Electro

Blog

HomeHome / Blog / Electro

Aug 18, 2023

Electro

Nature Biomedical Engineering (2023)Cite this article 1497 Accesses 111 Altmetric Metrics details The study of cardiac physiology is hindered by physiological differences between humans and

Nature Biomedical Engineering (2023)Cite this article

1497 Accesses

111 Altmetric

Metrics details

The study of cardiac physiology is hindered by physiological differences between humans and small-animal models. Here we report the generation of multi-chambered self-paced vascularized human cardiac organoids formed under anisotropic stress and their applicability to the study of cardiac arrhythmia. Sensors embedded in the cardiac organoids enabled the simultaneous measurement of oxygen uptake, extracellular field potentials and cardiac contraction at resolutions higher than 10 Hz. This microphysiological system revealed 1 Hz cardiac respiratory cycles that are coupled to the electrical rather than the mechanical activity of cardiomyocytes. This electro-mitochondrial coupling was driven by mitochondrial calcium oscillations driving respiration cycles. Pharmaceutical or genetic inhibition of this coupling results in arrhythmogenic behaviour. We show that the chemotherapeutic mitoxantrone induces arrhythmia through disruption of this pathway, a process that can be partially reversed by the co-administration of metformin. Our microphysiological cardiac systems may further facilitate the study of the mitochondrial dynamics of cardiac rhythms and advance our understanding of human cardiac physiology.

This is a preview of subscription content, access via your institution

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

$29.99 / 30 days

cancel any time

Subscribe to this journal

Receive 12 digital issues and online access to articles

$79.00 per year

only $6.58 per issue

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Sequencing data are available at NCBI GEO, under the accession number GSE234907. Data for the figures are provided with this paper. All data supporting the results of this study are available within the paper and its Supplementary Information. Source data are provided with this paper.

The custom analysis software is available at https://github.com/mohammadghosheh95/Heart-on-a-Chip.

Benjamin, E. J. et al. Heart disease and stroke statistics–2017 update: a report from the American Heart Association. Circulation 135, e146–e603 (2017).

Article PubMed PubMed Central Google Scholar

Benjamin, E. J. et al. Heart disease and stroke statistics–2019 update: a report from the American Heart Association. Circulation 139, e56–e528 (2019).

Article PubMed Google Scholar

Fryar, C. D., Chen, T. C. & Li, X. Prevalence of uncontrolled risk factors for cardiovascular disease: United States, 1999–2010. NCHS Data Brief 103, 1–8 (2012).

Huch, M., Knoblich, J. A., Lutolf, M. P. & Martinez-Arias, A. The hope and the hype of organoid research. Development 144, 938–941 (2017).

Article CAS PubMed Google Scholar

Bertero, E. & Maack, C. Metabolic remodelling in heart failure. Nat. Rev. Cardiol. 15, 457–470 (2018).

Article CAS PubMed Google Scholar

Rosano, G. M. & Vitale, C. Metabolic modulation of cardiac metabolism in heart failure. Card. Fail. Rev. 4, 99–103 (2018).

Article PubMed PubMed Central Google Scholar

Doehner, W., Frenneaux, M. & Anker, S. D. Metabolic impairment in heart failure: the myocardial and systemic perspective. J. Am. Coll. Cardiol. 64, 1388–1400 (2014).

Article PubMed Google Scholar

von Haehling, S., Doehner, W. & Anker, S. D. Nutrition, metabolism, and the complex pathophysiology of cachexia in chronic heart failure. Cardiovasc. Res. 73, 298–309 (2007).

Article Google Scholar

Bonnet, D. et al. Arrhythmias and conduction defects as presenting symptoms of fatty acid oxidation disorders in children. Circulation 100, 2248–2253 (1999).

Article CAS PubMed Google Scholar

Gheorghiade, M., Colucci, W. S. & Swedberg, K. Beta-blockers in chronic heart failure. Circulation 107, 1570–1575 (2003).

Article PubMed Google Scholar

Hamer, A. W., Arkles, L. B. & Johns, J. A. Beneficial effects of low dose amiodarone in patients with congestive cardiac failure: a placebo-controlled trial. J. Am. Coll. Cardiol. 14, 1768–1774 (1989).

Article CAS PubMed Google Scholar

Kloner, R. A., Shi, J. & Dai, W. New therapies for reducing post-myocardial left ventricular remodeling. Ann. Transl. Med. 3, 20 (2015).

PubMed PubMed Central Google Scholar

Brown, D. A. et al. Mitochondrial function as a therapeutic target in heart failure. Nat. Rev. Cardiol. 14, 238–250 (2017).

Article CAS PubMed Google Scholar

Aon, M. A., Cortassa, S., Marbán, E. & O’Rourke, B. Synchronized whole cell oscillations in mitochondrial metabolism triggered by a local release of reactive oxygen species in cardiac myocytes. J. Biol. Chem. 278, 44735–44744 (2003).

Article CAS PubMed Google Scholar

Romashko, D. N., Marban, E. & O’Rourke, B. Subcellular metabolic transients and mitochondrial redox waves in heart cells. Proc. Natl Acad. Sci. USA 95, 1618–1623 (1998).

Article CAS PubMed PubMed Central Google Scholar

Bers, D. M. Cardiac excitation–contraction coupling. Nature 415, 198–205 (2002).

