (P = .04) and physical height (P = .03) increased overtime. The dynamics of physical length and volume increases over time differed between tissue grafts and cell implants (P = .02 and 0.01 for sample type*time interactions, respectively). UBM-estimated volume was correlated with the post-recovery weight and volume of the grafts/implants (r = 0.98 and r = 0.99, respectively; P < .001). Pre- and post-recovery length and height of the grafts/implants were positively and strongly correlated (r = 0.50, P = .01; r = 0.70, P = .001) and so were the areas covered by cordal, non-cordal, or fluid-filled cavities between UBM and histology (r = 0.87, P < .001). DISCUSSION AND CONCLUSION UBM findings correlated with physical attributes of the grafts/implants, validating its use as a non-invasive high-fidelity tool to quantify the developmental changes in ectopic testis tissue grafts and cell implants, potentially leading to a reduction in the number of recipient mice needed for similar experiments.
Collapse
Affiliation(s)
- Mohammad Amin Fayaz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Awang Hazmi Awang-Junaidi
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jaswant Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ali Honaramooz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
10
|
Kumar P, Morton JS, Shah A, Do V, Sergi C, Serrano‐Lomelin J, Davidge ST, Beker D, Levasseur J, Hornberger LK. Intrauterine exposure to chronic hypoxia in the rat leads to progressive diastolic function and increased aortic stiffness from early postnatal developmental stages. Physiol Rep 2020; 8:e14327. [PMID: 31960611 PMCID: PMC6971413 DOI: 10.14814/phy2.14327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM We sought to explore whether fetal hypoxia exposure, an insult of placental insufficiency, is associated with left ventricular dysfunction and increased aortic stiffness at early postnatal ages. METHODS Pregnant Sprague Dawley rats were exposed to hypoxic conditions (11.5% FiO2 ) from embryonic day E15-21 or normoxic conditions (controls). After delivery, left ventricular function and aortic pulse wave velocity (measure of aortic stiffness) were assessed longitudinally by echocardiography from day 1 through week 8. A mixed ANOVA with repeated measures was performed to compare findings between groups across time. Myocardial hematoxylin and eosin and picro-sirius staining were performed to evaluate myocyte nuclear shape and collagen fiber characteristics, respectively. RESULTS Systolic function parameters transiently increased following hypoxia exposure primarily at week 2 (p < .008). In contrast, diastolic dysfunction progressed following fetal hypoxia exposure beginning weeks 1-2 with lower early inflow Doppler velocities, and less of an increase in early to late inflow velocity ratios and annular and septal E'/A' tissue velocities compared to controls (p < .008). As further evidence of altered diastolic function, isovolumetric relaxation time was significantly shorter relative to the cardiac cycle following hypoxia exposure from week 1 onward (p < .008). Aortic stiffness was greater following hypoxia from day 1 through week 8 (p < .008, except week 4). Hypoxia exposure was also associated with altered nuclear shape at week 2 and increased collagen fiber thickness at week 4. CONCLUSION Chronic fetal hypoxia is associated with progressive LV diastolic dysfunction, which corresponds with changes in nuclear shape and collagen fiber thickness, and increased aortic stiffness from early postnatal stages.
Collapse
Affiliation(s)
- Praveen Kumar
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
| | - Jude S. Morton
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Amin Shah
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Victor Do
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
| | - Consolato Sergi
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
- Department of Laboratory Medicine and PathologyUniversity of AlbertaEdmontonABCanada
| | - Jesus Serrano‐Lomelin
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
| | - Sandra T. Davidge
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Donna Beker
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Jody Levasseur
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| | - Lisa K. Hornberger
- Division of CardiologyDepartment of PediatricsUniversity of AlbertaEdmontonABCanada
- Women and Children’s Health Research InstituteUniversity of AlbertaEdmontonABCanada
- Department of Obstetrics/GynecologyUniversity of AlbertaEdmontonABCanada
- Cardiovascular Research Institute and Mazankowski Alberta Heart InstituteUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
11
|
Bowen ME, Selzman CH, McKellar SH. Right Ventricular Involution: Big Changes in Small Hearts. J Surg Res 2019; 243:255-264. [PMID: 31252349 DOI: 10.1016/j.jss.2019.05.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/14/2019] [Accepted: 05/29/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Before birth, the fetal right ventricle (RV) is the pump for the systemic circulation and is about as thick as the left ventricle (LV). After birth, the RV becomes the pump for the lower pressure pulmonary circulation, and the RV chamber elongates without change in its wall thickness. We hypothesize that the fetal RV may be a model of compensated RV hypertrophy, and understanding this process may aid in discovering therapeutic strategies for RV failure. METHODS We performed a literature review and identified pertinent articles from 1980 to present. RESULTS The following topics were identified to be most pertinent in right ventricular involution: morphologic and histologic changes of the RV, cellular proliferation and terminal differentiation, the effect of stress on RV development, excitation contraction coupling and inotropic response change over time, and the amount of apoptosis through RV development. CONCLUSIONS The RV changes on multiple levels after its transition from systemic to pulmonary circulation. Although published literature has variable results due partly from differences between animal models, the literature shows a clear need for more research in the field.
Collapse
Affiliation(s)
- Megan E Bowen
- University of Utah, School of Medicine, Salt Lake City, Utah; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, Utah.
| | - Craig H Selzman
- University of Utah, School of Medicine, Salt Lake City, Utah; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, Utah
| | - Stephen H McKellar
- University of Utah, School of Medicine, Salt Lake City, Utah; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, Utah
| |
Collapse
|
12
|
Guilbeau-Frugier C, Cauquil M, Karsenty C, Lairez O, Dambrin C, Payré B, Cassard H, Josse C, Seguelas MH, Allart S, Branchereau M, Heymes C, Mandel F, Delisle MB, Pathak A, Dague E, Sénard JM, Galés C. Structural evidence for a new elaborate 3D-organization of the cardiomyocyte lateral membrane in adult mammalian cardiac tissues. Cardiovasc Res 2019; 115:1078-1091. [PMID: 30329023 DOI: 10.1093/cvr/cvy256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/29/2018] [Accepted: 10/11/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS This study explored the lateral crest structures of adult cardiomyocytes (CMs) within healthy and diseased cardiac tissue. METHODS AND RESULTS Using high-resolution electron and atomic force microscopy, we performed an exhaustive quantitative analysis of the three-dimensional (3D) structure of the CM lateral surface in different cardiac compartments from various mammalian species (mouse, rat, cow, and human) and determined the technical pitfalls that limit its observation. Although crests were observed in nearly all CMs from all heart compartments in all species, we showed that their heights, dictated by the subsarcolemmal mitochondria number, substantially differ between compartments from one species to another and tightly correlate with the sarcomere length. Differences in crest heights also exist between species; for example, the similar cardiac compartments in cows and humans exhibit higher crests than rodents. Unexpectedly, we found that lateral surface crests establish tight junctional contacts with crests from neighbouring CMs. Consistently, super-resolution SIM or STED-based immunofluorescence imaging of the cardiac tissue revealed intermittent claudin-5-claudin-5 interactions in trans via their extracellular part and crossing the basement membrane. Finally, we found a loss of crest structures and crest-crest contacts in diseased human CMs and in an experimental mouse model of left ventricle barometric overload. CONCLUSION Overall, these results provide the first evidence for the existence of differential CM surface crests in the cardiac tissue as well as the existence of CM-CM direct physical contacts at their lateral face through crest-crest interactions. We propose a model in which this specific 3D organization of the CM lateral membrane ensures the myofibril/myofiber alignment and the overall cardiac tissue cohesion. A potential role in the control of sarcomere relaxation and of diastolic ventricular dysfunction is also discussed. Whether the loss of CM surface crests constitutes an initial and common event leading to the CM degeneration and the setting of heart failure will need further investigation.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cattle
- Cell Membrane/metabolism
- Cell Membrane/ultrastructure
- Claudin-5/metabolism
- Cryoelectron Microscopy
- Disease Models, Animal
- Female
- Humans
- Male
- Mice, Inbred C57BL
- Microscopy, Atomic Force
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Middle Aged
- Mitochondria, Heart/ultrastructure
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/ultrastructure
- Rats, Wistar
- Sarcomeres/ultrastructure
- Species Specificity
- Tight Junctions/metabolism
- Tight Junctions/ultrastructure
Collapse
Affiliation(s)
- Céline Guilbeau-Frugier
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
- Department of Forensic Medicine, Centre Hospitalier Universitaire de Toulouse, Université de Toulouse, Toulouse, France
- Centre de Microscopie Électronique Appliquée à la Biologie, Faculté de Médecine Rangueil, Université de Toulouse, Toulouse, France
| | - Marie Cauquil
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
| | - Clément Karsenty
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
- Department of Cardiology, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- CHU Toulouse, Pediatric and Congenital Cardiology, Children's Hospital, Université de Toulouse, Toulouse, France
| | - Olivier Lairez
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
- Department of Cardiology, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Camille Dambrin
- Department of Cardiology, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Bruno Payré
- Centre de Microscopie Électronique Appliquée à la Biologie, Faculté de Médecine Rangueil, Université de Toulouse, Toulouse, France
| | - Hervé Cassard
- UMR IHAP, Université de Toulouse, INRA, ENVT, Toulouse, France
| | - Claudie Josse
- Centre de MicroCaractérisation Raimond Castaing, UMS 3623, Toulouse, France
| | - Marie-Hélène Seguelas
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
| | - Sophie Allart
- Centre de Physiopathologie de Toulouse-Purpan, Université de Toulouse, INSERM, CNRS, Toulouse, France
| | - Maxime Branchereau
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
| | - Franck Mandel
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
| | - Marie-Bernadette Delisle
- Centre de Microscopie Électronique Appliquée à la Biologie, Faculté de Médecine Rangueil, Université de Toulouse, Toulouse, France
- Department of Histopathology, Centre Hospitalier Universitaire de Toulouse, Université de Toulouse, Toulouse, France
| | - Atul Pathak
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
- Department of Cardiovascular Medicine, Hypertension, Risk Factors and Heart Failure Unit, Clinique Pasteur, Toulouse, France
| | - Etienne Dague
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Jean-Michel Sénard
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
- Department of Clinical Pharmacology, Centre Hospitalier Universitaire de Toulouse, Université de Toulouse, Toulouse, France
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, INSERM U1048, I2MC, 1, Avenue Jean Poulhès-BP84225, Toulouse, France
| |
Collapse
|
13
|
Kant S, Freytag B, Herzog A, Reich A, Merkel R, Hoffmann B, Krusche CA, Leube RE. Desmoglein 2 mutation provokes skeletal muscle actin expression and accumulation at intercalated discs in murine hearts. J Cell Sci 2019; 132:jcs.199612. [PMID: 30659114 DOI: 10.1242/jcs.199612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/30/2018] [Indexed: 01/05/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is an incurable progressive disease that is linked to mutations in genes coding for components of desmosomal adhesions that are localized to the intercalated disc region, which electromechanically couples adjacent cardiomyocytes. To date, the underlying molecular dysfunctions are not well characterized. In two murine AC models, we find an upregulation of the skeletal muscle actin gene (Acta1), which is known to be a compensatory reaction to compromised heart function. Expression of this gene is elevated prior to visible morphological alterations and clinical symptoms, and persists throughout pathogenesis with an additional major rise during the chronic disease stage. We provide evidence that the increased Acta1 transcription is initiated through nuclear activation of the serum response transcription factor (SRF) by its transcriptional co-activator megakaryoblastic leukemia 1 protein (MKL1, also known as MRTFA). Our data further suggest that perturbed desmosomal adhesion causes Acta1 overexpression during the early stages of the disease, which is amplified by transforming growth factor β (TGFβ) release from fibrotic lesions and surrounding cardiomyocytes during later disease stages. These observations highlight a hitherto unknown molecular AC pathomechanism.
