1
|
Rivas VN, Vandewege MW, Ueda Y, Kaplan JL, Reader JR, Roberts JA, Stern JA. Transcriptomic and genetic profiling in a spontaneous non-human primate model of hypertrophic cardiomyopathy and sudden cardiac death. Sci Rep 2024; 14:31344. [PMID: 39733099 DOI: 10.1038/s41598-024-82770-4] [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: 09/13/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) afflicts humans, cats, pigs, and rhesus macaques. Disease sequelae include congestive heart failure, thromboembolism, and sudden cardiac death (SCD). Sarcomeric mutations explain some human and cat cases, however, the molecular basis in rhesus macaques remains unknown. RNA-Seq of the LV tissues of five HCM-affected and seven healthy control rhesus macaques was employed for differential transcriptomic analyses. DNA from 15 severely HCM-affected and 21 healthy geriatric rhesus macaques were selected for whole-genome sequencing. A genome-wide association study (GWAS) of disease status and SCD outcome was performed. 614 down- and 1,065 upregulated differentially expressed genes (DEGs) were identified between groups. The top DEG (MAFF) was overexpressed in affected animals (log2FoldChange = 4.71; PAdjusted-value = 1.14E-133). Channelopathy-associated enriched terms were identified in ~ 57% of downregulated DEGs providing transcriptomic evidence of hypertrophic and arrhythmic disease processes. For GWAS, no putative variant withstood segregation. Polygenic modeling analysis resulted in poor prediction power and burden testing could not explain HCM by an association of multiple variants in any gene. Neither single nor compound genetic variant(s), or identified polygenic profile, suggest complex genotype-phenotype interactions in rhesus macaques. Brought forth is an established dataset of robustly phenotyped rhesus macaques as an open-access resource for future cardiovascular disease genetic studies.
Collapse
Affiliation(s)
- Victor N Rivas
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Dr, Raleigh, NC, 27607, USA
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - Michael W Vandewege
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Dr, Raleigh, NC, 27607, USA
| | - Yu Ueda
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Dr, Raleigh, NC, 27607, USA
| | - Joanna L Kaplan
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - JRachel Reader
- California National Primate Research Center, University of California-Davis, Davis, CA, USA
| | - Jeffrey A Roberts
- California National Primate Research Center, University of California-Davis, Davis, CA, USA
| | - Joshua A Stern
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Dr, Raleigh, NC, 27607, USA.
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA.
| |
Collapse
|
2
|
Pius C, Niort B, Radcliffe EJ, Trafford AW. A refined, minimally invasive, reproducible ovine ischaemia-reperfusion-infarction model using implantable defibrillators: Methodology and validation. Exp Physiol 2024. [PMID: 39702979 DOI: 10.1113/ep091760] [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: 05/07/2024] [Accepted: 10/10/2024] [Indexed: 12/21/2024]
Abstract
Ischaemic heart disease remains a leading cause of premature mortality and morbidity. Understanding the associated pathophysiological mechanisms of cardiac dysfunction arising from ischaemic heart disease and the identification of sites for new therapeutic interventions requires a preclinical model that reproduces the key clinical characteristics of myocardial ischaemia, reperfusion and infarction. Here, we describe and validate a refined and minimally invasive translationally relevant approach to induce ischaemia, reperfusion and infarction in the sheep. The novelty and refinement in the procedure stems from utilization of implantable cardiac defibrillators prior to coronary engagement, balloon angioplasty to induce infarction, and intra-operative anti-arrhythmic drug protocols to reduce adverse arrhythmic events. The protocol is readily adoptable by researchers with access to standard fluoroscopic instrumentation, and it requires minimally invasive surgery. These refinements lead to a substantial reduction of intra-operative mortality to 6.7% from previously published values ranging between 13% and 43%. The model produces key characteristics associated with the fourth universal definition of myocardial infarction, including ECG changes, elevated cardiac biomarkers and cardiac wall motility defects. In conclusion, the model closely replicates the clinical paradigm of myocardial ischaemia, reperfusion and infarction in a translationally relevant large animal setting, and the applied refinements reduce the incidence of intra-operative mortality typically associated with preclinical myocardial infarction models.
Collapse
Affiliation(s)
- Charlene Pius
- Division of Cardiovascular Science, School of Medical Science, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Barbara Niort
- Division of Cardiovascular Science, School of Medical Science, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Emma J Radcliffe
- Division of Cardiovascular Science, School of Medical Science, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Andrew W Trafford
- Division of Cardiovascular Science, School of Medical Science, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
3
|
Lane KV, Dow LP, Castillo EA, Boros R, Feinstein SD, Pardon G, Pruitt BL. Cell Architecture and Dynamics of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPSC-CMs) on Hydrogels with Spatially Patterned Laminin and N-Cadherin. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39680735 DOI: 10.1021/acsami.4c11934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Controlling cellular shape with micropatterning extracellular matrix (ECM) proteins on hydrogels has been shown to improve the reproducibility of the cell structure, enhancing our ability to collect statistics on single-cell behaviors. Patterning methods have advanced efforts in developing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as a promising human model for studies of the heart structure, function, and disease. Patterned single hiPSC-CMs have exhibited phenotypes closer to mature, primary CMs across several metrics, including sarcomere alignment and contractility, area and aspect ratio, and force production. Micropatterning of hiPSC-CM pairs has shown further improvement of hiPSC-CM contractility compared to patterning single cells, suggesting that CM-CM interactions improve hiPSC-CM function. However, whether patterning single hiPSC-CMs on a protein associated with CM-CM adhesion, like N-cadherin, can drive similar enhancement of the hiPSC-CM structure and function has not been tested. To address this, we developed a novel dual-protein patterning process featuring covalent binding of proteins at the hydrogel surface to ensure robust force transfer and force sensing. The patterns comprised rectangular laminin islands for attachment across the majority of the cell area, with N-cadherin "end caps" to imitate CM-CM adherens junctions. We used this method to geometrically control single-cell CMs on deformable hydrogels suitable for traction force microscopy (TFM) to observe cellular dynamics. We seeded α-actinin::GFP-tagged hiPSC-CMs on dual-protein patterned hydrogels and verified the interaction between hiPSC-CMs and N-cadherin end caps via immunofluorescent staining. We found that hiPSC-CMs on dual-protein patterns exhibited higher cell area and contractility in the direction of sarcomere organization than those on laminin-only patterns but no difference in sarcomere organization or total force production. This work demonstrates a method for covalent patterning of multiple proteins on polyacrylamide hydrogels for mechanobiological studies. However, we conclude that N-cadherin only modestly improves single-cell patterned hiPSC-CM models and is not sufficient to elicit increases in contractility observed in hiPSC-CM pairs.
Collapse
Affiliation(s)
- Kerry V Lane
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Liam P Dow
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Erica A Castillo
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305, United States
| | - Rémi Boros
- Department of Physics, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Samuel D Feinstein
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Gaspard Pardon
- AGORA Cancer Research Center, Swiss Federal Institute of Technology of Lausanne, Lausanne CH-1011, Switzerland
| | - Beth L Pruitt
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
4
|
Williams J, Iheagwam FN, Maroney SP, Schmitt LR, Brown RD, Krafsur GM, Frid MG, McCabe MC, Gandjeva A, Williams KJ, Luyendyk JP, Saviola AJ, Tuder RM, Stenmark K, Hansen KC. A bovine model of hypoxia-induced pulmonary hypertension reveals a gradient of immune and matrisome response with a complement signature found in circulation. Am J Physiol Cell Physiol 2024; 327:C1666-C1680. [PMID: 39495247 DOI: 10.1152/ajpcell.00274.2024] [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: 04/29/2024] [Revised: 08/19/2024] [Accepted: 09/11/2024] [Indexed: 11/05/2024]
Abstract
Pulmonary hypertension (PH) is a progressive vascular disease characterized by vascular remodeling, stiffening, and luminal obstruction, driven by dysregulated cell proliferation, inflammation, and extracellular matrix (ECM) alterations. Despite the recognized contribution of ECM dysregulation to PH pathogenesis, the precise molecular alterations in the matrisome remain poorly understood. In this study, we employed a matrisome-focused proteomics approach to map the protein composition in a young bovine calf model of acute hypoxia-induced PH. Our findings reveal distinct alterations in the matrisome along the pulmonary vascular axis, with the most prominent changes observed in the main pulmonary artery. Key alterations included a strong immune response and wound repair signature, characterized by increased levels of complement components, coagulation cascade proteins, and provisional matrix markers. In addition, we observed upregulation of ECM-modifying enzymes, growth factors, and core ECM proteins implicated in vascular stiffening, such as collagens, periostin, tenascin-C, and fibrin(ogen). Notably, these alterations correlated with increased mean pulmonary arterial pressure and vascular remodeling. In the plasma, we identified increased levels of complement components, indicating a systemic inflammatory response accompanying the vascular remodeling. Our findings shed light on the dynamic matrisome remodeling in early-stage PH, implicating a wound-healing trajectory with distinct patterns from the main pulmonary artery to the distal vasculature. This study provides novel insights into the immune cell infiltration and matrisome alterations associated with PH pathogenesis and highlights potential biomarkers and therapeutic targets within the matrisome landscape.NEW & NOTEWORTHY Extensive immune cell infiltration and matrisome alterations associated with hypoxia-induced pulmonary hypertension in a large mammal model. Matrisome components correlate with increased resistance to identify candidate alterations that drive biomechanical manifestations of the disease.
Collapse
Affiliation(s)
- Jason Williams
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Franklyn N Iheagwam
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Sean P Maroney
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - R Dale Brown
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Greta M Krafsur
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Maxwell C McCabe
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Aneta Gandjeva
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kurt J Williams
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, United States
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, United States
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Rubin M Tuder
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Denver, Colorado, United States
| | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| |
Collapse
|
5
|
Grisorio L, Bongianino R, Gianeselli M, Priori SG. Gene therapy for cardiac diseases: methods, challenges, and future directions. Cardiovasc Res 2024; 120:1664-1682. [PMID: 39302117 DOI: 10.1093/cvr/cvae207] [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: 04/18/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 09/22/2024] Open
Abstract
Gene therapy is advancing at an unprecedented pace, and the recent success of clinical trials reinforces optimism and trust among the scientific community. Recently, the cardiac gene therapy pipeline, which had progressed more slowly than in other fields, has begun to advance, overcoming biological and technical challenges, particularly in treating genetic heart pathologies. The primary rationale behind the focus on monogenic cardiac diseases is the well-defined molecular mechanisms driving their phenotypes, directly linked to the pathogenicity of single genetic mutations. This aspect makes these conditions a remarkable example of 'genetically druggable' diseases. Unfortunately, current treatments for these life-threatening disorders are few and often poorly effective, underscoring the need to develop therapies to modulate or correct their molecular substrates. In this review we examine the latest advancements in cardiac gene therapy, discussing the pros and cons of different molecular approaches and delivery vectors, with a focus on their therapeutic application in cardiac inherited diseases. Additionally, we highlight the key factors that may enhance clinical translation, drawing insights from previous trials and the current prospects of cardiac gene therapy.
Collapse
Affiliation(s)
- Luca Grisorio
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
| | - Rossana Bongianino
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
| | - Matteo Gianeselli
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA
| | - Silvia Giuliana Priori
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
- Molecular Cardiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/ Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| |
Collapse
|
6
|
Namekata I, Seki M, Saito T, Odaka R, Hamaguchi S, Tanaka H. Automaticity of the Pulmonary Vein Myocardium and the Effect of Class I Antiarrhythmic Drugs. Int J Mol Sci 2024; 25:12367. [PMID: 39596432 PMCID: PMC11595185 DOI: 10.3390/ijms252212367] [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: 10/21/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The pulmonary vein wall contains a myocardial layer whose ectopic automaticity is the major cause of atrial fibrillation. This review summarizes the results obtained in isolated pulmonary vein myocardium from small experimental animals, focusing on the studies with the guinea pig. The diversity in the action potential waveform reflects the difference in the repolarizing potassium channel currents involved. The diastolic depolarization, the trigger of automatic action potentials, is caused by multiple membrane currents, including the Na+-Ca2+ exchanger current and late INa. The action potential waveform and automaticity are affected differentially by α- and β-adrenoceptor stimulation. Class I antiarrhythmic drugs block the propagation of ectopic electrical activity of the pulmonary vein myocardium through blockade of the peak INa. Some of the class I antiarrhythmic drugs block the late INa and inhibit pulmonary vein automaticity. The negative inotropic and chronotropic effects of class I antiarrhythmic drugs could be largely attributed to their blocking effect on the Ca2+ channel rather than the Na+ channel. Such a comprehensive understanding of pulmonary vein automaticity and class I antiarrhythmic drugs would lead to an improvement in pharmacotherapy and the development of novel therapeutic agents for atrial fibrillation.
Collapse
Affiliation(s)
| | | | | | | | | | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama Funabashi, Chiba 274-8510, Japan; (I.N.); (M.S.); (T.S.); (R.O.); (S.H.)
| |
Collapse
|
7
|
Ewoldt JK, DePalma SJ, Jewett ME, Karakan MÇ, Lin YM, Mir Hashemian P, Gao X, Lou L, McLellan MA, Tabares J, Ma M, Salazar Coariti AC, He J, Toussaint KC, Bifano TG, Ramaswamy S, White AE, Agarwal A, Lejeune E, Baker BM, Chen CS. Induced pluripotent stem cell-derived cardiomyocyte in vitro models: benchmarking progress and ongoing challenges. Nat Methods 2024:10.1038/s41592-024-02480-7. [PMID: 39516564 DOI: 10.1038/s41592-024-02480-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 09/15/2024] [Indexed: 11/16/2024]
Abstract
Recent innovations in differentiating cardiomyocytes from human induced pluripotent stem cells (hiPSCs) have unlocked a viable path to creating in vitro cardiac models. Currently, hiPSC-derived cardiomyocytes (hiPSC-CMs) remain immature, leading many in the field to explore approaches to enhance cell and tissue maturation. Here, we systematically analyzed 300 studies using hiPSC-CM models to determine common fabrication, maturation and assessment techniques used to evaluate cardiomyocyte functionality and maturity and compiled the data into an open-access database. Based on this analysis, we present the diversity of, and current trends in, in vitro models and highlight the most common and promising practices for functional assessments. We further analyzed outputs spanning structural maturity, contractile function, electrophysiology and gene expression and note field-wide improvements over time. Finally, we discuss opportunities to collectively pursue the shared goal of hiPSC-CM model development, maturation and assessment that we believe are critical for engineering mature cardiac tissue.
