1
|
Wu Z, Park J, Steiner PR, Zhu B, Zhang JXJ. A Graph-Based Machine-Learning Approach Combined with Optical Measurements to Understand Beating Dynamics of Cardiomyocytes. J Comput Biol 2025; 32:239-252. [PMID: 39648722 DOI: 10.1089/cmb.2024.0491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
The development of computational models for the prediction of cardiac cellular dynamics remains a challenge due to the lack of first-principled mathematical models. We develop a novel machine-learning approach hybridizing physics simulation and graph networks to deliver robust predictions of cardiomyocyte dynamics. Embedded with inductive physical priors, the proposed constraint-based interaction neural projection (CINP) algorithm can uncover hidden physical constraints from sparse image data on a small set of beating cardiac cells and provide robust predictions for heterogenous large-scale cell sets. We also implement an in vitro culture and imaging platform for cellular motion and calcium transient analysis to validate the model. We showcase our model's efficacy by predicting complex organoid cellular behaviors in both in silico and in vitro settings.
Collapse
Affiliation(s)
- Ziqian Wu
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Jiyoon Park
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Paul R Steiner
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Bo Zhu
- School of Interactive Computing, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - John X J Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
2
|
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
|
3
|
Manfra O, Louey S, Jonker SS, Perdreau-Dahl H, Frisk M, Giraud GD, Thornburg KL, Louch WE. Augmenting workload drives T-tubule assembly in developing cardiomyocytes. J Physiol 2024; 602:4461-4486. [PMID: 37128962 PMCID: PMC10854476 DOI: 10.1113/jp284538] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/11/2023] [Indexed: 05/03/2023] Open
Abstract
Contraction of cardiomyocytes is initiated at subcellular elements called dyads, where L-type Ca2+ channels in t-tubules are located within close proximity to ryanodine receptors in the sarcoplasmic reticulum. While evidence from small rodents indicates that dyads are assembled gradually in the developing heart, it is unclear how this process occurs in large mammals. We presently examined dyadic formation in fetal and newborn sheep (Ovis aries), and the regulation of this process by fetal cardiac workload. By employing advanced imaging methods, we demonstrated that t-tubule growth and dyadic assembly proceed gradually during fetal sheep development, from 93 days of gestational age until birth (147 days). This process parallels progressive increases in fetal systolic blood pressure, and includes step-wise colocalization of L-type Ca2+ channels and the Na+/Ca2+ exchanger with ryanodine receptors. These proteins are upregulated together with the dyadic anchor junctophilin-2 during development, alongside changes in the expression of amphiphysin-2 (BIN1) and its partner proteins myotubularin and dynamin-2. Increasing fetal systolic load by infusing plasma or occluding the post-ductal aorta accelerated t-tubule growth. Conversely, reducing fetal systolic load with infusion of enalaprilat, an angiotensin converting enzyme inhibitor, blunted t-tubule formation. Interestingly, altered t-tubule densities did not relate to changes in dyadic junctions, or marked changes in the expression of dyadic regulatory proteins, indicating that distinct signals are responsible for maturation of the sarcoplasmic reticulum. In conclusion, augmenting blood pressure and workload during normal fetal development critically promotes t-tubule growth, while additional signals contribute to dyadic assembly. KEY POINTS: T-tubule growth and dyadic assembly proceed gradually in cardiomyocytes during fetal sheep development, from 93 days of gestational age until the post-natal stage. Increasing fetal systolic load by infusing plasma or occluding the post-ductal aorta accelerated t-tubule growth and hypertrophy. In contrast, reducing fetal systolic load by enalaprilat infusion slowed t-tubule development and decreased cardiomyocyte size. Load-dependent modulation of t-tubule maturation was linked to altered expression patterns of the t-tubule regulatory proteins junctophilin-2 and amphiphysin-2 (BIN1) and its protein partners. Altered t-tubule densities did not influence dyadic formation, indicating that distinct signals are responsible for maturation of the sarcoplasmic reticulum.
Collapse
Affiliation(s)
- Ornella Manfra
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Samantha Louey
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, OR, USA
| | - Sonnet S Jonker
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, OR, USA
| | - Harmonie Perdreau-Dahl
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - George D Giraud
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, OR, USA
- VA Portland Health Care System Portland, OR, USA
| | - Kent L Thornburg
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, OR, USA
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
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
|
5
|
Lee SG, Rhee J, Seok J, Kim J, Kim MW, Song GE, Park S, Jeong KS, Lee S, Lee YH, Jeong Y, Kim CY, Chung HM. Promotion of maturation of human pluripotent stem cell-derived cardiomyocytes via treatment with the peroxisome proliferator-activated receptor alpha agonist Fenofibrate. Stem Cells Transl Med 2024; 13:750-762. [PMID: 38946019 PMCID: PMC11328931 DOI: 10.1093/stcltm/szae029] [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: 06/12/2023] [Accepted: 04/04/2024] [Indexed: 07/02/2024] Open
Abstract
As research on in vitro cardiotoxicity assessment and cardiac disease modeling becomes more important, the demand for human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is increasing. However, it has been reported that differentiated hPSC-CMs are in a physiologically immature state compared to in vivo adult CMs. Since immaturity of hPSC-CMs can lead to poor drug response and loss of acquired heart disease modeling, various approaches have been attempted to promote maturation of CMs. Here, we confirm that peroxisome proliferator-activated receptor alpha (PPARα), one of the representative mechanisms of CM metabolism and cardioprotective effect also affects maturation of CMs. To upregulate PPARα expression, we treated hPSC-CMs with fenofibrate (Feno), a PPARα agonist used in clinical hyperlipidemia treatment, and demonstrated that the structure, mitochondria-mediated metabolism, and electrophysiology-based functions of hPSC-CMs were all mature. Furthermore, as a result of multi electrode array (MEA)-based cardiotoxicity evaluation between control and Feno groups according to treatment with arrhythmia-inducing drugs, drug response was similar in a dose-dependent manner. However, main parameters such as field potential duration, beat period, and spike amplitude were different between the 2 groups. Overall, these results emphasize that applying matured hPSC-CMs to the field of preclinical cardiotoxicity evaluation, which has become an essential procedure for new drug development, is necessary.
Collapse
Affiliation(s)
- Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Gwangjin-Gu, Seoul 05029, Republic of Korea
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jooeon Rhee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Seok
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Gyeong-Eun Song
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Shinhye Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyu Sik Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Suemin Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Yun Hyeong Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Youngin Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Gwangjin-Gu, Seoul 05029, Republic of Korea
- Miraecell Bio Co. Ltd., Seoul 04795, Korea
| |
Collapse
|
6
|
Naumenko N, Koivumäki JT, Lunko O, Tuomainen T, Leigh R, Rabiee M, Laurila J, Oksanen M, Lehtonen S, Koistinaho J, Tavi P. Presenilin-1 ΔE9 mutation associated sarcoplasmic reticulum leak alters [Ca 2+] i distribution in human iPSC-derived cardiomyocytes. J Mol Cell Cardiol 2024; 193:78-87. [PMID: 38851626 DOI: 10.1016/j.yjmcc.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Mutations in ubiquitously expressed presenilin genes (PSENs) lead to early-onset familial Alzheimer's disease (FAD), but patients carrying the mutation also suffer from heart diseases. To elucidate the cardiac myocyte specific effects of PSEN ΔE9, we studied cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs) from patients carrying AD-causing PSEN1 exon 9 deletion (PSEN1 ΔE9). When compared with their isogenic controls, PSEN1 ΔE9 cardiomyocytes showed increased sarcoplasmic reticulum (SR) Ca2+ leak that was resistant to blockage of ryanodine receptors (RyRs) by tetracaine or inositol-3-reseceptors (IP3Rs) by 2-ABP. The SR Ca2+ leak did not affect electrophysiological properties of the hiPSC-CMs, but according to experiments and in silico simulations the leak induces a diastolic buildup of [Ca2+] near the perinuclear SR and reduces the releasable Ca2+ during systole. This demonstrates that PSEN1 ΔE9 induced SR Ca2+ leak has specific effects in iPSC-CMs, reflecting their unique structural and calcium signaling features. The results shed light on the physiological and pathological mechanisms of PSEN1 in cardiac myocytes and explain the intricacies of comorbidity associated with AD-causing mutations in PSEN1.
Collapse
Affiliation(s)
- Nikolay Naumenko
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jussi T Koivumäki
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olesia Lunko
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tomi Tuomainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Robert Leigh
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mina Rabiee
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jalmari Laurila
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna Oksanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sarka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland; Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland; Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Pasi Tavi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
7
|
Mensah IK, Gowher H. Epigenetic Regulation of Mammalian Cardiomyocyte Development. EPIGENOMES 2024; 8:25. [PMID: 39051183 PMCID: PMC11270418 DOI: 10.3390/epigenomes8030025] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The heart is the first organ formed during mammalian development and functions to distribute nutrients and oxygen to other parts of the developing embryo. Cardiomyocytes are the major cell types of the heart and provide both structural support and contractile function to the heart. The successful differentiation of cardiomyocytes during early development is under tight regulation by physical and molecular factors. We have reviewed current studies on epigenetic factors critical for cardiomyocyte differentiation, including DNA methylation, histone modifications, chromatin remodelers, and noncoding RNAs. This review also provides comprehensive details on structural and morphological changes associated with the differentiation of fetal and postnatal cardiomyocytes and highlights their differences. A holistic understanding of all aspects of cardiomyocyte development is critical for the successful in vitro differentiation of cardiomyocytes for therapeutic purposes.
Collapse
Affiliation(s)
| | - Humaira Gowher
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
8
|
Chen Z, Pan Z, Huang C, Zhu X, Li N, Huynh H, Xu J, Huang L, Vaz FM, Liu J, Han Z, Ouyang K. Cardiac lipidomic profiles in mice undergo changes from fetus to adult. Life Sci 2024; 341:122484. [PMID: 38311219 DOI: 10.1016/j.lfs.2024.122484] [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: 10/25/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
AIMS Lipids are essential cellular components with many important biological functions. Disturbed lipid biosynthesis and metabolism has been shown to cause cardiac developmental abnormality and cardiovascular diseases. In this study, we aimed to investigate the composition and the molecular profiles of lipids in mammalian hearts between embryonic and adult stages and uncover the underlying links between lipid and cardiac development and maturation. MATERIALS AND METHODS We collected mouse hearts at the embryonic day 11.5 (E11.5), E15.5, and the age of 2 months, 4 months and 10 months, and performed lipidomic analysis to determine the changes of the composition, molecular species, and relative abundance of cardiac lipids between embryonic and adult stages. Additionally, we also performed the electronic microscopy and RNA sequencing in both embryonic and adult mouse hearts. KEY FINDINGS The relative abundances of certain phospholipids and sphingolipids including cardiolipin, phosphatidylglycerol, phosphatidylethanolamine, and ceramide, are different between embryonic and adult hearts. Such lipidomic changes are accompanied with increased densities of mitochondrial membranes and elevated expression of genes related to mitochondrial formation in adult mouse hearts. We also analyzed individual molecular species of phospholipids and sphingolipids, and revealed that the composition and distribution of lipid molecular species in hearts also change with development. SIGNIFICANCE Our study provides not only a lipidomic view of mammalian hearts when developing from the embryonic to the adult stage, but also a potential pool of lipid indicators for cardiac cell development and maturation.