Article CAS PubMed Google Scholar

Gottlieb, R. A. & Bernstein, D. Mitochondria shape cardiac metabolism. Science 350, 1162–1163 (2015).

Article CAS PubMed Google Scholar

Wang, S. C. et al. Pathological basis of cardiac arrhythmias: vicious cycle of immune-metabolic dysregulation. Cardiovasc. Disord. Med. https://doi.org/10.15761/CDM.1000158 (2018).

Edwards, A. G. & Louch, W. E. Species-dependent mechanisms of cardiac arrhythmia: a cellular focus. Clin. Med. Insights Cardiol. 11, 1179546816686061 (2017).

Article PubMed PubMed Central Google Scholar

Milani-Nejad, N. & Janssen, P. M. L. Small and large animal models in cardiac contraction research: advantages and disadvantages. Pharmacol. Ther. 141, 235–249 (2014).

Article CAS PubMed Google Scholar

Ebert, A. et al. Proteasome-dependent regulation of distinct metabolic states during long-term culture of human iPSC-derived cardiomyocytes. Circ. Res. 125, 90–103 (2019).

Article CAS PubMed PubMed Central Google Scholar

Ronaldson-Bouchard, K. et al. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556, 239–243 (2018).

Article CAS PubMed PubMed Central Google Scholar

Giacomelli, E. et al. Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells. Development 144, 1008–1017 (2017).

CAS PubMed PubMed Central Google Scholar

Noor, N. et al. 3D printing of personalized thick and perfusable cardiac patches and hearts. Adv. Sci. 6, 1900344 (2019).

Article Google Scholar

Skylar-Scott, M. A. et al. Biomanufacturing of organ-specific tissues with high cellular density and embedded vascular channels. Sci. Adv. 5, eaaw2459 (2019).

Article CAS PubMed PubMed Central Google Scholar

Mills, R. J. et al. Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest. Proc. Natl Acad. Sci. USA 114, E8372–e8381 (2017).

Article CAS PubMed PubMed Central Google Scholar

Mills, R. J. et al. Drug screening in human PSC-cardiac organoids identifies pro-proliferative compounds acting via the mevalonate pathway. Cell Stem Cell 24, 895–907.e6 (2019).

Article CAS PubMed Google Scholar

Richards, D. J. et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat. Biomed. Eng. 4, 446–462 (2020).

Article CAS PubMed PubMed Central Google Scholar

Li, R. A. et al. Bioengineering an electro-mechanically functional miniature ventricular heart chamber from human pluripotent stem cells. Biomaterials 163, 116–127 (2018).

Article PubMed PubMed Central Google Scholar

Goldfracht, I. et al. Generating ring-shaped engineered heart tissues from ventricular and atrial human pluripotent stem cell-derived cardiomyocytes. Nat. Commun. 11, 75 (2020).

Article CAS PubMed PubMed Central Google Scholar

Zhao, Y. et al. A platform for generation of chamber-specific cardiac tissues and disease modeling. Cell 176, 913–927.e8 (2019).

Article CAS PubMed PubMed Central Google Scholar

Lind, J. U. et al. Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing. Nat. Mater. 16, 303–308 (2017).

Article CAS PubMed Google Scholar

Lind, J. U. et al. Cardiac microphysiological devices with flexible thin-film sensors for higher-throughput drug screening. Lab Chip 17, 3692–3703 (2017).

Article CAS PubMed PubMed Central Google Scholar

Oleaga, C. et al. Long-term electrical and mechanical function monitoring of a human-on-a-chip system. Adv. Funct. Mater. 29, 1805792 (2019).

Article PubMed Google Scholar

Maoz, B. M. et al. Organs-on-Chips with combined multi-electrode array and transepithelial electrical resistance measurement capabilities. Lab Chip 17, 2294–2302 (2017).

Article CAS PubMed Google Scholar

Tian, B. et al. Macroporous nanowire nanoelectronic scaffolds for synthetic tissues. Nat. Mater. 11, 986–994 (2012).

Article CAS PubMed PubMed Central Google Scholar

Feiner, R. et al. Engineered hybrid cardiac patches with multifunctional electronics for online monitoring and regulation of tissue function. Nat. Mater. 15, 679–685 (2016).

Article CAS PubMed PubMed Central Google Scholar

Drakhlis, L. et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat. Biotechnol. 39, 737–746 (2021).

Article CAS PubMed PubMed Central Google Scholar

Miquerol, L. & Kelly, R. G. Organogenesis of the vertebrate heart. Wiley Interdiscip. Rev. Dev. Biol. 2, 17–29 (2013).

Article CAS PubMed Google Scholar

Moorman, A., Webb, S., Brown, N. A., Lamers, W. & Anderson, R. H. Development of the heart: (1) formation of the cardiac chambers and arterial trunks. Heart 89, 806–814 (2003).

Article PubMed PubMed Central Google Scholar

Lin, C. J., Lin, C. Y., Chen, C. H., Zhou, B. & Chang, C. P. Partitioning the heart: mechanisms of cardiac septation and valve development. Development 139, 3277–3299 (2012).

Article CAS PubMed PubMed Central Google Scholar

Swift, J. et al. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341, 1240104 (2013).