Collapse
Affiliation(s)
- Jourdan K Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Maggie E Jewett
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - M Çağatay Karakan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Yih-Mei Lin
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA
| | - Paria Mir Hashemian
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Xining Gao
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Lihua Lou
- Department of Mechanical and Material Engineering, Florida International University, Miami, FL, USA
| | - Micheal A McLellan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jonathan Tabares
- Department of Physics, Florida International University, Miami, FL, USA
| | - Marshall Ma
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | | | - Jin He
- Department of Physics, Florida International University, Miami, FL, USA
| | - Kimani C Toussaint
- School of Engineering, Brown University, Providence, RI, USA
- Brown-Lifespan Center for Digital Health, Providence, RI, USA
| | - Thomas G Bifano
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Sharan Ramaswamy
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA
| | - Alice E White
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
- Division of Materials Science and Engineering, Boston University, Boston, MA, USA
- Department of Physics, Boston University, Boston, MA, USA
| | - Arvind Agarwal
- Department of Mechanical and Material Engineering, Florida International University, Miami, FL, USA
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
8
|
Hirakis SP, Bartol TM, Autin L, Amaro RE, Sejnowski TJ. Electrophysical cardiac remodeling at the molecular level: Insights into ryanodine receptor activation and calcium-induced calcium release from a stochastic explicit-particle model. Biophys J 2024; 123:3812-3831. [PMID: 39369273 PMCID: PMC11560313 DOI: 10.1016/j.bpj.2024.09.029] [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: 04/28/2023] [Revised: 07/03/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024] Open
Abstract
We present the first-ever, fully discrete, stochastic model of triggered cardiac Ca2+ dynamics. Using anatomically accurate subcellular cardiac myocyte geometries, we simulate the molecular players involved in Ca2+ handling using high-resolution stochastic and explicit-particle methods at the level of an individual cardiac dyadic junction. Integrating data from multiple experimental sources, the model not only replicates the findings of traditional in silico studies and complements in vitro experimental data but also reveals new insights into the molecular mechanisms driving cardiac dysfunction under stress and disease conditions. We improve upon older, nondiscrete models using the same realistic geometry by incorporating molecular mechanisms for spontaneous, as well as triggered calcium-induced calcium release (CICR). Action potentials are used to activate L-type calcium channels (LTCC), triggering CICR through ryanodine receptors (RyRs) on the surface of the sarcoplasmic reticulum. These improvements allow for the specific focus on the couplon: the structure-function relationship between LTCC and RyR. We investigate the electrophysical effects of normal and diseased action potentials on CICR and interrogate the effects of dyadic junction deformation through detubulation and orphaning of RyR. Our work demonstrates the importance of the electrophysical integrity of the calcium release unit on CICR fidelity, giving insights into the molecular basis of heart disease. Finally, we provide a unique, detailed, molecular view of the CICR process using advanced rendering techniques. This easy-to-use model comes complete with tutorials and the necessary software for use and analysis to maximize usability and reproducibility. Our work focuses on quantifying, qualifying, and visualizing the behavior of the molecular species that underlie the function and dysfunction of subcellular cardiomyocyte systems.
Collapse
Affiliation(s)
- Sophia P Hirakis
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California; Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California
| | - Thomas M Bartol
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California
| | - Ludovic Autin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California.
| | - Terrence J Sejnowski
- Computational Neurobiology Lab, The Salk Institute of Biological Studies, La Jolla, California; Department of Chemistry and Biochemistry, The University of California San Diego, La Jolla, California.
| |
Collapse
|
9
|
Hu Y, Zou Y, Qiao L, Lin L. Integrative proteomic and metabolomic elucidation of cardiomyopathy with in vivo and in vitro models and clinical samples. Mol Ther 2024; 32:3288-3312. [PMID: 39233439 PMCID: PMC11489546 DOI: 10.1016/j.ymthe.2024.08.030] [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: 04/30/2024] [Revised: 07/16/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Cardiomyopathy is a prevalent cardiovascular disease that affects individuals of all ages and can lead to life-threatening heart failure. Despite its variety in types, each with distinct characteristics and causes, our understanding of cardiomyopathy at a systematic biology level remains incomplete. Mass spectrometry-based techniques have emerged as powerful tools, providing a comprehensive view of the molecular landscape and aiding in the discovery of biomarkers and elucidation of mechanisms. This review highlights the significant potential of integrating proteomic and metabolomic approaches with specialized databases to identify biomarkers and therapeutic targets across different types of cardiomyopathies. In vivo and in vitro models, such as genetically modified mice, patient-derived or induced pluripotent stem cells, and organ chips, are invaluable in exploring the pathophysiological complexities of this disease. By integrating omics approaches with these sophisticated modeling systems, our comprehension of the molecular underpinnings of cardiomyopathy can be greatly enhanced, facilitating the development of diagnostic markers and therapeutic strategies. Among the promising therapeutic targets are those involved in extracellular matrix remodeling, sarcomere damage, and metabolic remodeling. These targets hold the potential to advance precision therapy in cardiomyopathy, offering hope for more effective treatments tailored to the specific molecular profiles of patients.
Collapse
Affiliation(s)
- Yiwei Hu
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China
| | - Yunzeng Zou
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Liang Qiao
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Ling Lin
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| |
Collapse
|
10
|
Vazquez-Gonzalez KI, Ochoa-Brust A, Rodriguez-Hernandez A, Ventura-Cisneros H, Lino-Lopez GJ, Barbosa-Valdovinos R, Rodriguez-Vazquez A, Vazquez-Vuelvas OF, Felix RA, Jiménez-Vargas JM, Melnikov V, Valdez-Velazquez LL. Cardiac alterations induced by Heloderma horridum horridum venom in rats: An experimental study with ECG analysis using a linear regression algorithm. Toxicon 2024; 249:108062. [PMID: 39127082 DOI: 10.1016/j.toxicon.2024.108062] [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: 02/22/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Envenomation by reptile venom, particularly from lizards, poses significant health risks and can lead to physiological and cardiovascular changes. The venom of Heloderma horridum horridum, endemic to Colima, Mexico, was tested on Wistar rats. Electrocardiographic (ECG) data were collected pre-treatment and at 5-min intervals for 1 h post-envenomation. A specially designed computational linear regression algorithm (LRA) was used for the segmentation analysis of the ECG data to improve the detection of fiducial points (P, Q, R, S, and T) in ECG waves. Additionally, heart tissue was analyzed for macroscopic and microscopic changes. The results revealed significant electrocardiographic alterations, including pacemaker migration, junctional extrasystoles, and intraventricular conduction aberrations. By applying a linear regression algorithm, the study compensated for noise and anomalies in the isoelectric line in an ECG signal, improving the detection of P and T waves and the QRS complex with an efficiency of 97.5%. Cardiac enzyme evaluation indicated no statistically significant differences between the control and experimental groups. Macroscopic and microscopic examination revealed no apparent signs of damage or inflammatory responses in heart tissues. This study enhances our understanding of the cardiovascular impact of Heloderma venom, suggesting a greater influence on changes in conduction and arrhythmias than on direct cardiac damage to the myocardium.
Collapse
Affiliation(s)
| | - Alberto Ochoa-Brust
- Facultad de Ingeniería Mecánica Eléctrica, Universidad de Colima, Kilometro 9 Carretera Colima-Coquimatlan, Coquimatlan, Colima, 28400, Mexico
| | | | - Hugo Ventura-Cisneros
- Clinica ISSTE Hospital Dr. Miguel Trejo Ochoa. Calle Ignacio Sandoval 867, Lomas de Circunvalación, 28010, Colima, Colima, Mexico
| | - Gisela Jareth Lino-Lopez
- Centro Nacional de Referencia de Control Biológico, Dirección General de Sanidad Vegetal SENASICA SADER, Colima, 28110, Mexico
| | | | - Armando Rodriguez-Vazquez
- Unidad de Manejo para el Aprovechamiento de la Vida Silvestre en el Parque del "Palapo" Domicilio Conocido P-75 Agua Zarca, Coquimatlan, 28400, Mexico
| | - Oscar F Vazquez-Vuelvas
- Facultad de Ciencias Químicas, Universidad de Colima, Kilometro 9 Carretera Colima-Coquimatlan, Coquimatlan, Colima, 28400, Mexico
| | - Ramon A Felix
- Facultad de Ingeniería Mecánica Eléctrica, Universidad de Colima, Kilometro 9 Carretera Colima-Coquimatlan, Coquimatlan, Colima, 28400, Mexico
| | - Juana M Jiménez-Vargas
- Facultad de Ciencias Químicas, Universidad de Colima, Kilometro 9 Carretera Colima-Coquimatlan, Coquimatlan, Colima, 28400, Mexico
| | - Valery Melnikov
- Facultad de Medicina, Universidad de Colima, Av. Universidad 333, Colonia las Viboras, Colima, 28040, Mexico.
| | - Laura Leticia Valdez-Velazquez
- Facultad de Medicina, Universidad de Colima, Av. Universidad 333, Colonia las Viboras, Colima, 28040, Mexico; Facultad de Ciencias Químicas, Universidad de Colima, Kilometro 9 Carretera Colima-Coquimatlan, Coquimatlan, Colima, 28400, Mexico.
| |
Collapse
|
11
|
Hamaguchi S, Agata N, Seki M, Namekata I, Tanaka H. Developmental Changes in the Excitation-Contraction Mechanisms of the Ventricular Myocardium and Their Sympathetic Regulation in Small Experimental Animals. J Cardiovasc Dev Dis 2024; 11:267. [PMID: 39330325 PMCID: PMC11432613 DOI: 10.3390/jcdd11090267] [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: 05/15/2024] [Revised: 08/02/2024] [Accepted: 08/11/2024] [Indexed: 09/28/2024] Open
Abstract
The developmental changes in the excitation-contraction mechanisms of the ventricular myocardium of small animals (guinea pig, rat, mouse) and their sympathetic regulation will be summarized. The action potential duration monotonically decreases during pre- and postnatal development in the rat and mouse, while in the guinea pig it decreases during the fetal stage but turns into an increase just before birth. Such changes can be attributed to changes in the repolarizing potassium currents. The T-tubule and the sarcoplasmic reticulum are scarcely present in the fetal cardiomyocyte, but increase during postnatal development. This causes a developmental shift in the Ca2+ handling from a sarcolemma-dependent mechanism to a sarcoplasmic reticulum-dependent mechanism. The sensitivity for beta-adrenoceptor-mediated positive inotropy decreases during early postnatal development, which parallels the increase in sympathetic nerve innervation. The alpha-adrenoceptor-mediated inotropy in the mouse changes from positive in the neonate to negative in the adult. This can be explained by the change in the excitation-contraction mechanism mentioned above. The shortening of the action potential duration enhances trans-sarcolemmal Ca2+ extrusion by the Na+-Ca2+ exchanger. The sarcoplasmic reticulum-dependent mechanism of contraction in the adult allows Na+-Ca2+ exchanger activity to cause negative inotropy, a mechanism not observed in neonatal myocardium. Such developmental studies would provide clues towards a more comprehensive understanding of cardiac function.
Collapse
Affiliation(s)
| | | | | | | | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University, Funabashi 274-8510, Japan; (S.H.); (N.A.); (M.S.); (I.N.)
| |
Collapse
|
12
|
Moiroux-Sahraoui A, Manicone F, Herpain A. How preclinical models help to improve outcome in cardiogenic shock. Curr Opin Crit Care 2024; 30:333-339. [PMID: 38841979 DOI: 10.1097/mcc.0000000000001170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW Preclinical experimentation of cardiogenic shock resuscitation on large animal models represents a powerful tool to decipher its complexity and improve its poor outcome, when small animal models are lacking external validation, and clinical investigation are limited due to technical and ethical constraints. This review illustrates the currently available preclinical models addressing reliably the physiopathology and hemodynamic phenotype of cardiogenic shock, highlighting on the opposite questionable translation based on low severity acute myocardial infarction (AMI) models. RECENT FINDINGS Three types of preclinical models replicate reliably AMI-related cardiogenic shock, either with coronary microembolization, coronary deoxygenated blood perfusion or double critical coronary sub-occlusion. These models overcame the pitfall of frequent periprocedural cardiac arrest and offer, to different extents, robust opportunities to investigate pharmacological and/or mechanical circulatory support therapeutic strategies, cardioprotective approaches improving heart recovery and mitigation of the systemic inflammatory reaction. They all came with their respective strengths and weaknesses, allowing the researcher to select the right preclinical model for the right clinical question. SUMMARY AMI-related cardiogenic shock preclinical models are now well established and should replace low severity AMI models. Technical and ethical constraints are not trivial, but this translational research is a key asset to build up meaningful future clinical investigations.
