Modelling myocardial ischemia/reperfusion injury with inflammatory response in human ventricular cardiac organoids

Laihai Zhang , Yun Jiang , Wenwen Jia , Wenjun Le , Jie Liu , Peng Zhang , Huangtian Yang , Zhongmin Liu , Yang Liu

Cell Proliferation ›› 2025, Vol. 58 ›› Issue (3) : e13762

PDF
Cell Proliferation ›› 2025, Vol. 58 ›› Issue (3) : e13762 DOI: 10.1111/cpr.13762
ORIGINAL ARTICLE

Modelling myocardial ischemia/reperfusion injury with inflammatory response in human ventricular cardiac organoids

Author information +
History +
PDF

Abstract

Current therapeutic drug exploring targeting at myocardial ischemia/reperfusion (I/R) injury is limited due to the lack of humanized cardiac models that resemble myocardial damage and inflammatory response. Herein, we develop ventricular cardiac organoids from human induced pluripotent stem cells (hiPSCs) and simulate I/R injury by hypoxia/reoxygenation (H/R), which results in increased cardiomyocytes apoptosis, elevated oxidative stress, disrupted morphological structure and decreased beat amplitude. RNA-seq reveals a potential role of type I interferon (IFN-I) in this I/R injury model. We then introduce THP-1 cells and reveal inflammatory responses between monocytes/macrophages and H/R-induced ventricular cardiac organoids. Furthermore, we demonstrate Anifrolumab, an FDA approved antagonist of IFN-I receptor, effectively decreases IFN-I secretion and related gene expression, attenuates H/R-induced inflammation and oxidative stress in the co-culture system. This study advances the modelling of myocardial I/R injury with inflammatory response in human cardiac organoids, which provides a reliable platform for preclinical study and drug screening.

Cite this article

Download citation ▾
Laihai Zhang, Yun Jiang, Wenwen Jia, Wenjun Le, Jie Liu, Peng Zhang, Huangtian Yang, Zhongmin Liu, Yang Liu. Modelling myocardial ischemia/reperfusion injury with inflammatory response in human ventricular cardiac organoids. Cell Proliferation, 2025, 58(3): e13762 DOI:10.1111/cpr.13762

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Reed GW, Rossi JE, Cannon CP. Acute myocardial infarction. Lancet. 2017; 389: 197-210.

[2]

Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: a continual challenge. Eur Heart J. 2017; 38: 774-784.

[3]

Fröhlich GM, Meier P, White SK, Yellon DM, Hausenloy DJ. Myocardial reperfusion injury: looking beyond primary PCI. Eur Heart J. 2013; 34: 1714-1722.

[4]

Cung TT, Morel O, Cayla G, et al. Cyclosporine before PCI in patients with acute myocardial infarction. N Engl J Med. 2015; 373: 1021-1031.

[5]

Hahn JY, Song YB, Kim EK, et al. Ischemic postconditioning during primary percutaneous coronary intervention: the effects of postconditioning on myocardial reperfusion in patients with ST-segment elevation myocardial infarction (POST) randomized trial. Circulation. 2013; 128: 1889-1896.

[6]

Engstrøm T, Kelbæk H, Helqvist S, et al. Effect of ischemic postconditioning during primary percutaneous coronary intervention for patients with ST-segment elevation myocardial infarction: a randomized clinical trial. JAMA Cardiol. 2017; 2: 490-497.

[7]

Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014; 32: 40-51.

[8]

Odom DT, Dowell RD, Jacobsen ES, et al. Tissue-specific transcriptional regulation has diverged significantly between human and mouse. Nat Genet. 2007; 39: 730-732.

[9]

Haghighi K, Kolokathis F, Pater L, et al. Human phospholamban null results in lethal dilated cardiomyopathy revealing a critical difference between mouse and human. J Clin Invest. 2003; 111: 869-876.

[10]

Rahman A, Li Y, Chan TK, et al. Large animal models of cardiac ischemia-reperfusion injury: where are we now? Zool Res. 2023; 44: 591-603.

[11]

Lian X, Zhang J, Azarin SM, et al. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nat Protoc. 2013; 8: 162-175.

