1
|
Panico C, Felicetta A, Kunderfranco P, Cremonesi M, Salvarani N, Carullo P, Colombo F, Idini A, Passaretti M, Doro R, Rubino M, Villaschi A, Da Rin G, Peano C, Kallikourdis M, Greco CM, Condorelli G. Single-Cell RNA Sequencing Reveals Metabolic Stress-Dependent Activation of Cardiac Macrophages in a Model of Dyslipidemia-Induced Diastolic Dysfunction. Circulation 2024; 150:1517-1532. [PMID: 38126199 DOI: 10.1161/circulationaha.122.062984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Metabolic distress is often associated with heart failure with preserved ejection fraction (HFpEF) and represents a therapeutic challenge. Metabolism-induced systemic inflammation links comorbidities with HFpEF. How metabolic changes affect myocardial inflammation in the context of HFpEF is not known. METHODS We found that ApoE knockout mice fed a Western diet recapitulate many features of HFpEF. Single-cell RNA sequencing was used for expression analysis of CD45+ cardiac cells to evaluate the involvement of inflammation in diastolic dysfunction. We focused bioinformatics analysis on macrophages, obtaining high-resolution identification of subsets of these cells in the heart, enabling us to study the outcomes of metabolic distress on the cardiac macrophage infiltrate and to identify a macrophage-to-cardiomyocyte regulatory axis. To test whether a clinically relevant sodium glucose cotransporter-2 inhibitor could ameliorate the cardiac immune infiltrate profile in our model, mice were randomized to receive the sodium glucose cotransporter-2 inhibitor dapagliflozin or vehicle for 8 weeks. RESULTS ApoE knockout mice fed a Western diet presented with reduced diastolic function, reduced exercise tolerance, and increased pulmonary congestion associated with cardiac lipid overload and reduced polyunsaturated fatty acids. The main immune cell types infiltrating the heart included 4 subpopulations of resident and monocyte-derived macrophages, determining a proinflammatory profile exclusively in ApoE knockout-Western diet mice. Lipid overload had a direct effect on inflammatory gene activation in macrophages, mediated through endoplasmic reticulum stress pathways. Investigation of the macrophage-to-cardiomyocyte regulatory axis revealed the potential effects on cardiomyocytes of multiple inflammatory cytokines secreted by macrophages, affecting pathways such as hypertrophy, fibrosis, and autophagy. Finally, we describe an anti-inflammatory effect of sodium glucose cotransporter-2 inhibition in this model. CONCLUSIONS Using single-cell RNA sequencing in a model of diastolic dysfunction driven by hyperlipidemia, we have determined the effects of metabolic distress on cardiac inflammatory cells, in particular on macrophages, and suggest sodium glucose cotransporter-2 inhibitors as potential therapeutic agents for the targeting of a specific phenotype of HFpEF.
Collapse
Affiliation(s)
- Cristina Panico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Arianna Felicetta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Paolo Kunderfranco
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Marco Cremonesi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Nicolò Salvarani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Pierluigi Carullo
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Federico Colombo
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Alessandra Idini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Mauro Passaretti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Riccardo Doro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Marcello Rubino
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Alessandro Villaschi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Giorgio Da Rin
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Carolina M Greco
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| |
Collapse
|
2
|
Kass DA. Assessing and interpreting diastolic function in animal models of heart disease. J Mol Cell Cardiol 2024; 197:1-4. [PMID: 39368650 DOI: 10.1016/j.yjmcc.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Increasing interest in identifying the causes of and treatments for heart failure with preserved ejection fraction and cardiac fibrosis has spawned a focus on measures of cardiac diastolic function. The methods, their underlying principals and mechanics, and caveats to their measurement were largely worked out decades ago, but some of this seems a bit forgotten as scientists working in the field now have backgrounds more in molecular and cellular biology. This perspective was spawned by seeing the growing number of studies where diastolic function analysis is a key parameter used to justify a given pre-clinical model or to show the consequences of a particular genetic or pharmacological therapy. The goals are to discuss what comprises and influences diastolic function, how it is measured, what the parameters mean and what their limitations are, and what comprises evidence for pathophysiologically meaningful diastolic dysfunction.
Collapse
Affiliation(s)
- David A Kass
- Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Durumutla HB, Prabakaran AD, Soussi FEA, Akinborewa O, Latimer H, McFarland K, Piczer K, Werbrich C, Jain MK, Haldar SM, Quattrocelli M. Glucocorticoid chrono-pharmacology unveils novel targets for the cardiomyocyte-specific GR-KLF15 axis in cardiac glucose metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572210. [PMID: 38187555 PMCID: PMC10769285 DOI: 10.1101/2023.12.18.572210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Circadian time-of-intake gates the cardioprotective effects of glucocorticoid administration in both healthy and infarcted hearts. The cardiomyocyte-specific glucocorticoid receptor (GR) and its co-factor, Krüppel-like factor (Klf15), play critical roles in maintaining normal heart function in the long-term and serve as pleiotropic regulators of cardiac metabolism. Despite this understanding, the cardiomyocyte-autonomous metabolic targets influenced by the concerted epigenetic action of GR-Klf15 axis remain undefined. Here, we demonstrate the critical roles of the cardiomyocyte-specific GR and Klf15 in orchestrating a circadian-dependent glucose oxidation program within the heart. Combining integrated transcriptomics and epigenomics with cardiomyocyte-specific inducible ablation of GR or Klf15, we identified their synergistic role in the activation of adiponectin receptor expression (Adipor1) and the mitochondrial pyruvate complex (Mpc1/2), thereby enhancing insulin-stimulated glucose uptake and pyruvate oxidation. Furthermore, in obese diabetic (db/db) mice exhibiting insulin resistance and impaired glucose oxidation, light-phase prednisone administration, as opposed to dark-phase prednisone dosing, effectively restored cardiomyocyte glucose oxidation and improved diastolic function towards control-like levels in a sex-independent manner. Collectively, our findings uncover novel cardiomyocyte-autonomous metabolic targets of the GR-Klf15 axis. This study highlights the circadian-dependent cardioprotective effects of glucocorticoids on cardiomyocyte glucose metabolism, providing critical insights into chrono-pharmacological strategies for glucocorticoid therapy in cardiovascular disease.
Collapse
Affiliation(s)
- Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ashok Daniel Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Olukunle Akinborewa
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mukesh K Jain
- Dept Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Saptarsi M Haldar
- Amgen Research, South San Francisco, CA, USA; Gladstone Institutes, San Francisco, CA, USA and Dept Medicine, Cardiology Division, UCSF, San Francisco, CA, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
4
|
Silva J, Azevedo T, Ginja M, Oliveira PA, Duarte JA, Faustino-Rocha AI. Realistic Aspects of Cardiac Ultrasound in Rats: Practical Tips for Improved Examination. J Imaging 2024; 10:219. [PMID: 39330439 PMCID: PMC11433567 DOI: 10.3390/jimaging10090219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Echocardiography is a reliable and non-invasive method for assessing cardiac structure and function in both clinical and experimental settings, offering valuable insights into disease progression and treatment efficacy. The successful application of echocardiography in murine models of disease has enabled the evaluation of disease severity, drug testing, and continuous monitoring of cardiac function in these animals. However, there is insufficient standardization of echocardiographic measurements for smaller animals. This article aims to address this gap by providing a guide and practical tips for the appropriate acquisition and analysis of echocardiographic parameters in adult rats, which may also be applicable in other small rodents used for scientific purposes, like mice. With advancements in technology, such as ultrahigh-frequency ultrasonic transducers, echocardiography has become a highly sophisticated imaging modality, offering high temporal and spatial resolution imaging, thereby allowing for real-time monitoring of cardiac function throughout the lifespan of small animals. Moreover, it allows the assessment of cardiac complications associated with aging, cancer, diabetes, and obesity, as well as the monitoring of cardiotoxicity induced by therapeutic interventions in preclinical models, providing important information for translational research. Finally, this paper discusses the future directions of cardiac preclinical ultrasound, highlighting the need for continued standardization to advance research and improve clinical outcomes to facilitate early disease detection and the translation of findings into clinical practice.
Collapse
Affiliation(s)
- Jessica Silva
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
| | - Tiago Azevedo
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Centro de Investigação de Montanha (CIMO), Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Mário Ginja
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - José Alberto Duarte
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences (IUCS), Advanced Polytechnic and University Cooperative (CESPU), 4585-116 Gandra, Portugal;
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory (1H-TOXRUN), University Institute of Health Sciences (IUCS), Advanced Polytechnic and University Cooperative (CESPU), 4585-116 Gandra, Portugal
| | - Ana I. Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal; (J.S.); (T.A.); (M.G.); (P.A.O.)
- Department of Zootechnics, School of Sciences and Technology, University of Évora, 7004-516 Évora, Portugal
- Comprehensive Health Research Center (CHRC), University of Évora, 7004-516 Évora, Portugal
| |
Collapse
|
5
|
Fiamingo M, Bailey A, Toler S, Lee K, Oshiro W, Yoo B, Krantz T, Evansky P, Davies D, Gilmour MI, Farraj A, Jaspers I, Hazari MS. Enriched housing differentially alters allostatic load and cardiopulmonary responses to wildfire-related smoke in male and female mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:561-578. [PMID: 38721998 PMCID: PMC11167957 DOI: 10.1080/15287394.2024.2346582] [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] [Indexed: 05/14/2024]
Abstract
Living conditions are an important modifier of individual health outcomes and may lead to higher allostatic load (AL). However, housing-induced cardiovascular and immune effects contributing to altered environmental responsiveness remain understudied. This investigation was conducted to examine the influence of enriched (EH) versus depleted housing (DH) conditions on cardiopulmonary functions, systemic immune responses, and allostatic load in response to a single wildfire smoke (WS) exposure in mice. Male and female C57BL/6J mice were divided into EH or DH for 22 weeks, and cardiopulmonary assessments measured before and after exposures to either one-hr filtered air (FA) or flaming eucalyptus WS exposure. Male and female DH mice exhibited increased heart rate (HR) and left ventricular mass (LVM), as well as reduced stroke volume and end diastolic volume (EDV) one week following exposure to WS. Female DH mice displayed significantly elevated levels of IL-2, IL-17, corticosterone and hemoglobin A1c (HbA1c) following WS, while female in EH mice higher epinephrine levels were detected. Female mice exhibited higher AL than males with DH, which was potentiated post-WS exposure. Thus, DH increased susceptibility to extreme air pollution in a gender-dependent manner suggesting that living conditions need to be evaluated as a modifier of toxicological responses.
Collapse
Affiliation(s)
- Michelle Fiamingo
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina – Chapel Hill, Chapel Hill, NC 27599
| | - Aleah Bailey
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina – Chapel Hill, Chapel Hill, NC 27599
| | - Sydnie Toler
- Gillings School of Global Public Health and School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaleb Lee
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830
| | - Wendy Oshiro
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Brendan Yoo
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Todd Krantz
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Paul Evansky
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - David Davies
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - M. Ian Gilmour
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Aimen Farraj
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| | - Ilona Jaspers
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina – Chapel Hill, Chapel Hill, NC 27599
| | - Mehdi S. Hazari
- Public Health Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Research Triangle Park, NC 27711
| |
Collapse
|
6
|
Axelsen JS, Andersen S, Ringgaard S, Smal R, Lluciá-Valldeperas A, Nielsen-Kudsk JE, de Man FS, Andersen A. Right ventricular diastolic adaptation to pressure overload in different rat strains. Physiol Rep 2024; 12:e16132. [PMID: 38993022 PMCID: PMC11239975 DOI: 10.14814/phy2.16132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Different rat strains are used in various animal models of pulmonary hypertension and right ventricular (RV) failure. No systematic assessment has been made to test differences in RV response to pressure overload between rat strains. We compared RV adaptation to pulmonary trunk banding (PTB) in Wistar (W), Sprague Dawley (SD), and Fischer344 (F) rats by hemodynamic profiling focusing on diastolic function. Age-matched male rat weanlings were randomized to sham surgery (W-sham, n = 5; SD-sham, n = 4; F-sham, n = 4) or PTB (W-PTB, n = 8; SD-PTB, n = 8; F-PTB, n = 8). RV function was evaluated after 5 weeks by echocardiography, cardiac MRI, and invasive pressure-volume measurements. PTB caused RV failure and increased RV systolic pressures four-fold in all three PTB groups compared with sham. W- and SD-PTB had a 2.4-fold increase in RV end-systolic volume index compared with sham, while F-PTB rats were less affected. Diastolic and right atrial impairment were evident by increased RV end-diastolic elastance, filling pressure, and E/e' in PTB rats compared with sham, again F-PTB the least affected. In conclusions, PTB caused RV failure with signs of diastolic dysfunction. Despite a similar increase in RV systolic pressure, F-PTB rats showed less RV dilatation and a more preserved diastolic function compared with W- and SD-PTB.
Collapse
Affiliation(s)
- Julie S Axelsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Stine Andersen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Rowan Smal
- Department of Pulmonary Medicine, PHEniX Laboratory, Amsterdam UMC, Locatie VUmc, Amsterdam, The Netherlands
| | - Aida Lluciá-Valldeperas
- Department of Pulmonary Medicine, PHEniX Laboratory, Amsterdam UMC, Locatie VUmc, Amsterdam, The Netherlands
| | - Jens Erik Nielsen-Kudsk
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Frances S de Man
- Department of Pulmonary Medicine, PHEniX Laboratory, Amsterdam UMC, Locatie VUmc, Amsterdam, The Netherlands
| | - Asger Andersen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
7
|
Berger JH, Shi Y, Matsuura TR, Batmanov K, Chen X, Tam K, Marshall M, Kue R, Patel J, Taing R, Callaway R, Griffin J, Kovacs A, Shanthappa DH, Miller R, Zhang BB, Roth Flach RJ, Kelly DP. Two-hit mouse model of heart failure with preserved ejection fraction combining diet-induced obesity and renin-mediated hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597821. [PMID: 38895483 PMCID: PMC11185718 DOI: 10.1101/2024.06.06.597821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasingly common but its pathogenesis is poorly understood. The ability to assess genetic and pharmacologic interventions is hampered by the lack of robust preclinical mouse models of HFpEF. We have developed a novel "2-hit" model, which combines obesity and insulin resistance with chronic pressure overload to recapitulate clinical features of HFpEF. C57BL6/NJ mice fed a high fat diet for >10 weeks were administered an AAV8-driven vector resulting in constitutive overexpression of mouse Renin1d . Control mice, HFD only, Renin only and HFD-Renin (aka "HFpEF") littermates underwent a battery of cardiac and extracardiac phenotyping. HFD-Renin mice demonstrated obesity and insulin resistance, a 2-3-fold increase in circulating renin levels that resulted in 30-40% increase in left ventricular hypertrophy, preserved systolic function, and diastolic dysfunction indicated by altered E/e', IVRT, and strain measurements; increased left atrial mass; elevated natriuretic peptides; and exercise intolerance. Transcriptomic and metabolomic profiling of HFD-Renin myocardium demonstrated upregulation of pro-fibrotic pathways and downregulation of metabolic pathways, in particular branched chain amino acid catabolism, similar to findings in human HFpEF. Treatment of these mice with the sodium-glucose cotransporter 2 inhibitor empagliflozin, an effective but incompletely understood HFpEF therapy, improved exercise tolerance, left heart enlargement, and insulin homeostasis. The HFD-Renin mouse model recapitulates key features of human HFpEF and will enable studies dissecting the contribution of individual pathogenic drivers to this complex syndrome. Addition of HFD-Renin mice to the preclinical HFpEF model platform allows for orthogonal studies to increase validity in assessment of interventions. NEW & NOTEWORTHY Heart failure with preserved ejection fraction (HFpEF) is a complex disease to study due to limited preclinical models. We rigorously characterize a new two-hit HFpEF mouse model, which allows for dissecting individual contributions and synergy of major pathogenic drivers, hypertension and diet-induced obesity. The results are consistent and reproducible in two independent laboratories. This high-fidelity pre-clinical model increases the available, orthogonal models needed to improve our understanding of the causes and assessment treatments for HFpEF.
