25
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      CRISPR/Cas9 editing in human pluripotent stem cell-cardiomyocytes highlights arrhythmias, hypocontractility, and energy depletion as potential therapeutic targets for hypertrophic cardiomyopathy

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Aims

          Sarcomeric gene mutations frequently underlie hypertrophic cardiomyopathy (HCM), a prevalent and complex condition leading to left ventricle thickening and heart dysfunction. We evaluated isogenic genome-edited human pluripotent stem cell-cardiomyocytes (hPSC-CM) for their validity to model, and add clarity to, HCM.

          Methods and results

          CRISPR/Cas9 editing produced 11 variants of the HCM-causing mutation c.C9123T-MYH7 [(p.R453C-β-myosin heavy chain (MHC)] in 3 independent hPSC lines. Isogenic sets were differentiated to hPSC-CMs for high-throughput, non-subjective molecular and functional assessment using 12 approaches in 2D monolayers and/or 3D engineered heart tissues. Although immature, edited hPSC-CMs exhibited the main hallmarks of HCM (hypertrophy, multi-nucleation, hypertrophic marker expression, sarcomeric disarray). Functional evaluation supported the energy depletion model due to higher metabolic respiration activity, accompanied by abnormalities in calcium handling, arrhythmias, and contraction force. Partial phenotypic rescue was achieved with ranolazine but not omecamtiv mecarbil, while RNAseq highlighted potentially novel molecular targets.

          Conclusion

          Our holistic and comprehensive approach showed that energy depletion affected core cardiomyocyte functionality. The engineered R453C-βMHC-mutation triggered compensatory responses in hPSC-CMs, causing increased ATP production and αMHC to energy-efficient βMHC switching. We showed that pharmacological rescue of arrhythmias was possible, while MHY7: MYH6 and mutant: wild-type MYH7 ratios may be diagnostic, and previously undescribed lncRNAs and gene modifiers are suggestive of new mechanisms.

          Related collections

          Most cited references64

          • Record: found
          • Abstract: found
          • Article: not found

          Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy.

          Characterized by ventricular dilatation, systolic dysfunction, and progressive heart failure, dilated cardiomyopathy (DCM) is the most common form of cardiomyopathy in patients. DCM is the most common diagnosis leading to heart transplantation and places a significant burden on healthcare worldwide. The advent of induced pluripotent stem cells (iPSCs) offers an exceptional opportunity for creating disease-specific cellular models, investigating underlying mechanisms, and optimizing therapy. Here, we generated cardiomyocytes from iPSCs derived from patients in a DCM family carrying a point mutation (R173W) in the gene encoding sarcomeric protein cardiac troponin T. Compared to control healthy individuals in the same family cohort, cardiomyocytes derived from iPSCs from DCM patients exhibited altered regulation of calcium ion (Ca(2+)), decreased contractility, and abnormal distribution of sarcomeric α-actinin. When stimulated with a β-adrenergic agonist, DCM iPSC-derived cardiomyocytes showed characteristics of cellular stress such as reduced beating rates, compromised contraction, and a greater number of cells with abnormal sarcomeric α-actinin distribution. Treatment with β-adrenergic blockers or overexpression of sarcoplasmic reticulum Ca(2+) adenosine triphosphatase (Serca2a) improved the function of iPSC-derived cardiomyocytes from DCM patients. Thus, iPSC-derived cardiomyocytes from DCM patients recapitulate to some extent the morphological and functional phenotypes of DCM and may serve as a useful platform for exploring disease mechanisms and for drug screening.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Cellular mechanisms of cardiomyopathy

            The heart exhibits remarkable adaptive responses to a wide array of genetic and extrinsic factors to maintain contractile function. When compensatory responses are not sustainable, cardiac dysfunction occurs, leading to cardiomyopathy. The many forms of cardiomyopathy exhibit a set of overlapping phenotypes reflecting the limited range of compensatory responses that the heart can use. These include cardiac hypertrophy, induction of genes normally expressed during development, fibrotic deposits that replace necrotic and apoptotic cardiomyocytes, and metabolic disturbances. The compensatory responses are mediated by signaling pathways that initially serve to maintain normal contractility; however, persistent activation of these pathways leads to cardiac dysfunction. Current research focuses on ways to target these specific pathways therapeutically.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Cardiomyocytes from human pluripotent stem cells: From laboratory curiosity to industrial biomedical platform☆