Collapse
Affiliation(s)
- Sebastian Kant
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Benjamin Freytag
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Antonia Herzog
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna Reich
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Rudolf Merkel
- Forschungszentrum Jülich, Institute of Complex Systems, ICS-7, Biomechanics, 52428 Jülich, Germany
| | - Bernd Hoffmann
- Forschungszentrum Jülich, Institute of Complex Systems, ICS-7, Biomechanics, 52428 Jülich, Germany
| | - Claudia A Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
14
|
Pascual F, Schisler JC, Grevengoed TJ, Willis MS, Coleman RA. Modeling the Transition From Decompensated to Pathological Hypertrophy. J Am Heart Assoc 2018; 7:JAHA.117.008293. [PMID: 29622588 PMCID: PMC6015423 DOI: 10.1161/jaha.117.008293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Long-chain acyl-CoA synthetases (ACSL) catalyze the conversion of long-chain fatty acids to fatty acyl-CoAs. Cardiac-specific ACSL1 temporal knockout at 2 months results in a shift from FA oxidation toward glycolysis that promotes mTORC1-mediated ventricular hypertrophy. We used unbiased metabolomics and gene expression analyses to examine the early effects of genetic inactivation of fatty acid oxidation on cardiac metabolism, hypertrophy development, and function. METHODS AND RESULTS Global cardiac transcriptional analysis revealed differential expression of genes involved in cardiac metabolism, fibrosis, and hypertrophy development in Acsl1H-/- hearts 2 weeks after Acsl1 ablation. Comparison of the 2- and 10-week transcriptional responses uncovered 137 genes whose expression was uniquely changed upon knockdown of cardiac ACSL1, including the distinct upregulation of fibrosis genes, a phenomenon not observed after complete ACSL1 knockout. Metabolomic analysis identified metabolites altered in hearts displaying partially reduced ACSL activity, and rapamycin treatment normalized the cardiac metabolomic fingerprint. CONCLUSIONS Short-term cardiac-specific ACSL1 inactivation resulted in metabolic and transcriptional derangements distinct from those observed upon complete ACSL1 knockout, suggesting heart-specific mTOR (mechanistic target of rapamycin) signaling that occurs during the early stages of substrate switching. The hypertrophy observed with partial Acsl1 ablation occurs in the context of normal cardiac function and is reminiscent of a physiological process, making this a useful model to study the transition from physiological to pathological hypertrophy.
Collapse
Affiliation(s)
- Florencia Pascual
- Department of Nutrition, University of North Carolina at Chapel Hill, NC
| | - Jonathan C Schisler
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, NC.,McAllister Heart Institute University of North Carolina at Chapel Hill, NC
| | | | - Monte S Willis
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, NC.,McAllister Heart Institute University of North Carolina at Chapel Hill, NC
| | - Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, NC
| |
Collapse
|
15
|
In Vivo Evaluation of the Cardiovascular System of Mouse Embryo and Fetus Using High Frequency Ultrasound. Methods Mol Biol 2018. [PMID: 29564759 DOI: 10.1007/978-1-4939-7714-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Genetically engineered mice have been widely used for studying cardiovascular development, physiology and diseases. In the past decade, high frequency ultrasound imaging technology has been significantly advanced and applied to observe the cardiovascular structure, function, and blood flow dynamics with high spatial and temporal resolution in mice. This noninvasive imaging approach has made possible longitudinal studies of the mouse embryo/fetus in utero. In this chapter, we describe detailed methods for: (1) the assessment of the structure, function, and flow dynamics of the developing heart of the mouse embryo during middle gestation (E10.5-E13.5); and (2) the measurement of flow distribution throughout the circulatory system of the mouse fetus at late gestation (E17.5). With the described protocols, we are able to illustrate the main cardiovascular structures and the corresponding functional and flow dynamic events at each stage of development, and generate baseline physiological information about the normal mouse embryo/fetus. These data will serve as the reference material for the identification of cardiovascular abnormalities in numerous mouse models with targeted genetic manipulations.
Collapse
|
16
|
Sun X, Hota SK, Zhou YQ, Novak S, Miguel-Perez D, Christodoulou D, Seidman CE, Seidman JG, Gregorio CC, Henkelman RM, Rossant J, Bruneau BG. Cardiac-enriched BAF chromatin-remodeling complex subunit Baf60c regulates gene expression programs essential for heart development and function. Biol Open 2018; 7:bio029512. [PMID: 29183906 PMCID: PMC5829499 DOI: 10.1242/bio.029512] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 11/21/2017] [Indexed: 01/01/2023] Open
Abstract
How chromatin-remodeling complexes modulate gene networks to control organ-specific properties is not well understood. For example, Baf60c (Smarcd3) encodes a cardiac-enriched subunit of the SWI/SNF-like BAF chromatin complex, but its role in heart development is not fully understood. We found that constitutive loss of Baf60c leads to embryonic cardiac hypoplasia and pronounced cardiac dysfunction. Conditional deletion of Baf60c in cardiomyocytes resulted in postnatal dilated cardiomyopathy with impaired contractile function. Baf60c regulates a gene expression program that includes genes encoding contractile proteins, modulators of sarcomere function, and cardiac metabolic genes. Many of the genes deregulated in Baf60c null embryos are targets of the MEF2/SRF co-factor Myocardin (MYOCD). In a yeast two-hybrid screen, we identified MYOCD as a BAF60c interacting factor; we showed that BAF60c and MYOCD directly and functionally interact. We conclude that Baf60c is essential for coordinating a program of gene expression that regulates the fundamental functional properties of cardiomyocytes.
Collapse
Affiliation(s)
- Xin Sun
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 1X8 Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Swetansu K Hota
- Gladstone Institutes, San Francisco, CA, 94158 USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yu-Qing Zhou
- The Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, M5G 1X8 Canada
| | - Stefanie Novak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Dario Miguel-Perez
- Gladstone Institutes, San Francisco, CA, 94158 USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, Gladstone Institutes, San Francisco, CA 94158, USA
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - J G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - R Mark Henkelman
- The Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, M5G 1X8 Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 1X8 Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, CA, 94158 USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
17
|
High-speed, high-frequency ultrasound, in utero vector-flow imaging of mouse embryos. Sci Rep 2017; 7:16658. [PMID: 29192281 PMCID: PMC5709407 DOI: 10.1038/s41598-017-16933-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/19/2017] [Indexed: 01/16/2023] Open
Abstract
Real-time imaging of the embryonic murine cardiovascular system is challenging due to the small size of the mouse embryo and rapid heart rate. High-frequency, linear-array ultrasound systems designed for small-animal imaging provide high-frame-rate and Doppler modes but are limited in regards to the field of view that can be imaged at fine-temporal and -spatial resolution. Here, a plane-wave imaging method was used to obtain high-speed image data from in utero mouse embryos and multi-angle, vector-flow algorithms were applied to the data to provide information on blood flow patterns in major organs. An 18-MHz linear array was used to acquire plane-wave data at absolute frame rates ≥10 kHz using a set of fixed transmission angles. After beamforming, vector-flow processing and image compounding, effective frame rates were on the order of 2 kHz. Data were acquired from the embryonic liver, heart and umbilical cord. Vector-flow results clearly revealed the complex nature of blood-flow patterns in the embryo with fine-temporal and -spatial resolution.
Collapse
|
18
|
Rahman A, Cahill LS, Zhou YQ, Hoggarth J, Rennie MY, Seed M, Macgowan CK, Kingdom JC, Adamson SL, Sled JG. A mouse model of antepartum stillbirth. Am J Obstet Gynecol 2017; 217:443.e1-443.e11. [PMID: 28619691 DOI: 10.1016/j.ajog.2017.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/01/2017] [Accepted: 06/06/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Many stillbirths of normally formed fetuses in the third trimester could be prevented via delivery if reliable means to anticipate this outcome existed. However, because the etiology of these stillbirths is often unexplained and although the underlying mechanism is presumed to be hypoxia from placental insufficiency, the placentas often appear normal on histopathological examination. Gestational age is a risk factor for antepartum stillbirth, with a rapid rise in stillbirth rates after 40 weeks' gestation. We speculate that a common mechanism may explain antepartum stillbirth in both the late-term and postterm periods. Mice also show increasing rates of stillbirth when pregnancy is artificially prolonged. The model therefore affords an opportunity to characterize events that precede stillbirth. OBJECTIVE The objective of the study was to prolong gestation in mice and monitor fetal and placental growth and cardiovascular changes. STUDY DESIGN From embryonic day 15.5 to embryonic day 18.5, pregnant CD-1 mice received daily progesterone injections to prolong pregnancy by an additional 24 hour period (to embryonic day 19.5). To characterize fetal and placental development, experimental assays were performed throughout late gestation (embryonic day 15.5 to embryonic day 19.5), including postnatal day 1 pups as controls. In addition to collecting fetal and placental weights, we monitored fetal blood flow using Doppler ultrasound and examined the fetoplacental arterial vascular geometry using microcomputed tomography. Evidence of hypoxic organ injury in the fetus was assessed using magnetic resonance imaging and pimonidazole immunohistochemistry. RESULTS At embryonic day 19.5, mean fetal weights were reduced by 14% compared with control postnatal day 1 pups. Ultrasound biomicroscopy showed that fetal heart rate and umbilical artery flow continued to increase at embryonic day 19.5. Despite this, the embryonic day 19.5 fetuses had significant pimonidazole staining in both brain and liver tissue, indicating fetal hypoxia. Placental weights at embryonic day 19.5 were 21% lower than at term (embryonic day 18.5). Microcomputed tomography showed no change in quantitative morphology of the fetoplacental arterial vasculature between embryonic day 18.5 and embryonic day 19.5. CONCLUSION Prolongation of pregnancy renders the murine fetus vulnerable to significant growth restriction and hypoxia because of differential loss of placental mass rather than any compromise in fetoplacental blood flow. Our data are consistent with a hypoxic mechanism of antepartum fetal death in human term and postterm pregnancy and validates the inability of umbilical artery Doppler to safely monitor such fetuses. New tests of placental function are needed to identify the late-term fetus at risk of hypoxia to intervene by delivery to avoid antepartum stillbirth.