Collapse
Affiliation(s)
- Alexander Moiroux-Sahraoui
- Experimental Laboratory of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
- Department of Cardiac Surgery, Institut de Cardiologie, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Francesca Manicone
- Experimental Laboratory of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
| | - Antoine Herpain
- Experimental Laboratory of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
- Department of Intensive Care, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
13
|
Sabe SA, Harris DD, Broadwin M, Sellke FW. Cardioprotection in cardiovascular surgery. Basic Res Cardiol 2024; 119:545-568. [PMID: 38856733 DOI: 10.1007/s00395-024-01062-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/11/2024]
Abstract
Since the invention of cardiopulmonary bypass, cardioprotective strategies have been investigated to mitigate ischemic injury to the heart during aortic cross-clamping and reperfusion injury with cross-clamp release. With advances in cardiac surgical and percutaneous techniques and post-operative management strategies including mechanical circulatory support, cardiac surgeons are able to operate on more complex patients. Therefore, there is a growing need for improved cardioprotective strategies to optimize outcomes in these patients. This review provides an overview of the basic principles of cardioprotection in the setting of cardiac surgery, including mechanisms of cardiac injury in the context of cardiopulmonary bypass, followed by a discussion of the specific approaches to optimizing cardioprotection in cardiac surgery, including refinements in cardiopulmonary bypass and cardioplegia, ischemic conditioning, use of specific anesthetic and pharmaceutical agents, and novel mechanical circulatory support technologies. Finally, translational strategies that investigate cardioprotection in the setting of cardiac surgery will be reviewed, with a focus on promising research in the areas of cell-based and gene therapy. Advances in this area will help cardiologists and cardiac surgeons mitigate myocardial ischemic injury, improve functional post-operative recovery, and optimize clinical outcomes in patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Rhode Island Hospital, Alpert Medical School of Brown University, 2 Dudley Street, MOC 360, Providence, RI, 02905, USA
| | - Dwight D Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Rhode Island Hospital, Alpert Medical School of Brown University, 2 Dudley Street, MOC 360, Providence, RI, 02905, USA
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Rhode Island Hospital, Alpert Medical School of Brown University, 2 Dudley Street, MOC 360, Providence, RI, 02905, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Rhode Island Hospital, Alpert Medical School of Brown University, 2 Dudley Street, MOC 360, Providence, RI, 02905, USA.
| |
Collapse
|
14
|
Singh M, Roubertie F, Ozturk C, Borchiellini P, Rames A, Bonnemain J, Gollob SD, Wang SX, Naulin J, El Hamrani D, Dugot-Senant N, Gosselin I, Grenet C, L'Heureux N, Roche ET, Kawecki F. Hemodynamic evaluation of biomaterial-based surgery for Tetralogy of Fallot using a biorobotic heart, in silico, and ovine models. Sci Transl Med 2024; 16:eadk2936. [PMID: 38985852 DOI: 10.1126/scitranslmed.adk2936] [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: 08/23/2023] [Revised: 03/12/2024] [Accepted: 06/20/2024] [Indexed: 07/12/2024]
Abstract
Tetralogy of Fallot is a congenital heart disease affecting newborns and involves stenosis of the right ventricular outflow tract (RVOT). Surgical correction often widens the RVOT with a transannular enlargement patch, but this causes issues including pulmonary valve insufficiency and progressive right ventricle failure. A monocusp valve can prevent pulmonary regurgitation; however, valve failure resulting from factors including leaflet design, morphology, and immune response can occur, ultimately resulting in pulmonary insufficiency. A multimodal platform to quantitatively evaluate the effect of shape, size, and material on clinical outcomes could optimize monocusp design. This study introduces a benchtop soft biorobotic heart model, a computational fluid model of the RVOT, and a monocusp valve made from an entirely biological cell-assembled extracellular matrix (CAM) to tackle the multifaceted issue of monocusp failure. The hydrodynamic and mechanical performance of RVOT repair strategies was assessed in biorobotic and computational platforms. The monocusp valve design was validated in vivo in ovine models through echocardiography, cardiac magnetic resonance, and catheterization. These models supported assessment of surgical feasibility, handling, suturability, and hemodynamic and mechanical monocusp capabilities. The CAM-based monocusp offered a competent pulmonary valve with regurgitation of 4.6 ± 0.9% and a transvalvular pressure gradient of 4.3 ± 1.4 millimeters of mercury after 7 days of implantation in sheep. The biorobotic heart model, in silico analysis, and in vivo RVOT modeling allowed iteration in monocusp design not now feasible in a clinical environment and will support future surgical testing of biomaterials for complex congenital heart malformations.
Collapse
Affiliation(s)
- Manisha Singh
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - François Roubertie
- IHU Liryc, Electrophysiology and Heart Modeling Institute, F-33604 Pessac, France
- Congenital Heart Diseases Department, CHU de Bordeaux, F-33604 Pessac, France
| | - Caglar Ozturk
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Paul Borchiellini
- University of Bordeaux, INSERM, BioTis, U1026, F-33000 Bordeaux, France
| | - Adeline Rames
- University of Bordeaux, INSERM, BioTis, U1026, F-33000 Bordeaux, France
| | - Jean Bonnemain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Adult Intensive Care Medicine, Lausanne University Hospital and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Samuel Dutra Gollob
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sophie X Wang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Jérôme Naulin
- IHU Liryc, Electrophysiology and Heart Modeling Institute, F-33604 Pessac, France
| | - Dounia El Hamrani
- IHU Liryc, Electrophysiology and Heart Modeling Institute, F-33604 Pessac, France
| | - Nathalie Dugot-Senant
- Plateforme d'histopathologie, TBMcore INSERM US005-CNRS 3427, F-33000 Bordeaux, France
| | - Isalyne Gosselin
- Plateforme d'histopathologie, TBMcore INSERM US005-CNRS 3427, F-33000 Bordeaux, France
| | - Célia Grenet
- University of Bordeaux, INSERM, BioTis, U1026, F-33000 Bordeaux, France
| | - Nicolas L'Heureux
- University of Bordeaux, INSERM, BioTis, U1026, F-33000 Bordeaux, France
| | - Ellen T Roche
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Fabien Kawecki
- University of Bordeaux, INSERM, BioTis, U1026, F-33000 Bordeaux, France
| |
Collapse
|
15
|
Mihaylova A, Shopova D, Parahuleva N, Yaneva A, Bakova D. (3D) Bioprinting-Next Dimension of the Pharmaceutical Sector. Pharmaceuticals (Basel) 2024; 17:797. [PMID: 38931464 PMCID: PMC11206453 DOI: 10.3390/ph17060797] [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: 05/11/2024] [Revised: 05/26/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
To create a review of the published scientific literature on the benefits and potential perspectives of the use of 3D bio-nitrification in the field of pharmaceutics. This work was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for reporting meta-analyses and systematic reviews. The scientific databases PubMed, Scopus, Google Scholar, and ScienceDirect were used to search and extract data using the following keywords: 3D bioprinting, drug research and development, personalized medicine, pharmaceutical companies, clinical trials, drug testing. The data points to several aspects of the application of bioprinting in pharmaceutics were reviewed. The main applications of bioprinting are in the development of new drug molecules as well as in the preparation of personalized drugs, but the greatest benefits are in terms of drug screening and testing. Growth in the field of 3D printing has facilitated pharmaceutical applications, enabling the development of personalized drug screening and drug delivery systems for individual patients. Bioprinting presents the opportunity to print drugs on demand according to the individual needs of the patient, making the shape, structure, and dosage suitable for each of the patient's physical conditions, i.e., print specific drugs for controlled release rates; print porous tablets to reduce swallowing difficulties; make transdermal microneedle patches to reduce patient pain; and so on. On the other hand, bioprinting can precisely control the distribution of cells and biomaterials to build organoids, or an Organ-on-a-Chip, for the testing of drugs on printed organs mimicking specified disease characteristics instead of animal testing and clinical trials. The development of bioprinting has the potential to offer customized drug screening platforms and drug delivery systems meeting a range of individualized needs, as well as prospects at different stages of drug development and patient therapy. The role of bioprinting in preclinical and clinical testing of drugs is also of significant importance in terms of shortening the time to launch a medicinal product on the market.
Collapse
Affiliation(s)
- Anna Mihaylova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Dobromira Shopova
- Department of Prosthetic Dentistry, Faculty of Dental Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Nikoleta Parahuleva
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Antoniya Yaneva
- Department of Medical Informatics, Biostatistics and eLearning, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Desislava Bakova
- Department of Healthcare Management, Faculty of Public Health, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| |
Collapse
|
16
|
Galloway CSL, Coates KD, Dawkins TG. Pace yourself: murine model insights into atrial fibrillation in endurance athletes. J Physiol 2024; 602:2691-2693. [PMID: 38717307 DOI: 10.1113/jp286420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
Affiliation(s)
- Camille S L Galloway
- School of Health and Exercise Sciences, University of British Columbia - Okanagan, Kelowna, Canada
| | - Kyla D Coates
- School of Health and Exercise Sciences, University of British Columbia - Okanagan, Kelowna, Canada
| | - Tony G Dawkins
- School of Health and Exercise Sciences, University of British Columbia - Okanagan, Kelowna, Canada
| |
Collapse
|
17
|
Zhao J, Qi L, Yuan S, Irving TC, Ma W. Differences in thick filament activation in fast rodent skeletal muscle and slow porcine cardiac muscle. J Physiol 2024; 602:2751-2762. [PMID: 38695322 PMCID: PMC11178443 DOI: 10.1113/jp286072] [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: 11/30/2023] [Accepted: 04/16/2024] [Indexed: 06/15/2024] Open
Abstract
There is a growing appreciation that regulation of muscle contraction requires both thin filament and thick filament activation in order to fully activate the sarcomere. The prevailing mechano-sensing model for thick filament activation was derived from experiments on fast-twitch muscle. We address the question whether, or to what extent, this mechanism can be extrapolated to the slow muscle in the hearts of large mammals, including humans. We investigated the similarities and differences in structural signatures of thick filament activation in porcine myocardium as compared to fast rat extensor digitorum longus (EDL) skeletal muscle under relaxed conditions and sub-maximal contraction using small angle X-ray diffraction. Thick and thin filaments were found to adopt different structural configurations under relaxing conditions, and myosin heads showed different changes in configuration upon sub-maximal activation, when comparing the two muscle types. Titin was found to have an X-ray diffraction signature distinct from those of the overall thick filament backbone, and its spacing change appeared to be positively correlated to the force exerted on the thick filament. Structural changes in fast EDL muscle were found to be consistent with the mechano-sensing model. In porcine myocardium, however, the structural basis of mechano-sensing is blunted suggesting the need for additional activation mechanism(s) in slow cardiac muscle. These differences in thick filament regulation can be related to their different physiological roles where fast muscle is optimized for rapid, burst-like, contractions, and the slow cardiac muscle in large mammalian hearts adopts a more finely tuned, graded response to allow for their substantial functional reserve. KEY POINTS: Both thin filament and thick filament activation are required to fully activate the sarcomere. Thick and thin filaments adopt different structural configurations under relaxing conditions, and myosin heads show different changes in configuration upon sub-maximal activation in fast extensor digitorum longus muscle and slow porcine cardiac muscle. Titin has an X-ray diffraction signature distinct from those of the overall thick filament backbone and this titin reflection spacing change appeared to be directly proportional to the force exerted on the thick filament. Mechano-sensing is blunted in porcine myocardium suggesting the need for additional activation mechanism(s) in slow cardiac muscle. Fast skeletal muscle is optimized for rapid, burst-like contractions, and the slow cardiac muscle in large mammalian hearts adopts a more finely tuned graded response to allow for their substantial functional reserve.
Collapse
Affiliation(s)
- Jing Zhao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Lin Qi
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Shengyao Yuan
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Thomas C Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL, USA
| | - Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
18
|
Haberbusch M, Kronsteiner B, Aigner P, Kiss A, Podesser BK, Moscato F. Importance of cardiac-synchronized vagus nerve stimulation parameters on the provoked chronotropic response for different levels of cardiac innervation. Front Physiol 2024; 15:1379936. [PMID: 38835728 PMCID: PMC11148559 DOI: 10.3389/fphys.2024.1379936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction The influence of vagus nerve stimulation (VNS) parameters on provoked cardiac effects in different levels of cardiac innervation is not well understood yet. This study examines the effects of VNS on heart rate (HR) modulation across a spectrum of cardiac innervation states, providing data for the potential optimization of VNS in cardiac therapies. Materials and Methods Utilizing previously published data from VNS experiments on six sheep with intact innervation, and data of additional experiments in five rabbits post bilateral rostral vagotomy, and four isolated rabbit hearts with additionally removed sympathetic influences, the study explored the impact of diverse VNS parameters on HR. Results Significant differences in physiological threshold charges were identified across groups: 0.09 ± 0.06 μC for intact, 0.20 ± 0.04 μC for vagotomized, and 9.00 ± 0.75 μC for isolated hearts. Charge was a key determinant of HR reduction across all innervation states, with diminishing correlations from intact (r = 0.7) to isolated hearts (r = 0.44). An inverse relationship was observed for the number of pulses, with its influence growing in conditions of reduced innervation (intact r = 0.11, isolated r = 0.37). Frequency and stimulation delay showed minimal correlations (r < 0.17) in all conditions. Conclusion Our study highlights for the first time that VNS parameters, including stimulation intensity, pulse width, and pulse number, crucially modulate heart rate across different cardiac innervation states. Intensity and pulse width significantly influence heart rate in innervated states, while pulse number is key in denervated states. Frequency and delay have less impact impact across all innervation states. These findings suggest the importance of customizing VNS therapy based on innervation status, offering insights for optimizing cardiac neuromodulation.
Collapse
Affiliation(s)
- Max Haberbusch
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Bettina Kronsteiner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Philipp Aigner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Bruno Karl Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Francesco Moscato
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
19
|
Hennis K, Piantoni C, Biel M, Fenske S, Wahl-Schott C. Pacemaker Channels and the Chronotropic Response in Health and Disease. Circ Res 2024; 134:1348-1378. [PMID: 38723033 PMCID: PMC11081487 DOI: 10.1161/circresaha.123.323250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Loss or dysregulation of the normally precise control of heart rate via the autonomic nervous system plays a critical role during the development and progression of cardiovascular disease-including ischemic heart disease, heart failure, and arrhythmias. While the clinical significance of regulating changes in heart rate, known as the chronotropic effect, is undeniable, the mechanisms controlling these changes remain not fully understood. Heart rate acceleration and deceleration are mediated by increasing or decreasing the spontaneous firing rate of pacemaker cells in the sinoatrial node. During the transition from rest to activity, sympathetic neurons stimulate these cells by activating β-adrenergic receptors and increasing intracellular cyclic adenosine monophosphate. The same signal transduction pathway is targeted by positive chronotropic drugs such as norepinephrine and dobutamine, which are used in the treatment of cardiogenic shock and severe heart failure. The cyclic adenosine monophosphate-sensitive hyperpolarization-activated current (If) in pacemaker cells is passed by hyperpolarization-activated cyclic nucleotide-gated cation channels and is critical for generating the autonomous heartbeat. In addition, this current has been suggested to play a central role in the chronotropic effect. Recent studies demonstrate that cyclic adenosine monophosphate-dependent regulation of HCN4 (hyperpolarization-activated cyclic nucleotide-gated cation channel isoform 4) acts to stabilize the heart rate, particularly during rapid rate transitions induced by the autonomic nervous system. The mechanism is based on creating a balance between firing and recently discovered nonfiring pacemaker cells in the sinoatrial node. In this way, hyperpolarization-activated cyclic nucleotide-gated cation channels may protect the heart from sinoatrial node dysfunction, secondary arrhythmia of the atria, and potentially fatal tachyarrhythmia of the ventricles. Here, we review the latest findings on sinoatrial node automaticity and discuss the physiological and pathophysiological role of HCN pacemaker channels in the chronotropic response and beyond.