Collapse
Affiliation(s)
- Ze'e Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Zhixiang Pan
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Helen Huynh
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Junjie Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, the Netherlands
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| |
Collapse
|
9
|
Li P, Zhang J, Hayashi H, Yue J, Li W, Yang C, Sun C, Shi J, Huberman-Shlaes J, Hibino N, Tian B. Monolithic silicon for high spatiotemporal translational photostimulation. Nature 2024; 626:990-998. [PMID: 38383782 PMCID: PMC11646366 DOI: 10.1038/s41586-024-07016-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 01/02/2024] [Indexed: 02/23/2024]
Abstract
Electrode-based electrical stimulation underpins several clinical bioelectronic devices, including deep-brain stimulators1,2 and cardiac pacemakers3. However, leadless multisite stimulation is constrained by the technical difficulties and spatial-access limitations of electrode arrays. Optogenetics offers optically controlled random access with high spatiotemporal capabilities, but clinical translation poses challenges4-6. Here we show tunable spatiotemporal photostimulation of cardiac systems using a non-genetic platform based on semiconductor-enabled biomodulation interfaces. Through spatiotemporal profiling of photoelectrochemical currents, we assess the magnitude, precision, accuracy and resolution of photostimulation in four leadless silicon-based monolithic photoelectrochemical devices. We demonstrate the optoelectronic capabilities of the devices through optical overdrive pacing of cultured cardiomyocytes (CMs) targeting several regions and spatial extents, isolated rat hearts in a Langendorff apparatus, in vivo rat hearts in an ischaemia model and an in vivo mouse heart model with transthoracic optical pacing. We also perform the first, to our knowledge, optical override pacing and multisite pacing of a pig heart in vivo. Our systems are readily adaptable for minimally invasive clinical procedures using our custom endoscopic delivery device, with which we demonstrate closed-thoracic operations and endoscopic optical stimulation. Our results indicate the clinical potential of the leadless, lightweight and multisite photostimulation platform as a pacemaker in cardiac resynchronization therapy (CRT), in which lead-placement complications are common.
Collapse
Affiliation(s)
- Pengju Li
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Jing Zhang
- The James Franck Institute, The University of Chicago, Chicago, IL, USA
| | - Hidenori Hayashi
- Section of Cardiac Surgery, Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Jiping Yue
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Wen Li
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Chuanwang Yang
- The James Franck Institute, The University of Chicago, Chicago, IL, USA
| | - Changxu Sun
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Jiuyun Shi
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | | | - Narutoshi Hibino
- Section of Cardiac Surgery, Department of Surgery, The University of Chicago, Chicago, IL, USA.
| | - Bozhi Tian
- The James Franck Institute, The University of Chicago, Chicago, IL, USA.
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
10
|
Yogendran V, Mele L, Prysyazhna O, Budhram-Mahadeo VS. Vascular dysfunction caused by loss of Brn-3b/POU4F2 transcription factor in aortic vascular smooth muscle cells is linked to deregulation of calcium signalling pathways. Cell Death Dis 2023; 14:770. [PMID: 38007517 PMCID: PMC10676411 DOI: 10.1038/s41419-023-06306-w] [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: 12/21/2022] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
Phenotypic and functional changes in vascular smooth muscle cells (VSMCs) contribute significantly to cardiovascular diseases (CVD) but factors driving early adverse vascular changes are poorly understood. We report on novel and important roles for the Brn-3b/POU4F2 (Brn-3b) transcription factor (TF) in controlling VSMC integrity and function. Brn-3b protein is expressed in mouse aorta with localisation to VSMCs. Male Brn-3b knock-out (KO) aortas displayed extensive remodelling with increased extracellular matrix (ECM) deposition, elastin fibre disruption and small but consistent narrowing/coarctation in the descending aortas. RNA sequencing analysis showed that these effects were linked to deregulation of genes required for calcium (Ca2+) signalling, vascular contractility, sarco-endoplasmic reticulum (S/ER) stress responses and immune function in Brn-3b KO aortas and validation studies confirmed changes in Ca2+ signalling genes linked to increased intracellular Ca2+ and S/ER Ca2+ depletion [e.g. increased, Cacna1d Ca2+ channels; ryanodine receptor 2, (RyR2) and phospholamban (PLN) but reduced ATP2a1, encoding SERCA1 pump] and chaperone proteins, Hspb1, HspA8, DnaJa1 linked to increased S/ER stress, which also contributes to contractile dysfunction. Accordingly, vascular rings from Brn-3b KO aortas displayed attenuated contractility in response to KCl or phenylephrine (PE) while Brn-3b KO-derived VSMC displayed abnormal Ca2+ signalling following ATP stimulation. This data suggests that Brn-3b target genes are necessary to maintain vascular integrity /contractile function and deregulation upon loss of Brn-3b will contribute to contractile dysfunction linked to CVD.
Collapse
Affiliation(s)
- Vaishaali Yogendran
- Molecular Biology Development and Disease, UCL Institute of Cardiovascular Science, London, UK
| | - Laura Mele
- Molecular Biology Development and Disease, UCL Institute of Cardiovascular Science, London, UK
| | - Oleksandra Prysyazhna
- Clinical Pharmacology Centre, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
11
|
Li J, Sundnes J, Hou Y, Laasmaa M, Ruud M, Unger A, Kolstad TR, Frisk M, Norseng PA, Yang L, Setterberg IE, Alves ES, Kalakoutis M, Sejersted OM, Lanner JT, Linke WA, Lunde IG, de Tombe PP, Louch WE. Stretch Harmonizes Sarcomere Strain Across the Cardiomyocyte. Circ Res 2023; 133:255-270. [PMID: 37401464 PMCID: PMC10355805 DOI: 10.1161/circresaha.123.322588] [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: 01/27/2023] [Revised: 06/07/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Increasing cardiomyocyte contraction during myocardial stretch serves as the basis for the Frank-Starling mechanism in the heart. However, it remains unclear how this phenomenon occurs regionally within cardiomyocytes, at the level of individual sarcomeres. We investigated sarcomere contractile synchrony and how intersarcomere dynamics contribute to increasing contractility during cell lengthening. METHODS Sarcomere strain and Ca2+ were simultaneously recorded in isolated left ventricular cardiomyocytes during 1 Hz field stimulation at 37 °C, at resting length and following stepwise stretch. RESULTS We observed that in unstretched rat cardiomyocytes, differential sarcomere deformation occurred during each beat. Specifically, while most sarcomeres shortened during the stimulus, ≈10% to 20% of sarcomeres were stretched or remained stationary. This nonuniform strain was not traced to regional Ca2+ disparities but rather shorter resting lengths and lower force production in systolically stretched sarcomeres. Lengthening of the cell recruited additional shortening sarcomeres, which increased contractile efficiency as less negative, wasted work was performed by stretched sarcomeres. Given the known role of titin in setting sarcomere dimensions, we next hypothesized that modulating titin expression would alter intersarcomere dynamics. Indeed, in cardiomyocytes from mice with titin haploinsufficiency, we observed greater variability in resting sarcomere length, lower recruitment of shortening sarcomeres, and impaired work performance during cell lengthening. CONCLUSIONS Graded sarcomere recruitment directs cardiomyocyte work performance, and harmonization of sarcomere strain increases contractility during cell stretch. By setting sarcomere dimensions, titin controls sarcomere recruitment, and its lowered expression in haploinsufficiency mutations impairs cardiomyocyte contractility.
Collapse
Affiliation(s)
- Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | | | - Yufeng Hou
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Martin Laasmaa
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Marianne Ruud
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Andreas Unger
- Institute of Physiology II, University of Münster, Germany (A.U., W.A.L.)
| | - Terje R. Kolstad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
| | | | - Ingunn E. Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Estela S. Alves
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.S.A., M.K., J.T.L.)
| | - Michaeljohn Kalakoutis
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.S.A., M.K., J.T.L.)
| | - Ole M. Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Johanna T. Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (E.S.A., M.K., J.T.L.)
| | - Wolfgang A. Linke
- Institute of Physiology II, University of Münster, Germany (A.U., W.A.L.)
| | - Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| | - Pieter P. de Tombe
- Department of Physiology and Biophysics, University of Illinois at Chicago (P.P.d.T.)
- Phymedexp, Université de Montpellier, INSERM, CNRS, France (P.P.d.T.)
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., P.A.N., I.E.S., O.M.S., I.G.L., W.E.L.)
- KG Jebsen Center for Cardiac Research, University of Oslo, Norway (J.L., Y.H., M.L., M.R., T.R.K., M.F., I.E.S., O.M.S., I.G.L., W.E.L.)
| |
Collapse
|
12
|
Grandi E, Navedo MF, Saucerman JJ, Bers DM, Chiamvimonvat N, Dixon RE, Dobrev D, Gomez AM, Harraz OF, Hegyi B, Jones DK, Krogh-Madsen T, Murfee WL, Nystoriak MA, Posnack NG, Ripplinger CM, Veeraraghavan R, Weinberg S. Diversity of cells and signals in the cardiovascular system. J Physiol 2023; 601:2547-2592. [PMID: 36744541 PMCID: PMC10313794 DOI: 10.1113/jp284011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023] Open
Abstract
This white paper is the outcome of the seventh UC Davis Cardiovascular Research Symposium on Systems Approach to Understanding Cardiovascular Disease and Arrhythmia. This biannual meeting aims to bring together leading experts in subfields of cardiovascular biomedicine to focus on topics of importance to the field. The theme of the 2022 Symposium was 'Cell Diversity in the Cardiovascular System, cell-autonomous and cell-cell signalling'. Experts in the field contributed their experimental and mathematical modelling perspectives and discussed emerging questions, controversies, and challenges in examining cell and signal diversity, co-ordination and interrelationships involved in cardiovascular function. This paper originates from the topics of formal presentations and informal discussions from the Symposium, which aimed to develop a holistic view of how the multiple cell types in the cardiovascular system integrate to influence cardiovascular function, disease progression and therapeutic strategies. The first section describes the major cell types (e.g. cardiomyocytes, vascular smooth muscle and endothelial cells, fibroblasts, neurons, immune cells, etc.) and the signals involved in cardiovascular function. The second section emphasizes the complexity at the subcellular, cellular and system levels in the context of cardiovascular development, ageing and disease. Finally, the third section surveys the technological innovations that allow the interrogation of this diversity and advancing our understanding of the integrated cardiovascular function and dysfunction.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Rose E. Dixon
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Ana M. Gomez
- Signaling and Cardiovascular Pathophysiology-UMR-S 1180, INSERM, Université Paris-Saclay, Orsay, France
| | - Osama F. Harraz
- Department of Pharmacology, Larner College of Medicine, and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - David K. Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Trine Krogh-Madsen
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, New York, USA
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Matthew A. Nystoriak
- Department of Medicine, Division of Environmental Medicine, Center for Cardiometabolic Science, University of Louisville, Louisville, KY, 40202, USA
| | - Nikki G. Posnack
- Department of Pediatrics, Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children’s National Heart Institute, Children’s National Hospital, Washington, DC, USA
| | | | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University – Wexner Medical Center, Columbus, OH, USA
| | - Seth Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University – Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
13
|
Kryzhanovskii SA, Zinchenko VP, Tsorin IB, Teplov IY, Vititnova MB, Mokrov GV, Stolyaruk VN. To the Mechanism of the Antiarrhythmic Action of Compound ALM-802: the Role of Ryanodine Receptors. Bull Exp Biol Med 2023; 174:734-737. [PMID: 37170020 DOI: 10.1007/s10517-023-05781-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Indexed: 05/13/2023]
Abstract
The effect of the compound N1-(2,3,4-trimethoxy)-N2-{2-[(2,3,4-trimethoxybenzyl)amino]ethyl}-1,2-ethane-diamine (code ALM-802) on the amplitude of the Ca2+ response in the cell was studied in in vitro experiments. The concentration of intracellular calcium was assessed using a Fura-2 two-wave probe. The experiments were performed on a culture of isolated rat hippocampal neurons. The effect of compound ALM-802 on the activity of ryanodine receptors (RyR2) was studied on an isolated strip of rat myocardium. The compound ALM-802 (69.8 μM) in hippocampal neurons causes a significant decrease in the amplitude of the Ca2+ response induced by addition of KCl to the medium. Experiments performed on an isolated myocardial strip showed that compound ALM-802 (10-5 M) almost completely blocked the positive inotropic reaction of the strip to the RyR2 agonist caffeine (5×10-5 M). The data obtained indicate that the decrease in the concentration of Ca2+ ions in the cell caused by ALM-802 is due to its ability to block RyR2 located on the membrane of the sarcoplasmic reticulum, which can be associated with the antiarrhythmic activity of the compound.