Article PubMed PubMed Central Google Scholar

Dupont, S. et al. Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011).

Article CAS PubMed Google Scholar

Gordan, R., Gwathmey, J. K. & Xie, L.-H. Autonomic and endocrine control of cardiovascular function. World J. Cardiol. 7, 204–214 (2015).

Article PubMed PubMed Central Google Scholar

Mackin, C. et al. Intravenous amiodarone and sotalol impair contractility and cardiac output, but procainamide does not: a Langendorff study. J. Cardiovasc. Pharmacol. Ther. 24, 288–297 (2019).

Article CAS PubMed Google Scholar

Karbassi, E. et al. Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat. Rev. Cardiol. 17, 341–359 (2020).

Article PubMed PubMed Central Google Scholar

Wegner, A., Meiser, J., Weindl, D. & Hiller, K. How metabolites modulate metabolic flux. Curr. Opin. Biotechnol. 34, 16–22 (2015).

Article CAS PubMed Google Scholar

Collins-Nakai, R. L., Noseworthy, D. & Lopaschuk, G. D. Epinephrine increases ATP production in hearts by preferentially increasing glucose metabolism. Am. J. Physiol. 267, H1862–H1871 (1994).

CAS PubMed Google Scholar

Itzhaki, I. et al. Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. J. Am. Coll. Cardiol. 60, 990–1000 (2012).

Article CAS PubMed Google Scholar

Lee, C. H. & Ruben, P. C. Interaction between voltage-gated sodium channels and the neurotoxin, tetrodotoxin. Channels 2, 407–412 (2008).

Article PubMed Google Scholar

Farman, G. P. et al. Blebbistatin: use as inhibitor of muscle contraction. Pflugers Arch. 455, 995–1005 (2008).

Article CAS PubMed Google Scholar

Taxin, Z. H., Neymotin, S. A., Mohan, A., Lipton, P. & Lytton, W. W. Modeling molecular pathways of neuronal ischemia. Prog. Mol. Biol. Transl. Sci. 123, 249–275 (2014).

Article CAS PubMed PubMed Central Google Scholar

Fonteriz, R. I. et al. Monitoring mitochondrial [Ca(2+)] dynamics with rhod-2, ratiometric pericam and aequorin. Cell Calcium 48, 61–69 (2010).

Article CAS PubMed Google Scholar

Tsien, R. Y. A non-disruptive technique for loading calcium buffers and indicators into cells. Nature 290, 527–528 (1981).

Article CAS PubMed Google Scholar

Berry, W., Dakhil, S., Modiano, M., Gregurich, M. & Asmar, L. Phase III study of mitoxantrone plus low dose prednisone versus low dose prednisone alone in patients with asymptomatic hormone refractory prostate cancer. J. Urol. 168, 2439–2443 (2002).

Article CAS PubMed Google Scholar

Evison, B. J., Sleebs, B. E., Watson, K. G., Phillips, D. R. & Cutts, S. M. Mitoxantrone, more than just another topoisomerase II poison. Med. Res. Rev. 36, 248–299 (2016).

Article CAS PubMed Google Scholar

Scott, L. J. & Figgitt, D. P. Mitoxantrone: a review of its use in multiple sclerosis. CNS Drugs 18, 379–396 (2004).

Article CAS PubMed Google Scholar

Ewer, M. S. & Ewer, S. M. Cardiotoxicity of anticancer treatments. Nat. Rev. Cardiol. 12, 547–558 (2015).

Article CAS PubMed Google Scholar

Romitan, D. M. et al. Cardiomyopathies and arrhythmias induced by cancer therapies. Biomedicines https://doi.org/10.3390/biomedicines8110496 (2020).

Guglin, M., Aljayeh, M., Saiyad, S., Ali, R. & Curtis, A. B. Introducing a new entity: chemotherapy-induced arrhythmia. Europace 11, 1579–1586 (2009).

Article PubMed Google Scholar

Neri, B., Cini-Neri, G. & D’Alterio, M. Effect of anthracyclines and mitoxantrone on oxygen uptake and ATP intracellular concentration in rat heart slices. Biochem. Biophys. Res. Commun. 125, 954–960 (1984).

Article CAS PubMed Google Scholar

Arduino, D. M. et al. Systematic identification of MCU modulators by orthogonal interspecies chemical screening. Mol. Cell 67, 711–723.e7 (2017).

Article CAS PubMed PubMed Central Google Scholar

De Mario, A. et al. Identification and functional validation of FDA-approved positive and negative modulators of the mitochondrial calcium uniporter. Cell Rep. 35, 109275 (2021).

Article PubMed PubMed Central Google Scholar

Loubiere, C. et al. The energy disruptor metformin targets mitochondrial integrity via modification of calcium flux in cancer cells. Sci. Rep. 7, 5040 (2017).

Article PubMed PubMed Central Google Scholar

Zhao, H. et al. AMPK-mediated activation of MCU stimulates mitochondrial Ca(2+) entry to promote mitotic progression. Nat. Cell Biol. 21, 476–486 (2019).

Article CAS PubMed Google Scholar

Moscovitz, H. L., Donoso, E., Gelb, I. J. & Welkowitz, W. Intracardiac phonocardiography: correlation of mechanical, acoustic and electric events of the cardiac cycle. Circulation 18, 983–988 (1958).