Collapse
Affiliation(s)
- Konstantin Hennis
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Chiara Piantoni
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Stefanie Fenske
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Christian Wahl-Schott
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
20
|
Steele AG, Taccola G, Frazier AM, Manzella M, Hogan M, Horner PJ, Faraji AH, Sayenko DG. Mapping lumbar efferent and afferent spinal circuitries via paddle array in a porcine model. J Neurosci Methods 2024; 405:110104. [PMID: 38447914 PMCID: PMC10990770 DOI: 10.1016/j.jneumeth.2024.110104] [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: 10/12/2023] [Revised: 02/04/2024] [Accepted: 03/03/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Preclinical models are essential for identifying changes occurring after neurologic injury and assessing therapeutic interventions. Yucatan miniature pigs (minipigs) have brain and spinal cord dimensions like humans and are useful for laboratory-to-clinic studies. Yet, little work has been done to map spinal sensorimotor distributions and identify similarities and differences between the porcine and human spinal cords. NEW METHODS To characterize efferent and afferent signaling, we implanted a conventional 32-contact, four-column array into the dorsal epidural space over the lumbosacral spinal cord, spanning the L5-L6 vertebrae, in two Yucatan minipigs. Spinally evoked motor potentials were recorded bilaterally in four hindlimb muscles during stimulation delivered from different array locations. Then, cord dorsum potentials were recorded via the array by stimulating the left and right tibial nerves. RESULTS Utilizing epidural spinal stimulation, we achieved selective left, right, proximal, and distal activation in the hindlimb muscles. We then determined the selectivity of each muscle as a function of stimulation location which relates to the distribution of the lumbar motor pools. COMPARISON WITH EXISTING METHODS Mapping motoneuron distribution to hindlimb muscles and recording responses to peripheral nerve stimulation in the dorsal epidural space reveals insights into ascending and descending signal propagation in the lumbar spinal cord. Clinical-grade arrays have not been utilized in a porcine model. CONCLUSIONS These results indicate that efferent and afferent spinal sensorimotor networks are spatially distinct, provide information about the organization of motor pools in the lumbar enlargement, and demonstrate the feasibility of using clinical-grade devices in large animal research.
Collapse
Affiliation(s)
- A G Steele
- Department of Neurosurgery, Center for Translational Neural Prosthetics and Interfaces, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States; Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States
| | - G Taccola
- Department of Neurosurgery, Center for Translational Neural Prosthetics and Interfaces, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States; Neuroscience Department, International School for Advanced Studies (SISSA), Bonomea, Trieste, Italy
| | - A M Frazier
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States
| | - M Manzella
- Bostion Scientific, Valencia, CA 91355, United States
| | - M Hogan
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States
| | - P J Horner
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States
| | - A H Faraji
- Department of Neurosurgery, Center for Translational Neural Prosthetics and Interfaces, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States; Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States
| | - D G Sayenko
- Department of Neurosurgery, Center for Translational Neural Prosthetics and Interfaces, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States; Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX 77030, United States.
| |
Collapse
|
21
|
Wilhelm TI, Lewalter T, Reiser J, Werner J, Keil A, Oesterlein T, Gleirscher L, Tiemann K, Jilek C. Influence of Heart Rate and Change in Wavefront Direction through Pacing on Conduction Velocity and Voltage Amplitude in a Porcine Model: A High-Density Mapping Study. J Pers Med 2024; 14:473. [PMID: 38793055 PMCID: PMC11122149 DOI: 10.3390/jpm14050473] [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: 02/27/2024] [Revised: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Understanding the dynamics of conduction velocity (CV) and voltage amplitude (VA) is crucial in cardiac electrophysiology, particularly for substrate-based catheter ablations targeting slow conduction zones and low voltage areas. This study utilizes ultra-high-density mapping to investigate the impact of heart rate and pacing location on changes in the wavefront direction, CV, and VA of healthy pig hearts. METHODS We conducted in vivo electrophysiological studies on four healthy juvenile pigs, involving various pacing locations and heart rates. High-resolution electroanatomic mapping was performed during intrinsic normal sinus rhythm (NSR) and electrical pacing. The study encompassed detailed analyses at three levels: entire heart cavities, subregions, and localized 5-mm-diameter circular areas. Linear mixed-effects models were used to analyze the influence of heart rate and pacing location on CV and VA in different regions. RESULTS An increase in heart rate correlated with an increase in conduction velocity and a decrease in voltage amplitude. Pacing influenced conduction velocity and voltage amplitude. Pacing also influenced conduction velocity and voltage amplitude, with varying effects observed based on the pacing location within different heart cavities. Pacing from the right atrium (RA) decreased CV in all heart cavities. The overall CV and VA changes in the whole heart cavities were not uniformly reflected in all subregions and subregional CV and VA changes were not always reflected in the overall analysis. Overall, there was a notable variability in absolute CV and VA changes attributed to pacing. CONCLUSIONS Heart rate and pacing location influence CV and VA within healthy juvenile pig hearts. Subregion analysis suggests that specific regions of the heart cavities are more susceptible to pacing. High-resolution mapping aids in detecting regional changes, emphasizing the substantial physiological variations in CV and VA.
Collapse
Affiliation(s)
- Theresa Isabelle Wilhelm
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (K.T.)
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Medical Graduate Center, TUM School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany
| | - Thorsten Lewalter
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (K.T.)
- Department of Medicine, University of Bonn, 53127 Bonn, Germany
| | - Judith Reiser
- Center for Preclinical Research, TUM School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany; (J.R.)
| | - Julia Werner
- Center for Preclinical Research, TUM School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany; (J.R.)
| | - Andreas Keil
- Boston Scientific Medizintechnik GmbH, 40468 Düsseldorf, Germany
| | | | - Lukas Gleirscher
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (K.T.)
| | - Klaus Tiemann
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (K.T.)
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany
| | - Clemens Jilek
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (K.T.)
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
22
|
Baron V, Sommer ST, Fiegle DJ, Pfeuffer AKM, Peyronnet R, Volk T, Seidel T. Effects of electro-mechanical uncouplers, hormonal stimulation and pacing rate on the stability and function of cultured rabbit myocardial slices. Front Bioeng Biotechnol 2024; 12:1363538. [PMID: 38646013 PMCID: PMC11026719 DOI: 10.3389/fbioe.2024.1363538] [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: 12/30/2023] [Accepted: 03/18/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction: Recent advances have enabled organotypic culture of beating human myocardial slices that are stable for weeks. However, human myocardial samples are rare, exhibit high variability and frequently originate from diseased hearts. Thus, there is a need to adapt long-term slice culture for animal myocardium. When applied to animal cardiac slices, studies in healthy or genetically modified myocardium will be possible. We present the culture of slices from rabbit hearts, which resemble the human heart in microstructure, electrophysiology and excitation-contraction coupling. Methods: Left ventricular myocardium from New Zealand White rabbits was cut using a vibratome and cultured in biomimetic chambers for up to 7 days (d). Electro-mechanical uncoupling agents 2,3-butanedione monoxime (BDM) and cytochalasin D (CytoD) were added during initiation of culture and effects on myocyte survival were quantified. We investigated pacing rates (0.5 Hz, 1 Hz, and 2 Hz) and hormonal supplements (cortisol, T3, catecholamines) at physiological plasma concentrations. T3 was buffered using BSA. Contractile force was recorded continuously. Glucose consumption and lactate production were measured. Whole-slice Ca2+ transients and action potentials were recorded. Effects of culture on microstructure were investigated with confocal microscopy and image analysis. Results: Protocols for human myocardial culture resulted in sustained contracture and myocyte death in rabbit slices within 24 h, which could be prevented by transient application of a combination of BDM and CytoD. Cortisol stabilized contraction amplitude and kinetics in culture. T3 and catecholaminergic stimulation did not further improve stability. T3 and higher pacing rates increased metabolic rate and lactate production. T3 stabilized the response to β-adrenergic stimulation over 7 d. Pacing rates above 1 Hz resulted in progredient decline in contraction force. Image analysis revealed no changes in volume fractions of cardiomyocytes or measures of fibrosis over 7 d. Ca2+ transient amplitudes and responsiveness to isoprenaline were comparable after 1 d and 7 d, while Ca2+ transient duration was prolonged after 7 d in culture. Conclusions: A workflow for rabbit myocardial culture has been established, preserving function for up to 7 d. This research underscores the importance of glucocorticoid signaling in maintaining tissue function and extending culture duration. Furthermore, BDM and CytoD appear to protect from tissue damage during the initiation phase of tissue culture.
Collapse
Affiliation(s)
- V. Baron
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - S. T. Sommer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - D. J. Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - A.-K. M. Pfeuffer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - R. Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - T. Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - T. Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
23
|
Tran P, Linekar A, Dandekar U, Barker T, Balasubramanian S, Bhaskara-Pillai J, Shelley S, Maddock H, Banerjee P. Profiling the Biomechanical Responses to Workload on the Human Myocyte to Explore the Concept of Myocardial Fatigue and Reversibility: Rationale and Design of the POWER Heart Failure Study. J Cardiovasc Transl Res 2024; 17:275-286. [PMID: 37126208 PMCID: PMC10150683 DOI: 10.1007/s12265-023-10391-9] [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: 11/24/2022] [Accepted: 04/20/2023] [Indexed: 05/02/2023]
Abstract
It remains unclear why some patients develop heart failure without evidence of structural damage. One theory relates to impaired myocardial energetics and ventricular-arterial decoupling as the heart works against adverse mechanical load. In this original study, we propose the novel concept of myocardial fatigue to capture this phenomenon and aim to investigate this using human cardiomyocytes subjected to a modern work-loop contractility model that closely mimics in vivo cardiac cycles. This proof-of-concept study (NCT04899635) will use human myocardial tissue samples from patients undergoing cardiac surgery to develop a reproducible protocol to isolate robust calcium-tolerant cardiomyocytes. Thereafter, work-loop contractility experiments will be performed over a range of preload, afterload and cycle frequency as a function of time to elicit any reversible reduction in contractile performance (i.e. fatigue). This will provide novel insight into mechanisms behind heart failure and myocardial recovery and serve as a valuable research platform in translational cardiovascular research.
Collapse
Affiliation(s)
- Patrick Tran
- Centre for Sport, Exercise & Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, UK.
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry, UK.
| | - Adam Linekar
- Centre for Sport, Exercise & Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, UK
- InoCardia Ltd, TechnoCentre, Puma Way, Coventry, UK
| | - Uday Dandekar
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Thomas Barker
- Centre for Sport, Exercise & Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, UK
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Sendhil Balasubramanian
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Jain Bhaskara-Pillai
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Sharn Shelley
- Centre for Sport, Exercise & Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, UK
- InoCardia Ltd, TechnoCentre, Puma Way, Coventry, UK
| | - Helen Maddock
- Centre for Sport, Exercise & Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, UK
- InoCardia Ltd, TechnoCentre, Puma Way, Coventry, UK
| | - Prithwish Banerjee
- Centre for Sport, Exercise & Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, UK
- Cardiology Department, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
24
|
van Doorn ECH, Amesz JH, Sadeghi AH, de Groot NMS, Manintveld OC, Taverne YJHJ. Preclinical Models of Cardiac Disease: A Comprehensive Overview for Clinical Scientists. Cardiovasc Eng Technol 2024; 15:232-249. [PMID: 38228811 PMCID: PMC11116217 DOI: 10.1007/s13239-023-00707-w] [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: 03/02/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
For recent decades, cardiac diseases have been the leading cause of death and morbidity worldwide. Despite significant achievements in their management, profound understanding of disease progression is limited. The lack of biologically relevant and robust preclinical disease models that truly grasp the molecular underpinnings of cardiac disease and its pathophysiology attributes to this stagnation, as well as the insufficiency of platforms that effectively explore novel therapeutic avenues. The area of fundamental and translational cardiac research has therefore gained wide interest of scientists in the clinical field, while the landscape has rapidly evolved towards an elaborate array of research modalities, characterized by diverse and distinctive traits. As a consequence, current literature lacks an intelligible and complete overview aimed at clinical scientists that focuses on selecting the optimal platform for translational research questions. In this review, we present an elaborate overview of current in vitro, ex vivo, in vivo and in silico platforms that model cardiac health and disease, delineating their main benefits and drawbacks, innovative prospects, and foremost fields of application in the scope of clinical research incentives.
