Single-cell atlas of multilineage cardiac organoids derived from human induced pluripotent stem cells

Fengzhi Zhang, Hui Qiu, Xiaohui Dong, Xiaoyan Zhang, Chunlan Wang, Xin Li, Xingwu Zhang, Jie Na, Jin Zhou, Changyong Wang

PDF(3141 KB)
PDF(3141 KB)
Life Medicine ›› 2022, Vol. 1 ›› Issue (2) : 179-195. DOI: 10.1093/lifemedi/lnac002
Article
Article

Single-cell atlas of multilineage cardiac organoids derived from human induced pluripotent stem cells

Author information +
History +

Abstract

Human induced pluripotent stem cell (hiPSC)-derived cardiac organoids can be used to model human heart development and cardiovascular disease, and provide therapeutic cells to repair the heart. We used single-cell transcriptome analysis to dissect the development of 3D mini-cardiac organoids (MCOs) consisting of hiPSC-derived cardiomyocytes, and endothelial and smooth muscle cells. We found that the 3D matrix-rich microenvironment significantly promoted the maturation of cardiomyocytes, and mixing endothelial and smooth muscle cells with cardiomyocytes led to the formation of cardiac fibroblast highly expressing DLK1. Modulation of DLK1 signaling affected immunomodulatory gene expression in 2D cultured cardiomyocytes. Transplantation of multilineage MCO into a rat model of myocardial infarction significantly improved cardiac function and reduced fibrosis in the infarcted area. Our single-cell analysis of MCO provided rich information about cell state and fate dynamics in the 3D multilineage microenvironment and brought new insight into the molecular mechanism that promotes cardiomyocyte maturation and heart repair.

Keywords

human induced pluripotent stem cells / cardiomyocytes / mini-cardiac organoid / single-cell analysis / myocardial infarction

Cite this article

Download citation ▾
Fengzhi Zhang, Hui Qiu, Xiaohui Dong, Xiaoyan Zhang, Chunlan Wang, Xin Li, Xingwu Zhang, Jie Na, Jin Zhou, Changyong Wang. Single-cell atlas of multilineage cardiac organoids derived from human induced pluripotent stem cells. Life Medicine, 2022, 1(2): 179‒195 https://doi.org/10.1093/lifemedi/lnac002

References

[1]
JugduttBI. Ventricular remodeling after infarction and the extracellular collagen matrix: when is enough enough? Circulation 2003;108:1395–403.
CrossRef Google scholar
[2]
PrabhuSD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction. Circ Res 2016;119:91–112.
CrossRef Google scholar
[3]
GaoL, Gregorich ZR, ZhuW, et al. Large cardiac muscle patches engineered from human induced-pluripotent stem cell-derived cardiac cells improve recovery from myocardial infarction in swine. Circulation 2018;137:1712–30.
CrossRef Google scholar
[4]
ShadrinIY, AllenBW, QianY, et al. Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues. Nat Commun 2017;8:1825.
CrossRef Google scholar
[5]
TiburcyM, HudsonJE, BalfanzP, et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair. Circulation 2017;135:1832–47.
CrossRef Google scholar
[6]
CaspiO, LesmanA, BasevitchY, et al. Tissue engineering of vascularized cardiac muscle from human embryonic stem cells. Circ Res 2007;100:263–72.
CrossRef Google scholar
[7]
XiongQ, YeL, ZhangP, et al. Bioenergetic and functional consequences of cellular therapy: activation of endogenous cardiovascular progenitor cells. Circ Res 2012;111:455–68.
CrossRef Google scholar
[8]
BurridgePW, MatsaE, ShuklaP, et al. Chemically defined generation of human cardiomyocytes. Nat Methods 2014;11:855–60.
CrossRef Google scholar
[9]
LianX, HsiaoC, WilsonG, et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci USA 2012;109:E1848–57.
CrossRef Google scholar
[10]
OwensGK, KumarMS, WamhoffBR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 2004;84:767–801.
CrossRef Google scholar
[11]
GaoL, KupferME, JungJP, et al. Myocardial tissue engineering with cells derived from human-induced pluripotent stem cells and a native-like, high-resolution, 3-dimensionally printed scaffold. Circ Res 2017;120:1318–25.
CrossRef Google scholar
[12]
ZhangD, Shadrin IY, LamJ, et al. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials 2013;34:5813–20.
CrossRef Google scholar
[13]
CambierL, PlateM, SucovHM, et al. Nkx2-5 regulates cardiac growth through modulation of Wnt signaling by R-spondin3. Development 2014;141:2959–71.
CrossRef Google scholar
[14]
ChalJ, Oginuma M, Al TanouryZ, et al. Differentiation of pluripotent stem cells to muscle fiber to model Duchenne muscular dystrophy. Nat Biotechnol 2015;33:962–9.
CrossRef Google scholar
[15]
ParkDS, Shekhar A, LiuF, et al. ETV1 is essential for Purkinje cell specification and rapid conduction in the vertebrate heart. J Clin Invest 2016;126:4444–59.
CrossRef Google scholar
[16]
ChurkoJM, GargP, TreutleinB, et al. Defining human cardiac transcription factor hierarchies using integrated single-cell heterogeneity analysis. Nat Commun 2018;9:4906.
CrossRef Google scholar
[17]
CuiY, ZhengY, LiuX, et al. Single-cell transcriptome analysis maps the developmental track of the human heart. Cell Rep 2019;26:1934–50.
CrossRef Google scholar
[18]
DeLaughterDM, BickAG, WakimotoH, et al. Single-cell resolution of temporal gene expression during heart development. Dev Cell 2016;39:480–90.
CrossRef Google scholar
[19]
MikiK, Deguchi K, Nakanishi-KoakutsuM, et al. ERRγ enhances cardiac maturation with T-tubule formation in human iPSC-derived cardiomyocytes. Nat Commun 2021;12:1–15.
CrossRef Google scholar
[20]
GrieskampT, RudatC, LüdtkeTHW, et al. Notch signaling regulates smooth muscle differentiation of epicardium-derived cells. Circ Res 2011;108:813–23.
CrossRef Google scholar
[21]
HewittKJ, ShamisY, KnightE, et al. PDGFRβ expression and function in fibroblasts derived from pluripotent cells is linked to DNA demethylation. J Cell Sci 2012;125:2276–87.
CrossRef Google scholar
[22]
DopplerSA, Carvalho C, LahmH, et al. Cardiac fibroblasts: more than mechanical support. J Thorac Dis 2017;9:S36–51.
CrossRef Google scholar
[23]
FurtadoMB, CostaMW, PranotoEA, et al. Cardiogenic genes expressed in cardiac fibroblasts contribute to heart development and repair. Circ Res 2014;114:1422–34.
CrossRef Google scholar
[24]
ZhangH, TianL, ShenM, et al. Generation of quiescent cardiac fibroblasts from human induced pluripotent stem cells for in vitro modeling of cardiac fibrosis. Circ Res 2019;125:552–66.
CrossRef Google scholar
[25]
FriedmanCE, NguyenQ, LukowskiSW, et al. Single-cell transcriptomic analysis of cardiac differentiation from human PSCs reveals HOPXdependent cardiomyocyte maturation. Cell Stem Cell 2018;23:586–98.
CrossRef Google scholar
[26]
TakeuchiO, AkiraS. Pattern recognition receptors and inflammation. Cell 2010;140:805–20.
CrossRef Google scholar
[27]
GiacomelliE, Meraviglia V, CampostriniG, et al. Human-iPSC-derived cardiac stromal cells enhance maturation in 3D cardiac microtissues and reveal non-cardiomyocyte contributions to heart disease. Cell Stem Cell 2020;26:862–79.
CrossRef Google scholar
[28]
Ronaldson-BouchardK, Ma SP, YeagerK, et al. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 2018;556:239–43.
CrossRef Google scholar
[29]
YangX, PabonL, MurryCE. Engineering adolescence: maturation of human pluripotent stem cell-derived cardiomyocytes. Circ Res 2014;114:511–23.
CrossRef Google scholar
[30]
TullochNL, Muskheli V, RazumovaMV, et al. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ Res 2011;109:47–59.
CrossRef Google scholar
[31]
WangY, YaoF, WangL, et al. Single-cell analysis of murine fibroblasts identifies neonatal to adult switching that regulates cardiomyocyte maturation. Nat Commun 2020;11:2585.
CrossRef Google scholar
[32]
RodriguezP, SassiY, TronconeL, et al. Deletion of delta-like 1 homologue accelerates fibroblast-myofibroblast differentiation and induces myocardial fibrosis. Eur Heart J 2019;40:967–78.
CrossRef Google scholar
[33]
DriskellRR, Lichtenberger BM, HosteE, et al. Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature 2013;504:277–81.
CrossRef Google scholar
[34]
TraustadóttirGA, Lagoni LV, AnkerstjerneLBS, et al. The imprinted gene Delta like non-canonical Notch ligand 1 (Dlk1)is conserved in mammals, and serves a growth modulatory role during tissue development and regeneration through Notch dependent and independent mechanisms. Cytokine Growth Factor Rev 2019;46:17–27.
CrossRef Google scholar
[35]
IchimTE, O’Heeron P, KesariS. Fibroblasts as a practical alternative to mesenchymal stem cells. J Transl Med 2018;16:1–9.
CrossRef Google scholar
[36]
Van LinthoutS, MitevaK, TschöpeC. Crosstalk between fibroblasts and inflammatory cells. Cardiovasc Res 2014;102:258–69.
CrossRef Google scholar
[37]
ShindeAV, Frangogiannis NG. Fibroblasts in myocardial infarction: a role in inflammation and repair. J Mol Cell Cardiol 2014;70:74–82.
CrossRef Google scholar
[38]
AbdallahBM, BoissyP, TanQ, et al. dlk1/FA1 regulates the function of human bone marrow mesenchymal stem cells by modulating gene expression of pro-inflammatory cytokines and immune response-related factors. J Biol Chem 2007;282:7339–51.
CrossRef Google scholar
[39]
GonzálezMJ, Ruiz-García A, MonsalveEM, et al. DLK1 is a novel inflammatory inhibitor which interferes with NOTCH1 signaling in TLR-activated murine macrophages. Eur J Immunol 2015;45:2615–27.
CrossRef Google scholar
[40]
FrangogiannisNG. Chemokines in the ischemic myocardium: from inflammation to fibrosis. Inflamm Res 2004;53:585–95.
CrossRef Google scholar
[41]
ChenJ, HeY, TuL, et al. Dual immune functions of IL-33 in inflammatory bowel disease. Histol Histopathol 2020;35:137–46.
[42]
DuanF, HuangR, ZhangF, et al. Biphasic modulation of insulin signaling enables highly efficient hematopoietic differentiation from human pluripotent stem cells. Stem Cell Res Ther 2018;9:1–16.
CrossRef Google scholar
[43]
ZhangF, WangL, LiY, et al. Optimizing mesoderm progenitor selection and three-dimensional microniche culture allows highly efficient endothelial differentiation and ischemic tissue repair from human pluripotent stem cells. Stem Cell Res Ther 2017;8:6.
CrossRef Google scholar
[44]
ZhangF, ZhuY, ChenJ, et al. Efficient endothelial and smooth muscle cell differentiation from human pluripotent stem cells through a simplified insulin-free culture system. Biomaterials 2021;271:120713.
CrossRef Google scholar
[45]
LüS-H, WangH-B, LiuH, et al. Reconstruction of engineered uterine tissues containing smooth muscle layer in Collagen/Matrigel scaffold in vitro. Tissue Eng Part A 2009;15:1611–8.
CrossRef Google scholar
[46]
TiroshI, IzarB, PrakadanSM, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 2016;189:96.
CrossRef Google scholar
[47]
PicelliS, Faridani OR, BjörklundAK, et al. Full-length RNA-seq from single cells using Smart-seq2. Nat Protoc 2014;9:171–81.
CrossRef Google scholar
[48]
MiyaharaY, NagayaN, KataokaM, et al. Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat Med 2006;12:459–65.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2022 The Author(s) 2022. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(3141 KB)

Accesses

Citations

Detail

Sections
Recommended

/