Collapse
|
19
|
Gerçek M, Gerçek M, Kant S, Simsekyilmaz S, Kassner A, Milting H, Liehn EA, Leube RE, Krusche CA. Cardiomyocyte Hypertrophy in Arrhythmogenic Cardiomyopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:752-766. [PMID: 28183531 DOI: 10.1016/j.ajpath.2016.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 11/23/2016] [Accepted: 12/20/2016] [Indexed: 12/28/2022]
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary disease leading to sudden cardiac death or heart failure. AC pathology is characterized by cardiomyocyte loss and replacement fibrosis. Our goal was to determine whether cardiomyocytes respond to AC progression by pathological hypertrophy. To this end, we examined tissue samples from AC patients with end-stage heart failure and tissue samples that were collected at different disease stages from desmoglein 2-mutant mice, a well characterized AC model. We find that cardiomyocyte diameters are significantly increased in right ventricles of AC patients. Increased mRNA expression of the cardiac stress marker natriuretic peptide B is also observed in the right ventricle of AC patients. Elevated myosin heavy chain 7 mRNA expression is detected in left ventricles. In desmoglein 2-mutant mice, cardiomyocyte diameters are normal during the concealed disease phase but increase significantly after acute disease onset on cardiomyocyte death and fibrotic myocardial remodeling. Hypertrophy progresses further during the chronic disease stage. In parallel, mRNA expression of myosin heavy chain 7 and natriuretic peptide B is up-regulated in both ventricles with right ventricular preference. Calcineurin/nuclear factor of activated T cells (Nfat) signaling, which is linked to pathological hypertrophy, is observed during AC progression, as evidenced by Nfatc2 and Nfatc3 mRNA in cardiomyocytes and increased mRNA of the Nfat target regulator of calcineurin 1. Taken together, we demonstrate that pathological hypertrophy occurs in AC and is secondary to cardiomyocyte loss and cardiac remodeling.
Collapse
Affiliation(s)
- Mustafa Gerçek
- Institutes for Molecular and Cellular Anatomy, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Muhammed Gerçek
- Institutes for Molecular and Cellular Anatomy, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Sebastian Kant
- Institutes for Molecular and Cellular Anatomy, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Sakine Simsekyilmaz
- Institute of Pharmacology and Clinical Pharmacology, Heinrich Heine University, Düsseldorf, Germany
| | - Astrid Kassner
- Heart and Diabetes Center North Rhine-Westphalia, Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Bad Oeynhausen, Germany
| | - Hendrik Milting
- Heart and Diabetes Center North Rhine-Westphalia, Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Bad Oeynhausen, Germany
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research and Interdisciplinary Center for Clinical Research Aachen, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Rudolf E Leube
- Institutes for Molecular and Cellular Anatomy, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Claudia A Krusche
- Institutes for Molecular and Cellular Anatomy, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany.
| |
Collapse
|
20
|
Kohut A, Patel N, Singh H. Comprehensive Echocardiographic Assessment of the Right Ventricle in Murine Models. J Cardiovasc Ultrasound 2016; 24:229-238. [PMID: 27721954 PMCID: PMC5050312 DOI: 10.4250/jcu.2016.24.3.229] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/03/2016] [Accepted: 08/03/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Non-invasive high-resolution echocardiography to evaluate cardiovascular function of small animals is increasingly being used due to availability of genetically engineered murine models. Even though guidelines and standard values for humans were revised by the American Society of Echocardiography, evaluations on murine models are not performed according to any standard protocols. These limitations are preventing translation of preclinical evaluations to clinical meaningful conclusions. We have assessed the right heart of two commonly used murine models according to standard clinical guidelines, and provided the practical guide and sample values for cardiac assessments. METHODS Right heart echocardiography evaluations of CD1 and C57BL/6 mice were performed under 1-3% isoflurane anesthesia using Vevo® 2100 Imaging System with a high-frequency (18-38 MHz) probe (VisualSonics MS400). We have provided a practical guide on how to image and assess the right heart of a mouse which is frequently used to evaluate development of right heart failure due to pulmonary hypertension. RESULTS Our results show significant differences between CD1 and C57BL/6 mice. Right ventricle structural assessment showed significantly larger (p < 0.05) size, and pulmonary artery diameter in CD1 mice (n = 11) compared to C57BL/6 mice (n = 15). Right heart systolic and diastolic functions were similar for both strains. CONCLUSION Our practical guide on how to image and assess the right heart of murine models provides the first comprehensive values which can be used for preclinical research studies using echocardiography. Additionally, our results indicate that there is a high variability between mouse species and experimental models should be carefully selected for cardiac evaluations.
Collapse
Affiliation(s)
- Andrew Kohut
- Division of Cardiology, Drexel University College of Medicine, Philadelphia, PA, USA.; Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Nishi Patel
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Harpreet Singh
- Division of Cardiology, Drexel University College of Medicine, Philadelphia, PA, USA.; Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA.; Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
21
|
Re A, Nanni S, Aiello A, Granata S, Colussi C, Campostrini G, Spallotta F, Mattiussi S, Pantisano V, D'Angelo C, Biroccio A, Rossini A, Barbuti A, DiFrancesco D, Trimarchi F, Pontecorvi A, Gaetano C, Farsetti A. Anacardic acid and thyroid hormone enhance cardiomyocytes production from undifferentiated mouse ES cells along functionally distinct pathways. Endocrine 2016; 53:681-8. [PMID: 26547215 DOI: 10.1007/s12020-015-0751-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/19/2015] [Indexed: 10/22/2022]
Abstract
The epigenetics of early commitment to embryonal cardiomyocyte is poorly understood. In this work, we compared the effect of thyroid hormone and that of anacardic acid, a naturally occurring histone acetylase inhibitor, or both in combination, on mouse embryonic stem cells (mES) differentiating into embryonal cardiomyocyte by embryoid bodies (EBs) formation. Although the results indicated that anacardic acid (AA) and thyroid hormone were both efficient in promoting cardiomyocyte differentiation, we noticed that a transient exposure of mES to AA alone was sufficient to enlarge the beating areas of EBs compared to those of untreated controls. This effect was associated with changes in the chromatin structure at the promoters of specific cardiomyogenic genes. Among them, a rapid induction of the transcription factor Castor 1 (CASZ1), important for cardiomyocytes differentiation and maturation during embryonic development, was observed in the presence of AA. In contrast, thyroid hormone (T 3) was more effective in stimulating spontaneous firing, thus suggesting a role in the production of a population of cardiomyocyte with pacemaker properties. In conclusion, AA and thyroid hormone both enhanced cardiomyocyte formation along in apparently distinct pathways.
Collapse
Affiliation(s)
- Agnese Re
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | - Simona Nanni
- Institute of Medical Pathology, Catholic University, Rome, Italy
| | - Aurora Aiello
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | - Serena Granata
- Institute of Medical Pathology, Catholic University, Rome, Italy
| | - Claudia Colussi
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | - Giulia Campostrini
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Internal Medicine Clinic III, Goethe University Frankfurt, 60590, Frankfurt, Germany
| | - Stefania Mattiussi
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | | | - Carmen D'Angelo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Annamaria Biroccio
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Rossini
- Bolzano Center for Biomedicine (Affiliated Institute of the University of Lübeck), European Academy Bozen/Bolzano (EURAC), Bolzano, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Andrea Barbuti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Dario DiFrancesco
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Francesco Trimarchi
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Internal Medicine Clinic III, Goethe University Frankfurt, 60590, Frankfurt, Germany.
| | - Antonella Farsetti
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy.
- Internal Medicine Clinic III, Goethe University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
22
|
Tanaka K, Valero-Muñoz M, Wilson RM, Essick EE, Fowler CT, Nakamura K, van den Hoff M, Ouchi N, Sam F. Follistatin like 1 Regulates Hypertrophy in Heart Failure with Preserved Ejection Fraction. JACC Basic Transl Sci 2016; 1:207-221. [PMID: 27430031 PMCID: PMC4944656 DOI: 10.1016/j.jacbts.2016.04.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for ∼50% of all clinical presentations of heart failure, (HF) and its prevalence is expected to increase. However, there are no evidence-based therapies for HFpEF; thus, HFpEF represents a major unmet need. Although hypertension is the single most important risk factor for HFpEF, with a prevalence of 60% to 89% from clinical trials and human HF registries, blood pressure therapy alone is insufficient to prevent and treat HFpEF. Follistatin-like 1 (Fstl1), a divergent member of the follistatin family of extracellular glycoproteins, has previously been shown to be elevated in HF with reduced ejection fraction and associated with increased left ventricular mass. In this study, blood levels of Fstl1 were increased in humans with HFpEF. This increase was also evident in mice with hypertension-induced HFpEF and adult rat ventricular myocytes stimulated with aldosterone. Treatment with recombinant Fstl1 abrogated aldosterone-induced cardiac myocyte hypertrophy, suggesting a role for Fstl1 in the regulation of hypertrophy in HFpEF. There was also a reduction in the E/A ratio, a measure of diastolic dysfunction. Furthermore, HFpEF induced in a mouse model that specifically ablates Fstl1 in cardiac myocytes (cardiac myocyte-specific Fstl1 knockout [cFstl1-KO]) showed exacerbation of HFpEF with worsened diastolic dysfunction. In addition, cFstl1-KO-HFpEF mice demonstrated more marked cardiac myocyte hypertrophy with increased molecular markers of atrial natriuretic peptide and brain natriuretic peptide expression. These findings indicate that Fstl1 exerts therapeutic effects by modulating cardiac hypertrophy in HFpEF. Fstl1, also known as transforming growth factor-β–stimulated clone 36, is an extra-cellular glycoprotein implicated in the pathophysiology of cardiac disease. Fstl1 acts in a noncanonical manner relative to other follistatin family members, but its functions remain poorly understood. Circulating Flst1 levels are increased in humans with chronic stable HFpEF. Fstl1 treatment modulates cardiomyocyte hypertrophy in vitro and in vivo. Cardiac myocyte deletion of Fstl1 worsens the HFpEF phenotype in mice. These studies indicate that Fstl1 may be therapeutically effective in HFpEF by modulating cardiac hypertrophy and improving parameters of diastolic dysfunction.