Collapse
|
8
|
Koay YC, Liu RP, McIntosh B, Vigder N, Lauren S, Bai AY, Tomita S, Li D, Harney D, Hunter B, Zhang Y, Yang J, Bannon P, Philp A, Philp A, Kaye DM, Larance M, Lal S, O’Sullivan JF. The Efficacy of Risk Factor Modification Compared to NAD + Repletion in Diastolic Heart Failure. JACC Basic Transl Sci 2024; 9:733-750. [PMID: 39070276 PMCID: PMC11282886 DOI: 10.1016/j.jacbts.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 07/30/2024]
Abstract
Heart failure (HF) with left ventricular diastolic dysfunction is a growing global concern. This study evaluated myocardial oxidized nicotinamide adenine dinucleotide (NAD+) levels in human systolic and diastolic HF and in a murine model of HF with preserved ejection fraction, exploring NAD+ repletion as therapy. We quantified myocardial NAD+ and nicotinamide phosphoribosyltransferase levels, assessing restoration with nicotinamide riboside (NR). Findings show significant NAD+ and nicotinamide phosphoribosyltransferase depletion in human diastolic HF myocardium, but NR successfully restored NAD+ levels. In murine HF with preserved ejection fraction, NR as preventive and therapeutic intervention improved metabolic and antioxidant profiles. This study underscores NAD+ repletion's potential in diastolic HF management.
Collapse
Affiliation(s)
- Yen Chin Koay
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Ren Ping Liu
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Bailey McIntosh
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Niv Vigder
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Serlin Lauren
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela Yu Bai
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Saki Tomita
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Desmond Li
- BCAL Diagnostics, National Innovation Centre, Eveleigh, New South Wales, Australia
| | - Dylan Harney
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Hunter
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory, The University of Sydney, New South Wales, Australia
| | - Yunwei Zhang
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Jean Yang
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul Bannon
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiothoracic Surgery, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Ashleigh Philp
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, New South Wales, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Andrew Philp
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Healthy Aging, Centenary Institute, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - David M. Kaye
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
- Faculty of Medicine, Monash University, Melbourne, Australia
| | - Mark Larance
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Sean Lal
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory, The University of Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F. O’Sullivan
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
McKay EJ, Luijten I, Broadway-Stringer S, Thomson A, Weng X, Gehmlich K, Gray GA, Semple RK. Female Alms1-deficient mice develop echocardiographic features of adult but not infantile Alström syndrome cardiomyopathy. Dis Model Mech 2024; 17:dmm050561. [PMID: 38756069 PMCID: PMC11225586 DOI: 10.1242/dmm.050561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/03/2024] [Indexed: 05/18/2024] Open
Abstract
Alström syndrome (AS), a multisystem disorder caused by biallelic ALMS1 mutations, features major early morbidity and mortality due to cardiac complications. The latter are biphasic, including infantile dilated cardiomyopathy and distinct adult-onset cardiomyopathy, and poorly understood. We assessed cardiac function of Alms1 knockout (KO) mice by echocardiography. Cardiac function was unaltered in Alms1 global KO mice of both sexes at postnatal day 15 (P15) and 8 weeks. At 23 weeks, female - but not male - KO mice showed increased left atrial area and decreased isovolumic relaxation time, consistent with early restrictive cardiomyopathy, as well as reduced ejection fraction. No histological or transcriptional changes were seen in myocardium of 23-week-old female Alms1 global KO mice. Female mice with Pdgfra-Cre-driven Alms1 deletion in cardiac fibroblasts and in a small proportion of cardiomyocytes did not recapitulate the phenotype of global KO at 23 weeks. In conclusion, only female Alms1-deficient adult mice show echocardiographic evidence of cardiac dysfunction, consistent with the cardiomyopathy of AS. The explanation for sexual dimorphism remains unclear but might involve metabolic or endocrine differences between sexes.
Collapse
Affiliation(s)
- Eleanor J. McKay
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ineke Luijten
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | - Adrian Thomson
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Xiong Weng
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Katya Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford OX3 9DU, UK
| | - Gillian A. Gray
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Robert K. Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| |
Collapse
|
10
|
Sun F, Yuan L, Wang Z, Cui X, Lv N, Zhang T, Zhang Y, Cai J. Cardiac sympathetic overdrive, M2 macrophage activation and fibroblast heterogeneity are associated with cardiac remodeling in a chronic pressure overload rat model of HFpEF. Front Pharmacol 2024; 15:1364758. [PMID: 38860171 PMCID: PMC11163040 DOI: 10.3389/fphar.2024.1364758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifaceted pathogenesis disease and the exact mechanisms driving HFpEF have not been completely elucidated. Pressure overload hypertrophy (POH) related fibroblasts and M2 macrophages in HFpEF myocardium have been recently identified and are now of great interest. Sympathetic overdrive has also been implicated in HFpEF. This study is designed to dynamically observe the potential roles of aforementioned mechanisms in pathological remodeling and cardiac dysfunction in chronic PO rats. Surgical constriction of the abdominal aorta was used for induction of HFpEF. Echocardiography, electrocardiogram, hemodynamic measurement, hematoxylin and eosin staining, Masson staining, immunohistochemistry and immunofluorescence were performed to assess the changes in heart dysfunction, cardiac remodeling and driving mechanisms at different time points (2, 18, 24 weeks). The PO induced HFpEF model was well established, which was confirmed by the persistent increase in carotid artery systolic and diastolic blood pressure, and left ventricle hypertrophy at the corresponding postoperative stage. Meanwhile, PO hypertrophy gradually developed into HFpEF, associated with QT and QTc intervals prolongation, normal systolic (EF was maintained at >50%) but impaired diastolic function (increasing LVEDP and LV -dP/dtmin, abnormal E/A ratio), increased myocytes size, and observed relatively slight inflammatory infiltration but robust reactive fibrosis. IHC staining further confirmed that macrophages (CD68) but not neutrophils (MPO) or T cells (CD3) accounted for a predominant proportion of infiltrating cells. Mechanistically, we found that the infiltrating macrophages in the heart expressed high levels of CD206 which was simultaneously adjacent to POH fibroblasts appeared to overexpression of α-SMA in PO rats at late stages. Interestingly, we distinguished two different POHF sub-populations during PO induced HFpEF development, according to non overlapping signals of α-SMA and PDGFRα/β proteins. Additionally, PO led to a pronounced exaggeration in sympathetic fibers at all time points. These findings suggest that the establishing model here begins with cardiac sympathetic overdrive, subsequently along with immune cells especially M2 macrophage accumulation and fibroblast heterogeneity at later stages is associated with the development of cardiac maladaptive remodeling and diastolic dysfunction thus further progression to HFpEF.
Collapse
Affiliation(s)
- Fengjiao Sun
- Cardiovascular and Cerebrovascular Drugs Research and Development Center, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Ling Yuan
- Department of Pathology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Zi Wang
- Cardiovascular and Cerebrovascular Drugs Research and Development Center, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Xiaoxue Cui
- Department of Pathology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Nan Lv
- Cardiovascular and Cerebrovascular Drugs Research and Development Center, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Ting Zhang
- Pharmaceutical Analysis Laboratory, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Yan Zhang
- Traditional Chinese Medicine Formulation Research Laboratory, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Jun Cai
- Department of Cancer Pharmacology, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| |
Collapse
|
11
|
Schröper T, Mehrkens D, Leiss V, Tellkamp F, Engelhardt S, Herzig S, Birnbaumer L, Nürnberg B, Matthes J. Protective effects of Gα i3 deficiency in a murine heart-failure model of β 1-adrenoceptor overexpression. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2401-2420. [PMID: 37843590 PMCID: PMC10933181 DOI: 10.1007/s00210-023-02751-8] [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: 08/08/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023]
Abstract
We have shown that in murine cardiomyopathy caused by overexpression of the β1-adrenoceptor, Gαi2-deficiency is detrimental. Given the growing evidence for isoform-specific Gαi-functions, we now examined the consequences of Gαi3 deficiency in the same heart-failure model. Mice overexpressing cardiac β1-adrenoceptors with (β1-tg) or without Gαi3-expression (β1-tg/Gαi3-/-) were compared to C57BL/6 wildtypes and global Gαi3-knockouts (Gαi3-/-). The life span of β1-tg mice was significantly shortened but improved when Gαi3 was lacking (95% CI: 592-655 vs. 644-747 days). At 300 days of age, left-ventricular function and survival rate were similar in all groups. At 550 days of age, β1-tg but not β1-tg/Gαi3-/- mice displayed impaired ejection fraction (35 ± 18% vs. 52 ± 16%) compared to wildtype (59 ± 4%) and Gαi3-/- mice (60 ± 5%). Diastolic dysfunction of β1-tg mice was prevented by Gαi3 deficiency, too. The increase of ANP mRNA levels and ventricular fibrosis observed in β1-tg hearts was significantly attenuated in β1-tg/Gαi3-/- mice. Transcript levels of phospholamban, ryanodine receptor 2, and cardiac troponin I were similar in all groups. However, Western blots and phospho-proteomic analyses showed that in β1-tg, but not β1-tg/Gαi3-/- ventricles, phospholamban protein was reduced while its phosphorylation increased. Here, we show that in mice overexpressing the cardiac β1-adrenoceptor, Gαi3 deficiency slows or even prevents cardiomyopathy and increases shortened life span. Previously, we found Gαi2 deficiency to aggravate cardiac dysfunction and mortality in the same heart-failure model. Our findings indicate isoform-specific interventions into Gi-dependent signaling to be promising cardio-protective strategies.
Collapse
Affiliation(s)
- Tobias Schröper
- Center of Pharmacology, Department II, University of Cologne and University Hospital Cologne, Cologne, Germany
- Department of Internal Medicine III, University Hospital of Cologne, Cologne, Germany and Centre for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Dennis Mehrkens
- Department of Internal Medicine III, University Hospital of Cologne, Cologne, Germany and Centre for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Veronika Leiss
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomics, and Interfaculty Centre for Pharmacogenomics and Drug Research, Eberhard Karls Universität, Tübingen, Germany
| | - Frederik Tellkamp
- CECAD Research Centre Institute for Genetics, University of Cologne, Cologne, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - Stefan Herzig
- Center of Pharmacology, Department II, University of Cologne and University Hospital Cologne, Cologne, Germany
- TH Köln-University of Applied Sciences, Cologne, Germany
| | - Lutz Birnbaumer
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina, USA
- Institute of Biomedical Research, School of Medical Sciences, Catholic University of Buenos Aires, Buenos Aires, Argentina
| | - Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute for Experimental and Clinical Pharmacology and Pharmacogenomics, and Interfaculty Centre for Pharmacogenomics and Drug Research, Eberhard Karls Universität, Tübingen, Germany
| | - Jan Matthes
- Center of Pharmacology, Department II, University of Cologne and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
12
|
Jacobsen JCB, Schubert IH, Larsen K, Terzic D, Thisted L, Thomsen MB. Preload dependence in an animal model of mild heart failure with preserved ejection fraction (HFpEF). Acta Physiol (Oxf) 2024; 240:e14099. [PMID: 38230889 DOI: 10.1111/apha.14099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/04/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024]
Abstract
AIM Heart Failure with preserved Ejection Fraction (HFpEF) is characterized by diastolic dysfunction and reduced cardiac output, but its pathophysiology remains poorly understood. Animal models of HFpEF are challenging due to difficulties in assessing the degree of heart failure in small animals. This study aimed at inducing HFpEF in a mouse model to probe preload-dependency. METHODS Increased body mass and arterial hypertension were induced in mice using a Western diet and NO synthase inhibition. Preload dependence was tested ex vivo. RESULTS Mice with obesity and hypertension exhibited reduced cardiac output, indicating a failing heart. Increased left ventricular filling pressure during diastole suggested reduced compliance. Notably, the ejection fraction was preserved, suggesting the development of HFpEF. Spontaneous physical activity at night was reduced in HFpEF mice, indicating exercise intolerance; however, the cardiac connective tissue content was comparable between HFpEF and control mice. The HFpEF mice showed increased vulnerability to reduced preload ex vivo, indicating that elevated left ventricular filling pressure compensated for the rigid left ventricle, preventing a critical decrease in cardiac output. CONCLUSION This animal model successfully developed mild HFpEF with a reduced pump function that was dependent on a high preload. A model of mild HFpEF may serve as a valuable tool for studying disease progression and interventions aimed at delaying or reversing symptom advancement, considering the slow development of HFpEF in patients.