[12]

Wnorowski A, Yang H, Wu JC. Progress, obstacles, and limitations in the use of stem cells in organ-on-a-chip models. Adv Drug Deliv Rev. 2019; 140: 3-11.

[13]

Hofbauer P, Jahnel SM, Papai N, et al. Cardioids reveal self-organizing principles of human cardiogenesis. Cell. 2021; 184: 3299-3317.e22.

[14]

Lewis-Israeli YR, Wasserman AH, Gabalski MA, et al. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat Commun. 2021; 12: 5142.

[15]

Lee A, Hudson AR, Shiwarski DJ, et al. 3D bioprinting of collagen to rebuild components of the human heart. Science. 2019; 365: 482-487.

[16]

Richards DJLY, Kerr CM, Yao J, Beeson GC, Coyle RC, et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng. 2020; 4: 446-462.

[17]

Hu W, Lazar MA. Modelling metabolic diseases and drug response using stem cells and organoids. Nat Rev Endocrinol. 2022; 18: 744-759.

[18]

Luo XL, Zhang P, Liu X, et al. Myosin light chain 2 marks differentiating ventricular cardiomyocytes derived from human embryonic stem cells. Pflug Arch Eur J Phy. 2021; 473: 991-1007.

[19]

Liu Y, Zhang Y, Mei T, et al. hESCs-Derived Early Vascular Cell Spheroids for Cardiac Tissue Vascular Engineering and Myocardial Infarction Treatment. Advanced Science. Adv Sci. 2022; 9: e2104299.

[20]

Qin XY, Shen HH, Zhang XY, et al. Hypoxia-mediated chemotaxis and residence of macrophage in decidua by secreting VEGFA and CCL2 during normal pregnancy. Reproduction. 2023; 165: 543-555.

[21]

Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15: 550.

[22]

Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium Nature Genetics. 2000; 25: 25-29.

[23]

Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000; 28: 27-30.

[24]

Lee JH, Protze SI, Laksman Z, Backx PH, Keller GM. Human pluripotent stem cell-derived atrial and ventricular cardiomyocytes develop from distinct mesoderm populations. Cell Stem Cell. 2017; 21: 179-194.e4.

[25]

Häkli M, Kreutzer J, Mäki AJ, et al. Human induced pluripotent stem cell-based platform for modeling cardiac ischemia. Sci Rep. 2021; 11: 4153.

[26]

Marinković G, Grauen Larsen H, Yndigegn T, et al. Inhibition of pro-inflammatory myeloid cell responses by short-term S100A9 blockade improves cardiac function after myocardial infarction. Eur Heart J. 2019; 40: 2713-2723.

[27]

Mullard A. FDA approves AstraZeneca's anifrolumab for lupus. Nat Rev Drug Discov. 2021; 20: 658.

[28]

Baker T, Sharifian H, Newcombe PJ, et al. Type I interferon blockade with anifrolumab in patients with systemic lupus erythematosus modulates key immunopathological pathways in a gene expression and proteomic analysis of two phase 3 trials. Ann Rheum Dis. 2024; 83: 1018-1027.

[29]

Wang H, Yang J, Cai Y, Zhao Y. Macrophages suppress cardiac reprogramming of fibroblasts in vivo via IFN-mediated intercellular self-stimulating circuit. Protein Cell. 2024:pwae013.

[30]

Riggs JM, Hanna RN, Rajan B, et al. Characterisation of anifrolumab, a fully human anti-interferon receptor antagonist antibody for the treatment of systemic lupus erythematosus. Lupus Sci Med. 2018; 5: e000261.

[31]

Zhang Q, Jiang J, Han P, et al. Direct differentiation of atrial and ventricular myocytes from human embryonic stem cells by alternating retinoid signals. Cell Res. 2011; 21: 579-587.

[32]

Feng W, Schriever H, Jiang S, et al. Computational profiling of hiPSC-derived heart organoids reveals chamber defects associated with NKX2-5 deficiency. Commun Bio. 2022; 5: 399.