Collapse
Affiliation(s)
| | - V P Zinchenko
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | - I B Tsorin
- V. V. Zakusov Research Institute of Pharmacology, Moscow, Russia.
| | - I Yu Teplov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | - M B Vititnova
- V. V. Zakusov Research Institute of Pharmacology, Moscow, Russia
| | - G V Mokrov
- V. V. Zakusov Research Institute of Pharmacology, Moscow, Russia
| | - V N Stolyaruk
- V. V. Zakusov Research Institute of Pharmacology, Moscow, Russia
| |
Collapse
|
14
|
Tabata T, Masumura Y, Higo S, Kunimatsu S, Kameda S, Inoue H, Okuno S, Ogawa S, Takashima S, Watanabe M, Miyagawa S, Hikoso S, Sakata Y. Multiplexed measurement of cell type-specific calcium kinetics using high-content image analysis combined with targeted gene disruption. Biochem Biophys Res Commun 2022; 637:40-49. [PMID: 36375249 DOI: 10.1016/j.bbrc.2022.10.088] [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: 10/14/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Kinetic analysis of intracellular calcium (Ca2+) in cardiomyocytes is commonly used to determine the pathogenicity of genetic mutations identified in patients with dilated cardiomyopathy (DCM). Conventional methods for measuring Ca2+ kinetics target whole-well cultured cardiomyocytes and therefore lack information concerning individual cells. Results are also affected by heterogeneity in cell populations. Here, we developed an analytical method using CRISPR/Cas9 genome editing combined with high-content image analysis (HCIA) that links cell-by-cell Ca2+ kinetics and immunofluorescence images in thousands of cardiomyocytes at a time. After transfecting cultured mouse cardiomyocytes that constitutively express Cas9 with gRNAs, we detected a prolonged action potential duration specifically in Serca2a-depleted ventricular cardiomyocytes in mixed culture. To determine the phenotypic effect of a frameshift mutation in PKD1 in a patient with DCM, we introduced the mutation into Cas9-expressing cardiomyocytes by gRNA transfection and found that it decreases the expression of PKD1-encoded PC1 protein that co-localizes specifically with Serca2a and L-type voltage-gated calcium channels. We also detected the suppression of Ca2+ amplitude in ventricular cardiomyocytes with decreased PC1 expression in mixed culture. Our HCIA method provides comprehensive kinetic and static information on individual cardiomyocytes and allows the pathogenicity of mutations to be determined rapidly.
Collapse
Affiliation(s)
- Tomoka Tabata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yuki Masumura
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shuichiro Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan; Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Suzuka Kunimatsu
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Satoshi Kameda
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Hiroyuki Inoue
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shota Okuno
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shou Ogawa
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Mikio Watanabe
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
15
|
Shiels HA. Avian cardiomyocyte architecture and what it reveals about the evolution of the vertebrate heart. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210332. [PMID: 36189815 PMCID: PMC9527935 DOI: 10.1098/rstb.2021.0332] [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] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022] Open
Abstract
Bird cardiomyocytes are long, thin and lack transverse (t)-tubules, which is akin to the cardiomyocyte morphology of ectothermic non-avian reptiles, who are typified by low maximum heart rates and low pressure development. However, birds can achieve greater contractile rates and developed pressures than mammals, whose wide cardiomyocytes contain a dense t-tubular network allowing for uniform excitation-contraction coupling and strong contractile force. To address this apparent paradox, this paper functionally links recent electrophysiological studies on bird cardiomyocytes with decades of ultrastructure measurements. It shows that it is the strong transsarcolemmal Ca2+ influx via the L-type Ca2+ current (ICaL) and the high gain of Ca2+-induced Ca2+ release from the sarcoplasmic reticulum (SR), coupled with an internal SR Ca2+ release relay system, that facilitates the strong fast contractions in the long thin bird cardiomyocytes, without the need for t-tubules. The maintenance of an elongated myocyte morphology following the post-hatch transition from ectothermy to endothermy in birds is discussed in relation to cardiac load, myocyte ploidy, and cardiac regeneration potential in adult cardiomyocytes. Overall, the paper shows how little we know about cellular Ca2+ dynamics in the bird heart and suggests how increased research efforts in this area would provide vital information in our quest to understand the role of myocyte architecture in the evolution of the vertebrate heart. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'. Please see glossary at the end of the paper for definitions of specialized terms.
Collapse
Affiliation(s)
- Holly A. Shiels
- Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
16
|
PGC-1α4 Interacts with REST to Upregulate Neuronal Genes and Augment Energy Consumption in Developing Cardiomyocytes. Cells 2022; 11:cells11192944. [PMID: 36230906 PMCID: PMC9564192 DOI: 10.3390/cells11192944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 02/02/2023] Open
Abstract
Transcriptional coactivator PGC-1α is a main regulator of cardiac energy metabolism. In addition to canonical PGC-1α1, other PGC-1α isoforms have been found to exert specific biological functions in a variety of tissues. We investigated the expression patterns and the biological effects of the non-canonical isoforms in the heart. We used RNA sequencing data to identify the expression patterns of PGC-1α isoforms in the heart. To evaluate the biological effects of the alternative isoform expression, we generated a transgenic mouse with cardiac-specific overexpression of PGC-1α4 and analysed the cardiac phenotype with a wide spectrum of physiological and biophysical tools. Our results show that non-canonical isoforms are expressed in the heart, and that the main variant PGC-1α4 is induced by β-adrenergic signalling in adult cardiomyocytes. Cardiomyocyte specific PGC-1α4 overexpression in mice relieves the RE1-Silencing Transcription factor (REST)-mediated suppression of neuronal genes during foetal heart development. The resulting de-repression of REST target genes induces a cardiac phenotype with increased cellular energy consumption, resulting in postnatal dilated cardiomyopathy. These results propose a new concept for actions of the PGC-1α protein family where activation of the Pgc-1α gene, through its isoforms, induces a phenotype with concurrent supply and demand for cellular energy. These data highlight the biological roles of the different PGC-1α isoforms, which should be considered when future therapies are developed.
Collapse
|
17
|
Shen S, Sewanan LR, Shao S, Halder SS, Stankey P, Li X, Campbell SG. Physiological calcium combined with electrical pacing accelerates maturation of human engineered heart tissue. Stem Cell Reports 2022; 17:2037-2049. [PMID: 35931080 PMCID: PMC9481907 DOI: 10.1016/j.stemcr.2022.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/24/2022] Open
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have wide potential application in basic research, drug discovery, and regenerative medicine, but functional maturation remains challenging. Here, we present a method whereby maturation of hiPSC-CMs can be accelerated by simultaneous application of physiological Ca2+ and frequency-ramped electrical pacing in culture. This combination produces positive force-frequency behavior, physiological twitch kinetics, robust β-adrenergic response, improved Ca2+ handling, and cardiac troponin I expression within 25 days. This study provides insights into the role of Ca2+ in hiPSC-CM maturation and offers a scalable platform for translational and clinical research.
Collapse
Affiliation(s)
- Shi Shen
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Lorenzo R Sewanan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephanie Shao
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Saiti S Halder
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Paul Stankey
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Xia Li
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, 55 Prospect St. MEC 211, New Haven, CT 06511, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
Hierarchically Structured Polystyrene-Based Surfaces Amplifying Fluorescence Signals: Cytocompatibility with Human Induced Pluripotent Stem Cell. Int J Mol Sci 2021; 22:ijms222111943. [PMID: 34769373 PMCID: PMC8584612 DOI: 10.3390/ijms222111943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/10/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
An innovative multi-step phase separation process was used to prepare tissue culture for the polystyrene-based, hierarchically structured substrates, which mimicked in vivo microenvironment and architecture. Macro- (pore area from 3000 to 18,000 µm2; roughness (Ra) 7.2 ± 0.1 µm) and meso- (pore area from 50 to 300 µm2; Ra 1.1 ± 0.1 µm) structured substrates covered with micro-pores (area around 3 µm2) were prepared and characterised. Both types of substrate were suitable for human-induced pluripotent stem cell (hiPSC) cultivation and were found to be beneficial for the induction of cardiomyogenesis in hiPSC. This was confirmed both by the number of promoted proliferated cells and the expressions of specific markers (Nkx2.5, MYH6, MYL2, and MYL7). Moreover, the substrates amplified the fluorescence signal when Ca2+ flow was monitored. This property, together with cytocompatibility, make this material especially suitable for in vitro studies of cell/material interactions within tissue-mimicking environments.
Collapse
|
19
|
Chaklader M, Rothermel BA. Calcineurin in the heart: New horizons for an old friend. Cell Signal 2021; 87:110134. [PMID: 34454008 PMCID: PMC8908812 DOI: 10.1016/j.cellsig.2021.110134] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 01/20/2023]
Abstract
Calcineurin, also known as PP2B or PPP3, is a member of the PPP family of protein phosphatases that also includes PP1 and PP2A. Together these three phosphatases carryout the majority of dephosphorylation events in the heart. Calcineurin is distinct in that it is activated by the binding of calcium/calmodulin (Ca2+/CaM) and therefore acts as a node for integrating Ca2+ signals with changes in phosphorylation, two fundamental intracellular signaling cascades. In the heart, calcineurin is primarily thought of in the context of pathological cardiac remodeling, acting through the Nuclear Factor of Activated T-cell (NFAT) family of transcription factors. However, calcineurin activity is also essential for normal heart development and homeostasis in the adult heart. Furthermore, it is clear that NFAT-driven changes in transcription are not the only relevant processes initiated by calcineurin in the setting of pathological remodeling. There is a growing appreciation for the diversity of calcineurin substrates that can impact cardiac function as well as the diversity of mechanisms for targeting calcineurin to specific sub-cellular domains in cardiomyocytes and other cardiac cell types. Here, we will review the basics of calcineurin structure, regulation, and function in the context of cardiac biology. Particular attention will be given to: the development of improved tools to identify and validate new calcineurin substrates; recent studies identifying new calcineurin isoforms with unique properties and targeting mechanisms; and the role of calcineurin in cardiac development and regeneration.
Collapse
Affiliation(s)
- Malay Chaklader
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Beverly A Rothermel
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
20
|
Setterberg IE, Le C, Frisk M, Li J, Louch WE. The Physiology and Pathophysiology of T-Tubules in the Heart. Front Physiol 2021; 12:718404. [PMID: 34566684 PMCID: PMC8458775 DOI: 10.3389/fphys.2021.718404] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
In cardiomyocytes, invaginations of the sarcolemmal membrane called t-tubules are critically important for triggering contraction by excitation-contraction (EC) coupling. These structures form functional junctions with the sarcoplasmic reticulum (SR), and thereby enable close contact between L-type Ca2+ channels (LTCCs) and Ryanodine Receptors (RyRs). This arrangement in turn ensures efficient triggering of Ca2+ release, and contraction. While new data indicate that t-tubules are capable of exhibiting compensatory remodeling, they are also widely reported to be structurally and functionally compromised during disease, resulting in disrupted Ca2+ homeostasis, impaired systolic and/or diastolic function, and arrhythmogenesis. This review summarizes these findings, while highlighting an emerging appreciation of the distinct roles of t-tubules in the pathophysiology of heart failure with reduced and preserved ejection fraction (HFrEF and HFpEF). In this context, we review current understanding of the processes underlying t-tubule growth, maintenance, and degradation, underscoring the involvement of a variety of regulatory proteins, including junctophilin-2 (JPH2), amphiphysin-2 (BIN1), caveolin-3 (Cav3), and newer candidate proteins. Upstream regulation of t-tubule structure/function by cardiac workload and specifically ventricular wall stress is also discussed, alongside perspectives for novel strategies which may therapeutically target these mechanisms.
Collapse
Affiliation(s)
- Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Christopher Le
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
21
|
Salem T, Frankman Z, Churko J. Tissue engineering techniques for iPSC derived three-dimensional cardiac constructs. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:891-911. [PMID: 34476988 PMCID: PMC9419978 DOI: 10.1089/ten.teb.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent developments in applied developmental physiology have provided well-defined methodologies for producing human stem cell derived cardiomyocytes. Cardiomyocytes produced in this way have become commonplace as cardiac physiology research models. This accessibility has also allowed for the development of tissue engineered human heart constructs for drug screening, surgical intervention, and investigating cardiac pathogenesis. However, cardiac tissue engineering is an interdisciplinary field that involves complex engineering and physiological concepts, which limits its accessibility. This review provides a readable, broad reaching, and thorough discussion of major factors to consider for the development of cardiovascular tissues from stem cell derived cardiomyocytes. This review will examine important considerations in undertaking a cardiovascular tissue engineering project, and will present, interpret, and summarize some of the recent advancements in this field. This includes reviewing different forms of tissue engineered constructs, a discussion on cardiomyocyte sources, and an in-depth discussion of the fabrication and maturation procedures for tissue engineered heart constructs.