Collapse
Affiliation(s)
- Jens C B Jacobsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irene H Schubert
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karin Larsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dijana Terzic
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Louise Thisted
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Salyer LG, Salhi HE, Brundage EA, Shettigar V, Sturgill SL, Zanella H, Templeton B, Abay E, Emmer KM, Lowe J, Rafael-Fortney JA, Parinandi N, Foster DB, McKinsey TA, Woulfe KC, Ziolo MT, Biesiadecki BJ. Troponin I Tyrosine Phosphorylation Beneficially Accelerates Diastolic Function. Circ Res 2024; 134:33-45. [PMID: 38095088 PMCID: PMC10872382 DOI: 10.1161/circresaha.123.323132] [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: 05/23/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND A healthy heart is able to modify its function and increase relaxation through post-translational modifications of myofilament proteins. While there are known examples of serine/threonine kinases directly phosphorylating myofilament proteins to modify heart function, the roles of tyrosine (Y) phosphorylation to directly modify heart function have not been demonstrated. The myofilament protein TnI (troponin I) is the inhibitory subunit of the troponin complex and is a key regulator of cardiac contraction and relaxation. We previously demonstrated that TnI-Y26 phosphorylation decreases calcium-sensitive force development and accelerates calcium dissociation, suggesting a novel role for tyrosine kinase-mediated TnI-Y26 phosphorylation to regulate cardiac relaxation. Therefore, we hypothesize that increasing TnI-Y26 phosphorylation will increase cardiac relaxation in vivo and be beneficial during pathological diastolic dysfunction. METHODS The signaling pathway involved in TnI-Y26 phosphorylation was predicted in silico and validated by tyrosine kinase activation and inhibition in primary adult murine cardiomyocytes. To investigate how TnI-Y26 phosphorylation affects cardiac muscle, structure, and function in vivo, we developed a novel TnI-Y26 phosphorylation-mimetic mouse that was subjected to echocardiography, pressure-volume loop hemodynamics, and myofibril mechanical studies. TnI-Y26 phosphorylation-mimetic mice were further subjected to the nephrectomy/DOCA (deoxycorticosterone acetate) model of diastolic dysfunction to investigate the effects of increased TnI-Y26 phosphorylation in disease. RESULTS Src tyrosine kinase is sufficient to phosphorylate TnI-Y26 in cardiomyocytes. TnI-Y26 phosphorylation accelerates in vivo relaxation without detrimental structural or systolic impairment. In a mouse model of diastolic dysfunction, TnI-Y26 phosphorylation is beneficial and protects against the development of disease. CONCLUSIONS We have demonstrated that tyrosine kinase phosphorylation of TnI is a novel mechanism to directly and beneficially accelerate myocardial relaxation in vivo.
Collapse
Affiliation(s)
- Lorien G Salyer
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Hussam E Salhi
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Elizabeth A Brundage
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Vikram Shettigar
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Sarah L Sturgill
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Helena Zanella
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Benjamin Templeton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Eaman Abay
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Kathryn M Emmer
- University Laboratory Animal Resources (K.M.E.), Ohio State University, Columbus
| | - Jeovanna Lowe
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Jill A Rafael-Fortney
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Narasimham Parinandi
- Division of Pulmonary, Critical Care and Sleep Medicine (N.P.), Ohio State University, Columbus
| | - D Brian Foster
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD (D.B.F.)
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology (T.A.M., K.C.W.), University of Colorado Anschutz Medical Campus, Aurora
- Consortium for Fibrosis Research and Translation (T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Kathleen C Woulfe
- Department of Medicine, Division of Cardiology (T.A.M., K.C.W.), University of Colorado Anschutz Medical Campus, Aurora
| | - Mark T Ziolo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| | - Brandon J Biesiadecki
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute (L.G.S., H.E.S., E.A.B., V.S., S.L.S., H.Z., B.T., E.A., J.L., J.A.R.-F., M.T.Z., B.J.B.), Ohio State University, Columbus
| |
Collapse
|
14
|
Gao S, Liu XP, Li TT, Chen L, Feng YP, Wang YK, Yin YJ, Little PJ, Wu XQ, Xu SW, Jiang XD. Animal models of heart failure with preserved ejection fraction (HFpEF): from metabolic pathobiology to drug discovery. Acta Pharmacol Sin 2024; 45:23-35. [PMID: 37644131 PMCID: PMC10770177 DOI: 10.1038/s41401-023-01152-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently a preeminent challenge for cardiovascular medicine. It has a poor prognosis, increasing mortality, and is escalating in prevalence worldwide. Despite accounting for over 50% of all HF patients, the mechanistic underpinnings driving HFpEF are poorly understood, thus impeding the discovery and development of mechanism-based therapies. HFpEF is a disease syndrome driven by diverse comorbidities, including hypertension, diabetes and obesity, pulmonary hypertension, aging, and atrial fibrillation. There is a lack of high-fidelity animal models that faithfully recapitulate the HFpEF phenotype, owing primarily to the disease heterogeneity, which has hampered our understanding of the complex pathophysiology of HFpEF. This review provides an updated overview of the currently available animal models of HFpEF and discusses their characteristics from the perspective of energy metabolism. Interventional strategies for efficiently utilizing energy substrates in preclinical HFpEF models are also discussed.
Collapse
Affiliation(s)
- Si Gao
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Xue-Ping Liu
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Ting-Ting Li
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Li Chen
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yi-Ping Feng
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yu-Kun Wang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yan-Jun Yin
- School of Pharmacy, Bengbu Medical College, Bengbu, 233000, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
| | - Xiao-Qian Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Suo-Wen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Xu-Dong Jiang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China.
| |
Collapse
|
15
|
Kacira E, Pittman M, Yang D, Sturgill SL, Ziolo MT, Han Y, Deschênes I, Fu JD. Long-Axis Biplane Echocardiography Sensitively Detects the Progressing Functional Deterioration of Mouse Heart. Circ Heart Fail 2024; 17:e011046. [PMID: 38054280 PMCID: PMC10872990 DOI: 10.1161/circheartfailure.123.011046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Affiliation(s)
- Ege Kacira
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Mitchell Pittman
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Dandan Yang
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Sarah L. Sturgill
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Mark T. Ziolo
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Yuchi Han
- Cardiovascular Division, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Isabelle Deschênes
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Ji-dong Fu
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
16
|
Flint G, Kooiker K, Moussavi-Harami F. Echocardiography to Assess Cardiac Structure and Function in Genetic Cardiomyopathies. Methods Mol Biol 2024; 2735:1-15. [PMID: 38038840 DOI: 10.1007/978-1-0716-3527-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Rodents are the most common experimental models used in cardiovascular research including studies of genetic cardiomyopathies. Genetic cardiomyopathies are characterized by changes in cardiac structure and function. Echocardiography allows for relatively inexpensive, non-invasive, reliable, and reproducible assessment of these changes. However, the fast heart and small size present unique challenges for investigators. To ensure accuracy and reproducibility of these measurements, investigators need to be familiar with standard practices in the field, normal values, and potential pitfalls. The goal of this chapter is to describe steps needed for reliable acquisition and analysis of echocardiography in rodent models. Additionally, we discuss some common pitfalls and challenges.
Collapse
Affiliation(s)
- Galina Flint
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kristina Kooiker
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Farid Moussavi-Harami
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Division of Cardiology, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Dorey TW, McRae MD, Belke DD, Rose RA. PDE4D mediates impaired β-adrenergic receptor signalling in the sinoatrial node in mice with hypertensive heart disease. Cardiovasc Res 2023; 119:2697-2711. [PMID: 37643895 PMCID: PMC10757582 DOI: 10.1093/cvr/cvad138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 06/06/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023] Open
Abstract
AIMS The sympathetic nervous system increases HR by activating β-adrenergic receptors (β-ARs) and increasing cAMP in sinoatrial node (SAN) myocytes while phosphodiesterases (PDEs) degrade cAMP. Chronotropic incompetence, the inability to regulate heart rate (HR) in response to sympathetic nervous system activation, is common in hypertensive heart disease; however, the basis for this is poorly understood. The objective of this study was to determine the mechanisms leading to chronotropic incompetence in mice with angiotensin II (AngII)-induced hypertensive heart disease. METHODS AND RESULTS C57BL/6 mice were infused with saline or AngII (2.5 mg/kg/day for 3 weeks) to induce hypertensive heart disease. HR and SAN function in response to the β-AR agonist isoproterenol (ISO) were studied in vivo using telemetry and electrocardiography, in isolated atrial preparations using optical mapping, in isolated SAN myocytes using patch-clamping, and using molecular biology. AngII-infused mice had smaller increases in HR in response to physical activity and during acute ISO injection. Optical mapping of the SAN in AngII-infused mice demonstrated impaired increases in conduction velocity and altered conduction patterns in response to ISO. Spontaneous AP firing responses to ISO in isolated SAN myocytes from AngII-infused mice were impaired due to smaller increases in diastolic depolarization (DD) slope, hyperpolarization-activated current (If), and L-type Ca2+ current (ICa,L). These changes were due to increased localization of PDE4D surrounding β1- and β2-ARs in the SAN, increased SAN PDE4 activity, and reduced cAMP generation in response to ISO. Knockdown of PDE4D using a virus-delivered shRNA or inhibition of PDE4 with rolipram normalized SAN sensitivity to β-AR stimulation in AngII-infused mice. CONCLUSIONS AngII-induced hypertensive heart disease results in impaired HR responses to β-AR stimulation due to up-regulation of PDE4D and reduced effects of cAMP on spontaneous AP firing in SAN myocytes.
Collapse
Affiliation(s)
- Tristan W Dorey
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
| | - Megan D McRae
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
| | - Darrell D Belke
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
| |
Collapse
|
18
|
Zalivina I, Barwari T, Yin X, Langley SR, Barallobre-Barreiro J, Wakimoto H, Zampetaki A, Mayr M, Avkiran M, Eminaga S. Inhibition of miR-199a-3p in a murine hypertrophic cardiomyopathy (HCM) model attenuates fibrotic remodeling. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 6:100056. [PMID: 38143961 PMCID: PMC10739604 DOI: 10.1016/j.jmccpl.2023.100056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
Background Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic disorder, characterized by cardiomyocyte hypertrophy, cardiomyocyte disarray and fibrosis, which has a prevalence of ∼1: 200-500 and predisposes individuals to heart failure and sudden death. The mechanisms through which diverse HCM-causing mutations cause cardiac dysfunction remain mostly unknown and their identification may reveal new therapeutic avenues. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression and disease phenotype in various pathologies. We explored whether miRNAs could play a role in HCM pathogenesis and offer potential therapeutic targets. Methods and results Using high-throughput miRNA expression profiling and qPCR analysis in two distinct mouse models of HCM, we found that miR-199a-3p expression levels are upregulated in mutant mice compared to age- and treatment-matched wild-type mice. We also found that miR-199a-3p expression is enriched in cardiac non-myocytes compared to cardiomyocytes. When we expressed miR-199a-3p mimic in cultured murine primary cardiac fibroblasts and analyzed the conditioned media by proteomics, we found that several extracellular matrix (ECM) proteins (e.g., TSP2, FBLN3, COL11A1, LYOX) were differentially secreted (data are available via ProteomeXchange with identifier PXD042904). We confirmed our proteomics findings by qPCR analysis of selected mRNAs and demonstrated that miR-199a-3p mimic expression in cardiac fibroblasts drives upregulation of ECM gene expression, including Tsp2, Fbln3, Pcoc1, Col1a1 and Col3a1. To examine the role of miR-199a-3p in vivo, we inhibited its function using lock-nucleic acid (LNA)-based inhibitors (antimiR-199a-3p) in an HCM mouse model. Our results revealed that progression of cardiac fibrosis is attenuated when miR-199a-3p function is inhibited in mild-to-moderate HCM. Finally, guided by computational target prediction algorithms, we identified mRNAs Cd151 and Itga3 as direct targets of miR-199a-3p and have shown that miR-199a-3p mimic expression negatively regulates AKT activation in cardiac fibroblasts. Conclusions Altogether, our results suggest that miR-199a-3p may contribute to cardiac fibrosis in HCM through its actions in cardiac fibroblasts. Thus, inhibition of miR-199a-3p in mild-to-moderate HCM may offer therapeutic benefit in combination with complementary approaches that target the primary defect in cardiac myocytes.
Collapse
Affiliation(s)
- Irina Zalivina
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Temo Barwari
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Xiaoke Yin
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Sarah R. Langley
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Anna Zampetaki
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Manuel Mayr
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Metin Avkiran
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Seda Eminaga
- King's College London, British Heart Foundation Centre of Research Excellence, London, United Kingdom
| |
Collapse
|
19
|
Kang R, Laborde C, Savchenko L, Swiader A, Pizzinat N, Marsal D, Sainte-Marie Y, Boal F, Tronchere H, Roncalli J, Kunduzova O. Age-Related Shift in Cardiac and Metabolic Phenotyping Linked to Inflammatory Cytokines and Antioxidant Status in Mice. Int J Mol Sci 2023; 24:15841. [PMID: 37958823 PMCID: PMC10650425 DOI: 10.3390/ijms242115841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/18/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Age-related alterations in cardiac function, metabolic, inflammatory and antioxidant profiles are associated with an increased risk of cardiovascular mortality and morbidity. Here, we examined cardiac and metabolic phenotypes in relation to inflammatory status and antioxidant capacity in young, middle-aged and old mice. Real-time reverse transcription-polymerase chain reactions were performed on myocardium and immunoassays on plasma. Left ventricular (LV) structure and function were assessed by echocardiography using high-frequency ultrasound. Middle-aged mice exhibited an altered metabolic profile and antioxidant capacity compared to young mice, whereas myocardial expression of inflammatory factors (TNFα, IL1β, IL6 and IL10) remained unchanged. In contrast, old mice exhibited increased expression of inflammatory cytokines and plasma levels of resistin compared to young and middle-aged mice (p < 0.05). The pro-inflammatory signature of aged hearts was associated with alterations in glutathione redox homeostasis and elevated contents of 4-hydroxynonenal (4-HNE), a marker of lipid peroxidation and oxidative stress. Furthermore, echocardiographic parameters of LV systolic and diastolic functions were significantly altered in old mice compared to young mice. Taken together, these findings suggest age-related shifts in cardiac phenotype encompass the spectrum of metabo-inflammatory abnormalities and altered redox homeostasis.