Collapse
Affiliation(s)
- Komei Tanaka
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - María Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Richard M Wilson
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Eric E Essick
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Conor T Fowler
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Kazuto Nakamura
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Maurice van den Hoff
- Academic Medical Center, Heart Failure Research Center, Department of Anatomy, Embryology & Physiology, Amsterdam, The Netherlands
| | - Noriyuki Ouchi
- Department of Molecular Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA.,Cardiovascular Section and Evans Department of Medicine, Heart Failure Program, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
23
|
Iorga A, Li J, Sharma S, Umar S, Bopassa JC, Nadadur RD, Centala A, Ren S, Saito T, Toro L, Wang Y, Stefani E, Eghbali M. Rescue of Pressure Overload-Induced Heart Failure by Estrogen Therapy. J Am Heart Assoc 2016; 5:e002482. [PMID: 26802104 PMCID: PMC4859364 DOI: 10.1161/jaha.115.002482] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/22/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Estrogen pretreatment has been shown to attenuate the development of heart hypertrophy, but it is not known whether estrogen could also rescue heart failure (HF). Furthermore, the heart has all the machinery to locally biosynthesize estrogen via aromatase, but the role of local cardiac estrogen synthesis in HF has not yet been studied. Here we hypothesized that cardiac estrogen is reduced in HF and examined whether exogenous estrogen therapy can rescue HF. METHODS AND RESULTS HF was induced by transaortic constriction in mice, and once mice reached an ejection fraction (EF) of ≈35%, they were treated with estrogen for 10 days. Cardiac structure and function, angiogenesis, and fibrosis were assessed, and estrogen was measured in plasma and in heart. Cardiac estrogen concentrations (6.18±1.12 pg/160 mg heart in HF versus 17.79±1.28 pg/mL in control) and aromatase transcripts (0.19±0.04, normalized to control, P<0.05) were significantly reduced in HF. Estrogen therapy increased cardiac estrogen 3-fold and restored aromatase transcripts. Estrogen also rescued HF by restoring ejection fraction to 53.1±1.3% (P<0.001) and improving cardiac hemodynamics both in male and female mice. Estrogen therapy stimulated angiogenesis as capillary density increased from 0.66±0.07 in HF to 2.83±0.14 (P<0.001, normalized to control) and reversed the fibrotic scarring observed in HF (45.5±2.8% in HF versus 5.3±1.0%, P<0.001). Stimulation of angiogenesis by estrogen seems to be one of the key mechanisms, since in the presence of an angiogenesis inhibitor estrogen failed to rescue HF (ejection fraction=29.3±2.1%, P<0.001 versus E2). CONCLUSIONS Estrogen rescues pre-existing HF by restoring cardiac estrogen and aromatase, stimulating angiogenesis, and suppressing fibrosis.
Collapse
MESH Headings
- Animals
- Aromatase/genetics
- Aromatase/metabolism
- Disease Models, Animal
- Estradiol/blood
- Estradiol/pharmacology
- Estrogen Receptor beta/drug effects
- Estrogen Receptor beta/metabolism
- Female
- Fibrosis
- Heart Failure/blood
- Heart Failure/drug therapy
- Heart Failure/genetics
- Heart Failure/pathology
- Heart Failure/physiopathology
- Male
- Mice, Inbred C57BL
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Neovascularization, Physiologic/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Recovery of Function
- Signal Transduction/drug effects
- Stroke Volume/drug effects
- Time Factors
- Ventricular Dysfunction, Left/blood
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/drug effects
Collapse
Affiliation(s)
- Andrea Iorga
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Jingyuan Li
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Salil Sharma
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Soban Umar
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Jean C. Bopassa
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Rangarajan D. Nadadur
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Alexander Centala
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Shuxun Ren
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Tomoaki Saito
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Ligia Toro
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
- Department of Molecular & Medical PharmacologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Yibin Wang
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
- Department of PhysiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Enrico Stefani
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
- Department of PhysiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| | - Mansoureh Eghbali
- Division of Molecular MedicineDepartment of AnesthesiologyDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCA
| |
Collapse
|
24
|
Stembridge M, Ainslie PN, Donnelly J, MacLeod NT, Joshi S, Hughes MG, Sherpa K, Shave R. Cardiac structure and function in adolescent Sherpa; effect of habitual altitude and developmental stage. Am J Physiol Heart Circ Physiol 2016; 310:H740-6. [PMID: 26801313 DOI: 10.1152/ajpheart.00938.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/16/2016] [Indexed: 12/14/2022]
Abstract
The purpose of this study was to examine ventricular structure and function in Sherpa adolescents to determine whether age-specific differences in oxygen saturation (SpO2 ) and pulmonary artery systolic pressure (PASP) influence cardiac adaptation to chronic hypoxia early in life. Two-dimensional, Doppler, and speckle-tracking echocardiography were performed on adolescent (9-16 yr) highland Sherpa (HLS; 3,840 m; n = 26) and compared with age-matched lowland Sherpa (LLS; 1,400 m; n = 10) and lowland Caucasian controls (LLC; sea level; n = 30). The HLS were subdivided into pre- and postadolescence; SpO2 was also recorded. Only HLS exhibited a smaller relative left ventricular (LV) end-diastolic volume; however, both HLS and LLS demonstrated a lower peak LV untwisting velocity compared with LLC (92 ± 26 and 100 ± 45 vs. 130 ± 43°/s, P < 0.05). Although SpO2 was similar between groups, PASP was higher in post- vs. preadolescent HLS (30 ± 5 vs. 25 ± 5 mmHg, P < 0.05), which negatively correlated with right ventricular strain rate (r = 0.50, P < 0.01). Much like their adult counterparts, HLS and LLS adolescents exhibit slower LV diastolic relaxation, despite residing at different altitudes. These findings suggest fundamental differences exist in the diastolic function of Sherpa that are present at an early age and may be retained after migration to lower altitudes. The higher PASP in postadolescent Sherpa is in contrast to previous reports of lowland children at high altitude and, unlike that in lowlanders, was not explained by differences in SpO2 ; thus different regulatory mechanisms seem to exist between these two distinct populations.
Collapse
Affiliation(s)
- Mike Stembridge
- Cardiff School of Sport, Cardiff Metropolitan University, Cardiff, United Kingdom;
| | - Philip N Ainslie
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia Okanagan Campus, Kelowna, British Columbia, Canada
| | - Joseph Donnelly
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Suchita Joshi
- Patan Academy of Health Sciences, Kathmandu, Nepal; and
| | - Michael G Hughes
- Cardiff School of Sport, Cardiff Metropolitan University, Cardiff, United Kingdom
| | | | - Rob Shave
- Cardiff School of Sport, Cardiff Metropolitan University, Cardiff, United Kingdom
| |
Collapse
|
25
|
Greco A, Coda ARD, Albanese S, Ragucci M, Liuzzi R, Auletta L, Gargiulo S, Lamagna F, Salvatore M, Mancini M. High-Frequency Ultrasound for the Study of Early Mouse Embryonic Cardiovascular System. Reprod Sci 2015; 22:1649-1655. [DOI: 10.1177/1933719115594017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
26
|
Lee J, Cao H, Kang BJ, Jen N, Yu F, Lee CA, Fei P, Park J, Bohlool S, Lash-Rosenberg L, Shung KK, Hsiai TK. Hemodynamics and ventricular function in a zebrafish model of injury and repair. Zebrafish 2015; 11:447-54. [PMID: 25237983 DOI: 10.1089/zeb.2014.1016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Myocardial infarction results in scar tissue and irreversible loss of ventricular function. Unlike humans, zebrafish has the capacity to remove scar tissue after injury. To assess ventricular function during repair, we synchronized microelectrocardiogram (μECG) signals with a high-frequency ultrasound pulsed-wave (PW) Doppler to interrogate cardiac hemodynamics. μECG signals allowed for identification of PW Doppler signals for passive (early [E]-wave velocity) and active ventricular filling (atrial [A]-wave velocity) during diastole. The A wave (9.0±1.2 cm·s(-1)) is greater than the E wave (1.1±0.4 cm·s(-1)), resulting in an E/A ratio <1 (0.12±0.05, n=6). In response to cryocauterization to the ventricular epicardium, the E-wave velocity increased, accompanied by a rise in the E/A ratio at 3 days postcryocauterization (dpc) (0.55±0.13, n=6, p<0.001 vs. sham). The E waves normalize toward the baseline, along with a reduction in the E/A ratio at 35 dpc (0.36±0.06, n=6, p<0.001 vs. sham) and 65 dpc (0.2±0.16, n=6, p<0.001 vs. sham). In zebrafish, E/A<1 at baseline is observed, suggesting the distinct two-chamber system in which the pressure gradient across the atrioventricular valve is higher compared with the ventriculobulbar valve. The initial rise and subsequent normalization of E/A ratios support recovery in the ventricular diastolic function.
Collapse
Affiliation(s)
- Juhyun Lee
- 1 Division of Cardiology, Department of Medicine, University of California , Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zheng D, Ma J, Yu Y, Li M, Ni R, Wang G, Chen R, Li J, Fan GC, Lacefield JC, Peng T. Silencing of miR-195 reduces diabetic cardiomyopathy in C57BL/6 mice. Diabetologia 2015; 58:1949-58. [PMID: 25994075 PMCID: PMC4499474 DOI: 10.1007/s00125-015-3622-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/14/2015] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS MicroRNAs (miRs) have been suggested as potential therapeutic targets for heart diseases. Inhibition of miR-195 prevents apoptosis in cardiomyocytes stimulated with palmitate and transgenic overexpression of miR-195 induces cardiac hypertrophy and heart failure. We investigated whether silencing of miR-195 reduces diabetic cardiomyopathy in a mouse model of streptozotocin (STZ)-induced type 1 diabetes. METHODS Type 1 diabetes was induced in C57BL/6 mice (male, 2 months old) by injections of STZ. RESULTS MiR-195 expression was increased and levels of its target proteins (B cell leukaemia/lymphoma 2 and sirtuin 1) were decreased in STZ-induced type 1 and db/db type 2 diabetic mouse hearts. Systemically delivering an anti-miR-195 construct knocked down miR-195 expression in the heart, reduced caspase-3 activity, decreased oxidative stress, attenuated myocardial hypertrophy and improved myocardial function in STZ-induced mice with a concurrent upregulation of B cell leukaemia/lymphoma 2 and sirtuin 1. Diabetes reduced myocardial capillary density and decreased maximal coronary blood flow in mice. Knockdown of miR-195 increased myocardial capillary density and improved maximal coronary blood flow in diabetic mice. Upregulation of miR-195 sufficiently induced apoptosis in cardiomyocytes and attenuated the angiogenesis of cardiac endothelial cells in vitro. Furthermore, inhibition of miR-195 prevented apoptosis in cardiac endothelial cells in response to NEFA, an important feature of diabetes. CONCLUSIONS/INTERPRETATION Therapeutic silencing of miR-195 reduces myocardial hypertrophy and improves coronary blood flow and myocardial function in diabetes, at least in part by reducing oxidative damage, inhibiting apoptosis and promoting angiogenesis. Thus, miR-195 may represent an alternative therapeutic target for diabetic heart diseases.
Collapse
Affiliation(s)
- Dong Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China 215123
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Jian Ma
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Yong Yu
- Zhongshan Hospital of Fudan University, Shanghai, China
| | - Minghui Li
- Zhongshan Hospital of Fudan University, Shanghai, China
| | - Rui Ni
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Grace Wang
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| | - Ruizhen Chen
- Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James C. Lacefield
- Electrical and Computer Engineering, Medical Biophysics, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Tianqing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China 215123
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, Canada N6A 4G5
- Department of Medicine, The University of Western Ontario, London, ON, Canada
- Department of Pathology, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
28
|
Murine left atrium and left atrial appendage structure and function: echocardiographic and morphologic evaluation. PLoS One 2015; 10:e0125541. [PMID: 25928887 PMCID: PMC4415937 DOI: 10.1371/journal.pone.0125541] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 03/25/2015] [Indexed: 11/19/2022] Open
Abstract
Aim of this study was to provide an echocardiographic protocol for the description of the normal murine venous reservoir (atrium, appendage and pulmonary veins) and to investigate the possibility to use this approach to discriminate changes on left atrium (LA) and left atrial appendage (LAA) in a stress-induced model such us myocardial infarction. Global left ventricular function and the venous reservoir were assessed by a Vevo2100 in 20 female C57BL/6N. LA and LAA were also studied in 10 CD-1 and 10 FVB mice, whereas modifications investigated in 15 C57BL/6N subjected to coronary artery ligation. Left ventricle function was evaluated as well as pulsed Doppler mitral valve, pulmonary vein, and LAA velocities. From 2D view monoplane LA volumes were obtained and LAA long axis measured. Macroscopic inspection with casts and immunohistochemistry were performed. Results show that compared to humans, in C57BL/6N mice left atrium was disproportionately smaller (5.2±1.4 μL) than the left ventricle (53±8 μL) and connected through a duct by a large LAA and posteriorly to three pulmonary veins. The LA volume increased 2-fold during reservoir with two distinct phases, early and late divided by a short pause. LAA long axis (4.1±0.5 mm) was almost 2 times longer than the LA. LAA flow volume together with LA volume reservoir account for about 36% of stroke volume and the rest was provided by conduit flow. Linear regressions showed that stroke volume was strongly influenced by LAA flow, LA early filling volume and left ventricle base descent. Moreover, we also report the ability to assess LA and LAA in other mice strains and discriminate size increase following myocardial infarction. In conclusion, we performed a complete characterization of murine left venous reservoir establishing an optimized protocol that can be used in both investigative and pharmacological studies requiring rapid and serial determination of cardiac structure and function.