Collapse
Affiliation(s)
- Tori Salem
- University of Arizona Medical Center - University Campus, 22165, Cellular and Molecular Medicine, Tucson, Arizona, United States;
| | - Zachary Frankman
- University of Arizona Medical Center - University Campus, 22165, Biomedical Engineering, Tucson, Arizona, United States;
| | - Jared Churko
- University of Arizona Medical Center - University Campus, 22165, 1501 N Campbell RD, SHC 6143, Tucson, Arizona, United States, 85724-5128;
| |
Collapse
|
22
|
Scalzo S, Afonso MQ, da Fonseca NJ, Jesus IC, Alves AP, Mendonça CAF, Teixeira VP, Biagi D, Cruvinel E, Santos AK, Miranda K, Marques FA, Mesquita ON, Kushmerick C, Campagnole-Santos MJ, Agero U, Guatimosim S. Dense optical flow software to quantify cellular contractility. CELL REPORTS METHODS 2021; 1:100044. [PMID: 35475144 PMCID: PMC9017166 DOI: 10.1016/j.crmeth.2021.100044] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/02/2021] [Accepted: 06/14/2021] [Indexed: 01/02/2023]
Abstract
Cell membrane deformation is an important feature that occurs during many physiological processes, and its study has been put to good use to investigate cardiomyocyte function. Several methods have been developed to extract information on cardiomyocyte contractility. However, no existing computational framework has provided, in a single platform, a straightforward approach to acquire, process, and quantify this type of cellular dynamics. For this reason, we develop CONTRACTIONWAVE, high-performance software written in Python programming language that allows the user to process large data image files and obtain contractility parameters by analyzing optical flow from images obtained with videomicroscopy. The software was validated by using neonatal, adult-, and human-induced pluripotent stem-cell-derived cardiomyocytes, treated or not with drugs known to affect contractility. Results presented indicate that CONTRACTIONWAVE is an excellent tool for examining changes to cardiac cellular contractility in animal models of disease and for pharmacological and toxicology screening during drug discovery.
Collapse
Affiliation(s)
- Sérgio Scalzo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Marcelo Q.L. Afonso
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Néli J. da Fonseca
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
- Cellular Structure and 3D Bioimaging, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Itamar C.G. Jesus
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Ana Paula Alves
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Carolina A.T. F. Mendonça
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Vanessa P. Teixeira
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Diogo Biagi
- PluriCell Biotech, São Paulo, SP 05508-000, Brazil
| | | | - Anderson K. Santos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Kiany Miranda
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Flavio A.M. Marques
- Departamento de Física, Instituto de Ciências Naturais, Universidade Federal de Lavras, Lavras, MG 37200-900, Brazil
| | - Oscar N. Mesquita
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Christopher Kushmerick
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Maria José Campagnole-Santos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Ubirajara Agero
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Silvia Guatimosim
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| |
Collapse
|
23
|
Knyrim M, Rabe S, Grossmann C, Gekle M, Schreier B. Influence of miR-221/222 on cardiomyocyte calcium handling and function. Cell Biosci 2021; 11:160. [PMID: 34404451 PMCID: PMC8369661 DOI: 10.1186/s13578-021-00676-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/02/2021] [Indexed: 11/10/2022] Open
Abstract
Background Cardiovascular disease is the leading cause of death worldwide. Cardiac electrical remodeling including altered ion channel expression and imbalance of calcium homeostasis can have detrimental effects on cardiac function. While it has been extensively reported that miR-221/222 are involved in structural remodeling, their role in electrical remodeling still has to be evaluated. We previously reported that subunits of the L-type Ca2+ channel (LTCC) are direct targets of miR-221/222. Furthermore, HL-1 cells transfected with miR-221 or -222 mimics showed a reduction in LTCC current density while the voltage-dependence of activation was not altered. The aim of the present study was to determine the influence of miR-221/222 on cardiomyocyte calcium handling and function. Results Transient transfection of HL-1 cells with miR-221/222 mimics led to slower depolarization-dependent Ca2+ entry and increased proportion of non-responding cells. Angiotensin II-induced Ca2+ release from the SR was not affected by miR-221/222. In miR-222-transfected neonatal cardiomyocytes the isoprenaline-induced positive inotropic effect on the intracellular Ca2+ transient was lost and the positive chronotropic effect on spontaneous beating activity was strongly reduced. This could have severe consequences for cardiomyocytes and could lead to a reduced contractility and systolic dysfunction of the whole heart. Conclusions This study adds a new role of miR-221/222 in cardiomyocytes by showing the impact on β-adrenergic regulation of LTCC function, calcium handling and beating frequency. Together with the previous report that miR-221/222 reduce GIRK1/4 function and LTCC current density, it expands our knowledge about the role of these miRs on cardiac ion channel regulation. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00676-4.
Collapse
Affiliation(s)
- Maria Knyrim
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle (Saale), Germany.
| | - Sindy Rabe
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle (Saale), Germany
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle (Saale), Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle (Saale), Germany
| | - Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle (Saale), Germany
| |
Collapse
|
24
|
Li Y, Zhang Z, Li S, Yu T, Jia Z. Therapeutic Effects of Traditional Chinese Medicine on Cardiovascular Diseases: the Central Role of Calcium Signaling. Front Pharmacol 2021; 12:682273. [PMID: 34305595 PMCID: PMC8299363 DOI: 10.3389/fphar.2021.682273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Calcium, as a second messenger, plays an important role in the pathogenesis of cardiovascular diseases (CVDs). The malfunction of calcium signaling in endothelial cells and vascular smooth muscle cells promotes hypertension. In cardiomyocytes, calcium overload induces apoptosis, leading to myocardial infarction and arrhythmias. Moreover, the calcium–calcineurin–nuclear factor of activated T cells (NFAT) pathway is essential for expressing the cardiac pro-hypertrophic gene. Heart failure is also characterized by reduced calcium transient amplitude and enhanced sarcoplasmic reticulum (SR) calcium leakage. Traditional Chinese medicine (TCM) has been used to treat CVDs for thousands of years in China. Because of its multicomponent and multitarget characteristics, TCM's unique advantages in CVD treatment are closely related to the modulation of multiple calcium handling proteins and calcium signaling pathways in different types of cells involved in distinct CVDs. Thus, we systematically review the diverse mechanisms of TCM in regulating calcium pathways to treat various types of CVDs, ranging from hypertrophic cardiomyopathy to diabetic heart disease.
Collapse
Affiliation(s)
- Yuxin Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhang Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Tingting Yu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhaoqi Jia
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Naumenko N, Mutikainen M, Holappa L, Ruas JL, Tuomainen T, Tavi P. PGC-1α deficiency reveals sex-specific links between cardiac energy metabolism and EC-coupling during development of heart failure in mice. Cardiovasc Res 2021; 118:1520-1534. [PMID: 34086875 PMCID: PMC9074965 DOI: 10.1093/cvr/cvab188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 06/03/2021] [Indexed: 12/24/2022] Open
Abstract
Aims Biological sex has fundamental effects on mammalian heart physiology and pathogenesis. While it has been established that female sex is a protective factor against most cardiovascular diseases (CVDs), this beneficial effect may involve pathways associated with cardiac energy metabolism. Our aim was to elucidate the role of transcriptional coactivator PGC-1α in sex dimorphism of heart failure (HF) development. Methods and results Here, we show that mice deficient in cardiac expression of the peroxisome proliferator-activated receptor gamma (PPAR-γ) coactivator-1α (PGC-1α) develop dilated HF associated with changes in aerobic and anaerobic metabolism, calcium handling, cell structure, electrophysiology, as well as gene expression. These cardiac changes occur in both sexes, but female mice develop an earlier and more severe structural and functional phenotype associated with dyssynchronous local calcium release resulting from disruption of t-tubular structures of the cardiomyocytes. Conclusions These data reveal that the integrity of the subcellular Ca2+ release and uptake machinery is dependent on energy metabolism and that female hearts are more prone to suffer from contractile dysfunction in conditions with compromised production of cellular energy. Furthermore, these findings suggest that PGC-1α is a central mediator of sex-specific differences in heart function and CVD susceptibility.
Collapse
Affiliation(s)
- Nikolay Naumenko
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maija Mutikainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lari Holappa
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Tomi Tuomainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi Tavi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
26
|
Iaparov BI, Zahradnik I, Moskvin AS, Zahradníková A. In silico simulations reveal that RYR distribution affects the dynamics of calcium release in cardiac myocytes. J Gen Physiol 2021; 153:211900. [PMID: 33735373 PMCID: PMC7980188 DOI: 10.1085/jgp.202012685] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 02/08/2021] [Indexed: 11/20/2022] Open
Abstract
The dyads of cardiac myocytes contain ryanodine receptors (RYRs) that generate calcium sparks upon activation. To test how geometric factors of RYR distribution contribute to the formation of calcium sparks, which cannot be addressed experimentally, we performed in silico simulations on a large set of models of calcium release sites (CRSs). Our models covered the observed range of RYR number, density, and spatial arrangement. The calcium release function of CRSs was modeled by RYR openings, with an open probability dependent on concentrations of free Ca2+ and Mg2+ ions, in a rapidly buffered system, with a constant open RYR calcium current. We found that simulations of spontaneous sparks by repeatedly opening one of the RYRs in a CRS produced three different types of calcium release events (CREs) in any of the models. Transformation of simulated CREs into fluorescence signals yielded calcium sparks with characteristics close to the observed ones. CRE occurrence varied broadly with the spatial distribution of RYRs in the CRS but did not consistently correlate with RYR number, surface density, or calcium current. However, it correlated with RYR coupling strength, defined as the weighted product of RYR vicinity and calcium current, so that CRE characteristics of all models followed the same state-response function. This finding revealed the synergy between structure and function of CRSs in shaping dyad function. Lastly, rearrangements of RYRs simulating hypothetical experiments on splitting and compaction of a dyad revealed an increased propensity to generate spontaneous sparks and an overall increase in calcium release in smaller and more compact dyads, thus underlying the importance and physiological role of RYR arrangement in cardiac myocytes.
Collapse
Affiliation(s)
- Bogdan I Iaparov
- Department of Cellular Cardiology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.,Research Institute of Physics and Applied Mathematics, and Department of Theoretical and Mathematical Physics, Ural Federal University, Ekaterinburg, Russia
| | - Ivan Zahradnik
- Department of Cellular Cardiology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alexander S Moskvin
- Research Institute of Physics and Applied Mathematics, and Department of Theoretical and Mathematical Physics, Ural Federal University, Ekaterinburg, Russia
| | - Alexandra Zahradníková
- Department of Cellular Cardiology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
27
|
Kay M, Soltani BM. LncRNAs in Cardiomyocyte Maturation: New Window for Cardiac Regenerative Medicine. Noncoding RNA 2021; 7:ncrna7010020. [PMID: 33802186 PMCID: PMC8005985 DOI: 10.3390/ncrna7010020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiomyocyte (CM) maturation, which is characterized by structural, functional, and metabolic specializations, is the last phase of CM development that prepares the cells for efficient and forceful contraction throughout life. Over the past decades, CM maturation has gained increased attention due to the fact that pluripotent stem cell-derived CMs are structurally, transcriptionally, and functionally immature and embryonic-like, which causes a defect in cell replacement therapy. The current challenge is to discover and understand the molecular mechanisms, which control the CM maturation process. Currently, emerging shreds of evidence emphasize the role of long noncoding RNAs (lncRNAs) in regulating different aspects of CM maturation, including myofibril maturation, electrophysiology, and Ca2+ handling maturation, metabolic maturation and proliferation to hypertrophy transition. Here, we describe the structural and functional characteristics of mature CMs. Furthermore, this review highlights the lncRNAs as crucial regulators of different aspects in CM maturation, which have the potential to be used for mature CM production. With the current advances in oligonucleotide delivery; lncRNAs may serve as putative therapeutic targets to produce highly mature CMs for research and regenerative medicine.