Collapse
Affiliation(s)
- Ryeonshi Kang
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | | | - Lesia Savchenko
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
- Department of Internal Medicine, Poltava State Medical University, 23 Shevchenko, 36000 Poltava, Ukraine
| | - Audrey Swiader
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Nathalie Pizzinat
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Dimitri Marsal
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Yannis Sainte-Marie
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Frederic Boal
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Helene Tronchere
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Jerome Roncalli
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- Department of Cardiology, University Hospital of Toulouse, CEDEX 9, 31400 Toulouse, France
| | - Oksana Kunduzova
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| |
Collapse
|
20
|
Smart CD, Fehrenbach DJ, Wassenaar JW, Agrawal V, Fortune NL, Dixon DD, Cottam MA, Hasty AH, Hemnes AR, Doran AC, Gupta DK, Madhur MS. Immune profiling of murine cardiac leukocytes identifies triggering receptor expressed on myeloid cells 2 as a novel mediator of hypertensive heart failure. Cardiovasc Res 2023; 119:2312-2328. [PMID: 37314125 PMCID: PMC10597637 DOI: 10.1093/cvr/cvad093] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/17/2023] [Accepted: 06/12/2023] [Indexed: 06/15/2023] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is characterized by diastolic dysfunction, microvascular dysfunction, and myocardial fibrosis with recent evidence implicating the immune system in orchestrating cardiac remodelling. METHODS AND RESULTS Here, we show the mouse model of deoxycorticosterone acetate (DOCA)-salt hypertension induces key elements of HFpEF, including diastolic dysfunction, exercise intolerance, and pulmonary congestion in the setting of preserved ejection fraction. A modified single-cell sequencing approach, cellular indexing of transcriptomes and epitopes by sequencing, of cardiac immune cells reveals an altered abundance and transcriptional signature in multiple cell types, most notably cardiac macrophages. The DOCA-salt model results in differential expression of several known and novel genes in cardiac macrophages, including up-regulation of Trem2, which has been recently implicated in obesity and atherosclerosis. The role of Trem2 in hypertensive heart failure, however, is unknown. We found that mice with genetic deletion of Trem2 exhibit increased cardiac hypertrophy, diastolic dysfunction, renal injury, and decreased cardiac capillary density after DOCA-salt treatment compared to wild-type controls. Moreover, Trem2-deficient macrophages have impaired expression of pro-angiogenic gene programmes and increased expression of pro-inflammatory cytokines. Furthermore, we found that plasma levels of soluble TREM2 are elevated in DOCA-salt treated mice and humans with heart failure. CONCLUSIONS Together, our data provide an atlas of immunological alterations that can lead to improved diagnostic and therapeutic strategies for HFpEF. We provide our dataset in an easy to explore and freely accessible web application making it a useful resource for the community. Finally, our results suggest a novel cardioprotective role for Trem2 in hypertensive heart failure.
Collapse
Affiliation(s)
- Charles Duncan Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA
| | - Daniel J Fehrenbach
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), 2215 Garland Avenue, P415D MRB IV, Nashville, TN 37232, USA
| | - Jean W Wassenaar
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), 1311 Medical Center Dr, Nashville, TN 37232, USA
| | - Vineet Agrawal
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), 1311 Medical Center Dr, Nashville, TN 37232, USA
| | - Niki L Fortune
- VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Debra D Dixon
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), 1311 Medical Center Dr, Nashville, TN 37232, USA
| | - Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Anna R Hemnes
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Amanda C Doran
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), 1311 Medical Center Dr, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center (VUMC), Medical Center North A-5121, 1161 21st Ave South, Nashville, TN 37232, USA
| | - Deepak K Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), 1311 Medical Center Dr, Nashville, TN 37232, USA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meena S Madhur
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), 2215 Garland Avenue, P415D MRB IV, Nashville, TN 37232, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center (VUMC), 1311 Medical Center Dr, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center (VUMC), Medical Center North A-5121, 1161 21st Ave South, Nashville, TN 37232, USA
| |
Collapse
|
21
|
Holmes JB, Lemieux ME, Stelzer JE. Torsional and strain dysfunction precede overt heart failure in a mouse model of dilated cardiomyopathy pathogenesis. Am J Physiol Heart Circ Physiol 2023; 325:H449-H467. [PMID: 37417875 PMCID: PMC10538988 DOI: 10.1152/ajpheart.00130.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/24/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Detailed assessments of whole heart mechanics are crucial for understanding the consequences of sarcomere perturbations that lead to cardiomyopathy in mice. Echocardiography offers an accessible and cost-effective method of obtaining metrics of cardiac function, but the most routine imaging and analysis protocols might not identify subtle mechanical deficiencies. This study aims to use advanced echocardiography imaging and analysis techniques to identify previously unappreciated mechanical deficiencies in a mouse model of dilated cardiomyopathy (DCM) before the onset of overt systolic heart failure (HF). Mice lacking muscle LIM protein expression (MLP-/-) were used to model DCM-linked HF pathogenesis. Left ventricular (LV) function of MLP-/- and wild-type (WT) controls were studied at 3, 6, and 10 wk of age using conventional and four-dimensional (4-D) echocardiography, followed by speckle-tracking analysis to assess torsional and strain mechanics. Mice were also studied with RNA-seq. Although 3-wk-old MLP-/- mice showed normal LV ejection fraction (LVEF), these mice displayed abnormal torsional and strain mechanics alongside reduced β-adrenergic reserve. Transcriptome analysis showed that these defects preceded most molecular markers of HF. However, these markers became upregulated as MLP-/- mice aged and developed overt systolic dysfunction. These findings indicate that subtle deficiencies in LV mechanics, undetected by LVEF and conventional molecular markers, may act as pathogenic stimuli in DCM-linked HF. Using these analyses in future studies will further help connect in vitro measurements of the sarcomere function to whole heart function.NEW & NOTEWORTHY A detailed study of how perturbations to sarcomere proteins impact whole heart mechanics in mouse models is a major yet challenging step in furthering our understanding of cardiovascular pathophysiology. This study uses advanced echocardiographic imaging and analysis techniques to reveal previously unappreciated subclinical whole heart mechanical defects in a mouse model of cardiomyopathy. In doing so, it offers an accessible set of measurements for future studies to use when connecting sarcomere and whole heart function.
Collapse
Affiliation(s)
- Joshua B Holmes
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, United States
| | | | - Julian E Stelzer
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
22
|
Pironti G. State-of-the-art methodologies used in preclinical studies to assess left ventricular diastolic and systolic function in mice, pitfalls and troubleshooting. Front Cardiovasc Med 2023; 10:1228789. [PMID: 37608817 PMCID: PMC10441126 DOI: 10.3389/fcvm.2023.1228789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/10/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVD) are still the leading cause of death worldwide. The improved survival of patients with comorbidities such as type 2 diabetes, hypertension, obesity together with the extension of life expectancy contributes to raise the prevalence of CVD in the increasingly aged society. Therefore, a translational research platform that enables precise evaluation of cardiovascular function in healthy and disease condition and assess the efficacy of novel pharmacological treatments, could implement basic science and contribute to reduce CVD burden. Heart failure is a deadly syndrome characterized by the inability of the heart to meet the oxygen demands of the body (unless there is a compensatory increased of filling pressure) and can manifest either with reduced ejection fraction (HFrEF) or preserved ejection fraction (HFpEF). The development and progression of HFrEF is mostly attributable to impaired contractile performance (systole), while in HFpEF the main problem resides in decreased ability of left ventricle to relax and allow the blood filling (diastole). Murine preclinical models have been broadly used in research to understand pathophysiologic mechanisms of heart failure and test the efficacy of novel therapies. Several methods have been employed to characterise cardiac systolic and diastolic function including Pressure Volume (PV) loop hemodynamic analysis, echocardiography and Magnetic Resonance Imaging (MRI). The choice of one methodology or another depends on many aspects including budget available, skills of the operator and design of the study. The aim of this review is to discuss the importance of several methodologies that are commonly used to characterise the cardiovascular phenotype of preclinical models of heart failure highlighting advantages and limitation of each procedure. Although it requires highly skilled operators for execution, PV loop analysis represents the "gold standard" methodology that enables the assessment of left ventricular performance also independently of vascular loading conditions and heart rate, which conferee a really high physiologic importance to this procedure.
Collapse
Affiliation(s)
- Gianluigi Pironti
- Cardiology Research Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Damen FW, Gramling DP, Ahlf Wheatcraft D, Wilpan RY, Costa MW, Goergen CJ. Application of 4-D ultrasound-derived regional strain and proteomics analysis in Nkx2-5-deficient male mice. Am J Physiol Heart Circ Physiol 2023; 325:H293-H310. [PMID: 37326999 PMCID: PMC10393333 DOI: 10.1152/ajpheart.00733.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/26/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023]
Abstract
The comprehensive characterization of cardiac structure and function is critical to better understanding various murine models of cardiac disease. We demonstrate here a multimodal analysis approach using high-frequency four-dimensional ultrasound (4DUS) imaging and proteomics to explore the relationship between regional function and tissue composition in a murine model of metabolic cardiomyopathy (Nkx2-5183P/+). The presented 4DUS analysis outlines a novel approach to mapping both circumferential and longitudinal strain profiles through a standardized framework. We then demonstrate how this approach allows for spatiotemporal comparisons of cardiac function and improved localization of regional left ventricular dysfunction. Guided by observed trends in regional dysfunction, our targeted Ingenuity Pathway Analysis (IPA) results highlight metabolic dysregulation in the Nkx2-5183P/+ model, including altered mitochondrial function and energy metabolism (i.e., oxidative phosphorylation and fatty acid/lipid handling). Finally, we present a combined 4DUS-proteomics z-score-based analysis that highlights IPA canonical pathways showing strong linear relationships with 4DUS biomarkers of regional cardiac dysfunction. The presented multimodal analysis methods aim to help future studies more comprehensively assess regional structure-function relationships in other preclinical models of cardiomyopathy.NEW & NOTEWORTHY A multimodal approach using both four-dimensional ultrasound (4DUS) and regional proteomics can help enhance our investigations of murine cardiomyopathy models. We present unique 4DUS-derived strain maps that provide a framework for both cross-sectional and longitudinal analysis of spatiotemporal cardiac function. We further detail and demonstrate an innovative 4DUS-proteomics z-score-based linear regression method, aimed at characterizing relationships between regional cardiac dysfunction and underlying mechanisms of disease.
Collapse
Affiliation(s)
- Frederick W Damen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States
- Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Daniel P Gramling
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States
| | | | | | - Mauro W Costa
- Jackson Laboratory, Bar Harbor, Maine, United States
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, United States
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States
- Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
24
|
Daniels LJ, Macindoe C, Koutsifeli P, Annandale M, James SL, Watson LE, Coffey S, Raaijmakers AJA, Weeks KL, Bell JR, Janssens JV, Curl CL, Delbridge LMD, Mellor KM. Myocardial deformation imaging by 2D speckle tracking echocardiography for assessment of diastolic dysfunction in murine cardiopathology. Sci Rep 2023; 13:12344. [PMID: 37524893 PMCID: PMC10390581 DOI: 10.1038/s41598-023-39499-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 07/26/2023] [Indexed: 08/02/2023] Open
Abstract
Diastolic dysfunction is increasingly identified as a key, early onset subclinical condition characterizing cardiopathologies of rising prevalence, including diabetic heart disease and heart failure with preserved ejection fraction (HFpEF). Diastolic dysfunction characterization has important prognostic value in management of disease outcomes. Validated tools for in vivo monitoring of diastolic function in rodent models of diabetes are required for progress in pre-clinical cardiology studies. 2D speckle tracking echocardiography has emerged as a powerful tool for evaluating cardiac wall deformation throughout the cardiac cycle. The aim of this study was to examine the applicability of 2D speckle tracking echocardiography for comprehensive global and regional assessment of diastolic function in a pre-clinical murine model of cardio-metabolic disease. Type 2 diabetes (T2D) was induced in C57Bl/6 male mice using a high fat high sugar dietary intervention for 20 weeks. Significant impairment in left ventricle peak diastolic strain rate was evident in longitudinal, radial and circumferential planes in T2D mice. Peak diastolic velocity was similarly impaired in the longitudinal and radial planes. Regional analysis of longitudinal peak diastolic strain rate revealed that the anterior free left ventricular wall is particularly susceptible to T2D-induced diastolic dysfunction. These findings provide a significant advance on characterization of diastolic dysfunction in a pre-clinical mouse model of cardiopathology and offer a comprehensive suite of benchmark values for future pre-clinical cardiology studies.
Collapse
Affiliation(s)
- L J Daniels
- Cellular and Molecular Cardiology Laboratory, Department of Physiology, University of Auckland, Auckland, New Zealand
- Radcliffe Department of Medicine, OCDEM, University of Oxford, Oxford, UK
| | - C Macindoe
- Cellular and Molecular Cardiology Laboratory, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - P Koutsifeli
- Cellular and Molecular Cardiology Laboratory, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - M Annandale
- Cellular and Molecular Cardiology Laboratory, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - S L James
- Cellular and Molecular Cardiology Laboratory, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - L E Watson
- Cellular and Molecular Cardiology Laboratory, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - S Coffey
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - A J A Raaijmakers
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia
| | - K L Weeks
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - J R Bell
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Australia
| | - J V Janssens
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia
| | - C L Curl
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia
| | - L M D Delbridge
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia
| | - Kimberley M Mellor
- Cellular and Molecular Cardiology Laboratory, Department of Physiology, University of Auckland, Auckland, New Zealand.
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia.
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
25
|
Dominic KL, Choi J, Holmes JB, Singh M, Majcher MJ, Stelzer JE. The contribution of N-terminal truncated cMyBPC to in vivo cardiac function. J Gen Physiol 2023; 155:e202213318. [PMID: 37067542 PMCID: PMC10114924 DOI: 10.1085/jgp.202213318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/13/2023] [Accepted: 03/24/2023] [Indexed: 04/18/2023] Open
Abstract
Cardiac myosin binding protein C (cMyBPC) is an 11-domain sarcomeric protein (C0-C10) integral to cardiac muscle regulation. In vitro studies have demonstrated potential functional roles for regions beyond the N-terminus. However, the in vivo contributions of these domains are mostly unknown. Therefore, we examined the in vivo consequences of expression of N-terminal truncated cMyBPC (C3C10). Neonatal cMyBPC-/- mice were injected with AAV9-full length (FL), C3C10 cMyBPC, or saline, and echocardiography was performed 6 wk after injection. We then isolated skinned myocardium from virus-treated hearts and performed mechanical experiments. Our results show that expression of C3C10 cMyBPC in cMyBPC-/- mice resulted in a 28% increase in systolic ejection fraction compared to saline-injected cMyBPC-/- mice and a 25% decrease in left ventricle mass-to-body weight ratio. However, unlike expression of FL cMyBPC, there was no prolongation of ejection time compared to saline-injected mice. In vitro mechanical experiments demonstrated that functional improvements in cMyBPC-/- mice expressing C3C10 were primarily due to a 35% reduction in the rate of cross-bridge recruitment at submaximal Ca2+ concentrations when compared to hearts from saline-injected cMyBPC-/- mice. However, unlike the expression of FL cMyBPC, there was no change in the rate of cross-bridge detachment when compared to saline-injected mice. Our data demonstrate that regions of cMyBPC beyond the N-terminus are important for in vivo cardiac function, and have divergent effects on cross-bridge behavior. Elucidating the molecular mechanisms of cMyBPC region-specific function could allow for development of targeted approaches to manipulate specific aspects of cardiac contractile function.