Collapse
|
29
|
Egemnazarov B, Schmidt A, Crnkovic S, Sydykov A, Nagy BM, Kovacs G, Weissmann N, Olschewski H, Olschewski A, Kwapiszewska G, Marsh LM. Pressure Overload Creates Right Ventricular Diastolic Dysfunction in a Mouse Model: Assessment by Echocardiography. J Am Soc Echocardiogr 2015; 28:828-43. [PMID: 25840639 DOI: 10.1016/j.echo.2015.02.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Indexed: 10/23/2022]
Abstract
BACKGROUND Noninvasive diagnostic tools for right ventricular (RV) dysfunction measurements are increasingly being used, although their association with the pathologic mechanisms of dysfunction is poorly understood. Although investigations have focused mainly on RV systolic function, RV diastolic function remains mostly neglected. The aim of this study was to test which echocardiographic parameters best reflect RV diastolic function in mice. METHODS Pulmonary artery banding (PAB) was used to induce RV pressure overload in mice. Transthoracic echocardiography and invasive hemodynamic measurements were performed after 3 weeks in PAB and sham-operated mice. Subsequently, the hearts were investigated by histology and analyzed for gene expression. RESULTS PAB-induced pressure overload (RV systolic pressure PAB 52.6 ± 11.8 mm Hg vs sham 27.0 ± 2.7 mm Hg) resulted in RV hypertrophy and remodeling, as reflected by increased Fulton index (PAB 0.37 ± 0.05 vs sham 0.25 ± 0.02, P = .001). Masson's trichrome staining revealed increased interstitial fibrosis (PAB 12.25 ± 3.12% vs sham 3.97 ± 1.58%, P = .002). This was associated with significant systolic RV dysfunction as demonstrated by reduced contractility index and diastolic dysfunction as demonstrated by end-diastolic pressure (PAB 2.66 ± 0.83 mm Hg vs sham 1.49 ± 0.50 mm Hg, P < .001) and τ (PAB 40.0 ± 16.1 msec vs sham 13.0 ± 3.5 msec, P < .001). Messenger ribonucleic acid expression of β-myosin heavy chain, atrial and brain natriuretic peptides, collagen family members was elevated, and the sarco/endoplasmic reticulum Ca(2+)-ATPase was decreased. Echocardiography revealed significant increases in RV free wall thickness and isovolumic relaxation time and a decrease in left ventricular eccentricity index, E', and tricuspid annular plane systolic excursion. Isovolumic relaxation time and E' were significantly correlated with end-diastolic pressure (rs = 0.511 and -0.451) and τ (rs = 0.739 and -0.445, respectively). Moreover, E' was negatively correlated with the degree of RV fibrosis (rs = -0.717). CONCLUSIONS Within 3 weeks, PAB causes pressure overload-induced RV hypertrophy and remodeling with compensated systolic and diastolic dysfunction in mice. RV free wall thickness, tricuspid annular plane systolic excursion, E', E/E' ratio, and isovolumic relaxation time appear to be the most reliable echocardiographic parameters for the assessment of RV dysfunction.
Collapse
Affiliation(s)
| | - Albrecht Schmidt
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Akylbek Sydykov
- University of Giessen and Marburg Lung Center, Excellence Cluster Cardio-Pulmonary System, Giessen, Germany
| | - Bence M Nagy
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Norbert Weissmann
- University of Giessen and Marburg Lung Center, Excellence Cluster Cardio-Pulmonary System, Giessen, Germany
| | - Horst Olschewski
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Experimental Anesthesiology, Department of Anesthesia and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Experimental Anesthesiology, Department of Anesthesia and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| |
Collapse
|
30
|
Kowalski WJ, Pekkan K, Tinney JP, Keller BB. Investigating developmental cardiovascular biomechanics and the origins of congenital heart defects. Front Physiol 2014; 5:408. [PMID: 25374544 PMCID: PMC4204442 DOI: 10.3389/fphys.2014.00408] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 10/02/2014] [Indexed: 11/24/2022] Open
Abstract
Innovative research on the interactions between biomechanical load and cardiovascular (CV) morphogenesis by multiple investigators over the past 3 decades, including the application of bioengineering approaches, has shown that the embryonic heart adapts both structure and function in order to maintain cardiac output to the rapidly growing embryo. Acute adaptive hemodynamic mechanisms in the embryo include the redistribution of blood flow within the heart, dynamic adjustments in heart rate and developed pressure, and beat to beat variations in blood flow and vascular resistance. These biomechanically relevant events occur coincident with adaptive changes in gene expression and trigger adaptive mechanisms that include alterations in myocardial cell growth and death, regional and global changes in myocardial architecture, and alterations in central vascular morphogenesis and remodeling. These adaptive mechanisms allow the embryo to survive these biomechanical stresses (environmental, maternal) and to compensate for developmental errors (genetic). Recent work from numerous laboratories shows that a subset of these adaptive mechanisms is present in every developing multicellular organism with a “heart” equivalent structure. This chapter will provide the reader with an overview of some of the approaches used to quantify embryonic CV functional maturation and performance, provide several illustrations of experimental interventions that explore the role of biomechanics in the regulation of CV morphogenesis including the role of computational modeling, and identify several critical areas for future investigation as available experimental models and methods expand.
Collapse
Affiliation(s)
- William J Kowalski
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA
| | - Kerem Pekkan
- Department of Biomedical Engineering, Carnegie Mellon University Pittsburgh, PA, USA
| | - Joseph P Tinney
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA
| | - Bradley B Keller
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA ; Department of Biomedical Engineering, Carnegie Mellon University Pittsburgh, PA, USA
| |
Collapse
|
31
|
Dweck D, Sanchez-Gonzalez MA, Chang AN, Dulce RA, Badger CD, Koutnik AP, Ruiz EL, Griffin B, Liang J, Kabbaj M, Fincham FD, Hare JM, Overton JM, Pinto JR. Long term ablation of protein kinase A (PKA)-mediated cardiac troponin I phosphorylation leads to excitation-contraction uncoupling and diastolic dysfunction in a knock-in mouse model of hypertrophic cardiomyopathy. J Biol Chem 2014; 289:23097-23111. [PMID: 24973218 DOI: 10.1074/jbc.m114.561472] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cardiac troponin I (cTnI) R21C (cTnI-R21C) mutation has been linked to hypertrophic cardiomyopathy and renders cTnI incapable of phosphorylation by PKA in vivo. Echocardiographic imaging of homozygous knock-in mice expressing the cTnI-R21C mutation shows that they develop hypertrophy after 12 months of age and have abnormal diastolic function that is characterized by longer filling times and impaired relaxation. Electrocardiographic analyses show that older R21C mice have elevated heart rates and reduced cardiovagal tone. Cardiac myocytes isolated from older R21C mice demonstrate that in the presence of isoproterenol, significant delays in Ca(2+) decay and sarcomere relaxation occur that are not present at 6 months of age. Although isoproterenol and stepwise increases in stimulation frequency accelerate Ca(2+)-transient and sarcomere shortening kinetics in R21C myocytes from older mice, they are unable to attain the corresponding WT values. When R21C myocytes from older mice are treated with isoproterenol, evidence of excitation-contraction uncoupling is indicated by an elevation in diastolic calcium that is frequency-dissociated and not coupled to shorter diastolic sarcomere lengths. Myocytes from older mice have smaller Ca(2+) transient amplitudes (2.3-fold) that are associated with reductions (2.9-fold) in sarcoplasmic reticulum Ca(2+) content. This abnormal Ca(2+) handling within the cell may be attributed to a reduction (2.4-fold) in calsequestrin expression in conjunction with an up-regulation (1.5-fold) of Na(+)-Ca(2+) exchanger. Incubation of permeabilized cardiac fibers from R21C mice with PKA confirmed that the mutation prevents facilitation of mechanical relaxation. Altogether, these results indicate that the inability to enhance myofilament relaxation through cTnI phosphorylation predisposes the heart to abnormal diastolic function, reduced accessibility of cardiac reserves, dysautonomia, and hypertrophy.
Collapse
Affiliation(s)
- David Dweck
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300
| | - Marcos A Sanchez-Gonzalez
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300,; Family Institute, Florida State University, Tallahassee, Florida 32306
| | - Audrey N Chang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Raul A Dulce
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - Crystal-Dawn Badger
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300
| | - Andrew P Koutnik
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300
| | - Edda L Ruiz
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300
| | - Brittany Griffin
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300
| | - Frank D Fincham
- Family Institute, Florida State University, Tallahassee, Florida 32306
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, and
| | - J Michael Overton
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306-4300,.
| |
Collapse
|
32
|
Paul DS, Grevengoed TJ, Pascual F, Ellis JM, Willis MS, Coleman RA. Deficiency of cardiac Acyl-CoA synthetase-1 induces diastolic dysfunction, but pathologic hypertrophy is reversed by rapamycin. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:880-7. [PMID: 24631848 DOI: 10.1016/j.bbalip.2014.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 02/17/2014] [Accepted: 03/03/2014] [Indexed: 12/14/2022]
Abstract
In mice with temporally-induced cardiac-specific deficiency of acyl-CoA synthetase-1 (Acsl1(H-/-)), the heart is unable to oxidize long-chain fatty acids and relies primarily on glucose for energy. These metabolic changes result in the development of both a spontaneous cardiac hypertrophy and increased phosphorylated S6 kinase (S6K), a substrate of the mechanistic target of rapamycin, mTOR. Doppler echocardiography revealed evidence of significant diastolic dysfunction, indicated by a reduced E/A ratio and increased mean performance index, although the deceleration time and the expression of sarco/endoplasmic reticulum calcium ATPase and phospholamban showed no difference between genotypes. To determine the role of mTOR in the development of cardiac hypertrophy, we treated Acsl1(H-/-) mice with rapamycin. Six to eight week old Acsl1(H-/-) mice and their littermate controls were given i.p. tamoxifen to eliminate cardiac Acsl1, then concomitantly treated for 10weeks with i.p. rapamycin or vehicle alone. Rapamycin completely blocked the enhanced ventricular S6K phosphorylation and cardiac hypertrophy and attenuated the expression of hypertrophy-associated fetal genes, including α-skeletal actin and B-type natriuretic peptide. mTOR activation of the related Acsl3 gene, usually associated with pathologic hypertrophy, was also attenuated in the Acsl1(H-/-) hearts, indicating that alternative pathways of fatty acid activation did not compensate for the loss of Acsl1. Compared to controls, Acsl1(H-/-) hearts exhibited an 8-fold higher uptake of 2-deoxy[1-(14)C]glucose and a 35% lower uptake of the fatty acid analog 2-bromo[1-(14)C]palmitate. These data indicate that Acsl1-deficiency causes diastolic dysfunction and that mTOR activation is linked to the development of cardiac hypertrophy in Acsl1(H-/-) mice.