Article CAS PubMed Google Scholar

Morad, M. & Orkand, R. K. Excitation-concentration coupling in frog ventricle: evidence from voltage clamp studies. J. Physiol. 219, 167–189 (1971).

Article CAS PubMed PubMed Central Google Scholar

Wendt-Gallitelli, M. F. & Isenberg, G. Total and free myoplasmic calcium during a contraction cycle: x-ray microanalysis in guinea-pig ventricular myocytes. J. Physiol. 435, 349–372 (1991).

Article CAS PubMed PubMed Central Google Scholar

Lee, K. S. A new technique for the simultaneous recording of oxygen consumption and contraction of muscle: the effect of ouabain on cat papillary muscle. J. Pharmacol. Exp. Ther. 109, 304–312 (1953).

CAS PubMed Google Scholar

Lentini, E. A. Myocardial oxygen consumption as influenced by isotonic contractions. Proc. Soc. Exp. Biol. Med. 109, 869–872 (1962).

Article CAS PubMed Google Scholar

Isenberg, G., Han, S., Schiefer, A. & Wendt-Gallitelli, M. F. Changes in mitochondrial calcium concentration during the cardiac contraction cycle. Cardiovasc. Res. 27, 1800–1809 (1993).

Article CAS PubMed Google Scholar

Reiermann, H. J., Herzig, J. W. & Rüegg, J. C. Ca++ activation of ATPase activity, ATP-Pi exchange, and tension in briefly glycerinated heart muscle. Basic Res. Cardiol. 72, 133–139 (1977).

Article CAS PubMed Google Scholar

O’Rourke, B., Ramza, B. M. & Marban, E. Oscillations of membrane current and excitability driven by metabolic oscillations in heart cells. Science 265, 962–966 (1994).

Article PubMed Google Scholar

Robert, V. et al. Beat-to-beat oscillations of mitochondrial [Ca2+] in cardiac cells. EMBO J. 20, 4998–5007 (2001).

Article CAS PubMed PubMed Central Google Scholar

Wescott, A. P., Kao, J. P. Y., Lederer, W. J. & Boyman, L. Voltage-energized calcium-sensitive ATP production by mitochondria. Nat. Metab. 1, 975–984 (2019).

Article CAS PubMed PubMed Central Google Scholar

Patterson, A. J. & Zhang, L. Hypoxia and fetal heart development. Curr. Mol. Med. 10, 653–666 (2010).

Article CAS PubMed PubMed Central Google Scholar

Bakkehaug, J. P. et al. Myosin activator omecamtiv mecarbil increases myocardial oxygen consumption and impairs cardiac efficiency mediated by resting myosin ATPase activity. Circ. Heart Fail. 8, 766–775 (2015).

Article CAS PubMed Google Scholar

Lewis, G. D. et al. Effect of omecamtiv mecarbil on exercise capacity in chronic heart failure with reduced ejection fraction: the METEORIC-HF randomized clinical trial. JAMA 328, 259–269 (2022).

Article CAS PubMed PubMed Central Google Scholar

Teerlink, J. R. et al. Chronic Oral Study of Myosin Activation to Increase Contractility in Heart Failure (COSMIC-HF): a phase 2, pharmacokinetic, randomised, placebo-controlled trial. Lancet 388, 2895–2903 (2016).

Article CAS PubMed Google Scholar

Bick, A. G. et al. Cardiovascular homeostasis dependence on MICU2, a regulatory subunit of the mitochondrial calcium uniporter. Proc. Natl Acad. Sci. USA 114, E9096–e9104 (2017).

Article CAS PubMed PubMed Central Google Scholar

Pan, X. et al. The physiological role of mitochondrial calcium revealed by mice lacking the mitochondrial calcium uniporter. Nat. Cell Biol. 15, 1464–1472 (2013).

Article CAS PubMed PubMed Central Google Scholar

Lambert, J. P. et al. MCUB regulates the molecular composition of the mitochondrial calcium uniporter channel to limit mitochondrial calcium overload during stress. Circulation 140, 1720–1733 (2019).

Article CAS PubMed PubMed Central Google Scholar

Wu, Y. et al. The mitochondrial uniporter controls fight or flight heart rate increases. Nat. Commun. 6, 6081 (2015).

Article CAS PubMed Google Scholar

Yellon, D. M. & Hausenloy, D. J. Myocardial reperfusion injury. N. Engl. J. Med. 357, 1121–1135 (2007).

Article CAS PubMed Google Scholar

Kwong, J. C. et al. Acute myocardial infarction after laboratory-confirmed influenza infection. N. Engl. J. Med. 378, 345–353 (2018).

Article PubMed Google Scholar

Tiburcy, M. et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair. Circulation 135, 1832–1847 (2017).

Article CAS PubMed PubMed Central Google Scholar

Fleischer, V. et al. Cardiotoxicity of mitoxantrone treatment in a German cohort of 639 multiple sclerosis patients. J. Clin. Neurol. 10, 289–295 (2014).

Article PubMed PubMed Central Google Scholar

Xie, A. et al. Mitochondrial Ca(2+) influx contributes to arrhythmic risk in nonischemic cardiomyopathy. J. Am. Heart Assoc. https://doi.org/10.1161/jaha.117.007805 (2018).