Collapse
|
28
|
Schranz D. Pharmacological Heart Failure Therapy in Children: Focus on Inotropic Support. Handb Exp Pharmacol 2020; 261:177-192. [PMID: 31707469 DOI: 10.1007/164_2019_267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Pediatric heart failure is a clinical syndrome, which needs to be distinctly defined and the pathophysiological consequences considered. Pharmacological treatment depends on the disease- and age-specific myocardial characteristics. Acute and chronic low cardiac output is the result of an inadequate heart rate (rhythm), myocardial contractility, preload and afterload, and also ventriculo-ventricular interaction, synchrony, atrio-ventricular and ventricular-arterial coupling. The treatment of choice is curing the cause of heart failure, if possible.Acute HF therapy is still based to the use of catecholamines and inodilators. The cornerstone of chronic HF treatment consists of blocking the endogenous, neuro-humoral axis, in particular the adrenergic and renin-angiotensin-aldosterone system.Before neprilysin inhibitors are used in young children, their potential side-effect for inducing Alzheimer disease needs to be clarified. The focus of the current review is put on the differential use of the inotropic drugs as epinephrine, norepinephrine, dopamine and dobutamine, and also the inodilators milrinone and levosimendan. Considering effects and side-effects of any cardiac stimulating treatment strategy, co-medication with ß-blockers, angiotensin converting inhibitors (ACEIs), angiotensin blockers (ARBs) and mineralocorticoid receptor antagonists (MRAs) is not a contradiction, but a senseful measure, even still during the acute inotropic treatment.Missing sophisticated clinical trials using accurate entry criteria and clinically relevant endpoints, there is especially in cardiovascular diagnosis and treatment of young children a compromise of evidence-based versus pathophysiology-based procedures. But based on the pharmacological and pathophysiological knowledge a hypothesis-driven individualized treatment is already currently possible and therefore indicated.
Collapse
Affiliation(s)
- Dietmar Schranz
- Pediatric Heart Center, Johann Wolfgang Goethe University Clinic, Frankfurt, Germany.
| |
Collapse
|
29
|
Montero P, Flandes-Iparraguirre M, Musquiz S, Pérez Araluce M, Plano D, Sanmartín C, Orive G, Gavira JJ, Prosper F, Mazo MM. Cells, Materials, and Fabrication Processes for Cardiac Tissue Engineering. Front Bioeng Biotechnol 2020; 8:955. [PMID: 32850768 PMCID: PMC7431658 DOI: 10.3389/fbioe.2020.00955] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease is the number one killer worldwide, with myocardial infarction (MI) responsible for approximately 1 in 6 deaths. The lack of endogenous regenerative capacity, added to the deleterious remodelling programme set into motion by myocardial necrosis, turns MI into a progressively debilitating disease, which current pharmacological therapy cannot halt. The advent of Regenerative Therapies over 2 decades ago kick-started a whole new scientific field whose aim was to prevent or even reverse the pathological processes of MI. As a highly dynamic organ, the heart displays a tight association between 3D structure and function, with the non-cellular components, mainly the cardiac extracellular matrix (ECM), playing both fundamental active and passive roles. Tissue engineering aims to reproduce this tissue architecture and function in order to fabricate replicas able to mimic or even substitute damaged organs. Recent advances in cell reprogramming and refinement of methods for additive manufacturing have played a critical role in the development of clinically relevant engineered cardiovascular tissues. This review focuses on the generation of human cardiac tissues for therapy, paying special attention to human pluripotent stem cells and their derivatives. We provide a perspective on progress in regenerative medicine from the early stages of cell therapy to the present day, as well as an overview of cellular processes, materials and fabrication strategies currently under investigation. Finally, we summarise current clinical applications and reflect on the most urgent needs and gaps to be filled for efficient translation to the clinical arena.
Collapse
Affiliation(s)
- Pilar Montero
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
| | - María Flandes-Iparraguirre
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
| | - Saioa Musquiz
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country – UPV/EHU, Vitoria-Gasteiz, Spain
| | - María Pérez Araluce
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country – UPV/EHU, Vitoria-Gasteiz, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- University Institute for Regenerative Medicine and Oral Implantology – UIRMI (UPV/EHU – Fundación Eduardo Anitua), Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, Singapore, Singapore
| | - Juan José Gavira
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Cardiology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
30
|
Ramadan M, Cooper B, Posnack NG. Bisphenols and phthalates: Plastic chemical exposures can contribute to adverse cardiovascular health outcomes. Birth Defects Res 2020; 112:1362-1385. [PMID: 32691967 DOI: 10.1002/bdr2.1752] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022]
Abstract
Phthalates and bisphenols are high production volume chemicals that are used in the manufacturing of consumer and medical products. Given the ubiquity of bisphenol and phthalate chemicals in the environment, biomonitoring studies routinely detect these chemicals in 75-90% of the general population. Accumulating evidence suggests that such chemical exposures may influence human health outcomes, including cardiovascular health. These associations are particularly worrisome for sensitive populations, including fetal, infant and pediatric groups-with underdeveloped metabolic capabilities and developing organ systems. In the presented article, we aimed to review the literature on environmental and clinical exposures to bisphenols and phthalates, highlight experimental work that suggests that these chemicals may exert a negative influence on cardiovascular health, and emphasize areas of concern that relate to vulnerable pediatric groups. Gaps in our current knowledge are also discussed, so that future endeavors may resolve the relationship between chemical exposures and the impact on pediatric cardiovascular physiology.
Collapse
Affiliation(s)
- Manelle Ramadan
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia, USA.,Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia, USA
| | - Blake Cooper
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia, USA
| | - Nikki Gillum Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia, USA.,Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington University, School of Medicine, Washington, District of Columbia, USA.,Department of Pharmacology & Physiology, George Washington University, School of Medicine, Washington, District of Columbia, USA
| |
Collapse
|
31
|
Structural variability of dyads relates to calcium release in rat ventricular myocytes. Sci Rep 2020; 10:8076. [PMID: 32415205 PMCID: PMC7229197 DOI: 10.1038/s41598-020-64840-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/11/2020] [Indexed: 12/30/2022] Open
Abstract
Cardiac excitation-contraction coupling relies on dyads, the intracellular calcium synapses of cardiac myocytes, where the plasma membrane contacts sarcoplasmic reticulum and where electrical excitation triggers calcium release. The morphology of dyads and dynamics of local calcium release vary substantially. To better understand the correspondence between the structure and the functionality of dyads, we estimated incidences of structurally different dyads and of kinetically different calcium release sites and tested their responsiveness to experimental myocardial injury in left ventricular myocytes of rats. According to the structure of dyads estimated in random electron microscopic images of myocardial tissue, the dyads were sorted into 'compact' or 'loose' types. The calcium release fluxes, triggered at local calcium release sites in patch-clamped ventricular myocytes and recorded by laser scanning confocal fluorescence microscopy, were decomposed into 'early' and 'late' components. ANOVA tests revealed very high correlation between the relative amplitudes of early and late calcium release flux components and the relative occurrences of compact and loose dyads in the control and in the injured myocardium. This finding ascertained the relationship between the structure of dyads and the functionality of calcium release sites and the responsiveness of calcium release sites to physical load in cardiac myocytes.
Collapse
|
32
|
Cong S, Ramachandra CJ, Mai Ja KPM, Yap J, Shim W, Wei L, Hausenloy DJ. Mechanisms underlying diabetic cardiomyopathy: From pathophysiology to novel therapeutic targets. CONDITIONING MEDICINE 2020; 3:82-97. [PMID: 34169234 PMCID: PMC8221238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Diabetic cardiomyopathy (DC) is defined as a clinical condition of cardiac dysfunction that occurs in the absence of coronary atherosclerosis, valvular disease, and hypertension in patients with diabetes mellitus (DM). Despite the increasing worldwide prevalence of DC, due to the global epidemic of DM, the underlying pathophysiology of DC has not been fully elucidated. In addition, the clinical criteria for diagnosing DC have not been established, and specific therapeutic options are not currently available. The current paradigm suggests the impaired cardiomyocyte function arises due to a number of DM-related metabolic disturbances including hyperglycemia, hyperinsulinemia, and hyperlipidemia, which lead to diastolic dysfunction and signs and symptoms of heart failure. Other factors, which have been implicated in the progression of DC, include mitochondrial dysfunction, increased oxidative stress, impaired calcium handling, inflammation, and cardiomyocyte apoptosis. Herein, we review the current theories surrounding the occurrence and progression of DC, and discuss the recent advances in diagnostic methodologies and therapeutic strategies. Moreover, apart from conventional animal DC models, we highlight alternative disease models for studying DC such as the use of patient-derived human induced pluripotent stem cells (hiPSCs) for studying the mechanisms underlying DC. The ability to obtain hiPSC-derived cardiomyocytes from DM patients with a DC phenotype could help identify novel therapeutic targets for preventing and delaying the progression of DC, and for improving clinical outcomes in DM patients.
Collapse
Affiliation(s)
- Shuo Cong
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Chrishan J.A. Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore
| | - KP Myu Mai Ja
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore
| | - Lai Wei
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Centre of Cardiac Valve, Shanghai, China
| | - Derek J. Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore
- Yong Loo Lin Medical School, National University of Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
- Cardiovascular Research Centre, College of Medical and Health Sciences, Asia University, Taiwan
| |
Collapse
|
33
|
Karbassi E, Fenix A, Marchiano S, Muraoka N, Nakamura K, Yang X, Murry CE. Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat Rev Cardiol 2020; 17:341-359. [PMID: 32015528 DOI: 10.1038/s41569-019-0331-x] [Citation(s) in RCA: 454] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 12/20/2022]
Abstract
Our knowledge of pluripotent stem cell (PSC) biology has advanced to the point where we now can generate most cells of the human body in the laboratory. PSC-derived cardiomyocytes can be generated routinely with high yield and purity for disease research and drug development, and these cells are now gradually entering the clinical research phase for the testing of heart regeneration therapies. However, a major hurdle for their applications is the immature state of these cardiomyocytes. In this Review, we describe the structural and functional properties of cardiomyocytes and present the current approaches to mature PSC-derived cardiomyocytes. To date, the greatest success in maturation of PSC-derived cardiomyocytes has been with transplantation into the heart in animal models and the engineering of 3D heart tissues with electromechanical conditioning. In conventional 2D cell culture, biophysical stimuli such as mechanical loading, electrical stimulation and nanotopology cues all induce substantial maturation, particularly of the contractile cytoskeleton. Metabolism has emerged as a potent means to control maturation with unexpected effects on electrical and mechanical function. Different interventions induce distinct facets of maturation, suggesting that activating multiple signalling networks might lead to increased maturation. Despite considerable progress, we are still far from being able to generate PSC-derived cardiomyocytes with adult-like phenotypes in vitro. Future progress will come from identifying the developmental drivers of maturation and leveraging them to create more mature cardiomyocytes for research and regenerative medicine.