Collapse
Affiliation(s)
- David S Paul
- McAllister Heart Institute, University of NC at Chapel Hill, 27599, USA.
| | | | - Florencia Pascual
- Department of Nutrition, University of NC at Chapel Hill, 27599, USA.
| | - Jessica M Ellis
- Department of Nutrition, University of NC at Chapel Hill, 27599, USA.
| | - Monte S Willis
- McAllister Heart Institute, University of NC at Chapel Hill, 27599, USA; Department of Pathology and Laboratory Medicine, University of NC at Chapel Hill, 27599, USA.
| | - Rosalind A Coleman
- Department of Nutrition, University of NC at Chapel Hill, 27599, USA; McAllister Heart Institute, University of NC at Chapel Hill, 27599, USA.
| |
Collapse
|
33
|
Echocardiographic assessment of embryonic and fetal mouse heart development: a focus on haemodynamics and morphology. ScientificWorldJournal 2014; 2014:531324. [PMID: 24707208 PMCID: PMC3951091 DOI: 10.1155/2014/531324] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/31/2013] [Indexed: 11/17/2022] Open
Abstract
Background. Heart development is a complex process, and abnormal development may result in congenital heart disease (CHD). Currently, studies on animal models mainly focus on cardiac morphology and the availability of hemodynamic data, especially of the right heart half, is limited. Here we aimed to assess the morphological and hemodynamic parameters of normal developing mouse embryos/fetuses by using a high-frequency ultrasound system. Methods. A timed breeding program was initiated with a WT mouse line (Swiss/129Sv background). All recordings were performed transabdominally, in isoflurane sedated pregnant mice, in hearts of sequential developmental stages: 12.5, 14.5, and 17.5 days after conception (n = 105). Results. Along development the heart rate increased significantly from 125 ± 9.5 to 219 ± 8.3 beats per minute. Reliable flow measurements could be performed across the developing mitral and tricuspid valves and outflow tract. M-mode measurements could be obtained of all cardiac compartments. An overall increase of cardiac systolic and diastolic function with embryonic/fetal development was observed. Conclusion. High-frequency echocardiography is a promising and useful imaging modality for structural and hemodynamic analysis of embryonic/fetal mouse hearts.
Collapse
|
34
|
Wu J, Zhou YQ, Zou Y, Henkelman M. Evaluation of bi-ventricular coronary flow patterns using high-frequency ultrasound in mice with transverse aortic constriction. ULTRASOUND IN MEDICINE & BIOLOGY 2013; 39:2053-2065. [PMID: 23932279 DOI: 10.1016/j.ultrasmedbio.2013.04.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 03/15/2013] [Accepted: 04/28/2013] [Indexed: 06/02/2023]
Abstract
Using high-frequency color and pulsed Doppler ultrasound, we evaluated the flow patterns of the left (LCA), septal (SCA) and right (RCA) coronary arteries in mice with and without transverse aortic constriction (TAC). Fifty-two male C57BL/6J mice were subjected to TAC or a corresponding sham operation. At 2 and 8 wk post-surgery, Doppler flow spectra from the three coronary arteries, together with morphologic and functional parameters of the left and right ventricles, were measured. Histology was performed to evaluate myocyte size and neo-angiogenesis in both ventricles. In sham-operated mice, the LCA and SCA both exhibited low-flow waveforms during systole and dominantly higher-flow waveforms during diastole. The RCA exhibited generally lower flow velocity, with similar systolic and diastolic waveforms. TAC significantly increased the systolic flow velocities of all coronary arteries, but enhanced the flow mainly in the LCA and SCA. In the left ventricle, coronary flow reserve was partially preserved 2 wk post-TAC, but decreased at 8 wk, consistent with changes in neo-angiogenesis and systolic function. In contrast, no significant change was found in the coronary flow reserve, structure or function of the right ventricle. This study has established a protocol for evaluating the flow pattern in three principal coronary arteries in mice using Doppler ultrasound and illustrated the difference among three vessels at baseline. In mice with TAC, the difference in the associating pattern of coronary flow dynamics with the myocardial structure and function between the left and right ventricles provides further insights into ventricular remodeling under pressure overload.
Collapse
Affiliation(s)
- Jian Wu
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
35
|
Gregg CL, Butcher JT. Translational paradigms in scientific and clinical imaging of cardiac development. ACTA ACUST UNITED AC 2013; 99:106-20. [PMID: 23897595 DOI: 10.1002/bdrc.21034] [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: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 01/25/2023]
Abstract
Congenital heart defects (CHD) are the most prevalent congenital disease, with 45% of deaths resulting from a congenital defect due to a cardiac malformation. Clinically significant CHD permit survival upon birth, but may become immediately life threatening. Advances in surgical intervention have significantly reduced perinatal mortality, but the outcome for many malformations is bleak. Furthermore, patients living while tolerating a CHD often acquire additional complications due to the long-term systemic blood flow changes caused by even subtle anatomical abnormalities. Accurate diagnosis of defects during fetal development is critical for interventional planning and improving patient outcomes. Advances in quantitative, multidimensional imaging are necessary to uncover the basic scientific and clinically relevant morphogenetic changes and associated hemodynamic consequences influencing normal and abnormal heart development. Ultrasound is the most widely used clinical imaging technology for assessing fetal cardiac development. Ultrasound-based fetal assessment modalities include motion mode (M-mode), two dimensional (2D), and 3D/4D imaging. These datasets can be combined with computational fluid dynamics analysis to yield quantitative, volumetric, and physiological data. Additional imaging modalities, however, are available to study basic mechanisms of cardiogenesis, including optical coherence tomography, microcomputed tomography, and magnetic resonance imaging. Each imaging technology has its advantages and disadvantages regarding resolution, depth of penetration, soft tissue contrast considerations, and cost. In this review, we analyze the current clinical and scientific imaging technologies, research studies utilizing them, and appropriate animal models reflecting clinically relevant cardiogenesis and cardiac malformations. We conclude with discussing the translational impact and future opportunities for cardiovascular development imaging research.
Collapse
Affiliation(s)
- Chelsea L Gregg
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
36
|
Scavone A, Capilupo D, Mazzocchi N, Crespi A, Zoia S, Campostrini G, Bucchi A, Milanesi R, Baruscotti M, Benedetti S, Antonini S, Messina G, DiFrancesco D, Barbuti A. Embryonic stem cell-derived CD166+ precursors develop into fully functional sinoatrial-like cells. Circ Res 2013; 113:389-98. [PMID: 23753573 DOI: 10.1161/circresaha.113.301283] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE A cell-based biological pacemaker is based on the differentiation of stem cells and the selection of a population displaying the molecular and functional properties of native sinoatrial node (SAN) cardiomyocytes. So far, such selection has been hampered by the lack of proper markers. CD166 is specifically but transiently expressed in the mouse heart tube and sinus venosus, the prospective SAN. OBJECTIVE We have explored the possibility of using CD166 expression for isolating SAN progenitors from differentiating embryonic stem cells. METHODS AND RESULTS We found that in embryonic day 10.5 mouse hearts, CD166 and HCN4, markers of the pacemaker tissue, are coexpressed. Sorting embryonic stem cells for CD166 expression at differentiation day 8 selects a population of pacemaker precursors. CD166+ cells express high levels of genes involved in SAN development (Tbx18, Tbx3, Isl-1, Shox2) and function (Cx30.2, HCN4, HCN1, CaV1.3) and low levels of ventricular genes (Cx43, Kv4.2, HCN2, Nkx2.5). In culture, CD166+ cells form an autorhythmic syncytium composed of cells morphologically similar to and with the electrophysiological properties of murine SAN myocytes. Isoproterenol increases (+57%) and acetylcholine decreases (-23%) the beating rate of CD166-selected cells, which express the β-adrenergic and muscarinic receptors. In cocultures, CD166-selected cells are able to pace neonatal ventricular myocytes at a rate faster than their own. Furthermore, CD166+ cells have lost pluripotency genes and do not form teratomas in vivo. CONCLUSIONS We demonstrated for the first time the isolation of a nonteratogenic population of cardiac precursors able to mature and form a fully functional SAN-like tissue.
Collapse
Affiliation(s)
- Angela Scavone
- Department of Biosciences, Università degli Studi di Milano, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Rog-Zielinska EA, Thomson A, Kenyon CJ, Brownstein DG, Moran CM, Szumska D, Michailidou Z, Richardson J, Owen E, Watt A, Morrison H, Forrester LM, Bhattacharya S, Holmes MC, Chapman KE. Glucocorticoid receptor is required for foetal heart maturation. Hum Mol Genet 2013; 22:3269-82. [PMID: 23595884 DOI: 10.1093/hmg/ddt182] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoids are vital for the structural and functional maturation of foetal organs, yet excessive foetal exposure is detrimental to adult cardiovascular health. To elucidate the role of glucocorticoid signalling in late-gestation cardiovascular maturation, we have generated mice with conditional disruption of glucocorticoid receptor (GR) in cardiomyocytes and vascular smooth muscle cells using smooth muscle protein 22-driven Cre recombinase (SMGRKO mice) and compared them with mice with global deficiency in GR (GR(-/-)). Echocardiography shows impaired heart function in both SMGRKO and GR(-/-) mice at embryonic day (E)17.5, associated with generalized oedema. Cardiac ultrastructure is markedly disrupted in both SMGRKO and GR(-/-) mice at E17.5, with short, disorganized myofibrils and cardiomyocytes that fail to align in the compact myocardium. Failure to induce critical genes involved in contractile function, calcium handling and energy metabolism underpins this common phenotype. However, although hearts of GR(-/-) mice are smaller, with 22% reduced ventricular volume at E17.5, SMGRKO hearts are normally sized. Moreover, while levels of mRNA encoding atrial natriuretic peptide are reduced in E17.5 GR(-/-) hearts, they are normal in foetal SMGRKO hearts. These data demonstrate that structural, functional and biochemical maturation of the foetal heart is dependent on glucocorticoid signalling within cardiomyocytes and vascular smooth muscle, though some aspects of heart maturation (size, ANP expression) are independent of GR at these key sites.