Collapse
Affiliation(s)
- Elisa C H van Doorn
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jorik H Amesz
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Amir H Sadeghi
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Natasja M S de Groot
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Yannick J H J Taverne
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
25
|
Ghita-Pettigrew M, Edgar KS, Kuburas R, Brown KH, Walls GM, Facchi C, Grieve DJ, Watson CJ, McWilliam A, van Herk M, Williams KJ, Butterworth KT. Dose-dependent changes in cardiac function, strain and remodelling in a preclinical model of heart base irradiation. Radiother Oncol 2024; 193:110113. [PMID: 38301958 DOI: 10.1016/j.radonc.2024.110113] [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: 08/15/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND AND PURPOSE Radiation induced cardiotoxicity (RICT) is as an important sequela of radiotherapy to the thorax for patients. In this study, we aim to investigate the dose and fractionation response of RICT. We propose global longitudinal strain (GLS) as an early indicator of RICT and investigate myocardial deformation following irradiation. METHODS RICT was investigated in female C57BL/6J mice in which the base of the heart was irradiated under image-guidance using a small animal radiation research platform (SARRP). Mice were randomly assigned to a treatment group: single-fraction dose of 16 Gy or 20 Gy, 3 consecutive fractions of 8.66 Gy, or sham irradiation; biological effective doses (BED) used were 101.3 Gy, 153.3 Gy and 101.3 Gy respectively. Longitudinal transthoracic echocardiography (TTE) was performed from baseline up to 50 weeks post-irradiation to detect structural and functional effects. RESULTS Irradiation of the heart base leads to BED-dependent changes in systolic and diastolic function 50 weeks post-irradiation. GLS showed significant decreases in a BED-dependent manner for all irradiated animals, as early as 10 weeks after irradiation. Early changes in GLS indicate late changes in cardiac function. BED-independent increases were observed in the left ventricle (LV) mass and volume and myocardial fibrosis. CONCLUSIONS Functional features of RICT displayed a BED dependence in this study. GLS showed an early change at 10 weeks post-irradiation. Cardiac remodelling was observed as increases in mass and volume of the LV, further supporting our hypothesis that dose to the base of the heart drives the global heart toxicity.
Collapse
Affiliation(s)
- Mihaela Ghita-Pettigrew
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom.
| | - Kevin S Edgar
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Refik Kuburas
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Kathryn H Brown
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Gerard M Walls
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom; Cancer Centre Belfast City Hospital, Belfast Health & Social Care Trust, Lisburn Road, Belfast, Northern Ireland
| | - Cecilia Facchi
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - David J Grieve
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Alan McWilliam
- Department of Radiotherapy Related Research, University of Manchester, Manchester, United Kingdom
| | - Marcel van Herk
- Department of Radiotherapy Related Research, University of Manchester, Manchester, United Kingdom
| | - Kaye J Williams
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - Karl T Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
26
|
He L. Biomaterials for Regenerative Cranioplasty: Current State of Clinical Application and Future Challenges. J Funct Biomater 2024; 15:84. [PMID: 38667541 PMCID: PMC11050949 DOI: 10.3390/jfb15040084] [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: 02/10/2024] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Acquired cranial defects are a prevalent condition in neurosurgery and call for cranioplasty, where the missing or defective cranium is replaced by an implant. Nevertheless, the biomaterials in current clinical applications are hardly exempt from long-term safety and comfort concerns. An appealing solution is regenerative cranioplasty, where biomaterials with/without cells and bioactive molecules are applied to induce the regeneration of the cranium and ultimately repair the cranial defects. This review examines the current state of research, development, and translational application of regenerative cranioplasty biomaterials and discusses the efforts required in future research. The first section briefly introduced the regenerative capacity of the cranium, including the spontaneous bone regeneration bioactivities and the presence of pluripotent skeletal stem cells in the cranial suture. Then, three major types of biomaterials for regenerative cranioplasty, namely the calcium phosphate/titanium (CaP/Ti) composites, mineralised collagen, and 3D-printed polycaprolactone (PCL) composites, are reviewed for their composition, material properties, and findings from clinical trials. The third part discusses perspectives on future research and development of regenerative cranioplasty biomaterials, with a considerable portion based on issues identified in clinical trials. This review aims to facilitate the development of biomaterials that ultimately contribute to a safer and more effective healing of cranial defects.
Collapse
Affiliation(s)
- Lizhe He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| |
Collapse
|
27
|
Roth K, Liu W, LeBar K, Ahern M, Wang Z. Establishment of a Biaxial Testing System for Characterization of Right Ventricle Viscoelasticity Under Physiological Loadings. Cardiovasc Eng Technol 2024:10.1007/s13239-024-00722-5. [PMID: 38468114 DOI: 10.1007/s13239-024-00722-5] [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: 03/10/2022] [Accepted: 02/19/2024] [Indexed: 03/13/2024]
Abstract
PURPOSE Prior studies have indicated an impact of cardiac muscle viscoelasticity on systolic and diastolic functions. However, the studies of ventricular free wall viscoelasticity, particularly for that of right ventricles (RV), are limited. Moreover, investigations on ventricular passive viscoelasticity have been restricted to large animals and there is a lack of data on rodent species. To fill this knowledge gap, this study aims to develop a biaxial tester that induces high-speed physiological deformations to characterize the passive viscoelasticity of rat RVs. METHODS The biaxial testing system was fabricated so that planar deformation of rat ventricle tissues at physiological strain rates was possible. The testing system was validated using isotropic polydimethylsiloxane (PDMS) sheets. Next, viscoelastic measurements were performed in healthy rat RV free walls by equibiaxial cyclic sinusoidal loadings and stress relaxation. RESULTS The biaxial tester's consistency, accuracy, and stability was confirmed from the PDMS samples measurements. Moreover, significant viscoelastic alterations of the RV were found between sub-physiological (0.1 Hz) and physiological frequencies (1-8 Hz). From hysteresis loop analysis, we found as the frequency increased, the elasticity and viscosity were increased in both directions. Interestingly, the ratio of storage energy to dissipated energy (Wd/Ws) remained constant at 0.1-5 Hz. We did not observe marked differences in healthy RV viscoelasticity between longitudinal and circumferential directions. CONCLUSION This work provides a new experimental tool to quantify the passive, biaxial viscoelasticity of ventricle free walls in both small and large animals. The dynamic mechanical tests showed frequency-dependent elastic and viscous behaviors of healthy rat RVs. But the ratio of dissipated energy to stored energy was maintained between frequencies. These findings offer novel baseline information on the passive viscoelasticity of healthy RVs in adult rats.
Collapse
Affiliation(s)
- Kellan Roth
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Wenqiang Liu
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Kristen LeBar
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Matt Ahern
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Zhijie Wang
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA.
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
28
|
Whitworth CP, Polacheck WJ. Vascular organs-on-chip made with patient-derived endothelial cells: technologies to transform drug discovery and disease modeling. Expert Opin Drug Discov 2024; 19:339-351. [PMID: 38117223 PMCID: PMC10922379 DOI: 10.1080/17460441.2023.2294947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Vascular diseases impart a tremendous burden on healthcare systems in the United States and across the world. Efforts to improve therapeutic interventions are hindered by limitations of current experimental models. The integration of patient-derived cells with organ-on-chip (OoC) technology is a promising avenue for preclinical drug screening that improves upon traditional cell culture and animal models. AREAS COVERED The authors review induced pluripotent stem cells (iPSC) and blood outgrowth endothelial cells (BOEC) as two sources for patient-derived endothelial cells (EC). They summarize several studies that leverage patient-derived EC and OoC for precision disease modeling of the vasculature, with a focus on applications for drug discovery. They also highlight the utility of patient-derived EC in other translational endeavors, including ex vivo organogenesis and multi-organ-chip integration. EXPERT OPINION Precision disease modeling continues to mature in the academic space, but end-use by pharmaceutical companies is currently limited. To fully realize their transformative potential, OoC systems must balance their complexity with their ability to integrate with the highly standardized and high-throughput experimentation required for drug discovery and development.
Collapse
Affiliation(s)
- Chloe P Whitworth
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
29
|
Chen L, Liu J, Rastegarpouyani H, Janssen PML, Pinto JR, Taylor KA. Structure of mavacamten-free human cardiac thick filaments within the sarcomere by cryoelectron tomography. Proc Natl Acad Sci U S A 2024; 121:e2311883121. [PMID: 38386705 PMCID: PMC10907299 DOI: 10.1073/pnas.2311883121] [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: 07/16/2023] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Heart muscle has the unique property that it can never rest; all cardiomyocytes contract with each heartbeat which requires a complex control mechanism to regulate cardiac output to physiological requirements. Changes in calcium concentration regulate the thin filament activation. A separate but linked mechanism regulates the thick filament activation, which frees sufficient myosin heads to bind the thin filament, thereby producing the required force. Thick filaments contain additional nonmyosin proteins, myosin-binding protein C and titin, the latter being the protein that transmits applied tension to the thick filament. How these three proteins interact to control thick filament activation is poorly understood. Here, we show using 3-D image reconstruction of frozen-hydrated human cardiac muscle myofibrils lacking exogenous drugs that the thick filament is structured to provide three levels of myosin activation corresponding to the three crowns of myosin heads in each 429Å repeat. In one crown, the myosin heads are almost completely activated and disordered. In another crown, many myosin heads are inactive, ordered into a structure called the interacting heads motif. At the third crown, the myosin heads are ordered into the interacting heads motif, but the stability of that motif is affected by myosin-binding protein C. We think that this hierarchy of control explains many of the effects of length-dependent activation as well as stretch activation in cardiac muscle control.
Collapse
Affiliation(s)
- Liang Chen
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
| | - Jun Liu
- Microbial Sciences Institute, Yale University, West Haven, CT06516
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT06536
| | - Hosna Rastegarpouyani
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Paul M. L. Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State College of Medicine, Florida State University, Tallahassee, FL32306
| | - Kenneth A. Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| |
Collapse
|
30
|
Smuder AJ. Doxorubicin-related effects on cardiorespiratory function and body composition. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 38:100360. [PMID: 38371271 PMCID: PMC10871714 DOI: 10.1016/j.ahjo.2024.100360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Affiliation(s)
- Ashley J. Smuder
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| |
Collapse
|
31
|
Li Y, Hu K, Li Y, Lu C, Guo Y, Wang W. The rodent models of arteriovenous fistula. Front Cardiovasc Med 2024; 11:1293568. [PMID: 38304139 PMCID: PMC10830807 DOI: 10.3389/fcvm.2024.1293568] [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: 09/13/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
Arteriovenous fistulas (AVFs) have long been used as dialysis access in patients with end-stage renal disease; however, their maturation and long-term patency still fall short of clinical needs. Rodent models are irreplaceable to facilitate the study of mechanisms and provide reliable insights into clinical problems. The ideal rodent AVF model recapitulates the major features and pathology of human disease as closely as possible, and pre-induction of the uremic milieu is an important addition to AVF failure studies. Herein, we review different surgical methods used so far to create AVF in rodents, including surgical suturing, needle puncture, and the cuff technique. We also summarize commonly used evaluations after AVF placement. The aim was to provide recent advances and ideas for better selection and induction of rodent AVF models. At the same time, further improvements in the models and a deeper understanding of AVF failure mechanisms are expected.
Collapse
Affiliation(s)
- Yuxuan Li
- Departmentof Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Hu
- Departmentof Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqing Li
- Departmentof Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chanjun Lu
- Department of General Vascular Surgery, Wuhan No.1 Hospital & Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Yi Guo
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weici Wang
- Departmentof Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Dos Santos JC, Alves JEF, de Azevedo RDS, de Lima ML, de Oliveira Silva MR, da Silva JG, da Silva JM, de Carvalho Correia AC, do Carmo Alves de Lima M, de Oliveira JF, de Moura RO, de Almeida SMV. Study of nitrogen heterocycles as DNA/HSA binder, topoisomerase inhibitors and toxicological safety. Int J Biol Macromol 2024; 254:127651. [PMID: 37949265 DOI: 10.1016/j.ijbiomac.2023.127651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
Four new nitrogen-containing heterocyclic derivatives (acridine, quinoline, indole, pyridine) were synthesized and their biological properties were evaluated. The compounds showed affinity for DNA and HSA, with CAIC and CAAC displaying higher binding constants (Kb) of 9.54 × 104 and 1.06 × 106, respectively. The fluorescence quenching assay (Ksv) revealed suppression values ranging from 0.34 to 0.64 × 103 M-1 for ethidium bromide (EB) and 0.1 to 0.34 × 103 M-1 for acridine orange (AO). Molecular docking confirmed the competition of the derivatives with intercalation probes at the same binding site. At 10 μM concentrations, the derivatives inhibited topoisomerase IIα activity. In the antiproliferative assays, the compounds demonstrated activity against MCF-7 and T47-D tumor cells and nonhemolytic profile. Regarding toxicity, no acute effects were observed in the embryos. However, some compounds caused enzymatic and cardiac changes, particularly the CAIC, which increased SOD activity and altered heart rate compared to the control. These findings suggest potential antitumor action of the derivatives and indicate that substituting the acridine core with different cores does not interfere with their interaction and topoisomerase inhibition. Further investigations are required to assess possible toxicological effects, including reactive oxygen species generation.
Collapse
Affiliation(s)
- Jéssica Celerino Dos Santos
- Molecular Biology Laboratory, University of Pernambuco (UPE), Multicampi Garanhuns, Garanhuns, PE 55290-000, Brazil
| | | | | | - Maksuelly Libanio de Lima
- Molecular Biology Laboratory, University of Pernambuco (UPE), Multicampi Garanhuns, Garanhuns, PE 55290-000, Brazil
| | | | - Josefa Gerlane da Silva
- Molecular Biology Laboratory, University of Pernambuco (UPE), Multicampi Garanhuns, Garanhuns, PE 55290-000, Brazil
| | - Jamire Muriel da Silva
- Department of Pharmacy, Laboratory of Synthesis and Vectorization of Molecules, State University of Paraíba (UEPB), Campus Campina Grande, 58429-500, PB, Brazil
| | | | - Maria do Carmo Alves de Lima
- Chemistry and Therapeutic Innovation Laboratory (LQIT), Department of Antibiotics, Federal University of Pernambuco, Recife, PE, Brazil
| | | | - Ricardo Olímpio de Moura
- Department of Pharmacy, Laboratory of Synthesis and Vectorization of Molecules, State University of Paraíba (UEPB), Campus Campina Grande, 58429-500, PB, Brazil
| | - Sinara Mônica Vitalino de Almeida
- Molecular Biology Laboratory, University of Pernambuco (UPE), Multicampi Garanhuns, Garanhuns, PE 55290-000, Brazil; Keizo Asami Immunopathology Laboratory (LIKA), Federal University of Pernambuco, Recife, PE, Brazil; Chemistry and Therapeutic Innovation Laboratory (LQIT), Department of Antibiotics, Federal University of Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
33
|
Deir S, Mozhdehbakhsh Mofrad Y, Mashayekhan S, Shamloo A, Mansoori-Kermani A. Step-by-step fabrication of heart-on-chip systems as models for cardiac disease modeling and drug screening. Talanta 2024; 266:124901. [PMID: 37459786 DOI: 10.1016/j.talanta.2023.124901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases are caused by hereditary factors, environmental conditions, and medication-related issues. On the other hand, the cardiotoxicity of drugs should be thoroughly examined before entering the market. In this regard, heart-on-chip (HOC) systems have been developed as a more efficient and cost-effective solution than traditional methods, such as 2D cell culture and animal models. HOCs must replicate the biology, physiology, and pathology of human heart tissue to be considered a reliable platform for heart disease modeling and drug testing. Therefore, many efforts have been made to find the best methods to fabricate different parts of HOCs and to improve the bio-mimicry of the systems in the last decade. Beating HOCs with different platforms have been developed and techniques, such as fabricating pumpless HOCs, have been used to make HOCs more user-friendly systems. Recent HOC platforms have the ability to simultaneously induce and record electrophysiological stimuli. Additionally, systems including both heart and cancer tissue have been developed to investigate tissue-tissue interactions' effect on cardiac tissue response to cancer drugs. In this review, all steps needed to be considered to fabricate a HOC were introduced, including the choice of cellular resources, biomaterials, fabrication techniques, biomarkers, and corresponding biosensors. Moreover, the current HOCs used for modeling cardiac diseases and testing the drugs are discussed. We finally introduced some suggestions for fabricating relatively more user-friendly HOCs and facilitating the commercialization process.