Collapse
Affiliation(s)
- Elaheh Karbassi
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Aidan Fenix
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Silvia Marchiano
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Naoto Muraoka
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA. .,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA. .,Department of Pathology, University of Washington, Seattle, WA, USA. .,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
34
|
Swift LM, Burke M, Guerrelli D, Reilly M, Ramadan M, McCullough D, Prudencio T, Mulvany C, Chaluvadi A, Jaimes R, Posnack NG. Age-dependent changes in electrophysiology and calcium handling: implications for pediatric cardiac research. Am J Physiol Heart Circ Physiol 2019; 318:H354-H365. [PMID: 31886723 DOI: 10.1152/ajpheart.00521.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Rodent models are frequently employed in cardiovascular research, yet our understanding of pediatric cardiac physiology has largely been deduced from more simplified two-dimensional cell studies. Previous studies have shown that postnatal development includes an alteration in the expression of genes and proteins involved in cell coupling, ion channels, and intracellular calcium handling. Accordingly, we hypothesized that postnatal cell maturation is likely to lead to dynamic alterations in whole heart electrophysiology and calcium handling. To test this hypothesis, we employed multiparametric imaging and electrophysiological techniques to quantify developmental changes from neonate to adult. In vivo electrocardiograms were collected to assess changes in heart rate, variability, and atrioventricular conduction (Sprague-Dawley rats). Intact, whole hearts were transferred to a Langendorff-perfusion system for multiparametric imaging (voltage, calcium). Optical mapping was performed in conjunction with an electrophysiology study to assess cardiac dynamics throughout development. Postnatal age was associated with an increase in the heart rate (181 ± 34 vs. 429 ± 13 beats/min), faster atrioventricular conduction (94 ± 13 vs. 46 ± 3 ms), shortened action potentials (APD80: 113 ± 18 vs. 60 ± 17 ms), and decreased ventricular refractoriness (VERP: 157 ± 45 vs. 57 ± 14 ms; neonatal vs. adults, means ± SD, P < 0.05). Calcium handling matured with development, resulting in shortened calcium transient durations (168 ± 18 vs. 117 ± 14 ms) and decreased propensity for calcium transient alternans (160 ± 18- vs. 99 ± 11-ms cycle length threshold; neonatal vs. adults, mean ± SD, P < 0.05). Results of this study can serve as a comprehensive baseline for future studies focused on pediatric disease modeling and/or preclinical testing.NEW & NOTEWORTHY This is the first study to assess cardiac electrophysiology and calcium handling throughout postnatal development, using both in vivo and whole heart models.
Collapse
Affiliation(s)
- Luther M Swift
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Morgan Burke
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Devon Guerrelli
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Marissa Reilly
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Manelle Ramadan
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Damon McCullough
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Tomas Prudencio
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Colm Mulvany
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Ashika Chaluvadi
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Rafael Jaimes
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia
| | - Nikki Gillum Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia.,Children's National Heart Institute, Children's National Health System, Washington, District of Columbia.,Department of Pediatrics and Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, District of Columbia
| |
Collapse
|
35
|
Soucy JR, Askaryan J, Diaz D, Koppes AN, Annabi N, Koppes RA. Glial cells influence cardiac permittivity as evidenced through in vitro and in silico models. Biofabrication 2019; 12:015014. [PMID: 31593932 PMCID: PMC11062241 DOI: 10.1088/1758-5090/ab4c0a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Excitation-contraction (EC) coupling in the heart has, until recently, been solely accredited to cardiomyocytes. The inherent complexities of the heart make it difficult to examine non-muscle contributions to contraction in vivo, and conventional in vitro models fail to capture multiple features and cellular heterogeneity of the myocardium. Here, we report on the development of a 3D cardiac μTissue to investigate changes in the cellular composition of native myocardium in vitro. Cells are encapsulated within micropatterned gelatin-based hydrogels formed via visible light photocrosslinking. This system enables spatial control of the microarchitecture, perturbation of the cellular composition, and functional measures of EC coupling via video microscopy and a custom algorithm to quantify beat frequency and degree of coordination. To demonstrate the robustness of these tools and evaluate the impact of altered cell population densities on cardiac μTissues, contractility and cell morphology were assessed with the inclusion of exogenous non-myelinating Schwann cells (SCs). Results demonstrate that the addition of exogenous SCs alter cardiomyocyte EC, profoundly inhibiting the response to electrical pacing. Computational modeling of connexin-mediated coupling suggests that SCs impact cardiomyocyte resting potential and rectification following depolarization. Cardiac μTissues hold potential for examining the role of cellular heterogeneity in heart health, pathologies, and cellular therapies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, United States of America
| | | | | | | | | | | |
Collapse
|
36
|
Kane C, Terracciano CM. Human Cardiac Fibroblasts Engage the Sarcoplasmic Reticulum in Induced Pluripotent Stem Cell-Derived Cardiomyocyte Excitation-Contraction Coupling. J Am Coll Cardiol 2019; 72:1061-1063. [PMID: 30139435 DOI: 10.1016/j.jacc.2018.06.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/30/2018] [Accepted: 06/10/2018] [Indexed: 11/30/2022]
|
37
|
Lemoine MD, Krause T, Koivumäki JT, Prondzynski M, Schulze ML, Girdauskas E, Willems S, Hansen A, Eschenhagen T, Christ T. Human Induced Pluripotent Stem Cell-Derived Engineered Heart Tissue as a Sensitive Test System for QT Prolongation and Arrhythmic Triggers. Circ Arrhythm Electrophysiol 2019; 11:e006035. [PMID: 29925535 DOI: 10.1161/circep.117.006035] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 05/08/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Cardiac repolarization abnormalities in drug-induced and genetic long-QT syndrome may lead to afterdepolarizations and life-threatening ventricular arrhythmias. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) should help to overcome the limitations of animal models based on species differences in repolarization reserve. Here, we compared head-to-head the contribution of IKs (long QT1) and IKr (long QT2) on action potentials (APs) in human left ventricular (LV) tissue and hiPSC-CM-derived engineered heart tissue (EHT). METHODS APs were measured with sharp microelectrodes in EHT from 3 different control hiPSC-CM lines and in tissue preparations from failing LV. RESULTS EHT from hiPSC-CMs showed spontaneous diastolic depolarization and AP generation that were sensitive to low concentrations of ivabradine. IKr block by E-4031 prolonged AP duration at 90% repolarization with similar half-effective concentration in EHT and LV but larger effect size in EHT (+281 versus +110 ms in LV). Although IKr block alone evoked early afterdepolarizations and triggered activity in 50% of EHTs, slow pacing, reduced extracellular K+, and blocking of IKr, IKs, and IK1 were necessary to induce early afterdepolarizations in LV. In accordance with their clinical safety, moxifloxacin and verapamil did not induce early afterdepolarizations in EHT. In both EHT and LV, IKs block by HMR-1556 prolonged AP duration at 90% repolarization slightly in the combined presence of E-4031 and isoprenaline. CONCLUSIONS EHT from hiPSC-CMs shows a lower repolarization reserve than human LV working myocardium and could thereby serve as a sensitive and specific human-based model for repolarization studies and arrhythmia, similar to Purkinje fibers. In both human LV and EHT, IKs only contributed to repolarization under adrenergic stimulation.
Collapse
Affiliation(s)
- Marc D Lemoine
- Department of Cardiology-Electrophysiology (M.D.L., S.W.) .,Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Tobias Krause
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Jussi T Koivumäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (J.T.K.).,BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Finland (J.T.K.)
| | - Maksymilian Prondzynski
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Mirja L Schulze
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Evaldas Girdauskas
- Department of Cardiovascular Surgery (E.G.), University Heart Center, Hamburg-Eppendorf, Germany
| | - Stephan Willems
- Department of Cardiology-Electrophysiology (M.D.L., S.W.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.).,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology, University Medical Center, Hamburg-Eppendorf, Germany (M.D.L., T.K., M.P., M.L.S., A.H., T.E., T.C.) .,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (M.D.L., T.K., M.P., M.L.S., S.W., A.H., T.E., T.C.)
| |
Collapse
|
38
|
Agata N, Kato Y, Hamaguchi S, Namekata I, Tanaka H. Resistance of Fetal Guinea Pig Ventricular Myocardium to Hypoxia: Maintained Intracellular ATP Prevents the Opening of ATP-Sensitive Potassium Channels. Biol Pharm Bull 2019; 42:268-272. [PMID: 30713257 DOI: 10.1248/bpb.b18-00762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The presence and function of the ATP-sensitive potassium channel current (IKATP) were examined in the guinea pig myocardium to clarify the mechanisms for the resistance of the fetal myocardium to hypoxia. Experimental hypoxia markedly reduced the action potential duration and contractile force in isolated ventricular myocardium from the adult, but only moderately in those from the fetus. In isolated ventricular cardiomyocytes, the density of the IKATP activated by cromakalim, as well as their sensitivity to intracellular ATP concentration, were not different between the fetus and adult. The tissue ATP content was similar between the fetal and adult myocardium under normal condition, but the hypoxia-induced decrease was smaller in the fetus. Confocal microscopic analysis revealed that the mitochondria in the fetal cardiomyocyte is less in quantity than that in the adult and is more localized to the cell center. These results indicate that IKATP in the fetal guinea pig myocardium has a current density and ATP sensitivity similar to those of the adult, but is not activated under hypoxic conditions because the energy metabolism of the fetal myocardium is less dependent on oxidative phosphorylation.
Collapse
Affiliation(s)
- Naoki Agata
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Yoshimitsu Kato
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Shogo Hamaguchi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Iyuki Namekata
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| |
Collapse
|
39
|
Liu C, Spinozzi S, Chen JY, Fang X, Feng W, Perkins G, Cattaneo P, Guimarães-Camboa N, Dalton ND, Peterson KL, Wu T, Ouyang K, Fu XD, Evans SM, Chen J. Nexilin Is a New Component of Junctional Membrane Complexes Required for Cardiac T-Tubule Formation. Circulation 2019; 140:55-66. [PMID: 30982350 PMCID: PMC6889818 DOI: 10.1161/circulationaha.119.039751] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/02/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Membrane contact sites are fundamental for transmission and translation of signals in multicellular organisms. The junctional membrane complexes in the cardiac dyads, where transverse (T) tubules are juxtaposed to the sarcoplasmic reticulum, are a prime example. T-tubule uncoupling and remodeling are well-known features of cardiac disease and heart failure. Even subtle alterations in the association between T-tubules and the junctional sarcoplasmic reticulum can cause serious cardiac disorders. NEXN (nexilin) has been identified as an actin-binding protein, and multiple mutations in the NEXN gene are associated with cardiac diseases, but the precise role of NEXN in heart function and disease is still unknown. METHODS Nexn global and cardiomyocyte-specific knockout mice were generated. Comprehensive phenotypic and RNA sequencing and mass spectrometry analyses were performed. Heart tissue samples and isolated single cardiomyocytes were analyzed by electron and confocal microscopy. RESULTS Global and cardiomyocyte-specific loss of Nexn in mice resulted in a rapidly progressive dilated cardiomyopathy. In vivo and in vitro analyses revealed that NEXN interacted with junctional sarcoplasmic reticulum proteins, was essential for optimal calcium transients, and was required for initiation of T-tubule invagination and formation. CONCLUSIONS These results demonstrated that NEXN is a pivotal component of the junctional membrane complex and is required for initiation and formation of T-tubules, thus providing insight into mechanisms underlying cardiomyopathy in patients with mutations in NEXN.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/metabolism
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cell Membrane/genetics
- Cell Membrane/metabolism
- Cell Membrane/pathology
- Cells, Cultured
- Intercellular Junctions/genetics
- Intercellular Junctions/metabolism
- Intercellular Junctions/pathology
- Mice
- Mice, Knockout
- Mice, Transgenic
- Microfilament Proteins/deficiency
- Microfilament Proteins/genetics
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
Collapse
Affiliation(s)
- Canzhao Liu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Simone Spinozzi
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jia-Yu Chen
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Wei Feng
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Paola Cattaneo
- National Research Council, Institute of Genetics and Biomedical Research, Milan 20138, Italy
- Humanitas Clinical and Research Center, Rozzano 20089, Italy
| | - Nuno Guimarães-Camboa
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Berlin 13347, Germany
| | - Nancy D. Dalton
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kirk L. Peterson
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tongbin Wu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kunfu Ouyang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Sylvia M. Evans
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
40
|
Škrabánek P, Zahradníková A. Automatic assessment of the cardiomyocyte development stages from confocal microscopy images using deep convolutional networks. PLoS One 2019; 14:e0216720. [PMID: 31145728 PMCID: PMC6542571 DOI: 10.1371/journal.pone.0216720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/26/2019] [Indexed: 11/23/2022] Open
Abstract
Computer assisted image acquisition techniques, including confocal microscopy, require efficient tools for an automatic sorting of vast amount of generated image data. The complexity of the classification process, absence of adequate tools, and insufficient amount of reference data has made the automated processing of images challenging. Mastering of this issue would allow implementation of statistical analysis in research areas such as in research on formation of t-tubules in cardiac myocytes. We developed a system aimed at automatic assessment of cardiomyocyte development stages (SAACS). The system classifies confocal images of cardiomyocytes with fluorescent dye stained sarcolemma. We based SAACS on a densely connected convolutional network (DenseNet) topology. We created a set of labelled source images, proposed an appropriate data augmentation technique and designed a class probability graph. We showed that the DenseNet topology, in combination with the augmentation technique is suitable for the given task, and that high-resolution images are instrumental for image categorization. SAACS, in combination with the automatic high-throughput confocal imaging, will allow application of statistical analysis in the research of the tubular system development or remodelling and loss.