Schweitzer, M. K. et al. Suppression of arrhythmia by enhancing mitochondrial Ca(2+) uptake in catecholaminergic ventricular tachycardia models. JACC Basic Transl. Sci. 2, 737–747 (2017).

Article PubMed PubMed Central Google Scholar

Burridge, P. W. et al. Chemically defined generation of human cardiomyocytes. Nat. Methods 11, 855–860 (2014).

Article CAS PubMed PubMed Central Google Scholar

Pires, R. H., Shree, N., Manu, E., Guzniczak, E. & Otto, O. Cardiomyocyte mechanodynamics under conditions of actin remodelling. Phil. Trans. R. Soc. Lond. B 374, 20190081 (2019).

Article CAS Google Scholar

Li, Z., Guo, X., Palmer, A. F., Das, H. & Guan, J. High-efficiency matrix modulus-induced cardiac differentiation of human mesenchymal stem cells inside a thermosensitive hydrogel. Acta Biomater. 8, 3586–3595 (2012).

Article CAS PubMed Google Scholar

Liu, M., Sun, J., Sun, Y., Bock, C. & Chen, Q. Thickness-dependent mechanical properties of polydimethylsiloxane membranes. J. Micromech. Microeng. 19, 035028 (2009).

Article Google Scholar

Soofi, S. S., Last, J. A., Liliensiek, S. J., Nealey, P. F. & Murphy, C. J. The elastic modulus of Matrigel as determined by atomic force microscopy. J. Struct. Biol. 167, 216–219 (2009).

Article CAS PubMed PubMed Central Google Scholar

Ebrahimi, A. P. Mechanical properties of normal and diseased cerebrovascular system. J. Vasc. Interv. Neurol. 2, 155–162 (2009).

PubMed PubMed Central Google Scholar

Dutta, D. et al. Non-invasive assessment of elastic modulus of arterial constructs during cell culture using ultrasound elasticity imaging. Ultrasound Med. Biol. 39, 2103–2115 (2013).

Article PubMed PubMed Central Google Scholar

Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

Article CAS PubMed PubMed Central Google Scholar

Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

Article CAS PubMed PubMed Central Google Scholar

Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

Article PubMed PubMed Central Google Scholar

Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

Article CAS PubMed Google Scholar

Mi, H., Muruganujan, A., Ebert, D., Huang, X. & Thomas, P. D. PANTHER version 14: more genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res. 47, D419–D426 (2018).

Article PubMed Central Google Scholar

Xia, J., Gill, E. E. & Hancock, R. E. W. NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nat. Protoc. 10, 823–844 (2015).

Article CAS PubMed Google Scholar

Ehrlich, A. et al. Microphysiological flux balance platform unravels the dynamics of drug induced steatosis. Lab Chip 18, 2510–2522 (2018).

Article CAS PubMed PubMed Central Google Scholar

Joung, J. et al. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening. Nat. Protoc. 12, 828–863 (2017).

Article CAS PubMed PubMed Central Google Scholar

Ou, Q. et al. Physiological biomimetic culture system for pig and human heart slices. Circ. Res. 125, 628–642 (2019).

Article CAS PubMed PubMed Central Google Scholar

Ludikhuize, M. C., Meerlo, M., Burgering, B. M. T. & Rodríguez Colman, M. J. Protocol to profile the bioenergetics of organoids using Seahorse. STAR Protoc. 2, 100386 (2021).

Article CAS PubMed PubMed Central Google Scholar

Sigg, C. & Buhmann, J. Expectation-maximization for sparse and non-negative PCA. In Proc. 25th International Conference on Machine Learning 960–967 (Association for Computing Machinery, 2008).

Wickham, H. & Sievert, C. ggplot2: Elegant Graphics for Data Analysis (Springer, 2016).

Download references

Funding was provided by the European Research Council Consolidator Grant OCLD (project no. 681870) and generous gifts from the Nikoh Foundation and the Sam and Rina Frankel Foundation. M.G. was supported by a Neubauer Foundation Graduate Fellowship. We thank O. Leitersdorf, Y. Kroiz, J. Gotlib, D. Viner, I. Shweky, H. Naimi, B. A. Berke, M. Ehrlich and S. Regenbaum. Figure 8a was generated using BioRender.com.

Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Jerusalem, Israel

Mohammad Ghosheh, Avner Ehrlich, Konstantinos Ioannidis, Muneef Ayyash, Merav Cohen & Yaakov Nahmias

The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel

Mohammad Ghosheh, Avner Ehrlich, Konstantinos Ioannidis, Muneef Ayyash, Merav Cohen & Yaakov Nahmias

Tissue Dynamics, LTD, Jerusalem, Israel

Avner Ehrlich, Konstantinos Ioannidis & Yaakov Nahmias

Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel

Idit Goldfracht & Lior Gepstein

Department of Cell and Developmental Biology, The Hebrew University of Jerusalem, Jerusalem, Israel

Merav Cohen & Yaakov Nahmias

Department of Biological Chemistry, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel

Amit Fischer

Department of General Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel

Yoav Mintz

Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel

Yoav Mintz

Cardiology Department, Rambam Health Care Campus, Haifa, Israel

Lior Gepstein

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

M.G., A.E. and Y.N. conceived the hypothesis. M.G., A.E., M.A., L.G. and Y.N. designed the experiment. M.G., A.E., M.A., K.I., M.C., A.F. and Y.N. performed the experiments. Y.M. provided the porcine heart and prepared the ex-vivo heart tissue. M.G. and A.E. analysed the results. M.G., A.E. and Y.M. wrote the manuscript. M.G., A.E. and I.G. built the system. M.C., L.G. and Y.N supervised the project. All authors read the manuscript and agree with its contents.