Collapse
Affiliation(s)
- Katherine L. Dominic
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Joohee Choi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Joshua B. Holmes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mandeep Singh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Michael J. Majcher
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
26
|
O'Riordan CE, Trochet P, Steiner M, Fuchs D. Standardisation and future of preclinical echocardiography. Mamm Genome 2023; 34:123-155. [PMID: 37160810 DOI: 10.1007/s00335-023-09981-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/31/2023] [Indexed: 05/11/2023]
Abstract
Echocardiography is a non-invasive imaging technique providing real-time information to assess the structure and function of the heart. Due to advancements in technology, ultra-high-frequency transducers have enabled the translation of ultrasound from humans to small animals due to resolutions down to 30 µm. Most studies are performed using mice and rats, with ages ranging from embryonic, to neonatal, and adult. In addition, alternative models such as zebrafish and chicken embryos are becoming more frequently used. With the achieved high temporal and spatial resolution in real-time, cardiac function can now be monitored throughout the lifespan of these small animals to investigate the origin and treatment of a range of acute and chronic pathological conditions. With the increased relevance of in vivo real-time imaging, there is still an unmet need for the standardisation of small animal echocardiography and the appropriate cardiac measurements that should be reported in preclinical cardiac models. This review focuses on the development of standardisation in preclinical echocardiography and reports appropriate cardiac measurements throughout the lifespan of rodents: embryonic, neonatal, ageing, and acute and chronic pathologies. Lastly, we will discuss the future of cardiac preclinical ultrasound.
Collapse
Affiliation(s)
| | | | | | - Dieter Fuchs
- FUJIFILM VisualSonics, Inc, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Wu H, Tang LX, Wang XM, Li LP, Chen XK, He YJ, Yang DZ, Shi Y, Shou JL, Zhang ZS, Wang L, Lu BJ, An SM, Zeng CY, Wang WE. Porcupine inhibitor CGX1321 alleviates heart failure with preserved ejection fraction in mice by blocking WNT signaling. Acta Pharmacol Sin 2023; 44:1149-1160. [PMID: 36473990 PMCID: PMC10203103 DOI: 10.1038/s41401-022-01025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 11/05/2022] [Indexed: 12/12/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is highly prevalent, and lacks effective treatment. The aberration of WNT pathway underlies many pathological processes including cardiac fibrosis and hypertrophy, while porcupine is an acyltransferase essential for the secretion of WNT ligands. In this study we investigated the role of WNT signaling pathway in HFpEF as well as whether blocking WNT signaling by a novel porcupine inhibitor CGX1321 alleviated HFpEF. We established two experimental HFpEF mouse models, namely the UNX/DOCA model and high fat diet/L-NAME ("two-hit") model. The UNX/DOCA and "two-hit" mice were treated with CGX1321 (3 mg·kg-1·d-1) for 4 and 10 weeks, respectively. We showed that CGX1321 treatment significantly alleviated cardiac hypertrophy and fibrosis, thereby improving cardiac diastolic function and exercise performance in both models. Furthermore, both canonical and non-canonical WNT signaling pathways were activated, and most WNT proteins, especially WNT3a and WNT5a, were upregulated during the development of HEpEF in mice. CGX1321 treatment inhibited the secretion of WNT ligands and repressed both canonical and non-canonical WNT pathways, evidenced by the reduced phosphorylation of c-Jun and the nuclear translocation of β-catenin and NFATc3. In an in vitro HFpEF model, MCM and ISO-treated cardiomyocytes, knockdown of porcupine by siRNA leads to a similar inhibitory effect on WNT pathways, cardiomyocyte hypertrophy and cardiac fibroblast activation as CGX1321 did, whereas supplementation of WNT3a and WNT5a reversed the anti-hypertrophy and anti-fibrosis effect of CGX1321. We conclude that WNT signaling activation plays an essential role in the pathogenesis of HFpEF, and porcupine inhibitor CGX1321 exerts a therapeutic effect on HFpEF in mice by attenuating cardiac hypertrophy, alleviating cardiac fibrosis and improving cardiac diastolic function.
Collapse
Affiliation(s)
- Hao Wu
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Lu-Xun Tang
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, 610083, China
| | - Xue-Mei Wang
- School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Liang-Peng Li
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Xiao-Kang Chen
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Yan-Ji He
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - De-Zhong Yang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Yu Shi
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Jia-Ling Shou
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Zong-Shu Zhang
- Guangzhou Curegenix Co. Ltd., International Business Incubator, Guangzhou Science City, Guangzhou, 510663, China
| | - Liang Wang
- Guangzhou Curegenix Co. Ltd., International Business Incubator, Guangzhou Science City, Guangzhou, 510663, China
| | - Bing-Jun Lu
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Songzhu Michael An
- Guangzhou Curegenix Co. Ltd., International Business Incubator, Guangzhou Science City, Guangzhou, 510663, China
- Curegenix, Inc., Burlingame, CA, 94010, USA
| | - Chun-Yu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China.
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, 400042, China.
- Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, 350001, China.
- Department of Cardiology, Chongqing General Hospital, Chongqing, 401147, China.
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, 400722, China.
| | - Wei Eric Wang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Military Medical University), Chongqing, 400042, China.
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China.
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
28
|
Zhang N, Harsch B, Zhang MJ, Gyberg DJ, Stevens JA, Wagner BM, Mendelson J, Patterson MT, Orchard DA, Healy CL, Williams JW, Townsend D, Shearer GC, Murphy KA, O'Connell TD. FFAR4 regulates cardiac oxylipin balance to promote inflammation resolution in HFpEF secondary to metabolic syndrome. J Lipid Res 2023; 64:100374. [PMID: 37075982 PMCID: PMC10209340 DOI: 10.1016/j.jlr.2023.100374] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex clinical syndrome, but a predominant subset of HFpEF patients has metabolic syndrome (MetS). Mechanistically, systemic, nonresolving inflammation associated with MetS might drive HFpEF remodeling. Free fatty acid receptor 4 (Ffar4) is a GPCR for long-chain fatty acids that attenuates metabolic dysfunction and resolves inflammation. Therefore, we hypothesized that Ffar4 would attenuate remodeling in HFpEF secondary to MetS (HFpEF-MetS). To test this hypothesis, mice with systemic deletion of Ffar4 (Ffar4KO) were fed a high-fat/high-sucrose diet with L-NAME in their water to induce HFpEF-MetS. In male Ffar4KO mice, this HFpEF-MetS diet induced similar metabolic deficits but worsened diastolic function and microvascular rarefaction relative to WT mice. Conversely, in female Ffar4KO mice, the diet produced greater obesity but no worsened ventricular remodeling relative to WT mice. In Ffar4KO males, MetS altered the balance of inflammatory oxylipins systemically in HDL and in the heart, decreasing the eicosapentaenoic acid-derived, proresolving oxylipin 18-hydroxyeicosapentaenoic acid (18-HEPE), while increasing the arachidonic acid-derived, proinflammatory oxylipin 12-hydroxyeicosatetraenoic acid (12-HETE). This increased 12-HETE/18-HEPE ratio reflected a more proinflammatory state both systemically and in the heart in male Ffar4KO mice and was associated with increased macrophage numbers in the heart, which in turn correlated with worsened ventricular remodeling. In summary, our data suggest that Ffar4 controls the proinflammatory/proresolving oxylipin balance systemically and in the heart to resolve inflammation and attenuate HFpEF remodeling.
Collapse
Affiliation(s)
- Naixin Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Brian Harsch
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Michael J Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Dylan J Gyberg
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jackie A Stevens
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Brandon M Wagner
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jenna Mendelson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | | | - Devin A Orchard
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Chastity L Healy
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jesse W Williams
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA; Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Gregory C Shearer
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA.
| | - Katherine A Murphy
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| | - Timothy D O'Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
29
|
Sharma A, Choi JSY, Watson AMD, Li L, Sonntag T, Lee MKS, Murphy AJ, De Blasio M, Head GA, Ritchie RH, de Haan JB. Cardiovascular characterisation of a novel mouse model that combines hypertension and diabetes co-morbidities. Sci Rep 2023; 13:8741. [PMID: 37253814 DOI: 10.1038/s41598-023-35680-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/22/2023] [Indexed: 06/01/2023] Open
Abstract
Epidemiologic data suggest that the prevalence of hypertension in patients with diabetes mellitus is ∼1.5-2.0 times greater than in matched non-diabetic patients. This co-existent disease burden exacerbates cardiac and vascular injury, leading to structural and functional changes to the myocardium, impaired cardiac function and heart failure. Oxidative stress and persistent low-grade inflammation underlie both conditions, and are identified as major contributors to pathological cardiac remodelling. There is an urgent need for effective therapies that specifically target oxidative stress and inflammation to protect against cardiac remodelling. Animal models are a valuable tool for testing emerging therapeutics, however, there is a notable lack of appropriate animal models of co-morbid diabetes and hypertension. In this study, we describe a novel preclinical mouse model combining diabetes and hypertension to investigate cardiac and vascular pathology of co-morbid disease. Type 1 diabetes was induced in spontaneously hypertensive, 8-week old, male Schlager (BPH/2) mice via 5 consecutive, daily injections of streptozotocin (55 mg/kg in citrate buffer; i.p.). Non-diabetic mice received citrate buffer only. After 10 weeks of diabetes induction, cardiac function was assessed by echocardiography prior to post-mortem evaluation of cardiomyocyte hypertrophy, interstitial fibrosis and inflammation by histology, RT-PCR and flow cytometry. We focussed on the oxidative and inflammatory stress pathways that contribute to cardiovascular remodelling. In particular, we demonstrate that markers of inflammation (monocyte chemoattractant protein; MCP-1), oxidative stress (urinary 8-isoprostanes) and fibrosis (connective tissue growth factor; CTGF) are significantly increased, whilst diastolic dysfunction, as indicated by prolonged isovolumic relaxation time (IVRT), is elevated in this diabetic and hypertensive mouse model. In summary, this pre-clinical mouse model provides researchers with a tool to test therapeutic strategies unique to co-morbid diabetes and hypertension, thereby facilitating the emergence of novel therapeutics to combat the cardiovascular consequences of these debilitating co-morbidities.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Judy S Y Choi
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Anna M D Watson
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, Australia
| | - Leila Li
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Thomas Sonntag
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Man K S Lee
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Andrew J Murphy
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Miles De Blasio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Geoffrey A Head
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Rebecca H Ritchie
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Judy B de Haan
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Australia
- Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
30
|
Federti E, Vinchi F, Iatcenko I, Ghigo A, Matte A, Toya SCM, Siciliano A, Chiabrando D, Tolosano E, Vance SZ, Riccardi V, Andolfo I, Iezzi M, Lamolinara A, Iolascon A, De Franceschi L. Duality of Nrf2 in iron-overload cardiomyopathy. Haematologica 2023; 108:1335-1348. [PMID: 36700398 PMCID: PMC10153524 DOI: 10.3324/haematol.2022.281995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Cardiomyopathy deeply affects quality of life and mortality of patients with b-thalassemia or with transfusion-dependent myelodysplastic syndromes. Recently, a link between Nrf2 activity and iron metabolism has been reported in liver ironoverload murine models. Here, we studied C57B6 mice as healthy control and nuclear erythroid factor-2 knockout (Nrf2-/-) male mice aged 4 and 12 months. Eleven-month-old wild-type and Nrf2-/- mice were fed with either standard diet or a diet containing 2.5% carbonyl-iron (iron overload [IO]) for 4 weeks. We show that Nrf2-/- mice develop an age-dependent cardiomyopathy, characterized by severe oxidation, degradation of SERCA2A and iron accumulation. This was associated with local hepcidin expression and increased serum non-transferrin-bound iron, which promotes maladaptive cardiac remodeling and interstitial fibrosis related to overactivation of the TGF-b pathway. When mice were exposed to IO diet, the absence of Nrf2 was paradoxically protective against further heart iron accumulation. Indeed, the combination of prolonged oxidation and the burst induced by IO diet resulted in activation of the unfolded protein response (UPR) system, which in turn promotes hepcidin expression independently from heart iron accumulation. In the heart of Hbbth3/+ mice, a model of b-thalassemia intermedia, despite the activation of Nrf2 pathway, we found severe protein oxidation, activation of UPR system and cardiac fibrosis independently from heart iron content. We describe the dual role of Nrf2 when aging is combined with IO and its novel interrelation with UPR system to ensure cell survival. We open a new perspective for early and intense treatment of cardiomyopathy in patients with b-thalassemia before the appearance of heart iron accumulation.
Collapse
Affiliation(s)
- Enrica Federti
- Department of Medicine, University of Verona and AOUI Verona, Verona
| | - Francesca Vinchi
- Iron Research Laboratory, Lindsley Kimball Research Institute, New York Blood Center, New York, NY, USA; Dept. of Pathology and Laboratory Medicine, Weill Cornell Medicine
| | - Iana Iatcenko
- Department of Medicine, University of Verona and AOUI Verona, Verona
| | - Alessandra Ghigo
- Department Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarrone", University of Torino, Torino
| | - Alessandro Matte
- Department of Medicine, University of Verona and AOUI Verona, Verona
| | | | - Angela Siciliano
- Department of Medicine, University of Verona and AOUI Verona, Verona
| | - Deborah Chiabrando
- Department Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarrone", University of Torino, Torino
| | - Emanuela Tolosano
- Department Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarrone", University of Torino, Torino
| | - Steven Zebulon Vance
- Iron Research Laboratory, Lindsley Kimball Research Institute, New York Blood Center, New York, NY
| | - Veronica Riccardi
- Department of Medicine, University of Verona and AOUI Verona, Verona
| | - Immacolata Andolfo
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples; CEINGE - Biotecnologie Avanzate, Naples
| | - Manuela Iezzi
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti, Chieti
| | - Alessia Lamolinara
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti, Chieti
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples; CEINGE - Biotecnologie Avanzate, Naples
| | | |
Collapse
|
31
|
Wang L, Tang XQ, Shi Y, Li HM, Meng ZY, Chen H, Li XH, Chen YC, Liu H, Hong Y, Xu HH, Liu L, Zhao L, Han WN, Liu X, Zhang Y. Tetrahydroberberrubine retards heart aging in mice by promoting PHB2-mediated mitophagy. Acta Pharmacol Sin 2023; 44:332-344. [PMID: 35948750 PMCID: PMC9889783 DOI: 10.1038/s41401-022-00956-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/07/2022] [Indexed: 02/08/2023] Open
Abstract
Heart aging is characterized by left ventricular hypertrophy and diastolic dysfunction, which in turn induces a variety of cardiovascular diseases. There is still no therapeutic drug to ameliorate cardiac abnormities in heart aging. In this study we investigated the protective effects of berberine (BBR) and its derivative tetrahydroberberrubine (THBru) against heart aging process. Heart aging was induced in mice by injection of D-galactose (D-gal, 120 mg · kg-1 · d-1, sc.) for 12 weeks. Meanwhile the mice were orally treated with berberine (50 mg · kg-1 · d-1) or THBru (25, 50 mg · kg-1 · d-1) for 12 weeks. We showed that BBR and THBru treatment significantly mitigated diastolic dysfunction and cardiac remodeling in D-gal-induced aging mice. Furthermore, treatment with BBR (40 μM) and THBru (20, 40 μM) inhibited D-gal-induced senescence in primary neonatal mouse cardiomyocytes in vitro. Overall, THBru exhibited higher efficacy than BBR at the same dose. We found that the levels of mitophagy were significantly decreased during the aging process in vivo and in vitro, THBru and BBR promoted mitophagy with different potencies. We demonstrated that the mitophagy-inducing effects of THBru resulted from increased mRNA stability of prohibitin 2 (PHB2), a pivotal factor during mitophagy, thereby upregulating PHB2 protein expression. Knockdown of PHB2 effectively reversed the antisenescence effects of THBru in D-gal-treated cardiomyocytes. On the contrary, overexpression of PHB2 promoted mitophagy and retarded cardiomyocyte senescence, as THBru did. In conclusion, this study identifies THBru as a potent antiaging medicine that induces PHB2-mediated mitophagy and suggests its clinical application prospects.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue-Qing Tang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Shi
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Hui-Min Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zi-Yu Meng
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Hui Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiao-Han Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yong-Chao Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Heng Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Hong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Heng-Hui Xu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ling Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Limin Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Wei-Na Han
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xin Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, Harbin, 150081, China.
| | - Yong Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, Harbin, 150081, China.