Collapse
Affiliation(s)
- Pavel Škrabánek
- Institute of Automation and Computer Science, Brno University of Technology, Brno, Czech Republic
| | - Alexandra Zahradníková
- Institute of Molecular Physiology and Genetics, Centre of Biosciences SAS, Bratislava, Slovakia
- Department of Cellular Cardiology, Inst. of Experimental Endocrinology, Biomedical Research Center SAS, Bratislava, Slovakia
| |
Collapse
|
41
|
Lipsett DB, Frisk M, Aronsen JM, Nordén ES, Buonarati OR, Cataliotti A, Hell JW, Sjaastad I, Christensen G, Louch WE. Cardiomyocyte substructure reverts to an immature phenotype during heart failure. J Physiol 2019; 597:1833-1853. [PMID: 30707448 PMCID: PMC6441900 DOI: 10.1113/jp277273] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022] Open
Abstract
Key points As reactivation of the fetal gene program has been implicated in pathological remodelling during heart failure (HF), we examined whether cardiomyocyte subcellular structure and function revert to an immature phenotype during this disease. Surface and internal membrane structures appeared gradually during development, and returned to a juvenile state during HF. Similarly, dyadic junctions between the cell membrane and sarcoplasmic reticulum were progressively ‘packed’ with L‐type Ca2+ channels and ryanodine receptors during development, and ‘unpacked’ during HF. Despite similarities in subcellular structure, dyads were observed to be functional from early developmental stages, but exhibited an impaired ability to release Ca2+ in failing cardiomyocytes. Thus, while immature and failing cardiomyocytes share similarities in subcellular structure, these do not fully account for the marked impairment of Ca2+ homeostasis observed in HF.
Abstract Reactivation of the fetal gene programme has been implicated as a driver of pathological cardiac remodelling. Here we examined whether pathological remodelling of cardiomyocyte substructure and function during heart failure (HF) reflects a reversion to an immature phenotype. Using scanning electron microscopy, we observed that Z‐grooves and t‐tubule openings at the cell surface appeared gradually during cardiac development, and disappeared during HF. Confocal and super‐resolution imaging within the cell interior revealed similar structural parallels; disorganization of t‐tubules in failing cells was strikingly reminiscent of the late stages of postnatal development, with fewer transverse elements and a high proportion of longitudinal tubules. Ryanodine receptors (RyRs) were observed to be laid down in advance of developing t‐tubules and similarly ‘orphaned’ in HF, although RyR distribution along Z‐lines was relatively sparse. Indeed, nanoscale imaging revealed coordinated packing of L‐type Ca2+ channels and RyRs into dyadic junctions during development, and orderly unpacking during HF. These findings support a ‘last in, first out’ paradigm, as the latest stages of dyadic structural development are reversed during disease. Paired imaging of t‐tubules and Ca2+ showed that the disorganized arrangement of dyads in immature and failing cells promoted desynchronized and slowed Ca2+ release in these two states. However, while developing cells exhibited efficient triggering of Ca2+ release at newly formed dyads, dyadic function was impaired in failing cells despite similar organization of Ca2+ handling proteins. Thus, pathologically deficient Ca2+ homeostasis during HF is only partly linked to the re‐emergence of immature subcellular structure, and additionally reflects lost dyadic functionality. As reactivation of the fetal gene program has been implicated in pathological remodelling during heart failure (HF), we examined whether cardiomyocyte subcellular structure and function revert to an immature phenotype during this disease. Surface and internal membrane structures appeared gradually during development, and returned to a juvenile state during HF. Similarly, dyadic junctions between the cell membrane and sarcoplasmic reticulum were progressively ‘packed’ with L‐type Ca2+ channels and ryanodine receptors during development, and ‘unpacked’ during HF. Despite similarities in subcellular structure, dyads were observed to be functional from early developmental stages, but exhibited an impaired ability to release Ca2+ in failing cardiomyocytes. Thus, while immature and failing cardiomyocytes share similarities in subcellular structure, these do not fully account for the marked impairment of Ca2+ homeostasis observed in HF.
Collapse
Affiliation(s)
- D B Lipsett
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - M Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - J M Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Bjørknes College, Oslo, Norway
| | - E S Nordén
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - O R Buonarati
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - A Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - J W Hell
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - I Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - G Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - W E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
42
|
Karppinen S, Hänninen SL, Rapila R, Tavi P. Sarcoplasmic reticulum Ca 2+ -induced Ca 2+ release regulates class IIa HDAC localization in mouse embryonic cardiomyocytes. Physiol Rep 2019; 6. [PMID: 29380950 PMCID: PMC5789715 DOI: 10.14814/phy2.13522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/28/2017] [Accepted: 10/30/2017] [Indexed: 11/24/2022] Open
Abstract
In embryonic cardiomyocytes, sarcoplasmic reticulum (SR)‐derived Ca2+ release is required to induce Ca2+ oscillations for contraction and to control cardiac development through Ca2+‐activated pathways. Here, our aim was to study how SR Ca2+ release regulates cytosolic and nuclear Ca2+ distribution and the subsequent effects on the Ca2+‐dependent localization of class IIa histone deacetylases (HDAC) and cardiac‐specific gene expression in embryonic cardiomyocytes. Confocal microscopy was used to study changes in Ca2+‐distribution and localization of immunolabeled HDAC4 and HDAC5 upon changes in SR Ca2+ release in mouse embryonic cardiomyocytes. Dynamics of translocation were also observed with a confocal microscope, using HDAC5‐green fluorescent protein transfected myocytes. Expression of class IIa HDACs in differentiating myocytes and changes in cardiac‐specific gene expression were studied using real‐time quantitative PCR. Inhibition of SR Ca2+ release caused a significant decrease in intranuclear Ca2+ concentration, a rapid nuclear import of HDAC5 and subnuclear redistribution of HDAC4. Endogenous localization of HDAC5 and HDAC4 was mostly cytosolic and at the nuclear periphery, respectively. Downregulated expression of cardiac‐specific genes was also observed upon SR Ca2+ release inhibition. Electrical stimulation of sarcolemmal Ca2+ influx was not sufficient to rescue either the HDAC localization or the gene expression changes. SR Ca2+ release controls subcellular Ca2+ distribution and regulates localization of HDAC4 and HDAC5 in embryonic cardiomyocytes. Changes in SR Ca2+ release also caused changes in expression of the developmental phase‐specific genes, which may be due to the changes in HDAC‐localization.
Collapse
Affiliation(s)
- Sari Karppinen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sandra L Hänninen
- Institute of Biomedicine, Department of Physiology and Biocenter Oulu, University of Oulu, Finland
| | - Risto Rapila
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
43
|
Sheard TMD, Kharche SR, Pinali C, Shiels HA. 3D ultrastructural organisation of calcium release units in the avian sarcoplasmic reticulum. J Exp Biol 2019; 222:jeb.197640. [DOI: 10.1242/jeb.197640] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/22/2019] [Indexed: 11/20/2022]
Abstract
Excitation-contraction coupling in vertebrate hearts is underpinned by calcium (Ca2+) release from Ca2+ release units (CRUs). CRUs are formed by clusters of channels called ryanodine receptors on the sarcoplasmic reticulum (SR) within the cardiomyocyte. Distances between CRUs influence the diffusion of Ca2+, thus influencing the rate and strength of excitation-contraction coupling. Avian myocytes lack T-tubules, thus Ca2+ from surface CRUs (peripheral couplings, PCs), must diffuse to internal CRU sites of the corbular SR (cSR) during centripetal propagation. Despite this, avian hearts achieve higher contractile rates and develop greater contractile strength than many mammalian hearts, which have T-tubules to provide simultaneous activation of the Ca2+ signal through the myocyte. We used 3D electron tomography to test the hypothesis that the intracellular distribution of CRUs in the avian heart permits faster and stronger contractions despite the absence T-tubules. Nearest edge-edge distances between PCs and cSR, and geometric information including surface area and volumes of individual cSR, were obtained for each cardiac chamber of the White Leghorn chicken. Computational modelling was then used to establish a relationship between CRUs distances and cell activation time in the avian heart. Our data suggest that cSR clustered close together along the Z-line is vital for rapid propagation of the Ca2+ signal from the cell periphery to the cell centre which would aid in the strong and fast contractions of the avian heart.
Collapse
Affiliation(s)
- Thomas M. D. Sheard
- University of Manchester, Faculty of Biology, Medicine and Health, Oxford Road, Manchester, M13 9PL, UK
| | - Sanjay R. Kharche
- University of Manchester, Faculty of Biology, Medicine and Health, Oxford Road, Manchester, M13 9PL, UK
- Department of Medical Biophysics, University of Western Ontario, London, N6A 3K7, Canada
- Lawson Health Research Institute, 800 Commissioners Road East, London, Ontario, N6C 2R5, Canada
| | - Christian Pinali
- University of Manchester, Faculty of Biology, Medicine and Health, Oxford Road, Manchester, M13 9PL, UK
| | - Holly A. Shiels
- University of Manchester, Faculty of Biology, Medicine and Health, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
44
|
Lv J, Deng C, Jiang S, Ji T, Yang Z, Wang Z, Yang Y. Blossoming 20: The Energetic Regulator's Birthday Unveils its Versatility in Cardiac Diseases. Am J Cancer Res 2019; 9:466-476. [PMID: 30809287 PMCID: PMC6376194 DOI: 10.7150/thno.29130] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022] Open
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) was first identified in 1998 as a PGC-1 family member that regulates adaptive thermogenesis and mitochondrial function following cold exposure in brown adipose tissue. The PGC-1 family has drawn widespread attention over the past two decades as the energetic regulator. We recently summarized a review regarding PGC-1 signaling pathway and its mechanisms in cardiac metabolism. In this review, we elaborate upon the PGC-1 signaling network and highlight the recent progress of its versatile roles in cardiac diseases, including myocardial hypertrophy, peripartum and diabetic cardiomyopathy, and heart failure. The information reviewed here may be useful in future studies, which may increase the potential of this energetic regulator as a therapeutic target.
Collapse
|
45
|
Jones PP, MacQuaide N, Louch WE. Dyadic Plasticity in Cardiomyocytes. Front Physiol 2018; 9:1773. [PMID: 30618792 PMCID: PMC6298195 DOI: 10.3389/fphys.2018.01773] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/23/2018] [Indexed: 11/13/2022] Open
Abstract
Contraction of cardiomyocytes is dependent on sub-cellular structures called dyads, where invaginations of the surface membrane (t-tubules) form functional junctions with the sarcoplasmic reticulum (SR). Within each dyad, Ca2+ entry through t-tubular L-type Ca2+ channels (LTCCs) elicits Ca2+ release from closely apposed Ryanodine Receptors (RyRs) in the SR membrane. The efficiency of this process is dependent on the density and macroscale arrangement of dyads, but also on the nanoscale organization of LTCCs and RyRs within them. We presently review accumulating data demonstrating the remarkable plasticity of these structures. Dyads are known to form gradually during development, with progressive assembly of both t-tubules and junctional SR terminals, and precise trafficking of LTCCs and RyRs. While dyads can exhibit compensatory remodeling when required, dyadic degradation is believed to promote impaired contractility and arrythmogenesis in cardiac disease. Recent data indicate that this plasticity of dyadic structure/function is dependent on the regulatory proteins junctophilin-2, amphiphysin-2 (BIN1), and caveolin-3, which critically arrange dyadic membranes while stabilizing the position and activity of LTCCs and RyRs. Indeed, emerging evidence indicates that clustering of both channels enables "coupled gating", implying that nanoscale localization and function are intimately linked, and may allow fine-tuning of LTCC-RyR crosstalk. We anticipate that improved understanding of dyadic plasticity will provide greater insight into the processes of cardiac compensation and decompensation, and new opportunities to target the basic mechanisms underlying heart disease.