Correspondence to Yaakov Nahmias.

Y.N. and A.E. are employees of Tissue Dynamics. M.G., A.E. and Y.N. filed a patent application through Hebrew University (US202163242091P; 2019, Israel). The other authors declare no competing interests.

Nature Biomedical Engineering thanks Alessandro Prigione and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

a, Representative time-lapse sequence confocal images depicting the formation of vascular networks in cardiac organoids formed from UN-1, ACS-1021, and ACS-1028 hiPSC-derived cardiomyocytes. Confocal microscopy shows the distribution of GFP-expressing rat microvascular cardiac endothelial cells (CECs) in the organoid. Vascular networks are apparent by day 10, stabilizing into a capillary-like network, distributed within the organoid at 25 d. UN-1 cardiac organoid images were taken from Fig.1b. Scale bar, 100 μm. b, Relative gene expression of the rat microvascular cardiac endothelial cells (CECs) and isolated rat primary endothelial cells. The CECs are showing expression signatures comparable with those of isolated rat primary endothelial cells across multiple endothelial markers. Anti-phospho-Nuclear Receptor (NR4A1), Cadherin 5 (CDH5), Von Willebrand factor (VWF), and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1) are similarly expressed among the CECs used and isolated rat primary endothelial cells.

Source data

a, Principal component analysis (PCA) of 513 genes differentially expressed between hiPS-derived cardiomyocytes and vascularized cardiac organoids (methods). Cardiac organoids cluster with adult, but not fetal cardiomyocytes. b, Principal component analysis (PCA) of 513 genes differentially expressed between hiPS-derived cardiomyocytes and vascularized cardiac organoids (methods) separated into PC components. PC1 gene set is enriched for angiogenesis and cell adhesion, clustering the non-beating AC16 cell line, the negative control, with the mature heart tissue. This clearly suggests that PC1 is less relevant for comparative analysis. c, Visual contraction analysis of UN-1 vascularized cardiac organoids treated with DMSO (Control), 10 µM amiodarone, or 100 µM epinephrine (Supplementary. Video 3), normalized to the highest and lowest signal recorded through the entire measurement duration (30 seconds; methods). Analysis shows that untreated organoids acquire a homogenous synchronized spontaneous beating of 66 ± 5 beats per minute. Stimulation with 100 µM epinephrine increases the contraction rate to 88 ± 7 bpm and relative contraction by 18% (n = 5, p < 0.001), while stimulation with 10 µM amiodarone decreased the rate to 52 ± 4 bpm and contraction by 28% (n = 5, p < 0.001), resulting in a physiological-like response to the drugs. Mean of 5 biological replicates; error bars, s.e.m. Significance was determined using a one-way ANOVA with Dunnett correction. Graphs were taken from Fig. 3d. d, Visual contraction analysis of UN-1 hiPSC-derived cardiomyocytes (hiPSC-CMs) treated with DMSO (Control), 10 µM amiodarone, or 100 µM epinephrine. Analysis shows that untreated hiPSC-CMs acquire a homogenous unsynchronized beating of 97 ± 4 beats per minute. Stimulation with 100 µM epinephrine increases the contraction rate to 106 ± 4 bpm and relative contraction by 18% (n = 3, p < 0.01), while stimulation with 10 µM amiodarone decreased the rate to 92 ± 3 bpm and contraction by 28% (n = 3, p < 0.01). Mean of 3 biological replicates; error bars, s.e.m. Significance was determined using a one-way ANOVA with Dunnett correction. e, Seahorse MitoStress assay nested analysis of UN-1 cardiac organoids, UN-1 hiPSC-derived cardiomyocytes (hiPSC-CMs), and cardiac endothelial cells (CECs). CECs show basal respiration equal to 4.5% of the basal respiration of the cardiac organoids and less than 2% of maximal respiration, indicating that changes in respiration are attributed to changes in the cardiomyocytes (n = 9, p < 0.001). Significance was determined using one-way ANOVA and Dunnett multiple comparison correction. Lines represent independent experiments, Error bars mark standard error of mean among n = 3 biological repeats.

Source data

a, Scheme and typical measurements depicting the advantages of using a 2-PMT system over a single PMT system. The addition of the second detector (cPMT), which measures the excitation signal, reduces the noise and enables accurate measurements at sub-second resolution. The second PMT also allows an emission-independent measurement of tissue contraction (methods). b, Representative calibration measurements of the reflected signal measured by the second PMT in different displacements. Curve fitting displays a sigmoidal relationship between the emission intensity, measured by peak-to-peak voltage (VP-P), and the sensor displacement. Cardiac displacement was measured by the embedded oxygen beads inside the cardiac organoids during a contraction when the beads move at different distances from the focal point. The Sigmoidal fit shows a correlation of R-square: 0.9835 and RMSE below 4. c, Representative continuous interstitial oxygen measurements in cardiac organoids measured continuously over 10 hours. Measurements show steady recordings that are unaffected by photobleaching or system sensitivity loss at prolonged measurements. Oxygen content and oscillation were unchanged even after 10 hours of measurement.

