Cardiopulmonary progenitors facilitate cardiac repair via exosomal transfer of miR-27b-3p targeting the SIK1-CREB1 axis
Ying-Ying Xiao, Luo-Xing Xia, Wen-Jing Jiang, Jian-Feng Qin, Li-Xin Zhao, Zhan Li, Li-Juan Huang, Ke-Xin Li, Peng-Jiu Yu, Li Wei, Xue-Yan Jiang, Zhe-Sheng Chen, Xi-Yong Yu
Cardiopulmonary progenitors facilitate cardiac repair via exosomal transfer of miR-27b-3p targeting the SIK1-CREB1 axis
Ischemic heart disease, especially myocardial infarction (MI), is one of the leading causes of death worldwide, and desperately needs effective treatments, such as cell therapy. Cardiopulmonary progenitors (CPPs) are stem cells for both heart and lung, but their repairing role in damaged heart is still unknown. Here, we obtained CPPs from E9.5 mouse embryos, maintained their stemness while expanding, and identified their characteristics by scRNA-seq, flow cytometry, quantitative reverse transcription-polymerase chain reaction, and differentiation assays. Moreover, we employed mouse MI model to investigate whether CPPs could repair the injured heart. Our data identified that CPPs exhibit hybrid fibroblastic, endothelial, and mesenchymal state, and they could differentiate into cell lineages within the cardiopulmonary system. Moreover, intramyocardial injection of CPPs improves cardiac function through CPPs exosomes (CPPs-Exo) by promotion of cardiomyocytic proliferation and vascularization. To uncover the underlying mechanism, we used miRNA-seq, bulk RNA-seq, and bioinformatic approaches, and found the highly expressed miR-27b-3p in CPPs-Exo and its target gene Sik1, which can influence the transcriptional activity of CREB1. Therefore, we postulate that CPPs facilitate cardiac repair partially through the SIK1-CREB1 axis via exosomal miR-27b-3p. Our study offers a novel insight into the role of CPPs-Exo in heart repair and highlights the potential of CPPs-Exo as a promising therapeutic strategy for MI.
[1] |
Collaborators GBDCoD. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden Of Disease Study 2017. Lancet. 2018;392:1736-1788.
|
[2] |
Roth GA, Johnson C, Abajobir A, et al. Global, regional, and National Burden of cardiovascular diseases for 10 causes, 1990 to 2015. J Am Coll Cardiol. 2017;70:1-25.
|
[3] |
Ebrahimi B. Cardiac progenitor reprogramming for heart regeneration. Cell Regen. 2018;7:1-6.
|
[4] |
Caporali A, Back M, Daemen MJ, et al. Future directions for therapeutic strategies in post-ischaemic vascularization: a position paper from European Society of Cardiology Working Group on Atherosclerosis and Vascular Biology. Cardiovasc Res. 2018;114:1411-1421.
|
[5] |
Ye L, Chang YH, Xiong Q, et al. Cardiac repair in a porcine model of acute myocardial infarction with human induced pluripotent stem cell-derived cardiovascular cells. Cell Stem Cell. 2014;15:750-761.
|
[6] |
Shiba Y, Fernandes S, Zhu WZ, et al. Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature. 2012;489:322-325.
|
[7] |
Fernandes S, Chong JJH, Paige SL, et al. Comparison of human embryonic stem cell-derived cardiomyocytes, cardiovascular progenitors, and bone marrow mononuclear cells for cardiac repair. Stem Cell Reports. 2015;5:753-762.
|
[8] |
Mao L, Li YD, Chen RL, et al. Heart-targeting exosomes from human cardiosphere-derived cells improve the therapeutic effect on cardiac hypertrophy. J Nanobiotechnology. 2022;20:435.
|
[9] |
Patel AN, Henry TD, Quyyumi AA, et al. Ixmyelocel-T for patients with ischaemic heart failure: a prospective randomised double-blind trial. Lancet. 2016;387:2412-2421.
|
[10] |
Mathiasen AB, Qayyum AA, Jorgensen E, et al. Bone marrow-derived mesenchymal stromal cell treatment in patients with ischaemic heart failure: final 4-year follow-up of the MSC-HF trial. Eur J Heart Fail. 2020;22:884-892.
|
[11] |
Bolli R, Mitrani RD, Hare JM, et al. A phase II study of autologous mesenchymal stromal cells and c-kit positive cardiac cells, alone or in combination, in patients with ischaemic heart failure: the CCTRN CONCERT-HF trial. Eur J Heart Fail. 2021;23:661-674.
|
[12] |
Borow KM, Yaroshinsky A, Greenberg B, Perin EC. Phase 3 DREAM-HF Trial of mesenchymal precursor cells in chronic heart failure. Circ Res. 2019;125:265-281.
|
[13] |
Kosyakovsky L. Stem cells did not decrease heart failure readmissions in DREAM-HF. Available from:
|
[14] |
Heldman AW, DiFede DL, Fishman JE, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. Jama. 2014;311:62-73.
|
[15] |
Bolli R, Tang XL. The sad plight of cell therapy for heart failure: causes and consequences. J Cardiovasc Aging. 2022;2:16.