[33]

Schmidt C, Deyett A, Ilmer T, et al. Multi-chamber cardioids unravel human heart development and cardiac defects. Cell. 2023; 186: 5587-5605.e27.

[34]

Song M, Choi DB, Im JS, et al. Modeling acute myocardial infarction and cardiac fibrosis using human induced pluripotent stem cell-derived multi-cellular heart organoids. Cell Death Dis. 2024; 15: 308.

[35]

Hidalgo A, Glass N, Ovchinnikov D, et al. Modelling ischemia-reperfusion injury (IRI) in vitro using metabolically matured induced pluripotent stem cell-derived cardiomyocytes. APL Bioeng. 2018; 2: 026102.

[36]

Ward MC, Gilad Y. A generally conserved response to hypoxia in iPSC-derived cardiomyocytes from humans and chimpanzees. Elife. 2019; 8: 8.

[37]

Sharma P, Liu Chung Ming C, Wang X, et al. Biofabrication of advanced in vitro 3D models to study ischaemic and doxorubicin-induced myocardial damage. Biofabrication. 2022; 14: 14.

[38]

Gunata M, Parlakpinar H. A review of myocardial ischaemia/reperfusion injury: pathophysiology, experimental models, biomarkers, genetics and pharmacological treatment. Cell Biochem Funct. 2021; 39: 190-217.

[39]

Ruparelia N, Godec J, Lee R, et al. Acute myocardial infarction activates distinct inflammation and proliferation pathways in circulating monocytes, prior to recruitment, and identified through conserved transcriptional responses in mice and humans. Eur Heart J. 2015; 36: 1923-1934.

[40]

Marsh SA, Park C, Redgrave RE, et al. Rapid fall in circulating non-classical monocytes in ST elevation myocardial infarction patients correlates with cardiac injury. FASEB J. 2021; 35: e21604.

[41]

Han Y, Liao X, Gao Z, et al. Cardiac troponin I exacerbates myocardial ischaemia/reperfusion injury by inducing the adhesion of monocytes to vascular endothelial cells via a TLR4/NF-κB-dependent pathway. Clin Sci. 2016; 130: 2279-2293.

[42]

King KR, Aguirre AD, Ye YX, et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat Med. 2017; 23: 1481-1487.

[43]

Lai L, Zhang A, Yang B, Charles EJ, Kron IL, Yang Z. Plasmacytoid dendritic cells mediate myocardial ischemia/reperfusion injury by secreting type I interferons. J Am Heart Assoc. 2021; 10: e020754.

[44]

Calcagno DM, Ng RP, Toomu A, et al. The myeloid type I interferon response to myocardial infarction begins in bone marrow and is regulated by Nrf2-activated macrophages. Sci Immun. 2020; 5: 5.

[45]

Luo XL, Jiang Y, Li Q, et al. hESC-Derived Epicardial Cells Promote Repair of Infarcted Hearts in Mouse and Swine. Adv Sci. 2023; 10: e2300470.

[46]

Parra-Izquierdo I, Sánchez-Bayuela T, López J, et al. Interferons are pro-inflammatory cytokines in sheared-stressed human aortic valve endothelial cells. Int J Mol Sci. 2021; 22: 22.

[47]

Miyazaki Y, Funada M, Nakayamada S, et al. Safety and efficacy of anifrolumab therapy in systemic lupus erythematosus in real-world clinical practice: LOOPS registry. Rheumatology (Oxford). 2024; 63(9): 2345-2354.

[48]

Bolivar S, Espitia-Corredor JA, Olivares-Silva F, et al. In cardiac fibroblasts, interferon-beta attenuates differentiation, collagen synthesis, and TGF-β1-induced collagen gel contraction. Cytokine. 2021; 138: 155359.

[49]

Wadman M. FDA no longer has to require animal testing for new drugs. Science. 2023; 379: 127-128.

[50]

Lyu Q, Gong S, Lees JG, et al. A soft and ultrasensitive force sensing diaphragm for probing cardiac organoids instantaneously and wirelessly. Nat Commun. 2022; 13: 7259.

RIGHTS & PERMISSIONS

2024 The Author(s). Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

15

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/