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, 150081, China.
| |
Collapse
|
32
|
Li Y, Zheng G, Salimova E, Broughton BRS, Ricardo SD, de Veer M, Samuel CS. Simultaneous late-gadolinium enhancement and T1 mapping of fibrosis and a novel cell-based combination therapy in hypertensive mice. Biomed Pharmacother 2023; 158:114069. [PMID: 36502754 DOI: 10.1016/j.biopha.2022.114069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Fibrosis is a hallmark of chronic hypertension and disrupts the viability of human bone marrow-derived mesenchymal stromal cells (BM-MSCs) post-transplantation. This study thus, determined whether the anti-fibrotic drug, serelaxin (RLX), could enhance the therapeutic effects of BM-MSCs or BM-MSC-derived exosomes (BM-MSC-EXO) in hypertensive mice. Left ventricular (LV) fibrosis in particular was assessed using conventional histological staining and non-invasive cardiac magnetic resonance imaging (CMRI). CMRI was employed using a novel magnetisation prepared 2 rapid acquisition gradient echo (MP2RAGE) sequence to simultaneously perform late gadolinium enhancement imaging and T1 mapping. Adult male C57BL/6 mice were uninephrectomised, received deoxycorticosterone acetate and saline to drink (1 K/DOCA/salt) for 21 days, whilst control mice were given normal drinking water for the same time-period. On day 14 post-injury, subgroups of 1 K/DOCA/salt-hypertensive mice were treated with RLX alone or in combination with BM-MSCs or BM-MSC-EXO; or the mineralocorticoid receptor antagonist, spironolactone. At day 21 post-injury, LV and kidney histopathology was assessed, whilst LV fibrosis and function were additionally analysed by CMRI and echocardiography. 1 K/DOCA/salt-hypertensive mice developed kidney tubular injury, inflammation, fibrosis, and more moderate LV hypertrophy, fibrosis and diastolic dysfunction. RLX and BM-MSCs combined provided optimal protection against these pathologies and significantly reduced picrosirius red-stained organ fibrosis and MP2RAGE analysis of LV fibrosis. A significant correlation between MP2RAGE analysis and histologically-stained interstitial LV fibrosis was detected. It was concluded that the MP2RAGE sequence enhanced the non-invasive CMRI detection of LV fibrosis. Furthermore, combining RLX and BM-MSCs may represent a promising treatment option for hypertensive cardiorenal syndrome.
Collapse
Affiliation(s)
- Yifang Li
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Gang Zheng
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Ekaterina Salimova
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Brad R S Broughton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Stem Cells and Development Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Michael de Veer
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Stem Cells and Development Program, Monash Biomedicine Discovery Institute (BDI) and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
33
|
Xu R, Ding Z, Li H, Shi J, Cheng L, Xu H, Wu J, Zou Y. Identification of early cardiac dysfunction and heterogeneity after pressure and volume overload in mice by high-frequency echocardiographic strain imaging. Front Cardiovasc Med 2023; 9:1071249. [PMID: 36712248 PMCID: PMC9880208 DOI: 10.3389/fcvm.2022.1071249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
Object Aortic stenosis and regurgitation are clinically important conditions characterized with different hypertrophic types induced by pressure or volume overload, respectively, but with comparable cardiac function in compensated stage. Speckle-tracking based strain imaging has been applied to assess subtle alterations in cardiac abnormality, but its application in differentiating these two types of ventricular hypertrophy is still sparse. Here, we performed strain imaging analysis of cardiac remodeling in these two loading conditions. Methods C57BL/6J mice were subjected to transverse aortic constriction (TAC)-induced pressure overload or aortic regurgitation (AR)-induced volume overload. Conventional echocardiography and strain imaging were comprehensively assessed to detect stimulus-specific alterations in TAC and AR hearts. Results Conventional echocardiography did not detect significant changes in left ventricular systolic (ejection fraction and fractional shortening) and diastolic (E/E') function in either TAC or AR mice. On the contrary, global strain analysis revealed global longitudinal strain and strain rate were remarkably impaired in TAC while preserved in AR mice, although global radial, and circumferential strain and strain rate were significantly reduced in both models. Regional strain analysis in the long axis demonstrated that longitudinal strain and strain rate in all or most segments were decreased in TAC but maintained or slightly dented in AR mice, while radial strain and strain rate indicated overt decline in both models. Moreover, decreased radial and circumferential strain and strain rate were observed in most segments of TAC and AR mice in the short axis. Conclusion Strain imaging is superior to conventional echocardiography to detect subtle changes in myocardial deformation, with longitudinal strain and strain rate indicating distinct functional changes in pressure versus volume overload myocardial hypertrophy, making it potentially an advanced approach for early detection and differential diagnosis of cardiac dysfunction.
Collapse
Affiliation(s)
- Ran Xu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiwen Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hao Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Shi
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Leilei Cheng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,*Correspondence: Jian Wu,
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,Yunzeng Zou,
| |
Collapse
|
34
|
Leyba K, Paiyabhroma N, Salvas JP, Damen FW, Janvier A, Zub E, Bernis C, Rouland R, Dubois CJ, Badaut J, Richard S, Marchi N, Goergen CJ, Sicard P. Neurovascular hypoxia after mild traumatic brain injury in juvenile mice correlates with heart-brain dysfunctions in adulthood. Acta Physiol (Oxf) 2023; 238:e13933. [PMID: 36625322 DOI: 10.1111/apha.13933] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/20/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
AIM Retrospective studies suggest that mild traumatic brain injury (mTBI) in pediatric patients may lead to an increased risk of cardiac events. However, the exact functional and temporal dynamics and the associations between heart and brain pathophysiological trajectories are not understood. METHODS A single impact to the left somatosensory cortical area of the intact skull was performed on juvenile mice (17 days postnatal). Cerebral 3D photoacoustic imaging was used to measure the oxygen saturation (sO2 ) in the impacted area 4 h after mTBI followed by 2D and 4D echocardiography at days 7, 30, 90, and 190 post-impact. At 8 months, we performed a dobutamine stress test to evaluate cardiac function. Lastly, behavioral analyses were conducted 1 year after initial injury. RESULTS We report a rapid and transient decrease in cerebrovascular sO2 and increased hemoglobin in the impacted left brain cortex. Cardiac analyses showed long-term diastolic dysfunction and a diminished systolic strain response under stress in the mTBI group. At the molecular level, cardiac T-p38MAPK and troponin I expression was pathologic modified post-mTBI. We found linear correlations between brain sO2 measured immediately post-mTBI and long-term cardiac strain after 8 months. We report that initial cerebrovascular hypoxia and chronic cardiac dysfunction correlated with long-term behavioral changes hinting at anxiety-like and memory maladaptation. CONCLUSION Experimental juvenile mTBI induces time-dependent cardiac dysfunction that corresponds to the initial neurovascular sO2 dip and is associated with long-term behavioral modifications. These imaging biomarkers of the heart-brain axis could be applied to improve clinical pediatric mTBI management.
Collapse
Affiliation(s)
- Katherine Leyba
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Nitchawat Paiyabhroma
- PhyMedExp, INSERM/CNRS/Université de Montpellier, IPAM/Biocampus, Montpellier, France
| | - John P Salvas
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Frederick W Damen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Alicia Janvier
- Institute de Genomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Emma Zub
- Institute de Genomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Corinne Bernis
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Inserm/Université Paul Sabatier UMR1048, Toulouse, France
| | | | | | - Jerome Badaut
- Univ. Bordeaux, CNRS, CRMSB, UMR 5536, Bordeaux, France
| | - Sylvain Richard
- PhyMedExp, INSERM/CNRS/Université de Montpellier, IPAM/Biocampus, Montpellier, France
| | - Nicola Marchi
- Institute de Genomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Pierre Sicard
- PhyMedExp, INSERM/CNRS/Université de Montpellier, IPAM/Biocampus, Montpellier, France
| |
Collapse
|
35
|
IL-1β neutralization prevents diastolic dysfunction development, but lacks hepatoprotective effect in an aged mouse model of NASH. Sci Rep 2023; 13:356. [PMID: 36611037 PMCID: PMC9825403 DOI: 10.1038/s41598-022-26896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/21/2022] [Indexed: 01/09/2023] Open
Abstract
Interleukin-1β (IL-1β) is a key mediator of non-alcoholic steatohepatitis (NASH), a chronic liver disease, and of systemic inflammation-driven aging. IL-1β contributes to cardio-metabolic decline, and may promote hepatic oncogenic transformation. Therefore, IL-1β is a potential therapeutic target in these pathologies. We aimed to investigate the hepatic and cardiac effects of an IL-1β targeting monoclonal antibody in an aged mouse model of NASH. 24 months old male C57Bl/6J mice were fed with control or choline deficient (CDAA) diet and were treated with isotype control or anti-IL-1β Mab for 8 weeks. Cardiac functions were assessed by conventional-and 2D speckle tracking echocardiography. Liver samples were analyzed by immunohistochemistry and qRT-PCR. Echocardiography revealed improved cardiac diastolic function in anti-IL-1β treated mice with NASH. Marked hepatic fibrosis developed in CDAA-fed group, but IL-1β inhibition affected fibrosis only at transcriptomic level. Hepatic inflammation was not affected by the IL-1β inhibitor. PCNA staining revealed intensive hepatocyte proliferation in CDAA-fed animals, which was not influenced by neutralization of IL-1β. IL-1β inhibition increased hepatic expression of Pd-1 and Ctla4, while Pd-l1 expression increased in NASH. In conclusion, IL-1β inhibition improved cardiac diastolic function, but did not ameliorate features of NASH; moreover, even promoted hepatic immune checkpoint expression, with concomitant NASH-related hepatocellular proliferation.
Collapse
|
36
|
Saito T, Mizobuchi M, Kato T, Ogata H, Koiwa F, Honda H. Fibroblast Growth Factor 23 Exacerbates Cardiac Fibrosis in Deoxycorticosterone Acetate-Salt Mice With Hypertension. J Transl Med 2023; 103:100003. [PMID: 36748187 DOI: 10.1016/j.labinv.2022.100003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 01/18/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is associated with cardiovascular disease in patients with chronic kidney disease; however, the mechanisms underlying the effect of FGF23 on cardiac function remain to be investigated. Herein, we studied the effect of continuous intravenous (CIV) FGF23 loading in a deoxycorticosterone acetate (DOCA)-salt mouse model with mild chronic kidney disease and hypertension as well as heart failure with a preserved ejection fraction. Wild-type male mice were randomly allocated to 4 groups: normal control, vehicle-treated DOCA-salt mice, FGF23-treated DOCA-salt mice, and FGF23- and calcitriol-treated DOCA-salt mice. The DOCA-salt mice received the agents via the CIV route for 10 days using an infusion minipump. DOCA-salt mice that received FGF23 showed a marked increase in the serum FGF23 level, and echocardiography in these mice revealed heart failure with a preserved ejection fraction. These mice also showed exacerbation of myocardial fibrosis, concomitant with an inverse and significant correlation with Cyp27b1 expression. Calcitriol treatment attenuated FGF23-induced cardiac fibrosis and improved diastolic function via inhibition of transforming growth factor-β signaling. This effect was independent of the systemic and local levels of FGF23. These results suggest that CIV FGF23 loading exacerbates cardiac fibrosis and that locally abnormal vitamin D metabolism is involved in this mechanism. Calcitriol attenuates this exacerbation by mediating transforming growth factor-β signaling independently of the FGF23 levels.
Collapse
Affiliation(s)
- Tomohiro Saito
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masahide Mizobuchi
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Tadashi Kato
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroaki Ogata
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Fumihiko Koiwa
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
37
|
Wang X, Zhu X, Shi L, Wang J, Xu Q, Yu B, Qu A. A time-series minimally invasive transverse aortic constriction mouse model for pressure overload-induced cardiac remodeling and heart failure. Front Cardiovasc Med 2023; 10:1110032. [PMID: 36891245 PMCID: PMC9986492 DOI: 10.3389/fcvm.2023.1110032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/19/2023] [Indexed: 02/22/2023] Open
Abstract
Transverse aortic constriction (TAC) is a widely-used animal model for pressure overload-induced cardiac hypertrophy and heart failure (HF). The severity of TAC-induced adverse cardiac remodeling is correlated to the degree and duration of aorta constriction. Most studies of TAC are performed with a 27-gauge needle, which is easy to cause a tremendous left ventricular overload and leads to a rapid HF, but it is accompanied by higher mortality attributed to tighter aortic arch constriction. However, a few studies are focusing on the phenotypes of TAC applied with a 25-gauge needle, which produces a mild overload to induce cardiac remodeling and has low post-operation mortality. Furthermore, the specific timeline of HF induced by TAC applied with a 25-gauge needle in C57BL/6 J mice remains unclear. In this study, C57BL/6 J mice were randomly subjected to TAC with a 25-gauge needle or sham surgery. Echocardiography, gross morphology, and histopathology were applied to evaluate time-series phenotypes in the heart after 2, 4, 6, 8, and 12 weeks. The survival rate of mice after TAC was more than 98%. All mice subjected to TAC maintained compensated cardiac remodeling during the first two weeks and began to exhibit heart failure characteristics after 4 weeks upon TAC. At 8 weeks post-TAC, the mice showed severe cardiac dysfunction, hypertrophy, and cardiac fibrosis compared to sham mice. Moreover, the mice raised a severe dilated HF at 12 weeks. This study provides an optimized method of the mild overload TAC-induced cardiac remodeling from the compensatory period to decompensatory HF in C57BL/6 J mice.