|
[16] |
Barreto S, Hamel L, Schiatti T, Yang Y, George V. Cardiac progenitor cells from stem cells: learning from genetics and biomaterials. Cells. 2019;8:1536.
|
[17] |
Peng T, Tian Y, Boogerd CJ, et al. Coordination of heart and lung co-development by a multipotent cardiopulmonary progenitor. Nature. 2013;500:589-592.
|
[18] |
Steimle JD, Rankin SA, Slagle CE, et al. Evolutionarily conserved Tbx5-Wnt2/2b pathway orchestrates cardiopulmonary development. Proc Natl Acad Sci U S A. 2018;115:E10615-E10624.
|
[19] |
Qi H, Liu H, Pullamsetti SS, et al. Epigenetic regulation by Suv4-20h1 in cardiopulmonary progenitor cells is required to prevent pulmonary hypertension and chronic obstructive pulmonary disease. Circulation. 2021;144:1042-1058.
|
[20] |
Witman N, Sahara M. Cardiac progenitor cells in basic biology and regenerative medicine. Stem Cells Int. 2018;2018:8283648.
|
[21] |
Madonna R, Van Laake LW, Davidson SM, et al. Position paper of the European Society of Cardiology Working Group Cellular Biology of the heart: cell-based therapies for myocardial repair and regeneration in ischemic heart disease and heart failure. Eur Heart J. 2016;37:1789-1798.
|
[22] |
Sullivan R, Maresh G, Zhang X, et al. The emerging roles of extracellular vesicles as communication vehicles within the tumor microenvironment and beyond. Front Endocrinol (Lausanne). 2017;8:194.
|
[23] |
Yanez-Mo M, Siljander PR, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066.
|
[24] |
Zhang J, Li S, Li L, et al. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics Proteomics Bioinformatics. 2015;13(1):17-24.
|
[25] |
Schulte C, Zeller T. microRNA-based diagnostics and therapy in cardiovascular disease-summing up the facts. Cardiovasc Diagn Ther. 2015;5:17-36.
|
[26] |
Wang Y, Chen J, Cowan DB, Wang DZ. Non-coding RNAs in cardiac regeneration: mechanism of action and therapeutic potential. Semin Cell Dev Biol. 2021;118:150-162.
|
[27] |
Mayourian J, Ceholski DK, Gorski PA, et al. Exosomal microRNA-21-5p mediates mesenchymal stem cell paracrine effects on human cardiac tissue contractility. Circ Res. 2018;122:933-944.
|
[28] |
Xiao C, Wang K, Xu Y, et al. Transplanted mesenchymal stem cells reduce autophagic flux in infarcted hearts via the Exosomal transfer of miR-125b. Circ Res. 2018;123:564-578.
|
[29] |
Xiao J, Pan Y, Li XH, et al. Cardiac progenitor cell-derived exosomes prevent cardiomyocytes apoptosis through exosomal miR-21 by targeting PDCD4. Cell Death Dis. 2016;7:e2277.
|
[30] |
Ibrahim AG, Cheng K, Marban E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Reports. 2014;2:606-619.
|
[31] |
Khan M, Nickoloff E, Abramova T, et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ Res. 2015;117:52-64.
|
[32] |
Maeda K, Enomoto A, Hara A, et al. Identification of Meflin as a potential marker for mesenchymal stromal cells. Sci Rep. 2016;6:22288.
|
[33] |
Hendry CE, Vanslambrouck JM, Ineson J, et al. Direct transcriptional reprogramming of adult cells to embryonic nephron progenitors. J Am Soc Nephrol. 2013;24:1424-1434.
|
[34] |
Gulati GS, Sikandar SS, Wesche DJ, et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science. 2020;367:405-411.
|
[35] |
Hwang T, Parker SS, Hill SM, et al. Native proline-rich motifs exploit sequence context to target actin-remodeling Ena/VASP protein ENAH. Elife. 2022;11:70680.
|
[36] |
Cascone I, Selimoglu R, Ozdemir C, et al. Distinct roles of RalA and RalB in the progression of cytokinesis are supported by distinct Ral-GEFs. EMBO J. 2008;27:2375-2387.
|
[37] |
Timon-Gomez A, Nyvltova E, Abriata LA, Vila AJ, Hosler J, Barrientos A. Mitochondrial cytochrome c oxidase biogenesis: recent developments. Semin Cell Dev Biol. 2018;76:163-178.
|
[38] |
Zhang S, Cui Y, Ma X, et al. Single-cell transcriptomics identifies divergent developmental lineage trajectories during human pituitary development. Nat Commun. 2020;11:5275.
|
[39] |
Jagannath A, Butler R, Godinho SIH, et al. The CRTC1-SIK1 pathway regulates entrainment of the circadian clock. Cell. 2013;154:1100-1111.
|
[40] |
Conkright MD, Canettieri G, Screaton R, et al. TORCs: transducers of regulated CREB activity. Mol Cell. 2003;12:413-423.