Collapse
Affiliation(s)
- Eva A Rog-Zielinska
- Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh EH16 4TJ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Willis MS, Dyer LA, Ren R, Lockyer P, Moreno-Miralles I, Schisler JC, Patterson C. BMPER regulates cardiomyocyte size and vessel density in vivo. Cardiovasc Pathol 2012. [PMID: 23200275 DOI: 10.1016/j.carpath.2012.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND BMPER, an orthologue of Drosophila melanogaster Crossveinless-2, is a secreted factor that regulates bone morphogenetic protein activity in endothelial cell precursors and during early cardiomyocyte differentiation. Although previously described in the heart, the role of BMPER in cardiac development and function remain unknown. METHODS BMPER-deficient hearts were phenotyped histologically and functionally using echocardiography and Doppler analysis. Since BMPER -/- mice die perinatally, adult BMPER +/- mice were challenged to pressure-overload-induced cardiac hypertrophy and hindlimb ischemia to determine changes in angiogenesis and regulation of cardiomyocyte size. RESULTS We identify for the first time the cardiac phenotype associated with BMPER haploinsufficiency. BMPER messenger RNA and protein are present in the heart during cardiac development through at least E14.5 but is lost by E18.5. BMPER +/- ventricles are thinner and less compact than sibling wild-type hearts. In the adult, BMPER +/- hearts present with decreased anterior and posterior wall thickness, decreased cardiomyocyte size and an increase in cardiac vessel density. Despite these changes, BMPER +/- mice respond to pressure-overload-induced cardiac hypertrophy challenge largely to the same extent as wild-type mice. CONCLUSION BMPER appears to play a role in regulating both vessel density and cardiac development in vivo; however, BMPER haploinsufficiency does not result in marked effects on cardiac function or adaptation to pressure overload hypertrophy.
Collapse
Affiliation(s)
- Monte S Willis
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Histological and ultrastructural abnormalities in murine desmoglein 2-mutant hearts. Cell Tissue Res 2012; 348:249-59. [PMID: 22293975 DOI: 10.1007/s00441-011-1322-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 12/16/2011] [Indexed: 12/21/2022]
Abstract
Mice carrying a deletion of the adhesive extracellular domain of the desmosomal cadherin desmoglein 2 develop an arrhythmogenic right ventricular cardiomyopathylike phenotype with ventricular dilation, fibrosis and arrhythmia. To unravel the sequence of myocardial alterations and to identify potential pathomechanisms, histological analyses were performed on mutant hearts from the juvenile to the adult state, i.e., between 2 and 13 weeks. At an age of 2 weeks 30% of mutants presented lesions,which were visible as white plaques on the heart surface or in the septum. From 4 weeks onwards, all mutants displayed a cardiac phenotype. Dying cardiomyocytes with calcification were found in lesions of all ages. But lesions of young mutant animals contained high amounts of CD45+ immune cells and little collagen fibers, whereas lesions of the older animals were collagen-rich and harbored only a small but still significantly increased number of CD45+ cells. Electron microscopy further showed that distinct desmosomes cannot be distinguished in intercalated discs of mutant hearts. Widening of the intercellular cleft and even complete dissociation of intercalated discs were often observed close to lesions. Disturbed sarcomer structure, altered Z-discs, multiple autophagic vacuoles and swollen mitochondria were other prominent pathological features. Taken together, the following scenario is suggested: mutant desmoglein 2 cannot fully support the increased mechanical requirements placed on intercalated disc adhesion during postnatal heart development, resulting in compromised adhesion and cell stress. This induces cardiomyocyte death, aseptic inflammation and fibrotic replacement. The acute stage of scar formation is followed by permanent impairment of the cardiac function.
Collapse
|
40
|
Garcia AG, Wilson RM, Heo J, Murthy NR, Baid S, Ouchi N, Sam F. Interferon-γ ablation exacerbates myocardial hypertrophy in diastolic heart failure. Am J Physiol Heart Circ Physiol 2012; 303:H587-96. [PMID: 22730392 DOI: 10.1152/ajpheart.00298.2012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Diastolic heart failure (HF) accounts for up to 50% of all HF admissions, with hypertension being the major cause of diastolic HF. Hypertension is characterized by left ventricular (LV) hypertrophy (LVH). Proinflammatory cytokines are increased in LVH and hypertension, but it is unknown if they mediate the progression of hypertension-induced diastolic HF. We sought to determine if interferon-γ (IFNγ) plays a role in mediating the transition from hypertension-induced LVH to diastolic HF. Twelve-week old BALB/c (WT) and IFNγ-deficient (IFNγKO) mice underwent either saline (n = 12) or aldosterone (n = 16) infusion, uninephrectomy, and fed 1% salt water for 4 wk. Tail-cuff blood pressure, echocardiography, and gene/protein analyses were performed. Isolated adult rat ventricular myocytes were treated with IFNγ (250 U/ml) and/or aldosterone (1 μM). Hypertension was less marked in IFNγKO-aldosterone mice than in WT-aldosterone mice (127 ± 5 vs. 136 ± 4 mmHg; P < 0.01), despite more LVH (LV/body wt ratio: 4.9 ± 0.1 vs. 4.3 ± 0.1 mg/g) and worse diastolic dysfunction (peak early-to-late mitral inflow velocity ratio: 3.1 ± 0.1 vs. 2.8 ± 0.1). LV ejection fraction was no different between IFNγKO-aldosterone vs. WT-aldosterone mice. LV end systolic dimensions were decreased significantly in IFNγKO-aldosterone vs. WT-aldosterone hearts (1.12 ± 0.1 vs. 2.1 ± 0.3 mm). Myocardial fibrosis and collagen expression were increased in both IFNγKO-aldosterone and WT-aldosterone hearts. Myocardial autophagy was greater in IFNγKO-aldosterone than WT-aldosterone mice. Conversely, tumor necrosis factor-α and interleukin-10 expressions were increased only in WT-aldosterone hearts. Recombinant IFNγ attenuated cardiac hypertrophy in vivo and modulated aldosterone-induced hypertrophy and autophagy in cultured cardiomyocytes. Thus IFNγ is a regulator of cardiac hypertrophy in diastolic HF and modulates cardiomyocyte size possibly by regulating autophagy. These findings suggest that IFNγ may mediate adaptive downstream responses and challenge the concept that inflammatory cytokines mediate only adverse effects.
Collapse
Affiliation(s)
- Anthony G Garcia
- Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Gregg CL, Butcher JT. Quantitative in vivo imaging of embryonic development: opportunities and challenges. Differentiation 2012; 84:149-62. [PMID: 22695188 DOI: 10.1016/j.diff.2012.05.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 05/03/2012] [Accepted: 05/04/2012] [Indexed: 10/28/2022]
Abstract
Animal models are critically important for a mechanistic understanding of embryonic morphogenesis. For decades, visualizing these rapid and complex multidimensional events has relied on projection images and thin section reconstructions. While much insight has been gained, fixed tissue specimens offer limited information on dynamic processes that are essential for tissue assembly and organ patterning. Quantitative imaging is required to unlock the important basic science and clinically relevant secrets that remain hidden. Recent advances in live imaging technology have enabled quantitative longitudinal analysis of embryonic morphogenesis at multiple length and time scales. Four different imaging modalities are currently being used to monitor embryonic morphogenesis: optical, ultrasound, magnetic resonance imaging (MRI), and micro-computed tomography (micro-CT). Each has its advantages and limitations with respect to spatial resolution, depth of field, scanning speed, and tissue contrast. In addition, new processing tools have been developed to enhance live imaging capabilities. In this review, we analyze each type of imaging source and its use in quantitative study of embryonic morphogenesis in small animal models. We describe the physics behind their function, identify some examples in which the modality has revealed new quantitative insights, and then conclude with a discussion of new research directions with live imaging.
Collapse
Affiliation(s)
- Chelsea L Gregg
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
42
|
Wu J, You J, Li L, Ma H, Jia J, Jiang G, Chen Z, Ye Y, Gong H, Bu L, Ge J, Zou Y. Early estimation of left ventricular systolic pressure and prediction of successful aortic constriction in a mouse model of pressure overload by ultrasound biomicroscopy. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:1030-1039. [PMID: 22425378 DOI: 10.1016/j.ultrasmedbio.2012.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 12/29/2011] [Accepted: 01/22/2012] [Indexed: 05/31/2023]
Abstract
Elevation of left ventricular end-systolic pressure (LVESP) and hypertrophic response in mice varies after transverse aorta constriction (TAC). Micromanometric catheterization, conventionally used to select mice with successful TAC, is invasive and nonreusable. We aimed to establish noninvasive imaging protocols for early estimation of successful TAC by ultrasound biomicroscopy (UBM). Out of 55 C57BL/6J mice, we randomly selected 45 as TAC group and 10 as controls. UMB was performed before TAC and, at day 3 and day 14, after TAC. In all mice, LVESP was measured with a Millar conductance catheter at day 14. With LVESP ≥ 150 mm Hg set as indicator of successful TAC (TAC+) and LVESP < 150 mm Hg as unsuccessful (TAC-), receiver operating characteristic curve analysis demonstrated that postoperative inner diameter at aortic banding site (IDb), peak flow velocity at aortic banding site (PVb) and peak flow velocity of right/left common carotid artery (PVr/l) at day 3 served as most effective predictors for LVESP at day 14 (area under curve = 0.9016, 0.9143, 0.8254, respectively. p < 0.01 for all). Among all UBM parameters at day 3, IDb, PVb, right common carotid artery peak flow velocity (PVr) and PVr/l correlated best with LVESP at day 14 (R(2) = 0.5740, 0.6549, 0.5208, 0.2274, respectively. p < 0.01 for all). Furthermore, IDb, PVb, and PVr/l at day 3 most effectively predict long-term cardiac hypertrophy, using the cut-off values of 0.45 mm, 2698.00 mm/s, 3.08, respectively. UBM can be a noninvasive and effective option for early prediction of successful TAC.
Collapse
Affiliation(s)
- Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Application of echocardiography on transgenic mice with cardiomyopathies. Biochem Res Int 2012; 2012:715197. [PMID: 22675635 PMCID: PMC3363992 DOI: 10.1155/2012/715197] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/12/2012] [Indexed: 11/30/2022] Open
Abstract
Cardiomyopathies are common cardiac disorders that primarily affect cardiac muscle resulting in cardiac dysfunction and heart failure. Transgenic mouse disease models have been developed to investigate the cellular mechanisms underlying heart failure and sudden cardiac death observed in cardiomyopathy cases and to explore the therapeutic outcomes in experimental animals in vivo. Echocardiography is an essential diagnostic tool for accurate and noninvasive assessment of cardiac structure and function in experimental animals. Our laboratory has been among the first to apply high-frequency research echocardiography on transgenic mice with cardiomyopathies. In this work, we have summarized our and other studies on assessment of systolic and diastolic dysfunction using conventional echocardiography, pulsed Doppler, and tissue Doppler imaging in transgenic mice with various cardiomyopathies. Estimation of embryonic mouse hearts has been performed as well using this high-resolution echocardiography. Some technical considerations in mouse echocardiography have also been discussed.