Collapse
Affiliation(s)
- Xia Wang
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Xinxin Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Li Shi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Jingjing Wang
- Laboratory of Animal Facility, Capital Medical University, Beijing, China
| | - Qing Xu
- Core Facility Centre, Capital Medical University, Beijing, China
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| |
Collapse
|
38
|
Zhang L, Smyth D, Al-Khalaf M, Blet A, Du Q, Bernick J, Gong M, Chi X, Oh Y, Roba-Oshin M, Coletta E, Feletou M, Gramolini AO, Kim KH, Coutinho T, Januzzi JL, Tyl B, Ziegler A, Liu PP. Insulin-like growth factor-binding protein-7 (IGFBP7) links senescence to heart failure. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1195-1214. [PMID: 39196168 PMCID: PMC11358005 DOI: 10.1038/s44161-022-00181-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/02/2022] [Indexed: 08/29/2024]
Abstract
Heart failure (HF) is a rising global cardiovascular epidemic driven by aging and chronic inflammation. As elderly populations continue to increase, precision treatments for age-related cardiac decline are urgently needed. Here we report that cardiac and blood expression of IGFBP7 is robustly increased in patients with chronic HF and in an HF mouse model. In a pressure overload mouse HF model, Igfbp7 deficiency attenuated cardiac dysfunction by reducing cardiac inflammatory injury, tissue fibrosis and cellular senescence. IGFBP7 promoted cardiac senescence by stimulating IGF-1R/IRS/AKT-dependent suppression of FOXO3a, preventing DNA repair and reactive oxygen species (ROS) detoxification, thereby accelerating the progression of HF. In vivo, AAV9-shRNA-mediated cardiac myocyte Igfbp7 knockdown indicated that myocardial IGFBP7 directly regulates pathological cardiac remodeling. Moreover, antibody-mediated IGFBP7 neutralization in vivo reversed IGFBP7-induced suppression of FOXO3a, restored DNA repair and ROS detoxification signals and attenuated pressure-overload-induced HF in mice. Consequently, selectively targeting IGFBP7-regulated senescence pathways may have broad therapeutic potential for HF.
Collapse
Affiliation(s)
- Liyong Zhang
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - David Smyth
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | | | - Alice Blet
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Qiujiang Du
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Jordan Bernick
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Michael Gong
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Xu Chi
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Yena Oh
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | | | | | - Michel Feletou
- Cardiovascular and Metabolic Disease Center for Therapeutic Innovation, Institut de Recherches Internationales Servier, Suresnes, France
| | - Anthony O Gramolini
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Kyoung-Han Kim
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Thais Coutinho
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Baim Institute for Clinical Research, Boston, MA, USA
| | - Benoit Tyl
- Cardiovascular and Metabolic Disease Center for Therapeutic Innovation, Institut de Recherches Internationales Servier, Suresnes, France
| | - Andre Ziegler
- Roche Diagnostics International, Ltd., Rotkreuz, Switzerland
| | - Peter P Liu
- University of Ottawa Heart Institute, Ottawa, ON, Canada.
- Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
39
|
Greenwell AA, Tabatabaei Dakhili SA, Gopal K, Saed CT, Chan JSF, Kazungu Mugabo N, Zhabyeyev P, Eaton F, Kruger J, Oudit GY, Ussher JR. Stimulating myocardial pyruvate dehydrogenase activity fails to alleviate cardiac abnormalities in a mouse model of human Barth syndrome. Front Cardiovasc Med 2022; 9:997352. [PMID: 36211560 PMCID: PMC9537754 DOI: 10.3389/fcvm.2022.997352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Barth syndrome (BTHS) is a rare genetic disorder due to mutations in the TAFAZZIN gene, leading to impaired maturation of cardiolipin and thereby adversely affecting mitochondrial function and energy metabolism, often resulting in cardiomyopathy. In a murine model of BTHS involving short-hairpin RNA mediated knockdown of Tafazzin (TazKD mice), myocardial glucose oxidation rates were markedly reduced, likely secondary to an impairment in the activity of pyruvate dehydrogenase (PDH), the rate-limiting enzyme of glucose oxidation. Furthermore, TazKD mice exhibited cardiac hypertrophy with minimal cardiac dysfunction. Because the stimulation of myocardial glucose oxidation has been shown to alleviate diabetic cardiomyopathy and heart failure, we hypothesized that stimulating PDH activity would alleviate the cardiac hypertrophy present in TazKD mice. In order to address our hypothesis, 6-week-old male TazKD mice and their wild-type (WT) littermates were treated with dichloroacetate (DCA; 70 mM in the drinking water), which stimulates PDH activity via inhibiting PDH kinase to prevent inhibitory phosphorylation of PDH. We utilized ultrasound echocardiography to assess cardiac function and left ventricular wall structure in all mice prior to and following 6-weeks of treatment. Consistent with systemic activation of PDH and glucose oxidation, DCA treatment improved glycemia in both TazKD mice and their WT littermates, and decreased PDH phosphorylation equivalently at all 3 of its inhibitory sites (serine 293/300/232). However, DCA treatment had no impact on left ventricular structure, or systolic and diastolic function in TazKD mice. Therefore, it is unlikely that stimulating glucose oxidation is a viable target to improve BTHS-related cardiomyopathy.
Collapse
Affiliation(s)
- Amanda A. Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Seyed Amirhossein Tabatabaei Dakhili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Christina T. Saed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Jordan S. F. Chan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Nick Kazungu Mugabo
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Pavel Zhabyeyev
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Farah Eaton
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Jennifer Kruger
- Health Sciences Laboratory Animal Services, University of Alberta, Edmonton, AB, Canada
| | - Gavin Y. Oudit
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - John R. Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
- *Correspondence: John R. Ussher
| |
Collapse
|
40
|
Mohamed BA, Elkenani M, Mobarak S, Marques Rodrigues D, Annamalai K, Schnelle M, Bader M, Hasenfuss G, Toischer K. Hemodynamic stress-induced cardiac remodelling is not modulated by ablation of phosphodiesterase 4D interacting protein. J Cell Mol Med 2022; 26:4440-4452. [PMID: 35860864 PMCID: PMC9357604 DOI: 10.1111/jcmm.17468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/13/2022] [Accepted: 06/19/2022] [Indexed: 11/28/2022] Open
Abstract
Adrenergic stimulation in the heart activates the protein kinase A (PKA), which phosphorylates key proteins involved in intracellular Ca2+ handling. PKA is held in proximity to its substrates by protein scaffolds, the A kinase anchoring proteins (AKAPs). We have previously identified the transcript of phosphodiesterase 4D interacting protein (Pde4dip; also known as myomegalin), one of the sarcomeric AKAPs, as being differentially expressed following hemodynamic overload, a condition inducing hyperadrenergic state in the heart. Here, we addressed whether PDE4DIP is involved in the adverse cardiac remodelling following hemodynamic stress. Homozygous Pde4dip knockout (KO) mice, generated by CRISPR-Cas9 technology, and wild-type (WT) littermates were exposed to aortocaval shunt (shunt) or transthoracic aortic constriction (TAC) to induce hemodynamic volume overload (VO) or pressure overload (PO), respectively. The mortality, cardiac structure, function and pathological cardiac remodelling were followed up after hemodynamic injuries. The PDE4DIP protein level was markedly downregulated in volume-overloaded- but upregulated in pressure-overloaded-WT hearts. Following shunt or TAC, mortality rates were comparably increased in both genotypes. Twelve weeks after shunt or TAC, Pde4dip-KO animals showed a similar degree of cardiac hypertrophy, dilatation and dysfunction as WT mice. Cardiomyocyte hypertrophy, myocardial fibrosis, reactivation of cardiac stress genes and downregulation of ATPase, Ca2+ transporting, cardiac muscle, slow twitch 2 transcript did not differ between WT and Pde4dip-KO hearts following shunt or TAC. In summary, despite a differential expression of PDE4DIP protein in remodelled WT hearts, Pde4dip deficiency does not modulate adverse cardiac remodelling after hemodynamic VO or PO.
Collapse
Affiliation(s)
- Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Manar Elkenani
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Sherok Mobarak
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Daniel Marques Rodrigues
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Karthika Annamalai
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Moritz Schnelle
- DZHK (German Centre for Cardiovascular Research), Göttingen, Germany.,Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin-Buch, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Charité Universitätsmedizin, Berlin, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| |
Collapse
|
41
|
Abstract
Major advances in biomedical imaging have occurred over the last 2 decades and now allow many physiological, cellular, and molecular processes to be imaged noninvasively in small animal models of cardiovascular disease. Many of these techniques can be also used in humans, providing pathophysiological context and helping to define the clinical relevance of the model. Ultrasound remains the most widely used approach, and dedicated high-frequency systems can obtain extremely detailed images in mice. Likewise, dedicated small animal tomographic systems have been developed for magnetic resonance, positron emission tomography, fluorescence imaging, and computed tomography in mice. In this article, we review the use of ultrasound and positron emission tomography in small animal models, as well as emerging contrast mechanisms in magnetic resonance such as diffusion tensor imaging, hyperpolarized magnetic resonance, chemical exchange saturation transfer imaging, magnetic resonance elastography and strain, arterial spin labeling, and molecular imaging.
Collapse
Affiliation(s)
- David E Sosnovik
- Cardiology Division, Cardiovascular Research Center (D.E.S.), Massachusetts General Hospital and Harvard Medical School, Boston.,A.A. Martinos Center for Biomedical Imaging (D.E.S.), Massachusetts General Hospital and Harvard Medical School, Boston.,Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Cambridge (D.E.S.)
| | - Marielle Scherrer-Crosbie
- Cardiology Division, Hospital of the University of Pennsylvania and Perelman School of Medicine, Philadelphia (M.S.-C)
| |
Collapse
|
42
|
Romaine A, Melleby AO, Alam J, Lobert VH, Lu N, Lockwood FE, Hasic A, Lunde IG, Sjaastad I, Stenmark H, Herum KM, Gullberg D, Christensen G. Integrin α11β1 and syndecan-4 dual receptor ablation attenuates cardiac hypertrophy in the pressure overloaded heart. Am J Physiol Heart Circ Physiol 2022; 322:H1057-H1071. [PMID: 35522553 DOI: 10.1152/ajpheart.00635.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pathological myocardial hypertrophy in response to an increase in left ventricular (LV) afterload may ultimately lead to heart failure. Cell surface receptors bridge the interface between the cell and the ECM in cardiac myocytes and cardiac fibroblasts, and have been suggested to be important mediators of pathological myocardial hypertrophy. We identify for the first time that integrin α11 (α11) is preferentially upregulated amongst integrin beta 1 heterodimer-forming α subunits in response to increased afterload induced by aortic banding (AB) in wild type mice (WT). Mice were anesthetized in a chamber with 4% isoflurane and 95% oxygen before being intubated and ventilated with 2.5% isoflurane and 97% oxygen. For pre- and post-operative analgesia, animals were administered 0.02 mL buprenorphine (0.3 mg/mL) subcutaneously. Surprisingly, mice lacking α11 develop myocardial hypertrophy following AB comparable to WT. In the mice lacking α11, we further show a compensatory increase in the expression of another mechanoreceptor, syndecan-4, following AB compared to WT AB mice, indicating that syndecan-4 compensated for lack of α11. Intriguingly, mice lacking mechanoreceptors α11 and syndecan-4 show ablated myocardial hypertrophy following AB compared to WT mice. Expression of the main cardiac collagen isoforms col1a2 and col3a1 was significantly reduced in AB mice lacking mechanoreceptors α11 and syndecan-4 compared to WT AB.
Collapse
Affiliation(s)
- Andreas Romaine
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Arne Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.,Section of Physiology, Department of Molecular Medicine, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jahedul Alam
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Ning Lu
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Francesca E Lockwood
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Almira Hasic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Harald Stenmark
- Institute for Cancer Research, Oslo University Hospital, Norway
| | - Kate M Herum
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway.,Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Donald Gullberg
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
43
|
Li H, Xia YY, Xia CL, Li Z, Shi Y, Li XB, Zhang JX. Mimicking Metabolic Disturbance in Establishing Animal Models of Heart Failure With Preserved Ejection Fraction. Front Physiol 2022; 13:879214. [PMID: 35592030 PMCID: PMC9110887 DOI: 10.3389/fphys.2022.879214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/30/2022] [Indexed: 01/10/2023] Open
Abstract
Heart failure (HF), the terminal state of different heart diseases, imposed a significant health care burden worldwide. It is the last battlefield in dealing with cardiovascular diseases. HF with preserved ejection fraction (HFpEF) is a type of HF in which the symptoms and signs of HF are mainly ascribed to diastolic dysfunction of left ventricle, whereas systolic function is normal or near-normal. Compared to HF with reduced ejection fraction (HFrEF), the diagnosis and treatment of HFpEF have made limited progress, partly due to the lack of suitable animal models for translational studies in the past. Given metabolic disturbance and inflammatory burden contribute to HFpEF pathogenesis, recent years have witnessed emerging studies focusing on construction of animal models with HFpEF phenotype by mimicking metabolic disorders. These models prefer to recapitulate the metabolic disorders and endothelial dysfunction, leading to the more detailed understanding of the entity. In this review, we summarize the currently available animal models of HFpEF with metabolic disorders, as well as their advantages and disadvantages as tools for translational studies.