Source data

a, Representative simultaneous measurements and (b) Fast Fourier Transformation (FFT) analysis of contraction, field potential, and interstitial oxygen during spontaneous beating of cardiac organoids formed from UN-1, ACS-1021, and ACS-1028 hiPSC-derived cardiomyocytes. Interstitial oxygen concentration shows oscillatory behaviour during the cardiac cycle, yielding distinct single-frequency peaks in FFT analysis correlated to the mechanical and electrical behaviour of the cardiac tissue. UN-1 cardiac organoid graphs were taken from Fig. 4f–h. Nested analysis of the organoid’s contraction and oxygen oscillation (c) frequency and (d) amplitude in UN-1, ACS-1021, and ACS-1028 cardiac organoids. Treatment with 10 μM of myosin II inhibitor Blebbistatin. Blebbistatin completely inhibits the contraction of vascularized cardiac organoids (n = 3, p < 0.001) but does not affect the field potential or oxygen oscillation (n = 3, p > 0.05). Treatment with 25 μM of Nav channel inhibitor Tetrodotoxin (TTX) resulted in a complete loss of field potential generation, the coupled mechanical contraction (n = 3, p < 0.001), and a concurrent loss of oxygen oscillations (n = 3, p < 0.001). Middle represents mean of 3 biological repeats for each line; error bars,s.e.m. Significance was determined using a two-tailed nested t-test. e, Representative graph of the interstitial oxygen behaviours in ACS-1021 cardiac organoids following treatment with 25 μM of Nav channel inhibitor Tetrodotoxin (TTX). After 7 minutes of exposure to TTX, a complete loss of field potential generation occurs, coupled with decay in oxygen oscillations (n = 3). Graph was taken from Fig. 4l. Middle represents mean of 3 biological repeats for each line; error bars,s.e.m. Significance was determined using a two-tailed nested t-test.

Source data

Nested analysis of the organoid’s contraction and oxygen oscillation (a) frequency and (b) amplitude during prolonged stimulation by 100 µM epinephrine in UN-1, ACS-1021, and ACS-1028 cardiac organoids. Analysis shows that epinephrine exposure significantly increases the frequency of cardiac contraction and oxygen oscillations (n = 3, p < 0.001), and the amplitude of cardiac contraction and oxygen oscillation amplitudes (n = 3, p < 0.001). c, Analysis of the kinetic behaviour of the organoid’s contraction rate during prolonged stimulation by 100 µM epinephrine. Kinetic analysis suggests that epinephrine stimulation results in a sigmoidal-like change in the organoid contraction rate. Representative relation graphs between (d) contraction amplitude (contractility) to contraction rate, and (e) interstitial oxygen content to contraction rate during prolonged epinephrine stimulation. Analysis suggests a correlation between an increase in cardiac organoid contractility and oxygen consumption. f, Representative frequency histograms of interstitial oxygen measurements at 0, 15, and 90 minutes after stimulation with 100 µM epinephrine. Analysis shows that an increase in oxygen consumption correlates to an increase in interstitial oxygen content variability correlative to the increased oxygen amplitudes measured. Representative correlation analysis between (g) oxygen oscillation frequency to contraction frequency and (h) oxygen oscillation amplitude to contractility reveals a direct linear correlation between the oscillatory behaviour of the interstitial oxygen and organoid contraction. i, Representative graphs of the cardiac organoid’s contraction and interstitial oxygen content 60 mins prior to epinephrine stimulation and 5 hours post-stimulation with 100 µM epinephrine. Oxygen returns to the baseline value after 300 minutes, indicating hypoxia, stimulation-induced or ischemia-like injuries did not occur. Middle represents mean of 3 biological repeats for each line; error bars,s.e.m. Significance was determined using a two-tailed nested t-test.

Source data

a, Immunofluorescent micrograph of mitochondrial membrane potential measured using live imaging of TMRE dye (Supplementary. Video 2; methods). b, Kinetic analysis of mitochondrial membrane potential using TMRE. The mitochondrial membrane potential of hiPSC-derived cardiomyocytes oscillates in the frequency of contraction. The mitochondrial membrane potential of non-beating cells did not oscillate and was lower overall. Rainbow heatmaps of (c) mean mitochondrial membrane potential and (d) major oscillating frequency of (MMP) measured using TMRE (methods). The heatmaps suggest a correlation between areas with high mean mitochondrial membrane potential and oscillation frequency. e, Immunofluorescent micrograph of mitochondrial membrane potential measured using live imaging of JC-1 dye (Supplementary. Video 3; methods). f, Kinetic analysis of mitochondrial membrane potential following aggregation of JC-1 dye. Mitochondrial membrane potential showed distinct polarization peaks in contracting cells, while non-beating cells did not oscillate and show lower mitochondrial membrane potential overall. Rainbow heatmaps of (g) mean mitochondrial membrane potential and (h) major oscillating frequency of (MMP) measured using JC-1 (methods). Similar to the behaviour measured by TMRE, JC-1 heatmaps suggest a correlation between areas with high mean mitochondrial membrane potential and oscillation frequency. Scale bar, 25 μm.