Collapse
|
44
|
Nieman BJ, Wong MD, Henkelman RM. Genes into geometry: imaging for mouse development in 3D. Curr Opin Genet Dev 2011; 21:638-46. [PMID: 21907568 DOI: 10.1016/j.gde.2011.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/19/2011] [Accepted: 08/23/2011] [Indexed: 02/07/2023]
Abstract
Mammalian development is a sophisticated program coordinated by a complex set of genetic and physiological factors. Alterations in anatomy or morphology provide intrinsic measures of progress in or deviations from this program. Emerging three-dimensional imaging methods now allow for more sophisticated morphological assessment than ever before, enabling comprehensive phenotyping, visualization of anatomical context and patterns, automated and quantitative morphological analysis, as well as improved understanding of the developmental time course. Furthermore, these imaging tools are becoming increasingly available and will consequently play a prominent role in elucidating the factors that direct and influence mammalian development.
Collapse
Affiliation(s)
- Brian J Nieman
- Mouse Imaging Centre, Hospital for Sick Children, and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
45
|
van Raaij ME, Lindvere L, Dorr A, He J, Sahota B, Foster FS, Stefanovic B. Functional micro-ultrasound imaging of rodent cerebral hemodynamics. Neuroimage 2011; 58:100-8. [PMID: 21704715 DOI: 10.1016/j.neuroimage.2011.05.088] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/11/2011] [Accepted: 05/23/2011] [Indexed: 11/16/2022] Open
Abstract
Healthy cerebral microcirculation is crucial to neuronal functioning. We present a new method to investigate microvascular hemodynamics in living rodent brain through a focal cranial window based on high-frequency ultrasound imaging. The method has a temporal resolution of 40ms, and a 100μm in-plane and 600μm through-plane spatial resolution. We use a commercially available high-frequency ultrasound imaging system to quantify changes in the relative cerebral blood volume (CBV) by measuring the scattered signal intensity from an ultrasound contrast agent circulating in the vasculature. Generalized linear model analysis is then used to produce effect size and significance maps of changes in cerebral blood volume upon electrical stimulation of the forepaw. We observe larger CBV increases in the forelimb representation of the primary somatosensory cortex than in the deep gray matter with stimuli as short as 2s (5.1 ± 1.3% vs. 3.3 ± 0.6%). We also investigate the temporal evolution of the blood volume changes in cortical and subcortical gray matter, pial vessels and subcortical major vessels, and show shorter response onset times in the parenchymal regions than in the neighboring large vessels (1.6 ± 1.0s vs. 2.6 ± 1.3s in the cortex for a 10 second stimulus protocol). This method, which we termed functional micro-ultrasound imaging or fMUS, is a novel, highly accessible, and cost-effective way of imaging rodent brain microvascular topology and hemodynamics in vivo at 100micron resolution over a 1-by-1cm field of view with 10s-100s frames per second that opens up a new set of questions regarding brain function in preclinical models of health and disease.
Collapse
Affiliation(s)
- Martijn E van Raaij
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
46
|
Foster FS, Hossack J, Adamson SL. Micro-ultrasound for preclinical imaging. Interface Focus 2011; 1:576-601. [PMID: 22866232 DOI: 10.1098/rsfs.2011.0037] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 05/13/2011] [Indexed: 12/19/2022] Open
Abstract
Over the past decade, non-invasive preclinical imaging has emerged as an important tool to facilitate biomedical discovery. Not only have the markets for these tools accelerated, but the numbers of peer-reviewed papers in which imaging end points and biomarkers have been used have grown dramatically. High frequency 'micro-ultrasound' has steadily evolved in the post-genomic era as a rapid, comparatively inexpensive imaging tool for studying normal development and models of human disease in small animals. One of the fundamental barriers to this development was the technological hurdle associated with high-frequency array transducers. Recently, new approaches have enabled the upper limits of linear and phased arrays to be pushed from about 20 to over 50 MHz enabling a broad range of new applications. The innovations leading to the new transducer technology and scanner architecture are reviewed. Applications of preclinical micro-ultrasound are explored for developmental biology, cancer, and cardiovascular disease. With respect to the future, the latest developments in high-frequency ultrasound imaging are described.
Collapse
Affiliation(s)
- F Stuart Foster
- Sunnybrook and Health Sciences Centre , University of Toronto , Toronto, Ontario , Canada
| | | | | |
Collapse
|
47
|
Wang Q, Lin JLC, Reinking BE, Feng HZ, Chan FC, Lin CI, Jin JP, Gustafson-Wagner EA, Scholz TD, Yang B, Lin JJC. Essential roles of an intercalated disc protein, mXinbeta, in postnatal heart growth and survival. Circ Res 2010; 106:1468-78. [PMID: 20360251 DOI: 10.1161/circresaha.109.212787] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE The Xin repeat-containing proteins mXinalpha and mXinbeta localize to the intercalated disc of mouse heart and are implicated in cardiac development and function. The mXinalpha directly interacts with beta-catenin, p120-catenin, and actin filaments. Ablation of mXinalpha results in adult late-onset cardiomyopathy with conduction defects. An upregulation of the mXinbeta in mXinalpha-deficient hearts suggests a partial compensation. OBJECTIVE The essential roles of mXinbeta in cardiac development and intercalated disc maturation were investigated. METHODS AND RESULTS Ablation of mXinbeta led to abnormal heart shape, ventricular septal defects, severe growth retardation, and postnatal lethality with no upregulation of the mXinalpha. Postnatal upregulation of mXinbeta in wild-type hearts, as well as altered apoptosis and proliferation in mXinbeta-null hearts, suggests that mXinbeta is required for postnatal heart remodeling. The mXinbeta-null hearts exhibited a misorganized myocardium as detected by histological and electron microscopic studies and an impaired diastolic function, as suggested by echocardiography and a delay in switching off the slow skeletal troponin I. Loss of mXinbeta resulted in the failure of forming mature intercalated discs and the mislocalization of mXinalpha and N-cadherin. The mXinbeta-null hearts showed upregulation of active Stat3 (signal transducer and activator of transcription 3) and downregulation of the activities of Rac1, insulin-like growth factor 1 receptor, protein kinase B, and extracellular signal-regulated kinases 1 and 2. CONCLUSIONS These findings identify not only an essential role of mXinbeta in the intercalated disc maturation but also mechanisms of mXinbeta modulating N-cadherin-mediated adhesion signaling and its crosstalk signaling for postnatal heart growth and animal survival.
Collapse
Affiliation(s)
- Qinchuan Wang
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sam F, Duhaney TAS, Sato K, Wilson RM, Ohashi K, Sono-Romanelli S, Higuchi A, De Silva DS, Qin F, Walsh K, Ouchi N. Adiponectin deficiency, diastolic dysfunction, and diastolic heart failure. Endocrinology 2010; 151:322-31. [PMID: 19850745 PMCID: PMC2803144 DOI: 10.1210/en.2009-0806] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aldosterone infusion results in left ventricular hypertrophy (LVH) and hypertension and may involve profibrotic and proinflammatory mechanisms. In turn, hypertension is the major cause of diastolic heart failure (HF). Adiponectin, an adipose-derived plasma protein, exerts antiinflammatory and anti-hypertrophic effects and is implicated in the development of hypertension and systolic HF. We thus tested the hypothesis that hypoadiponectinemia in aldosterone-induced hypertension exacerbated cardiac remodeling and diastolic HF. Wild-type (WT) or adiponectin-deficient (APNKO) mice underwent saline or aldosterone infusion and uninephrectomy and were fed 1% salt water for 4 wk. Blood pressure was increased in aldosterone-infused WT (132 +/- 2 vs. 109 +/- 3 mm Hg; P < 0.01) and further augmented in APNKO mice (140 +/- 3 mm Hg; P < 0.05 vs. aldosterone-infused WT). LVH was increased in aldosterone-infused WT vs. WT mice (LV/body weight ratio, 4.8 +/- 0.2 vs. 4.1 +/- 0.2 mg/g) and further increased in aldosterone-infused APNKO mice (LV/body weight ratio, 6.0 +/- 0.4 mg/g). Left ventricular ejection fraction was not decreased in either aldosterone-infused WT or APNKO hearts. Pulmonary congestion however was worse in APNKO mice (P < 0.01). The ratio of early ventricular filling over late ventricular filling (E/A) and the ratio of mitral peak velocity of early filling to early diastolic mitral annular velocity (E/e'), measures of diastolic function, were increased in aldosterone-infused WT hearts and further increased in APNKO hearts (P < 0.05 for both). Renal function and cardiac fibrosis were no different between both aldosterone-infused groups. Aldosterone increased matrix metalloproteinase-2 expression in WT hearts (P < 0.05 vs. WT and P < 0.01 vs. APNKO). Myocardial atrial natriuretic peptide, interferon-gamma, and TNF-alpha expression were increased in aldosterone-infused WT hearts. Expression of these proteins was further increased in aldosterone-infused APNKO hearts. Therefore, hypoadiponectinemia in hypertension-induced diastolic HF exacerbates LVH, diastolic dysfunction, and diastolic HF. Whether or not adiponectin replacement prevents the progression to diastolic HF will warrant further study.
Collapse
Affiliation(s)
- Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, Boston, Massachusetts 02118, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vongvatcharanon U, Udomuksorn W, Vongvatcharanon S, Sobhon P. Age-related changes in parvalbumin in the heart of female rats. Acta Histochem 2010; 112:96-100. [PMID: 18977019 DOI: 10.1016/j.acthis.2008.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Revised: 07/22/2008] [Accepted: 07/24/2008] [Indexed: 10/21/2022]
Abstract
Changes of parvalbumin protein levels and immunolocalisation during the postnatal development of the female rat heart were investigated in order to determine if they were correlated with age-related changes in cardiac function. Hearts from newborn, 3-month-old (young), 6-month-old (young adult) and 12-month-old (adult) female Wistar rats were processed for immunohistochemical localization of parvalbumin and for Western blotting assay. Parvalbumin was detected by both methods in all age groups from newborn to 12-month-old rats. In the newborn rat heart, parvalbumin immunoreactivity did not fully fill the sarcoplasm of the cardiac myocytes and the amount of parvalbumin was low compared to the adult levels. In contrast, in 3-12-month-old rats, strong parvalbumin immunoreactivity was detected throughout the sarcoplasm of all cardiac myocytes and the amount of parvalbumin increased with increasing age (from newborn to adult). Our study indicates that an increase of parvalbumin levels in the female rat heart with increasing age may be associated with maintenance of proper relaxation of the cardiac myocytes needed to cope with the increasing workload of the heart during postnatal growth.
Collapse
|
50
|
Abstract
Ultrasound biomicroscopy (UBM) and magnetic resonance microimaging (micro-MRI) provide noninvasive, high-resolution images in mouse embryos and neonates, enabling volumetric and functional analyses of phenotypes, including longitudinal imaging of individual mice over critical stages of in utero and early-postnatal development. In this chapter, we describe the underlying principles of UBM and micro-MRI, including the advantages and limitations of these approaches for studies of mouse development, and providing a number of examples to illustrate their use. To date, most imaging studies have focused on the developing nervous and cardiovascular systems, which are also reflected in the examples shown in this chapter, but we also discuss the future application of these methods to other organ systems.
Collapse
Affiliation(s)
- Brian J Nieman
- Mouse Imaging Centre, Hospital for Sick Children, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | |
Collapse
|