Collapse
Affiliation(s)
- Hui Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi-Yuan Xia
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chun-Lei Xia
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Intensive Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Shi
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Bo Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Xiao-Bo Li, ; Jun-Xia Zhang,
| | - Jun-Xia Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Xiao-Bo Li, ; Jun-Xia Zhang,
| |
Collapse
|
44
|
Getiye Y, Peterson MR, Phillips BD, Carrillo D, Bisha B, He G. E-cigarette exposure with or without heating the e-liquid induces differential remodeling in the lungs and right heart of mice. J Mol Cell Cardiol 2022; 168:83-95. [PMID: 35489388 DOI: 10.1016/j.yjmcc.2022.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/18/2022] [Accepted: 04/23/2022] [Indexed: 01/12/2023]
Abstract
Various cardiopulmonary pathologies associated with electronic cigarette (EC) vaping have been reported. This study investigated the differential adverse effects of heating-associated by-products versus the intact components of EC aerosol to the lungs and heart of mice. We further dissected the roles of caspase recruitment domain-containing protein 9 (CARD9)-associated innate immune response and NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome in EC exposure-induced cardiopulmonary injury. C57BL/6 wild type (WT), CARD9-/-, and NLRP3-/- mice were exposed to EC aerosol 3 h/day, 5 days/week for 6 month with or without heating the e-liquid with exposure to ambient air as the control. In WT mice, EC exposure with heating (EwH) significantly increased right ventricle (RV) free wall thickness at systole and diastole. However, EC exposure without heating (EwoH) caused a significant decrease in the wall thickness at systole. RV fractional shortening was also markedly reduced following EwH in WT and NLRP3-/- mice. Further, EwH activated NF-κB and p38 MAPK inflammatory signaling in the lungs, but not in the RV, in a CARD9- and NLRP3-dependent manner. Levels of circulatory inflammatory mediators were also elevated following EwH, indicating systemic inflammation. Moreover, EwoH activated TGF-β1/SMAD2/3/α-SMA fibrosis signaling in the lungs but not the RV of WT mice. In conclusion, EC aerosol exposure following EwH or EwoH induced differential cardiopulmonary remodeling and CARD9 innate immune and NLRP3 inflammasome contributed to the adverse effects.
Collapse
Affiliation(s)
- Yohannes Getiye
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, WY 82071, USA
| | - Matthew R Peterson
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, WY 82071, USA
| | - Brandon D Phillips
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, WY 82071, USA
| | - Daniel Carrillo
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, WY 82071, USA
| | - Bledar Bisha
- Department of Animal Science, University of Wyoming, Laramie, WY 82071, USA
| | - Guanglong He
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, WY 82071, USA.
| |
Collapse
|
45
|
Can E, Smith M, Boukens BJ, Coronel R, Buffenstein R, Riegler J. Naked mole-rats maintain cardiac function and body composition well into their fourth decade of life. GeroScience 2022; 44:731-746. [PMID: 35107705 PMCID: PMC9135933 DOI: 10.1007/s11357-022-00522-6] [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: 06/15/2021] [Accepted: 01/21/2022] [Indexed: 11/04/2022] Open
Abstract
The prevalence of cardiovascular disease increases exponentially with age, highlighting the contribution of aging mechanisms to cardiac diseases. Although model organisms which share human disease pathologies can elucidate mechanisms driving disease, they do not provide us with innate examples how cardiac aging might be slowed or attenuated. The identification of animal models that preserve cardiac function throughout most of life offers an alternative approach to study mechanisms which might slow cardiac aging. One such species may be the naked mole-rat (NMR), a mouse-sized (40 g) rodent with extraordinary longevity (> 37 years), and constant mortality hazard over its four decades of life. We used a cross-sectional study design to measure a range of physiological parameters in NMRs between 2 and 34 years of age and compared these findings with those of mice aged between 3 months and 2.5 years. We observed a rapid decline in body fat content and bone mineral density in old mice, but no changes in NMRs. Similarly, rhythm disorders (premature atrial and ventricular complexes) occurred in aged mice but not in NMRs. Magnetic resonance and ultrasound imaging showed age-dependent increases in cardiac hypertrophy and diastolic dysfunction in mice which were absent in NMRs. Finally, cardiac stress tests showed an age-dependent decline in normalized cardiac output in mice, which was absent in NMRs. Unlike mice, that manifest several aspects of human cardiac aging, NMRs maintain cardiac function and reserve capacity throughout their long lives and may offer insights on how to delay or prevent cardiac aging.
Collapse
Affiliation(s)
- Emine Can
- Calico Life Sciences LLC, 1170 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Megan Smith
- Calico Life Sciences LLC, 1170 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, 1105, AZ, Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6200, MD, Maastricht, The Netherlands
| | - Ruben Coronel
- Department of Experimental Cardiology, Heart Center, Academic University Medical Centers, University of Amsterdam, 1105, AZ, Amsterdam, The Netherlands
| | - Rochelle Buffenstein
- Calico Life Sciences LLC, 1170 Veterans Blvd, South San Francisco, CA, 94080, USA.
- Department of Biology, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| | - Johannes Riegler
- Calico Life Sciences LLC, 1170 Veterans Blvd, South San Francisco, CA, 94080, USA.
| |
Collapse
|
46
|
Narayan P, Trikantzopoulos E, Mezzaroma E, Mauro AG, Vohra H, Abbate A, Toldo S. The interleukin-1 receptor type I promotes the development of aging-associated cardiomyopathy in mice. Cytokine 2022; 151:155811. [DOI: 10.1016/j.cyto.2022.155811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 11/03/2022]
|
47
|
Ren D, Fedorova J, Davitt K, Van Le TN, Griffin JH, Liaw PC, Esmon CT, Rezaie AR, Li J. Activated Protein C Strengthens Cardiac Tolerance to Ischemic Insults in Aging. Circ Res 2022; 130:252-272. [PMID: 34930019 PMCID: PMC8882057 DOI: 10.1161/circresaha.121.319044] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND APC (activated protein C) is a plasma serine protease with anticoagulant and anti-inflammatory activities. EPCR (Endothelial protein C receptor) is associated with APC's activity and mediates its downstream signaling events. APC exerts cardioprotective effects during ischemia and reperfusion (I/R). This study aims to characterize the role of the APC-EPCR axis in ischemic insults in aging. METHODS Young (3-4 months) and aged (24-26 months) wild-type C57BL/6J mice, as well as EPCR point mutation (EPCRR84A/R84A) knockin C57BL/6J mice incapable of interaction with APC and its wild type of littermate C57BL/6J mice, were subjected to I/R. Wild-type APC, signaling-selective APC-2Cys, or anticoagulant-selective APC-E170A were administrated before reperfusion. RESULTS The results demonstrated that cardiac I/R reduces APC activity, and the APC activity was impaired in the aged versus young hearts possibly attributable to the declined EPCR level with aging. Serum EPCR measurement showed that I/R triggered the shedding of membrane EPCR into circulation, while administration of APC attenuated the I/R-induced EPCR shedding in both young and aged hearts. Subsequent echocardiography showed that APC and APC-2Cys but not APC-E170A ameliorated cardiac dysfunction during I/R in both young and aged mice. Importantly, APC elevated the resistance of the aged heart to ischemic insults through stabilizing EPCR. However, all these cardioprotective effects of APC were blunted in the EPCRR84A/R84A mice versus its wild-type littermates. The ex vivo working heart and metabolomics results demonstrated that AMPK (AMP-activated protein kinase) mediates acute adaptive response while AKT (protein kinase B) is involved in chronic metabolic programming in the hearts with APC treatment. CONCLUSIONS I/R stress causes shedding of the membrane EPCR in the heart, and administration of APC prevents I/R-induced cardiac EPCR shedding that is critical for limiting cardiac damage in aging.
Collapse
Affiliation(s)
- Di Ren
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Julia Fedorova
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Kayla Davitt
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Tran Ngoc Van Le
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Patricia C. Liaw
- Thrombosis and Atherosclerosis Research Institute, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Charles T. Esmon
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Alireza R. Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| |
Collapse
|
48
|
Alsalman AW, Farhan H, Qasim M. Echocardiographic assessment of left ventricle diastolic dysfunction using transmitral doppler acceleration rate of mitral inflow E-Wave. MUSTANSIRIYA MEDICAL JOURNAL 2022. [DOI: 10.4103/mj.mj_37_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
49
|
Barallobre-Barreiro J, Radovits T, Fava M, Mayr U, Lin WY, Ermolaeva E, Martínez-López D, Lindberg EL, Duregotti E, Daróczi L, Hasman M, Schmidt LE, Singh B, Lu R, Baig F, Siedlar AM, Cuello F, Catibog N, Theofilatos K, Shah AM, Crespo-Leiro MG, Doménech N, Hübner N, Merkely B, Mayr M. Extracellular Matrix in Heart Failure: Role of ADAMTS5 in Proteoglycan Remodeling. Circulation 2021; 144:2021-2034. [PMID: 34806902 PMCID: PMC8687617 DOI: 10.1161/circulationaha.121.055732] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/20/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Remodeling of the extracellular matrix (ECM) is a hallmark of heart failure (HF). Our previous analysis of the secretome of murine cardiac fibroblasts returned ADAMTS5 (a disintegrin and metalloproteinase with thrombospondin motifs 5) as one of the most abundant proteases. ADAMTS5 cleaves chondroitin sulfate proteoglycans such as versican. The contribution of ADAMTS5 and its substrate versican to HF is unknown. METHODS Versican remodeling was assessed in mice lacking the catalytic domain of ADAMTS5 (Adamts5ΔCat). Proteomics was applied to study ECM remodeling in left ventricular samples from patients with HF, with a particular focus on the effects of common medications used for the treatment of HF. RESULTS Versican and versikine, an ADAMTS-specific versican cleavage product, accumulated in patients with ischemic HF. Versikine was also elevated in a porcine model of cardiac ischemia/reperfusion injury and in murine hearts after angiotensin II infusion. In Adamts5ΔCat mice, angiotensin II infusion resulted in an aggravated versican build-up and hyaluronic acid disarrangement, accompanied by reduced levels of integrin β1, filamin A, and connexin 43. Echocardiographic assessment of Adamts5ΔCat mice revealed a reduced ejection fraction and an impaired global longitudinal strain on angiotensin II infusion. Cardiac hypertrophy and collagen deposition were similar to littermate controls. In a proteomics analysis of a larger cohort of cardiac explants from patients with ischemic HF (n=65), the use of β-blockers was associated with a reduction in ECM deposition, with versican being among the most pronounced changes. Subsequent experiments in cardiac fibroblasts confirmed that β1-adrenergic receptor stimulation increased versican expression. Despite similar clinical characteristics, patients with HF treated with β-blockers had a distinct cardiac ECM profile. CONCLUSIONS Our results in animal models and patients suggest that ADAMTS proteases are critical for versican degradation in the heart and that versican accumulation is associated with impaired cardiac function. A comprehensive characterization of the cardiac ECM in patients with ischemic HF revealed that β-blockers may have a previously unrecognized beneficial effect on cardiac chondroitin sulfate proteoglycan content.
Collapse
Affiliation(s)
- Javier Barallobre-Barreiro
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Tamás Radovits
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Budapest, Hungary (T.R., L.D., B.M.)
| | - Marika Fava
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Ursula Mayr
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Wen-Yu Lin
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
- Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (W.-Y.L.)
| | - Elizaveta Ermolaeva
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Diego Martínez-López
- IIS-Fundación Jiménez Díaz–Universidad Autónoma and CIBERCV, Madrid, Spain (D.M.-L.)
| | - Eric L. Lindberg
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (E.L.L., N.H.)
| | - Elisa Duregotti
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - László Daróczi
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Budapest, Hungary (T.R., L.D., B.M.)
| | - Maria Hasman
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Lukas E. Schmidt
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Bhawana Singh
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Ruifang Lu
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Ferheen Baig
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Aleksandra Malgorzata Siedlar
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, German Center for Heart Research (DZHK), Hamburg, Germany (F.C.)
| | - Norman Catibog
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Konstantinos Theofilatos
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Ajay M. Shah
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| | - Maria G. Crespo-Leiro
- Instituto de Investigación Biomédica de A Coruña (INIBIC)–CIBERCV, Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña, Spain (M.G.C.-L., N.D.)
| | - Nieves Doménech
- Instituto de Investigación Biomédica de A Coruña (INIBIC)–CIBERCV, Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña, Spain (M.G.C.-L., N.D.)
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany (E.L.L., N.H.)
- Charité-Universitätsmedizin, Berlin, Germany (N.H.)
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany (N.H.)
| | - Béla Merkely
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Budapest, Hungary (T.R., L.D., B.M.)
| | - Manuel Mayr
- King’s BHF Centre of Research Excellence, London, UK (J.B.-B., M.F., U.M., W.-Y.L., E.E., E.D., M.H., L.E.S., B.S., R.L., F.B., A.M.S., N.C., K.T., A.M.S., M.M.)
| |
Collapse
|
50
|
Ishikita A, Matsushima S, Ikeda S, Okabe K, Nishimura R, Tadokoro T, Enzan N, Yamamoto T, Sada M, Tsutsui Y, Miyake R, Ikeda M, Ide T, Kinugawa S, Tsutsui H. GFAT2 mediates cardiac hypertrophy through HBP-O-GlcNAcylation-Akt pathway. iScience 2021; 24:103517. [PMID: 34934932 DOI: 10.1016/j.isci.2021.103517] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 09/25/2021] [Accepted: 11/23/2021] [Indexed: 01/14/2023] Open
Abstract
Molecular mechanisms mediating cardiac hypertrophy by glucose metabolism are incompletely understood. Hexosamine biosynthesis pathway (HBP), an accessory pathway of glycolysis, is known to be involved in the attachment of O-linked N-acetylglucosamine motif (O-GlcNAcylation) to proteins, a post-translational modification. We here demonstrate that glutamine-fructose-6-phosphate amidotransferase 2 (GFAT2), a critical HBP enzyme, is a major isoform of GFAT in the heart and is increased in response to several hypertrophic stimuli, including isoproterenol (ISO). Knockdown of GFAT2 suppresses ISO-induced cardiomyocyte hypertrophy, accompanied by suppression of Akt O-GlcNAcylation and activation. Knockdown of GFAT2 does not affect anti-hypertrophic effect by Akt inhibition. Administration of glucosamine, a substrate of HBP, induces protein O-GlcNAcylation, Akt activation, and cardiomyocyte hypertrophy. In mice, 6-diazo-5-oxo-L-norleucine, an inhibitor of GFAT, attenuates ISO-induced protein O-GlcNAcylation, Akt activation, and cardiac hypertrophy. Our results demonstrate that GFAT2 mediates cardiomyocyte hypertrophy by HBP-O-GlcNAcylation-Akt pathway and could be a critical therapeutic target of cardiac hypertrophy.
Collapse
Affiliation(s)
- Akihito Ishikita
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Kosuke Okabe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryohei Nishimura
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Nobuyuki Enzan
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taishi Yamamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Sada
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitomo Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Miyake
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomomi Ide
- Department of Experimental and Clinical Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka 812-8582, Japan.,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|