Collapse
Affiliation(s)
- Sara Deir
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Yasaman Mozhdehbakhsh Mofrad
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Amir Shamloo
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran.
| | | |
Collapse
|
34
|
Guo Y, Gharibani P, Agarwal P, Cho S, Thakor NV, Geocadin RG. Hyperacute autonomic and cortical function recovery following cardiac arrest resuscitation in a rodent model. Ann Clin Transl Neurol 2023; 10:2223-2237. [PMID: 37776065 PMCID: PMC10723251 DOI: 10.1002/acn3.51907] [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: 08/04/2023] [Revised: 08/29/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
OBJECTIVE There is a complex interaction between nervous and cardiovascular systems, but sparse data exist on brain-heart electrophysiological responses to cardiac arrest resuscitation. Our aim was to investigate dynamic changes in autonomic and cortical function during hyperacute stage post-resuscitation. METHODS Ten rats were resuscitated from 7-min cardiac arrest, as indicators of autonomic response, heart rate (HR), and its variability (HRV) were measured. HR was monitored through continuous electrocardiography, while HRV was assessed via spectral analysis, whereby the ratio of low-/high-frequency (LF/HF) power indicates the balance between sympathetic/parasympathetic activities. Cortical response was evaluated by continuous electroencephalography and quantitative analysis. Parameters were quantified at 5-min intervals over the first-hour post-resuscitation. Neurological outcome was assessed by Neurological Deficit Score (NDS, range 0-80, higher = better outcomes) at 4-h post-resuscitation. RESULTS A significant increase in HR was noted over 15-30 min post-resuscitation (p < 0.01 vs.15-min, respectively) and correlated with higher NDS (rs = 0.56, p < 0.01). LF/HF ratio over 15-20 min was positively correlated with NDS (rs = 0.75, p < 0.05). Gamma band power surged over 15-30 min post-resuscitation (p < 0.05 vs. 0-15 min, respectively), and gamma band fraction during this period was associated with NDS (rs ≥0.70, p < 0.05, respectively). Significant correlations were identified between increased HR and gamma band power during 15-30 min (rs ≥0.83, p < 0.01, respectively) and between gamma band fraction and LF/HF ratio over 15-20 min post-resuscitation (rs = 0.85, p < 0.01). INTERPRETATIONS Hyperacute recovery of autonomic and cortical function is associated with favorable functional outcomes. While this observation needs further validation, it presents a translational opportunity for better autonomic and neurologic monitoring during early periods post-resuscitation to develop novel interventions.
Collapse
Affiliation(s)
- Yu Guo
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Payam Gharibani
- Division of Neuroimmunology, Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Prachi Agarwal
- Department of Electrical and Computer EngineeringJohns Hopkins Whiting School of EngineeringBaltimoreMarylandUSA
| | - Sung‐Min Cho
- Departments of Neurology, Anesthesiology‐Critical Care Medicine and NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Nitish V. Thakor
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Romergryko G. Geocadin
- Departments of Neurology, Anesthesiology‐Critical Care Medicine and NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
35
|
Siamwala JH, Pagano FS, Dubielecka PM, Ivey MJ, Guirao-Abad JP, Zhao A, Chen S, Granston H, Jeong JY, Rounds S, Kanisicak O, Sadayappan S, Gilbert RJ. IL-1β-mediated adaptive reprogramming of endogenous human cardiac fibroblasts to cells with immune features during fibrotic remodeling. Commun Biol 2023; 6:1200. [PMID: 38001239 PMCID: PMC10673909 DOI: 10.1038/s42003-023-05463-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/13/2023] [Indexed: 11/26/2023] Open
Abstract
The source and roles of fibroblasts and T-cells during maladaptive remodeling and myocardial fibrosis in the setting of pulmonary arterial hypertension (PAH) have been long debated. We demonstrate, using single-cell mass cytometry, a subpopulation of endogenous human cardiac fibroblasts expressing increased levels of CD4, a helper T-cell marker, in addition to myofibroblast markers distributed in human fibrotic RV tissue, interstitial and perivascular lesions in SUGEN/Hypoxia (SuHx) rats, and fibroblasts labeled with pdgfrα CreERt2/+ in R26R-tdTomato mice. Recombinant IL-1β increases IL-1R, CCR2 receptor expression, modifies the secretome, and differentiates cardiac fibroblasts to form CD68-positive cell clusters. IL-1β also activates stemness markers, such as NANOG and SOX2, and genes involved in dedifferentiation, lymphoid cell function and metabolic reprogramming. IL-1β induction of lineage traced primary mouse cardiac fibroblasts causes these cells to lose their fibroblast identity and acquire an immune phenotype. Our results identify IL-1β induced immune-competency in human cardiac fibroblasts and suggest that fibroblast secretome modulation may constitute a therapeutic approach to PAH and other diseases typified by inflammation and fibrotic remodeling.
Collapse
Affiliation(s)
- Jamila H Siamwala
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA.
- Warren Alpert Medical School of Brown University, Providence VA Medical Center, Providence, RI, USA.
| | - Francesco S Pagano
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA
| | - Patrycja M Dubielecka
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Malina J Ivey
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jose Pedro Guirao-Abad
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Alexander Zhao
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA
| | - Sonja Chen
- Warren Alpert Medical School of Brown University, Providence VA Medical Center, Providence, RI, USA
- Department of Pathology & Laboratory Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Haley Granston
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA
| | - Jae Yun Jeong
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI, USA
| | - Sharon Rounds
- Warren Alpert Medical School of Brown University, Providence VA Medical Center, Providence, RI, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Onur Kanisicak
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Richard J Gilbert
- Ocean State Research Institute, Providence VA Medical Center, Providence, RI, USA
| |
Collapse
|
36
|
Shi R, Reichardt M, Fiegle DJ, Küpfer LK, Czajka T, Sun Z, Salditt T, Dendorfer A, Seidel T, Bruegmann T. Contractility measurements for cardiotoxicity screening with ventricular myocardial slices of pigs. Cardiovasc Res 2023; 119:2469-2481. [PMID: 37934066 PMCID: PMC10651213 DOI: 10.1093/cvr/cvad141] [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: 11/08/2022] [Revised: 05/22/2023] [Accepted: 07/10/2023] [Indexed: 11/08/2023] Open
Abstract
AIMS Cardiotoxicity is one major reason why drugs do not enter or are withdrawn from the market. Thus, approaches are required to predict cardiotoxicity with high specificity and sensitivity. Ideally, such methods should be performed within intact cardiac tissue with high relevance for humans and detect acute and chronic side effects on electrophysiological behaviour, contractility, and tissue structure in an unbiased manner. Herein, we evaluate healthy pig myocardial slices and biomimetic cultivation setups (BMCS) as a new cardiotoxicity screening approach. METHODS AND RESULTS Pig left ventricular samples were cut into slices and spanned into BMCS with continuous electrical pacing and online force recording. Automated stimulation protocols were established to determine the force-frequency relationship (FFR), frequency dependence of contraction duration, effective refractory period (ERP), and pacing threshold. Slices generated 1.3 ± 0.14 mN/mm2 force at 0.5 Hz electrical pacing and showed a positive FFR and a shortening of contraction duration with increasing pacing rates. Approximately 62% of slices were able to contract for at least 6 days while showing stable ERP, contraction duration-frequency relationship, and preserved cardiac structure confirmed by confocal imaging and X-ray diffraction analysis. We used specific blockers of the most important cardiac ion channels to determine which analysis parameters are influenced. To validate our approach, we tested five drug candidates selected from the Comprehensive in vitro Proarrhythmia Assay list as well as acetylsalicylic acid and DMSO as controls in a blinded manner in three independent laboratories. We were able to detect all arrhythmic drugs and their respective mode of action on cardiac tissue including inhibition of Na+, Ca2+, and hERG channels as well as Na+/Ca2+ exchanger. CONCLUSION We systematically evaluate this approach for cardiotoxicity screening, which is of high relevance for humans and can be upscaled to medium-throughput screening. Thus, our approach will improve the predictive value and efficiency of preclinical cardiotoxicity screening.
Collapse
Affiliation(s)
- Runzhu Shi
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Göttingen, Germany
- International Research Training Group 1816, University Medical Center Göttingen, Göttingen, Germany
| | - Marius Reichardt
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Göttingen, Germany
- Institute for X-ray Physics, University of Göttingen, Göttingen, Germany
| | - Dominik J Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Linda K Küpfer
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Titus Czajka
- Institute for X-ray Physics, University of Göttingen, Göttingen, Germany
| | - Zhengwu Sun
- Walter-Brendel-Centre of Experimental Medicine, Hospital of the University Munich, Munich, Germany
| | - Tim Salditt
- Institute for X-ray Physics, University of Göttingen, Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| | - Andreas Dendorfer
- Walter-Brendel-Centre of Experimental Medicine, Hospital of the University Munich, Munich, Germany
- German Centre of Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Bruegmann
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| |
Collapse
|
37
|
Fujiwara Y, Miki K, Deguchi K, Naka Y, Sasaki M, Sakoda A, Narita M, Imaichi S, Sugo T, Funakoshi S, Nishimoto T, Imahashi K, Yoshida Y. ERRγ agonist under mechanical stretching manifests hypertrophic cardiomyopathy phenotypes of engineered cardiac tissue through maturation. Stem Cell Reports 2023; 18:2108-2122. [PMID: 37802074 PMCID: PMC10679535 DOI: 10.1016/j.stemcr.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 10/08/2023] Open
Abstract
Engineered cardiac tissue (ECT) using human induced pluripotent stem cell-derived cardiomyocytes is a promising tool for modeling heart disease. However, tissue immaturity makes robust disease modeling difficult. Here, we established a method for modeling hypertrophic cardiomyopathy (HCM) malignant (MYH7 R719Q) and nonmalignant (MYBPC3 G115∗) pathogenic sarcomere gene mutations by accelerating ECT maturation using an ERRγ agonist, T112, and mechanical stretching. ECTs treated with T112 under 10% elongation stimulation exhibited more organized and mature characteristics. Whereas matured ECTs with the MYH7 R719Q mutation showed broad HCM phenotypes, including hypertrophy, hypercontraction, diastolic dysfunction, myofibril misalignment, fibrotic change, and glycolytic activation, matured MYBPC3 G115∗ ECTs displayed limited phenotypes, which were primarily observed only under our new maturation protocol (i.e., hypertrophy). Altogether, ERRγ activation combined with mechanical stimulation enhanced ECT maturation, leading to a more accurate manifestation of HCM phenotypes, including non-cardiomyocyte activation, consistent with clinical observations.
Collapse
Affiliation(s)
- Yuya Fujiwara
- Center for iPS Cells Research and Application, Kyoto University, Kyoto, Japan; Takeda-CiRA Joint Program, Fujisawa, Japan
| | - Kenji Miki
- Center for iPS Cells Research and Application, Kyoto University, Kyoto, Japan; Center for Organ Engineering, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA.
| | - Kohei Deguchi
- Takeda-CiRA Joint Program, Fujisawa, Japan; T-CiRA Discovery, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuki Naka
- Center for iPS Cells Research and Application, Kyoto University, Kyoto, Japan; Takeda-CiRA Joint Program, Fujisawa, Japan
| | - Masako Sasaki
- Center for iPS Cells Research and Application, Kyoto University, Kyoto, Japan; Takeda-CiRA Joint Program, Fujisawa, Japan
| | - Ayaka Sakoda
- Takeda-CiRA Joint Program, Fujisawa, Japan; T-CiRA Discovery, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Megumi Narita
- Center for iPS Cells Research and Application, Kyoto University, Kyoto, Japan
| | - Sachiko Imaichi
- Pharmaceutical Science, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | | | - Shunsuke Funakoshi
- Center for iPS Cells Research and Application, Kyoto University, Kyoto, Japan; Takeda-CiRA Joint Program, Fujisawa, Japan
| | | | - Kenichi Imahashi
- Takeda-CiRA Joint Program, Fujisawa, Japan; T-CiRA Discovery, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yoshinori Yoshida
- Center for iPS Cells Research and Application, Kyoto University, Kyoto, Japan; Takeda-CiRA Joint Program, Fujisawa, Japan.
| |
Collapse
|
38
|
Ghosheh M, Ehrlich A, Ioannidis K, Ayyash M, Goldfracht I, Cohen M, Fischer A, Mintz Y, Gepstein L, Nahmias Y. Electro-metabolic coupling in multi-chambered vascularized human cardiac organoids. Nat Biomed Eng 2023; 7:1493-1513. [PMID: 37550423 DOI: 10.1038/s41551-023-01071-9] [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: 09/09/2021] [Accepted: 06/27/2023] [Indexed: 08/09/2023]
Abstract
The study of cardiac physiology is hindered by physiological differences between humans and small-animal models. Here we report the generation of multi-chambered self-paced vascularized human cardiac organoids formed under anisotropic stress and their applicability to the study of cardiac arrhythmia. Sensors embedded in the cardiac organoids enabled the simultaneous measurement of oxygen uptake, extracellular field potentials and cardiac contraction at resolutions higher than 10 Hz. This microphysiological system revealed 1 Hz cardiac respiratory cycles that are coupled to the electrical rather than the mechanical activity of cardiomyocytes. This electro-mitochondrial coupling was driven by mitochondrial calcium oscillations driving respiration cycles. Pharmaceutical or genetic inhibition of this coupling results in arrhythmogenic behaviour. We show that the chemotherapeutic mitoxantrone induces arrhythmia through disruption of this pathway, a process that can be partially reversed by the co-administration of metformin. Our microphysiological cardiac systems may further facilitate the study of the mitochondrial dynamics of cardiac rhythms and advance our understanding of human cardiac physiology.