              Cardiomyocytes from human pluripotent stem cells (hPSCs-CMs) could revolutionise biomedicine. Global burden of heart failure will soon reach USD $90bn, while unexpected cardiotoxicity underlies 28% of drug withdrawals. Advances in hPSC isolation, Cas9/CRISPR genome engineering and hPSC-CM differentiation have improved patient care, progressed drugs to clinic and opened a new era in safety pharmacology. Nevertheless, predictive cardiotoxicity using hPSC-CMs contrasts from failure to almost total success. Since this likely relates to cell immaturity, efforts are underway to use biochemical and biophysical cues to improve many of the ~ 30 structural and functional properties of hPSC-CMs towards those seen in adult CMs. Other developments needed for widespread hPSC-CM utility include subtype specification, cost reduction of large scale differentiation and elimination of the phenotyping bottleneck. This review will consider these factors in the evolution of hPSC-CM technologies, as well as their integration into high content industrial platforms that assess structure, mitochondrial function, electrophysiology, calcium transients and contractility. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
                Bookmark

                Author and article information

                Journal
                Eur Heart J
                Eur. Heart J
                eurheartj
                European Heart Journal
                Oxford University Press
                0195-668X
                1522-9645
                14 November 2018
                08 May 2018
                08 May 2018
                : 39
                : 43 , Focus Issue on Heart Failure
                : 3879-3892
                Affiliations
                [1 ]Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, UK
                [2 ]Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
                [3 ]Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), Hamburg, Germany
                [4 ]The Heart Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London, UK
                [5 ]Molecular Therapeutics and Formulation. School of Pharmacy, University of Nottingham, UK
                [6 ]Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
                Author notes
                Corresponding authors. Tel: +44(0)115 8231236, Fax: +44(0)115 8231230, Email: diogo.mosqueira@ 123456nottingham.ac.uk ; chris.denning@ 123456nottingham.ac.uk
                Article
                ehy249
                10.1093/eurheartj/ehy249
                6234851
                29741611
                984b9f1c-47e3-4b86-94f3-08e30cfb6c55
                © The Author(s) 2018. Published by Oxford University Press on behalf of the European Society of Cardiology.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 12 January 2018
                : 22 February 2018
                : 11 April 2018
                Page count
                Pages: 16
                Funding
                Funded by: British Heart Foundation 10.13039/501100000274
                Award ID: SP/15/9/31605
                Award ID: RG/15/6/31436
                Award ID: PG/14/59/31000
                Award ID: RG/14/1/30588
                Award ID: RM/13/30157
                Award ID: P47352/CRM
                Funded by: Britain Israel Research and Academic Exchange Partnership
                Award ID: 04BX14CDLG
                Funded by: Medical Research Council 10.13039/501100000265
                Award ID: MR/M017354/1
                Funded by: MICA
                Funded by: Development of Metrics and Quality Standards for Scale up of Human Pluripotent Stem Cells
                Funded by: National Centre for the Replacement, Refinement & Reduction of Animals in Research
                Award ID: CRACK-IT:35911-259146
                Award ID: NC/K000225/1
                Funded by: German Research Foundation 10.13039/501100001659
                Award ID: DFG-Es-88/12-1
                Award ID: HA3423/5-1
                Funded by: European Research Council 10.13039/100010663
                Funded by: European Commission 10.13039/501100000780
                Funded by: German Centre for Cardiovascular Research 10.13039/100010447
                Funded by: DZHK 10.13039/100010447
                Funded by: German Ministry of Education and Research
                Funded by: Freie und Hansestadt Hamburg
                Categories
                Basic Science
                Heart Failure/Cardiomyopathy

                Cardiovascular Medicine
                hypertrophic cardiomyopathy,disease modeling ,crispr/cas9 ,genome-edited human pluripotent stem cell-cardiomyocytes ,r453c-βmhc

                Comments

                Comment on this article

                scite_

                Similar content195

                Cited by91

                Most referenced authors1,190