Collapse
Affiliation(s)
- Peter P. Jones
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Niall MacQuaide
- Institute of Cardiovascular Sciences, University of Glasgow, Glasgow, United Kingdom
- Clyde Biosciences, Glasgow, United Kingdom
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
46
|
Mhatre KN, Wakula P, Klein O, Bisping E, Völkl J, Pieske B, Heinzel FR. Crosstalk between FGF23- and angiotensin II-mediated Ca 2+ signaling in pathological cardiac hypertrophy. Cell Mol Life Sci 2018; 75:4403-4416. [PMID: 30062428 PMCID: PMC11105615 DOI: 10.1007/s00018-018-2885-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 07/16/2018] [Accepted: 07/19/2018] [Indexed: 02/07/2023]
Abstract
Heart failure (HF) manifestation and progression are driven by systemic activation of neuroendocrine signaling cascades, such as the renin-angiotensin aldosterone system (RAAS). Fibroblast growth factor 23 (FGF23), an endocrine hormone, is linked to HF and cardiovascular mortality. It is also a mediator of left-ventricular hypertrophy (LVH). In vivo, high circulating levels of FGF23 are associated with an altered systemic RAAS response. FGF23 is proposed to trigger pathological signaling mediated by Ca2+-regulated transcriptional pathways. In the present study, we investigated Ca2+-dependent signaling of FGF23 in ventricular cardiomyocytes and its association with angiotensin II (ATII). In neonatal rat ventricular myocytes (NRVMs), both ATII and FGF23 induced hypertrophy as observed by an increase in cell area and hypertrophic gene expression. Furthermore, FGF23 activates nuclear Ca2+-regulated CaMKII-HDAC4 pathway, similar to ATII. In addition to a global increase in cytoplasmic Ca2+, FGF23, like ATII, induced inositol 1, 4, 5-triphosphate (IP3)-induced Ca2+ release from the nucleoplasmic Ca2+ store, associated with cellular hypertrophy. Interestingly, ATII receptor antagonist, losartan, significantly attenuated FGF23-induced changes in Ca2+ homeostasis and cellular hypertrophy suggesting an involvement of ATII receptor-mediated signaling. In addition, application of FGF23 increased intracellular expression of ATII peptide and its secretion in NRVMs, confirming the participation of ATII. In conclusion, FGF23 and ATII share a common mechanism of IP3-nuclear Ca2+-dependent cardiomyocyte hypertrophy. FGF23-mediated cellular hypertrophy is associated with increased production and secretion of ATII by cardiomyocytes. These findings indicate a pathophysiological role of the cellular angiotensin system in FGF23-induced hypertrophy in ventricular cardiomyocytes.
Collapse
Affiliation(s)
- Ketaki N Mhatre
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, 13353, Berlin, Germany
- Department of Cardiology, Medical University Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Paulina Wakula
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, 13353, Berlin, Germany
| | - Oliver Klein
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, 13353, Berlin, Germany
| | - Egbert Bisping
- Department of Cardiology, Medical University Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Jakob Völkl
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, 13353, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, 13353, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Internal Medicine and Cardiology, German Heart Center, 13353, Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, 13353, Berlin, Germany.
- German Center for Cardiovascular Research (DZHK), Berlin, Germany.
| |
Collapse
|
47
|
Jiang Y, Zhou Y, Bao X, Chen C, Randolph LN, Du J, Lian XL. An Ultrasensitive Calcium Reporter System via CRISPR-Cas9-Mediated Genome Editing in Human Pluripotent Stem Cells. iScience 2018; 9:27-35. [PMID: 30368079 PMCID: PMC6203247 DOI: 10.1016/j.isci.2018.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/26/2018] [Accepted: 10/08/2018] [Indexed: 12/21/2022] Open
Abstract
Genetically encoded calcium indicator (GCaMP) proteins have been reported for imaging cardiac cell activity based on intracellular calcium transients. To bring human pluripotent stem cell (hPSC)-derived cardiomyocytes (CMs) to the clinic, it is critical to evaluate the functionality of CMs. Here, we show that GCaMP6s-expressing hPSCs can be generated and used for CM characterization. By leveraging CRISPR-Cas9 genome editing tools, we generated a knockin cell line that constitutively expresses GCaMP6s, an ultrasensitive calcium sensor protein. We further showed that this clone maintained pluripotency and cardiac differentiation potential. These knockin hPSC-derived CMs exhibited sensitive fluorescence fluctuation with spontaneous contraction. We then compared the fluorescence signal with mechanical contraction signal. The knockin hPSC-derived CMs also showed sensitive response to isoprenaline treatment in a concentration-dependent manner. Therefore, the GCaMP6s knockin hPSC line provides a non-invasive, sensitive, and economic approach to characterize the functionality of hPSC-derived CMs.
Collapse
Affiliation(s)
- Yuqian Jiang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yuxiao Zhou
- Department of Mechanical and Nuclear Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Xiaoping Bao
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Chuanxin Chen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Lauren N Randolph
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jing Du
- Department of Mechanical and Nuclear Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Xiaojun Lance Lian
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
48
|
Taha MF, Javeri A, Karimipour M, Yamaghani MS. Priming with oxytocin and relaxin improves cardiac differentiation of adipose tissue-derived stem cells. J Cell Biochem 2018; 120:5825-5834. [PMID: 30362159 DOI: 10.1002/jcb.27868] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 09/20/2018] [Indexed: 11/07/2022]
Abstract
Previous studies have identified the heart as a source and a target tissue for oxytocin and relaxin hormones. These hormones play important roles in the regulation of cardiovascular function and repair of ischemic heart injury. In the current study, we examined the impact of oxytocin and relaxin on the development of cardiomyocytes from mesenchymal stem cells. For this purpose, mouse adipose tissue-derived stem cells (ADSCs) were treated with different concentrations of oxytocin or relaxin for 4 days. Three weeks after initiation of cardiac induction, differentiated ADSCs expressed cardiac-specific genes, Gata4, Mef2c, Nkx2.5, Tbx5, α- and β-Mhc, Mlc2v, Mlc2a and Anp, and cardiac proteins including connexin 43, desmin and α-actinin. 10 -7 M oxytocin and 50 ng/mL relaxin induced the maximum upregulation in the expression of cardiac markers. A combination of oxytocin and relaxin induced cardiomyocyte differentiation more potently than the individual factors. In our experiment, oxytocin-relaxin combination increased the population of cardiac troponin I-expressing cells to 6.84% as compared with 2.36% for the untreated ADSCs, 3.7% for oxytocin treatment and 3.41% for relaxin treatment groups. In summary, the results of this study indicated that oxytocin and relaxin hormones individually and in combination can improve cardiac differentiation of ADSCs, and treatment of the ADSCs and possibly other mesenchymal stem cells with these hormones may enhance their cardiogenic differentiation and survival after transplantation into the ischemic heart tissue.
Collapse
Affiliation(s)
- Masoumeh Fakhr Taha
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Arash Javeri
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Mojtaba Karimipour
- Department of Anatomy, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | |
Collapse
|
49
|
Iguchi T, Fujimoto K, Nakamura S, Kishino H, Niino N, Mori K. Establishment of an in vitro cytotoxicity assay platform using primary monkey cardiomyocytes. Toxicol In Vitro 2018; 54:130-136. [PMID: 30261314 DOI: 10.1016/j.tiv.2018.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 11/18/2022]
Abstract
To establish an in vitro cytotoxicity assay platform using monkey cardiomyocytes, we isolated primary cardiomyocytes from fetal cynomolgus monkeys at different gestation days (from day 39 to 90) using the trypsin and collagenase digestion method, which was identical to the standard procedure for rat cardiomyocytes. Under these conditions, the primary cells obtained from monkeys at gestation day 63 or earlier showed spontaneous beating, with >80% cells being viable from all fetuses. Transcriptome analysis of the monkey cardiomyocytes indicated that the cells have essential components of cardiac functions, such as myosins, α-actin, cardiac troponins, and calcium-related molecules. The susceptibility to doxorubicin-induced cytotoxicity in monkey cardiomyocytes was comparable to that in rat cardiomyocytes, as evaluated based on intracellular ATP levels. Microarray analysis with Ingenuity Pathway Analysis revealed that doxorubicin predominantly increased the expression of several key genes involved in the endoplasmic reticulum stress pathway in monkey cardiomyocytes than in rat cardiomyocytes. In conclusion, we isolated primary monkey cardiomyocytes that showed similar sensitivity to doxorubicin as compared with rat cardiomyocytes. This in vitro monkey cardiomyocyte assay platform would serve as a powerful tool for the investigation of the interspecies differences in drug-induced cardiotoxicity and its underlying mechanism.
Collapse
Affiliation(s)
- Takuma Iguchi
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Kazunori Fujimoto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Shinichiro Nakamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga 520-2192, Japan.
| | - Hiroyuki Kishino
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Noriyo Niino
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Kazuhiko Mori
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan.
| |
Collapse
|
50
|
Ramadan M, Sherman M, Jaimes R, Chaluvadi A, Swift L, Posnack NG. Disruption of neonatal cardiomyocyte physiology following exposure to bisphenol-a. Sci Rep 2018; 8:7356. [PMID: 29743542 PMCID: PMC5943481 DOI: 10.1038/s41598-018-25719-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/27/2018] [Indexed: 12/21/2022] Open
Abstract
Bisphenol chemicals are commonly used in the manufacturing of polycarbonate plastics, polyvinyl chloride plastics, resins, and thermal printing applications. Humans are inadvertently exposed to bisphenols through contact with consumer products and/or medical devices. Recent reports have shown a link between bisphenol-a (BPA) exposure and adverse cardiovascular outcomes; although these studies have been limited to adult subjects and models. Since cardiac physiology differs significantly between the developing and adult heart, we aimed to assess the impact of BPA exposure on cardiac function, using a neonatal cardiomyocyte model. Neonatal rat ventricular myocytes were monitored to assess cell viability, spontaneous beating rate, beat rate variability, and calcium-handling parameters in the presence of control or bisphenol-supplemented media. A range of doses were tested to mimic environmental exposure (10-9-10-8M), maximum clinical exposure (10-5M), and supraphysiological exposure levels (10-4M). Acute BPA exposure altered cardiomyocyte functionality, resulting in a slowed spontaneous beating rate and increased beat rate variability. BPA exposure also impaired intracellular calcium handling, resulting in diminished calcium transient amplitudes, prolonged calcium transient upstroke and duration time. Alterations in calcium handling also increased the propensity for alternans and skipped beats. Notably, the effect of BPA-treatment on calcium handling was partially reversible. Our data suggest that acute BPA exposure could precipitate secondary adverse effects on contractile performance and/or electrical alternans, both of which are dependent on intracellular calcium homeostasis.
Collapse
Affiliation(s)
- Manelle Ramadan
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children's National Health System, Washington, USA
- Children's National Heart Institute, Children's National Health System, Washington, USA
| | - Meredith Sherman
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children's National Health System, Washington, USA
| | - Rafael Jaimes
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children's National Health System, Washington, USA
- Children's National Heart Institute, Children's National Health System, Washington, USA
| | - Ashika Chaluvadi
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children's National Health System, Washington, USA
| | - Luther Swift
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children's National Health System, Washington, USA
- Children's National Heart Institute, Children's National Health System, Washington, USA
| | - Nikki Gillum Posnack
- Sheikh Zayed Institute for Pediatric and Surgical Innovation, Children's National Health System, Washington, USA.
- Children's National Heart Institute, Children's National Health System, Washington, USA.
- Department of Pediatrics, Department of Pharmacology & Physiology, School of Medicine and Health Sciences, George Washington University, Washington, USA.
| |
Collapse
|