Collapse
Affiliation(s)
- Mohammad Ghosheh
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avner Ehrlich
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Tissue Dynamics, LTD, Jerusalem, Israel
| | - Konstantinos Ioannidis
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Tissue Dynamics, LTD, Jerusalem, Israel
| | - Muneef Ayyash
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Idit Goldfracht
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
| | - Merav Cohen
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amit Fischer
- Department of Biological Chemistry, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yoav Mintz
- Department of General Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lior Gepstein
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
- Cardiology Department, Rambam Health Care Campus, Haifa, Israel
| | - Yaakov Nahmias
- Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Tissue Dynamics, LTD, Jerusalem, Israel.
- Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
39
|
Balderas-Villalobos J, Medina-Contreras JML, Lynch C, Kabadi R, Hayles J, Ramirez RJ, Tan AY, Kaszala K, Samsó M, Huizar JF, Eltit JM. Mechanisms of adaptive hypertrophic cardiac remodeling in a large animal model of premature ventricular contraction-induced cardiomyopathy. IUBMB Life 2023; 75:926-940. [PMID: 37427864 PMCID: PMC10592397 DOI: 10.1002/iub.2765] [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: 03/05/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023]
Abstract
Frequent premature ventricular contractions (PVCs) promoted eccentric cardiac hypertrophy and reduced ejection fraction (EF) in a large animal model of PVC-induced cardiomyopathy (PVC-CM), but the molecular mechanisms and markers of this hypertrophic remodeling remain unexplored. Healthy mongrel canines were implanted with pacemakers to deliver bigeminal PVCs (50% burden with 200-220 ms coupling interval). After 12 weeks, left ventricular (LV) free wall samples were studied from PVC-CM and Sham groups. In addition to reduced LV ejection fraction (LVEF), the PVC-CM group showed larger cardiac myocytes without evident ultrastructural alterations compared to the Sham group. Biochemical markers of pathological hypertrophy, such as store-operated Ca2+ entry, calcineurin/NFAT pathway, β-myosin heavy chain, and skeletal type α-actin were unaltered in the PVC-CM group. In contrast, pro-hypertrophic and antiapoptotic pathways including ERK1/2 and AKT/mTOR were activated and/or overexpressed in the PVC-CM group, which appeared counterbalanced by an overexpression of protein phosphatase 1 and a borderline elevation of the anti-hypertrophic factor atrial natriuretic peptide. Moreover, the potent angiogenic and pro-hypertrophic factor VEGF-A and its receptor VEGFR2 were significantly elevated in the PVC-CM group. In conclusion, a molecular program is in place to keep this structural remodeling associated with frequent PVCs as an adaptive pathological hypertrophy.
Collapse
Affiliation(s)
| | - JML Medina-Contreras
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Christopher Lynch
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Rajiv Kabadi
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Janée Hayles
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rafael J. Ramirez
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Alex Y. Tan
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, United States of America
| | - Karoly Kaszala
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, United States of America
| | - Montserrat Samsó
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| | - Jose F. Huizar
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States of America
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, United States of America
| | - Jose M. Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University
| |
Collapse
|
40
|
Kwon K, Kim JU, Won SM, Zhao J, Avila R, Wang H, Chun KS, Jang H, Lee KH, Kim JH, Yoo S, Kang YJ, Kim J, Lim J, Park Y, Lu W, Kim TI, Banks A, Huang Y, Rogers JA. A battery-less wireless implant for the continuous monitoring of vascular pressure, flow rate and temperature. Nat Biomed Eng 2023; 7:1215-1228. [PMID: 37037964 DOI: 10.1038/s41551-023-01022-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/13/2023] [Indexed: 04/12/2023]
Abstract
Devices for monitoring blood haemodynamics can guide the perioperative management of patients with cardiovascular disease. Current technologies for this purpose are constrained by wired connections to external electronics, and wireless alternatives are restricted to monitoring of either blood pressure or blood flow. Here we report the design aspects and performance parameters of an integrated wireless sensor capable of implantation in the heart or in a blood vessel for simultaneous measurements of pressure, flow rate and temperature in real time. The sensor is controlled via long-range communication through a subcutaneously implanted and wirelessly powered Bluetooth Low Energy system-on-a-chip. The device can be delivered via a minimally invasive transcatheter procedure or it can be mounted on a passive medical device such as a stent, as we show for the case of the pulmonary artery in a pig model and the aorta and left ventricle in a sheep model, where the device performs comparably to clinical tools for monitoring of blood flow and pressure. Battery-less and wireless devices such as these that integrate capabilities for flow, pressure and temperature sensing offer the potential for continuous monitoring of blood haemodynamics in patients.
Collapse
Affiliation(s)
- Kyeongha Kwon
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| | - Jong Uk Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Sang Min Won
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jianzhong Zhao
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, China
- Department of Civil and Environmental Engineering, Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Raudel Avila
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Heling Wang
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, China
- Department of Civil and Environmental Engineering, Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Keum San Chun
- Electrical and Computer Engineering, the University of Texas at Austin, Austin, TX, USA
| | - Hokyung Jang
- Department of Electrical & Computer Engineering, University of Wisconsin, Madison, WI, USA
| | | | - Jae-Hwan Kim
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Seonggwang Yoo
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Youn J Kang
- Department of Ocean System Engineering, Jeju National University, Jeju, Republic of Korea
| | - Joohee Kim
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Jaeman Lim
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Yoonseok Park
- Department of Advanced Materials Engineering for Information and Electronics, Kyung Hee University, Yongin, Republic of Korea
| | - Wei Lu
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Tae-Il Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Anthony Banks
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Wearifi, Inc., Evanston, IL, USA
| | - Yonggang Huang
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, China
- Department of Civil and Environmental Engineering, Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - John A Rogers
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA.
- Wearifi, Inc., Evanston, IL, USA.
- Department of Biomedical Engineering, Neurological Surgery, Chemistry, Electrical Engineering and Computer Science, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
41
|
Kim HM, Kim Y, Kim Y, Kim YJ, Ko KS. Organoid Establishment of Long-Term Culture Using Primary Mouse Hepatocytes and Evaluation of Liver Function. Prev Nutr Food Sci 2023; 28:360-369. [PMID: 37842244 PMCID: PMC10567590 DOI: 10.3746/pnf.2023.28.3.360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 10/17/2023] Open
Abstract
Primary hepatocytes and various animal models have traditionally been used in liver function tests to assess the effects of nutrients. However, these approaches present several limitations such as time consumption, high cost, the need for facilities, and ethical issues in primary mouse hepatocytes and animal models. In this study, we constructed liver organoids from primary mouse hepatocytes (OrgPH) to replace primary hepatocytes and animal models. We isolated primary mouse hepatocytes from 6- to 10-week-old male C57BL/6J mice using the two-step collagenase method, and generated liver organoids by clustering the cells in Matrigel. To assess the hepatic function of OrgPH, we examined specific liver markers and gene expressions related to hepatic glucose, ethanol, and cholesterol metabolism. Over a 28-day culture period, liver-specific markers, including Alb, Arg1, G6pc, and Cyp1a1, increased or remained stable in the OrgPH. However, they eventually decreased in primary hepatocytes. Glucose and ethanol metabolism-related gene expression levels exhibited a similar tendency in AML12 cells and OrgPH. However, the expression levels of cholesterol metabolism-related genes displayed an opposite trend in OrgPH compared with those in AML12 cells. These results agree with those of previous studies involving in vivo models. In conclusion, our study indicates that OrgPH can retain liver function and mimic the hepatocytic physiology of mouse in vivo models. Therefore, organoids originating from primary mouse hepatocytes are potentially useful as an animal-free method for evaluating the safety and toxicity of health functional foods and a replacement for animal models.
Collapse
Affiliation(s)
- Hye Mi Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Yerin Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Department of Medicine, Catholic Kwandong University College of Medicine, Gangwon 25601, Korea
| | - Yuri Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Young Jun Kim
- Department of Food and Biotechnology and Food and Regulatory Science, Korea University, Sejong 30019, Korea
| | - Kwang Suk Ko
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
42
|
Mahadevan A, Tipler A, Jones H. Shared developmental pathways of the placenta and fetal heart. Placenta 2023; 141:35-42. [PMID: 36604258 DOI: 10.1016/j.placenta.2022.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
Congenital heart defects (CHD) remain the most common class of birth defect worldwide, affecting 1 in every 110 live births. A host of clinical and morphological indicators of placental dysfunction are observed in pregnancies complicated by fetal CHD and, with the recent emergence of single-cell sequencing capabilities, the molecular and physiological associations between the embryonic heart and developing placenta are increasingly evident. In CHD pregnancies, a hostile intrauterine environment may negatively influence and alter fetal development. Placental maldevelopment and dysfunction creates this hostile in-utero environment and may manifest in the development of various subtypes of CHD, with downstream perfusion and flow-related alterations leading to yet further disruption in placental structure and function. The adverse in-utero environment of CHD-complicated pregnancies is well studied, however the specific etiological role that the placenta plays in CHD development remains unclear. Many mouse and rat models have been used to characterize the relationship between CHD and placental dysfunction, but these paradigms present substantial limitations in the assessment of both the heart and placenta. Improvements in non-invasive placental assessment can mitigate these limitations and drive human-specific investigation in relation to fetal and placental development. Here, we review the clinical, structural, and molecular relationships between CHD and placental dysfunction, the CHD subtype-dependence of these changes, and the future of Placenta-Heart axis modeling and investigation.
Collapse
Affiliation(s)
- Aditya Mahadevan
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA
| | - Alyssa Tipler
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA
| | - Helen Jones
- Physiology and Aging, University of Florida College of Medicine, USA; Center for Research in Perinatal Outcomes, University of Florida, USA.
| |
Collapse
|
43
|
Frankowski J, Kurzątkowska M, Sobczak M, Piotrowska U. Utilization of 3D bioprinting technology in creating human tissue and organoid models for preclinical drug research - State-of-the-art. Int J Pharm 2023; 644:123313. [PMID: 37579828 DOI: 10.1016/j.ijpharm.2023.123313] [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: 05/25/2023] [Revised: 07/28/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Rapid development of tissue engineering in recent years has increased the importance of three-dimensional (3D) bioprinting technology as novel strategy for fabrication functional 3D tissue and organoid models for pharmaceutical research. 3D bioprinting technology gives hope for eliminating many problems associated with traditional cell culture methods during drug screening. However, there is a still long way to wider clinical application of this technology due to the numerous difficulties associated with development of bioinks, advanced printers and in-depth understanding of human tissue architecture. In this review, the work associated with relatively well-known extrusion-based bioprinting (EBB), jetting-based bioprinting (JBB), and vat photopolymerization bioprinting (VPB) is presented and discussed with the latest advances and limitations in this field. Next we discuss state-of-the-art research of 3D bioprinted in vitro models including liver, kidney, lung, heart, intestines, eye, skin as well as neural and bone tissue that have potential applications in the development of new drugs.
Collapse
Affiliation(s)
- Joachim Frankowski
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Matylda Kurzątkowska
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Marcin Sobczak
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Urszula Piotrowska
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland.
| |
Collapse
|
44
|
Bachmann JC, Kirchhoff JE, Napolitano JE, Sorota S, Gordon WM, Feric N, Aschar‐Sobbi R, Lv J, Cao Z, Coppieters K, Borghetti G, Nyberg M. C-type natriuretic peptide induces inotropic and lusitropic effects in human 3D-engineered cardiac tissue: Implications for the regulation of cardiac function in humans. Exp Physiol 2023; 108:1172-1188. [PMID: 37493451 PMCID: PMC10988518 DOI: 10.1113/ep091303] [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: 05/17/2023] [Accepted: 07/06/2023] [Indexed: 07/27/2023]
Abstract
The role of C-type natriuretic peptide (CNP) in the regulation of cardiac function in humans remains to be established as previous investigations have been confined to animal model systems. Here, we used well-characterized engineered cardiac tissues (ECTs) generated from human stem cell-derived cardiomyocytes and fibroblasts to study the acute effects of CNP on contractility. Application of CNP elicited a positive inotropic response as evidenced by increases in maximum twitch amplitude, maximum contraction slope and maximum calcium amplitude. This inotropic response was accompanied by a positive lusitropic response as demonstrated by reductions in time from peak contraction to 90% of relaxation and time from peak calcium transient to 90% of decay that paralleled increases in maximum contraction decay slope and maximum calcium decay slope. To establish translatability, CNP-induced changes in contractility were also assessed in rat ex vivo (isolated heart) and in vivo models. Here, the effects on force kinetics observed in ECTs mirrored those observed in both the ex vivo and in vivo model systems, whereas the increase in maximal force generation with CNP application was only detected in ECTs. In conclusion, CNP induces a positive inotropic and lusitropic response in ECTs, thus supporting an important role for CNP in the regulation of human cardiac function. The high degree of translatability between ECTs, ex vivo and in vivo models further supports a regulatory role for CNP and expands the current understanding of the translational value of human ECTs. NEW FINDINGS: What is the central question of this study? What are the acute responses to C-type natriuretic peptide (CNP) in human-engineered cardiac tissues (ECTs) on cardiac function and how well do they translate to matched concentrations in animal ex vivo and in vivo models? What is the main finding and its importance? Acute stimulation of ECTs with CNP induced positive lusitropic and inotropic effects on cardiac contractility, which closely reflected the changes observed in rat ex vivo and in vivo cardiac models. These findings support an important role for CNP in the regulation of human cardiac function and highlight the translational value of ECTs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Juan Lv
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | - Zhiyou Cao
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | - Ken Coppieters
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | | | - Michael Nyberg
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| |
Collapse
|
45
|
Kakimoto Y, Ueda A, Ito M, Tanaka M, Kubota T, Isozaki S, Osawa M. Proteomic profiling of sudden cardiac death with acquired cardiac hypertrophy. Int J Legal Med 2023; 137:1453-1461. [PMID: 37284852 PMCID: PMC10421815 DOI: 10.1007/s00414-023-03038-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/01/2023] [Indexed: 06/08/2023]
Abstract
BACKGROUND Cardiac hypertrophy, which develops in middle-aged and older individuals as a consequence of hypertension and obesity, is an established risk factor for sudden cardiac death (SCD). However, it is sometimes difficult to differentiate SCD with acquired cardiac hypertrophy (SCH) from compensated cardiac hypertrophy (CCH), at autopsy. We aimed to elucidate the proteomic alteration in SCH, which can be a guideline for future postmortem diagnosis. METHODS Cardiac tissues were sampled at autopsy. SCH group consisted of ischemic heart failure, hypertensive heart failure, and aortic stenosis. CCH group included cases of non-cardiac death with cardiac hypertrophy. The control group comprised cases of non-cardiac death without cardiac hypertrophy. All patients were aged > 40 years, and hypertrophic cardiomyopathy was not included in this study. We performed histological examination and shotgun proteomic analysis, followed by quantitative polymerase chain reaction analysis. RESULTS Significant obesity and myocardial hypertrophy, and mild myocardial fibrosis were comparable in SCH and CCH cases compared to control cases. The proteomic profile of SCH cases was distinguishable from those of CCH and control cases, and many sarcomere proteins were increased in SCH cases. Especially, the protein and mRNA levels of MYH7 and MYL3 were significantly increased in SCH cases. CONCLUSION This is the first report of cardiac proteomic analysis in SCH and CCH cases. The stepwise upregulation of sarcomere proteins may increase the risk for SCD in acquired cardiac hypertrophy before cardiac fibrosis progresses significantly. These findings can possibly aid in the postmortem diagnosis of SCH in middle-aged and older individuals.
Collapse
Affiliation(s)
- Yu Kakimoto
- Department of Forensic Medicine, Tokai University School of Medicine, Kanagawa, Japan.
| | - Atsushi Ueda
- Department of Forensic Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Masatoshi Ito
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Masayuki Tanaka
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Tomoko Kubota
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Shotaro Isozaki
- Department of Forensic Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Motoki Osawa
- Department of Forensic Medicine, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
46
|
Gao B, Abi-Gerges N, Truong K, Stafford A, Nguyen W, Sutherland W, Vargas HM, Qu Y. Assessment of sarcomere shortening and calcium transient in primary human and dog ventricular myocytes. J Pharmacol Toxicol Methods 2023; 123:107278. [PMID: 37268094 DOI: 10.1016/j.vascn.2023.107278] [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: 03/23/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/04/2023]
Abstract
Understanding translation from preclinical observations to clinical findings is important for evaluating the efficacy and safety of novel compounds. Of relevance to cardiac safety is profiling drug effects on cardiomyocyte (CM) sarcomere shortening and intracellular Ca2+ dynamics. Although CM from different animal species have been used to assess such effects, primary human CM isolated from human organ donor heart represent an ideal non-animal alternative approach. We performed a study to evaluate primary human CM and have them compared to freshly isolated dog cardiomyocytes for their basic function and responses to positive inotropes with well-known mechanisms. Our data showed that simultaneous assessment of sarcomere shortening and Ca2+-transient can be performed with both myocytes using the IonOptix system. Amplitude of sarcomere shortening and Ca2+-transient (CaT) were significantly higher in dog compared to human CM in the basic condition (absence of treatment), while longer duration of sarcomere shortening and CaT were observed in human cells. We observed that human and dog CMs have similar pharmacological responses to five inotropes with different mechanisms, including dobutamine and isoproterenol (β-adrenergic stimulation), milrinone (PDE3 inhibition), pimobendan and levosimendan (increase of Ca2+sensitization as well as PDE3 inhibition). In conclusion, our study suggests that myocytes obtained from both human donor hearts and dog hearts can be used to simultaneously assess drug-induced effects on sarcomere shortening and CaT using the IonOptix platform.
Collapse
Affiliation(s)
- BaoXi Gao
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Ky Truong
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Alexa Stafford
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - William Nguyen
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Weston Sutherland
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Hugo M Vargas
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Yusheng Qu
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| |
Collapse
|
47
|
Wilhelm TI, Lewalter T, Fischer J, Reiser J, Werner J, Baumgartner C, Gleirscher L, Hoppmann P, Kupatt C, Tiemann K, Jilek C. Electroanatomical Conduction Characteristics of Pig Myocardial Tissue Derived from High-Density Mapping. J Clin Med 2023; 12:5598. [PMID: 37685665 PMCID: PMC10488835 DOI: 10.3390/jcm12175598] [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: 05/25/2023] [Revised: 08/19/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Ultra-high-density mapping systems allow more precise measurement of the heart chambers at corresponding conduction velocities (CVs) and voltage amplitudes (VAs). Our aim for this study was to define and compare a basic value set for unipolar CV and VA in all four heart chambers and their separate walls in healthy, juvenile porcine hearts using ultra-high-density mapping. METHODS We used the Rhythmia Mapping System to create electroanatomical maps of four pig hearts in sinus rhythm. CVs and VAs were calculated for chambers and wall segments with overlapping circular areas (radius of 5 mm). RESULTS We analysed 21 maps with a resolution of 1.4 points/mm2. CVs were highest in the left atrium (LA), followed by the left ventricle (LV), right ventricle (RV), and right atrium (RA). As for VA, LV was highest, followed by RV, LA, and RA. The left chambers had a higher overall CV and VA than the right. Within the chambers, CV varied more in the right than in the left chambers, and VA varied in the ventricles but not in the atria. There was a slightly positive correlation between CVs and VAs at velocity values of <1.5 m/s. CONCLUSIONS In healthy porcine hearts, the left chambers showed higher VAs and CVs than the right. CV differs mainly within the right chambers and VA differs only within the ventricles. A slightly positive linear correlation was found between slow CVs and low VAs.
Collapse
Affiliation(s)
- Theresa Isabelle Wilhelm
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (T.L.)
- Medical Graduate Center, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Thorsten Lewalter
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (T.L.)
| | - Johannes Fischer
- Center for Preclinical Research, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Judith Reiser
- Center for Preclinical Research, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Julia Werner
- Center for Preclinical Research, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Christine Baumgartner
- Center for Preclinical Research, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Lukas Gleirscher
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (T.L.)
| | - Petra Hoppmann
- Department of Internal Medicine I, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Christian Kupatt
- Department of Internal Medicine I, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Klaus Tiemann
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (T.L.)
- Department of Internal Medicine I, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Clemens Jilek
- Peter-Osypka Heart Centre Munich, Internistisches Klinikum München Süd, 81379 Munich, Germany (T.L.)
- Department of Internal Medicine I, University Hospital Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
48
|
Klösener L, Samolovac S, Barnekow I, König J, Moussavi A, Boretius S, Fuchs D, Haegens A, Hinkel R, Mietsch M. Functional Cardiovascular Characterization of the Common Marmoset ( Callithrix jacchus). BIOLOGY 2023; 12:1123. [PMID: 37627007 PMCID: PMC10452209 DOI: 10.3390/biology12081123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
Appropriate cardiovascular animal models are urgently needed to investigate genetic, molecular, and therapeutic approaches, yet the translation of results from the currently used species is difficult due to their genetic distance as well as their anatomical or physiological differences. Animal species that are closer to the human situation might help to bridge this translational gap. The common marmoset (Callithrix jacchus) is an interesting candidate to investigate certain heart diseases and cardiovascular comorbidities, yet a basic functional characterization of its hemodynamic system is still missing. Therefore, cardiac functional analyses were performed by utilizing the invasive intracardiac pressure-volume loops (PV loop) system in seven animals, magnetic resonance imaging (MRI) in six animals, and echocardiography in five young adult male common marmosets. For a direct comparison between the three methods, only data from animals for which all three datasets could be acquired were selected. All three modalities were suitable for characterizing cardiac function, though with some systemic variations. In addition, vena cava occlusions were performed to investigate the load-independent parameters collected with the PV loop system, which allowed for a deeper analysis of the cardiac function and for a more sensitive detection of the alterations in a disease state, such as heart failure or certain cardiovascular comorbidities.
Collapse
Affiliation(s)
- Lina Klösener
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany (M.M.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, University of Veterinary Medicine, 30173 Hannover, Germany
| | - Sabine Samolovac
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany (M.M.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Ina Barnekow
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Jessica König
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Amir Moussavi
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Susann Boretius
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
- Johann-Friedrich-Blumenbach Institute of Zoology and Anthropology, Georg August University, 37077 Göttingen, Germany
| | - Dieter Fuchs
- FUJIFILM VisualSonics Inc., 1114 AB Amsterdam, The Netherlands
| | | | - Rabea Hinkel
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany (M.M.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, University of Veterinary Medicine, 30173 Hannover, Germany
| | - Matthias Mietsch
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany (M.M.)
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
49
|
Bayne EF, Rossler KJ, Gregorich ZR, Aballo TJ, Roberts DS, Chapman EA, Guo W, Palecek SP, Ralphe JC, Kamp TJ, Ge Y. Top-down proteomics of myosin light chain isoforms define chamber-specific expression in the human heart. J Mol Cell Cardiol 2023; 181:89-97. [PMID: 37327991 PMCID: PMC10528938 DOI: 10.1016/j.yjmcc.2023.06.003] [Citation(s) in RCA: 6] [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: 01/28/2023] [Revised: 05/27/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Myosin functions as the "molecular motor" of the sarcomere and generates the contractile force necessary for cardiac muscle contraction. Myosin light chains 1 and 2 (MLC-1 and -2) play important functional roles in regulating the structure of the hexameric myosin molecule. Each of these light chains has an 'atrial' and 'ventricular' isoform, so called because they are believed to exhibit chamber-restricted expression in the heart. However, recently the chamber-specific expression of MLC isoforms in the human heart has been questioned. Herein, we analyzed the expression of MLC-1 and -2 atrial and ventricular isoforms in each of the four cardiac chambers in adult non-failing donor hearts using top-down mass spectrometry (MS)-based proteomics. Strikingly, we detected an isoform thought to be ventricular, MLC-2v (gene: MYL2), in the atria and confirmed the protein sequence using tandem MS (MS/MS). For the first time, a putative deamidation post-translation modification (PTM) located on MLC-2v in atrial tissue was localized to amino acid N13. MLC-1v (MYL3) and MLC-2a (MYL7) were the only MLC isoforms exhibiting chamber-restricted expression patterns across all donor hearts. Importantly, our results unambiguously show that MLC-1v, not MLC-2v, is ventricle-specific in adult human hearts. Moreover, we found elevated MLC-2 phosphorylation in male hearts compared to female hearts across each cardiac chamber. Overall, top-down proteomics allowed an unbiased analysis of MLC isoform expression throughout the human heart, uncovering previously unexpected isoform expression patterns and PTMs.
Collapse
Affiliation(s)
- Elizabeth F Bayne
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kalina J Rossler
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachery R Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Timothy J Aballo
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David S Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Emily A Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
50
|
GOTO A, KAMBAYASHI R, SAITO H, AKIE Y, IZUMI-NAKASEKO H, TAKEI Y, SUGIYAMA A. Characterization of the electrocardiogram of microminipig in comparison with that of Clawn miniature swine: impacts of miniaturization of body size on electrocardiographic indices. J Vet Med Sci 2023; 85:828-831. [PMID: 37302820 PMCID: PMC10466054 DOI: 10.1292/jvms.23-0049] [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: 01/29/2023] [Accepted: 05/31/2023] [Indexed: 06/13/2023] Open
Abstract
Effects of body size reduction on electrocardiographic indices were examined using microminipigs in comparison with Clawn miniature swine (Clawn). Electrocardiogram was recorded using Holter electrocardiograph in conscious state for 24 hr for microminipigs (male: 11.6 ± 0.1 kg, 12-17 months, n=5; and female: 9.9 ± 0.4 kg, 6 months, n=5) and Clawn (female: 20.3 ± 0.4 kg, 8-9 months, n=8). Microminipig had shorter PR interval and QRS width than Clawn, whereas no significant difference was detected in JTcF/QTcF between them. Ratios of PR interval, QRS width, and body weight cubic root for microminipigs to Clawn ranged between 0.713 and 0.830. These findings indicate that PR interval and QRS width will depend on distance for excitatory current propagation, whereas JTcF/QTcF may be governed by local electrical activities.
Collapse
Affiliation(s)
- Ai GOTO
- Department of Pharmacology, Faculty of Medicine, Toho
University, Tokyo, Japan
| | - Ryuichi KAMBAYASHI
- Department of Pharmacology, Faculty of Medicine, Toho
University, Tokyo, Japan
| | - Hiroyuki SAITO
- Department of Pharmacology, Faculty of Medicine, Toho
University, Tokyo, Japan
- CMIC Bioresearch Center, CMIC Pharma Science Co., Ltd.,
Yamanashi, Japan
| | - Yasuki AKIE
- Department of Pharmacology, Faculty of Medicine, Toho
University, Tokyo, Japan
- CMIC Bioresearch Center, CMIC Pharma Science Co., Ltd.,
Yamanashi, Japan
| | | | - Yoshinori TAKEI
- Department of Pharmacology, Faculty of Medicine, Toho
University, Tokyo, Japan
| | - Atsushi SUGIYAMA
- Department of Pharmacology, Faculty of Medicine, Toho
University, Tokyo, Japan
| |
Collapse
|