|
[41] |
Menasche P, Vanneaux V, Hagege A, et al. Transplantation of human embryonic stem cell-derived cardiovascular progenitors for severe ischemic left ventricular dysfunction. J Am Coll Cardiol. 2018;71:429-438.
|
[42] |
Wu Q, Wang J, Tan WLW, et al. Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis. Cell Death Dis. 2020;11:354.
|
[43] |
Beliën H, Evens L, Hendrikx M, Bito V, Bronckaers A. Combining stem cells in myocardial infarction: the road to superior repair? Med Res Rev. 2022;42(1):343-373.
|
[44] |
Jones SP, Tang XL, Guo Y, et al. The NHLBI-sponsored consortium for preclinicAl assESsment of cARdioprotective therapies (CAESAR): a new paradigm for rigorous, accurate, and reproducible evaluation of putative infarct-sparing interventions in mice, rabbits, and pigs. Circ Res. 2015;116(4):572-586.
|
[45] |
Gallet R, Dawkins J, Valle J, et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2017;38:201-211.
|
[46] |
Tang S, Fan C, Iroegbu CD, et al. TMSB4 overexpression enhances the potency of marrow mesenchymal stromal cells for myocardial repair. Front Cell Dev Biol. 2021;9:670913.
|
[47] |
Klopsch C, Skorska A, Ludwig M, et al. Intramyocardial angiogenetic stem cells and epicardial erythropoietin save the acute ischemic heart. Dis Model Mech. 2018;11(6):dmm033282.
|
[48] |
Tan SH, Loo SJ, Gao Y, et al. Thymosin β4 increases cardiac cell proliferation, cell engraftment, and the reparative potency of human induced-pluripotent stem cell-derived cardiomyocytes in a porcine model of acute myocardial infarction. Theranostics. 2021;11(16):7879-7895.
|
[49] |
Gong XH, Liu H, Wang SJ, Liang SW, Wang GG. Exosomes derived from SDF1-overexpressing mesenchymal stem cells inhibit ischemic myocardial cell apoptosis and promote cardiac endothelial microvascular regeneration in mice with myocardial infarction. J Cell Physiol. 2019;234:13878-13893.
|
[50] |
Liu B, Lee BW, Nakanishi K, et al. Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat Biomed Eng. 2018;2:293-303.
|
[51] |
Nakamura Y, Kita S, Tanaka Y, et al. Adiponectin stimulates exosome release to enhance mesenchymal stem-cell-driven therapy of heart failure in mice. Mol Ther. 2020;28:2203-2219.
|
[52] |
El Harane N, Kervadec A, Bellamy V, et al. Acellular therapeutic approach for heart failure: in vitro production of extracellular vesicles from human cardiovascular progenitors. Eur Heart J. 2018;39:1835-1847.
|
[53] |
Poch CM, Foo KS, De Angelis MT, et al. Migratory and anti-fibrotic programmes define the regenerative potential of human cardiac progenitors. Nat Cell Biol. 2022;24:659-671.
|
[54] |
Taubel J, Hauke W, Rump S, et al. Novel antisense therapy targeting microRNA-132 in patients with heart failure: results of a first-in-human phase 1b randomized, double-blind, placebo-controlled study. Eur Heart J. 2021;42:178-188.
|
[55] |
Chen X, Wang L, Huang R, et al. Dgcr8 deletion in the primitive heart uncovered novel microRNA regulating the balance of cardiac-vascular gene program. Protein Cell. 2019;10:327-346.
|
[56] |
Wang Y, Liu L, Gu JH, et al. Salt-inducible kinase 1-CREB-regulated transcription coactivator 1 signalling in the paraventricular nucleus of the hypothalamus plays a role in depression by regulating the hypothalamic-pituitary-adrenal axis. Mol Psychiatry. 2022;Advance online publication.
|
[57] |
Cardoso BR, Hare DJ, Bush AI, Roberts BR. Glutathione peroxidase 4: a new player in neurodegeneration? Mol Psychiatry. 2017;22:328-335.
|
[58] |
Jiang Y, Qiao Y, He D, Tian A, Li Z. Adaptor protein HIP-55-mediated signalosome protects against ferroptosis in myocardial infarction. Cell Death Differ. 2023;30:825-838.
|
[59] |
O'Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol. 2018;9:402.
|
[60] |
Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez-Cabo F, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat Commun. 2013;4:2980.
|
[61] |
Wang XL, Zhao YY, Sun L, et al. Exosomes derived from human umbilical cord mesenchymal stem cells improve myocardial repair via upregulation of Smad7. Int J Mol Med. 2018;41:3063-3072.
|
[62] |
Zhu W, Sun L, Zhao P, et al. Macrophage migration inhibitory factor facilitates the therapeutic efficacy of mesenchymal stem cells derived exosomes in acute myocardial infarction through upregulating miR-133a-3p. J Nanobiotechnology. 2021;19:61.
|
[63] |
Han C, Yang J, Sun J, Qin G. Extracellular vesicles in cardiovascular disease: biological functions and therapeutic implications. Pharmacol Ther. 2022;233:108025.
|
/
〈 | 〉 |