Source data

a, Kinetic measurements of mitochondrial calcium in beating 2D-cultured hiPSC-derived cardiomyocytes. Non-targeting sgRNA had no effect on [Ca2+]m showing a dominant frequency of 0.8 Hz, while MCU knockout (MCUKO) showed a 35–50% decrease in [Ca2+]m and oscillation magnitude while increasing oscillation rate to 1.3–1.4 Hz. b, Immunofluorescent confocal microscopy demonstrated a marked reduction in MCU expression in the MCU knockout (MCUKO) compared to the non-targeting control. Scale bar, 100 µm. c, Relative gene expression of the MCU gene was markedly reduced by 46% (n = 3, p < 0.001) and 27% (n = 3, p < 0.1) in the MCU knockout MCUKO cardiac organoids compared to the non-targeting cardiac organoids in ACS-1021 and UN-1 cell lines respectively. Mean of 3 biological replicates; error bars, s.e.m. d, Interstitial oxygen showed a 78% (n = 3, p < 0.001) and 126% (n = 4, p < 0.001) increase in the MCUKO cardiac organoids compared to the non-targeting organoids in ACS-1021 and UN-1 cell lines, respectively. Mean of 3 biological replicates; error bars,s.e.m. Significance was determined using a one-way ANOVA with Dunnett correction. UN-1 cardiac organoid graphs were taken from Fig. 6a.

Source data

a, Representative kinetic measurements of interstitial oxygen content and mean oxygen content analysis in non-homogenous MCUKO or Non-targeting sgRNA cardiac organoids. Analysis shows a decrease in oscillation amplitude and an increase in oscillation frequency in the MCUKO organoids, coupled with an increase in mean oxygen content (n = 3, p < 0.001). Mean of 3 biological replicates; error bars,s.e.m. Significance was determined using two tailed t-test. Cardiac organoid graphs were taken from Fig. 6c. b, Representative kinetic measurements of interstitial oxygen content and mean oxygen content analysis in cardiac organoids treated with DMSO (Control), 10 μM mitoxantrone (Mitoxantrone) or 10 μM mitoxantrone, and 100 μM AMP-activated protein kinase (AMPK) activator metformin (Mitoxantrone + Metformin). Analysis shows a decrease in oscillation amplitude and an increase in oscillation frequency in the mitoxantrone-treated organoids, coupled with an increase in mean oxygen content (n = 3, p < 0.001). Metformin reverts these changes, restoring mean oxygen content to levels not significantly different than the control (n = 3, p > 0.05). Cardiac organoid graphs were taken from Fig. 6e. Mean of 3 biological replicates; error bars,s.e.m. Significance was determined using two tailed t-test. c, Representative kinetic measurements of interstitial oxygen content and mean oxygen content analysis in porcine cardiac tissue exposed to DMSO (control), 10 μM of blebbistatin, 10 µM mitoxantrone, or 10 µM mitoxantrone, and 100 μM metformin (mitoxantrone + metformin). Analysis shows that blebbistatin does not change oscillation amplitude and oscillation frequency or mean oxygen content (n = 3, p > 0.05). Mitoxantrone-treated organoids show a decrease in oscillation amplitude and an increase in oscillation frequency, coupled with an increase in mean oxygen content (n = 3, p < 0.001). Metformin partly reverts these changes, increasing mean oxygen content by 29% (n = 3, p < 0.001). Porcine cardiac tissue graphs were taken from Fig. 8d. Mean of 3 biological replicates; error bars,s.e.m. Significance was determined using one-way ANOVA with Dunnett multiple comparison correction.

Source data

Supplementary figures.

Representative time-lapse sequence of brightfield images showing vascularized cardiac organoids during organoid formation.

Synchronized beating cardiac organoid embedded with oxygen sensors.

The effect of pharmaceutical stimulation of epinephrine and amiodarone on the contraction rate and contractility of a cardiac organoid.

Immunofluorescent live imaging of mitochondrial membrane-potential oscillations by TMRE stain in homogeneous beating hiPSC-derived cardiomyocytes.

Beating cardiac organoids under epinephrine stimulation at different timepoints.

MATLAB projection of the desynchronization of cardiac organoids due to MCU inhibition by KB-R7943.

Beating cardiac organoids embedded with oxygen sensors on an MEA chip.

Non-targeting CRISPR knockout cardiomyocyte wells display cardiac contractions, whereas none of the MCU CRISPR KO wells display cardiac contraction.

Time-lapse sequence of brightfield and fluorescent images of cardiac organoids loaded with the cell-impermeable Dextran-Texas Red.

Source data and statistics.

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and Permissions

Ghosheh, M., Ehrlich, A., Ioannidis, K. et al. Electro-metabolic coupling in multi-chambered vascularized human cardiac organoids. Nat. Biomed. Eng (2023). https://doi.org/10.1038/s41551-023-01071-9

Download citation

Received: 09 September 2021

Accepted: 27 June 2023

Published: 07 August 2023

DOI: https://doi.org/10.1038/s41551-023-01071-9

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative