Skip to main content
Erschienen in: Basic Research in Cardiology 1/2021

Open Access 01.12.2021 | Editorial

Cardioprotection vs. regeneration: the case of extracellular vesicle-derived microRNAs

verfasst von: Kai C. Wollert, Loren J. Field

Erschienen in: Basic Research in Cardiology | Ausgabe 1/2021

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN
Hinweise
This comment refers to the article available at https://​doi.​org/​10.​1007/​s00395-021-00858-8.
Cardiomyocyte renewal in the adult mammalian heart occurs via proliferation of pre-existing cardiomyocytes, as opposed to differentiation from endogenous cardiomyogenic stem cells [7]. Unfortunately, the intrinsic renewal capacity of adult mammalian cardiomyocytes is limited, with similarly low rates reported for rodents and humans [3, 17]. Consequently, injuries associated with massive cardiomyocyte death, like an acute myocardial infarction (MI), promote scarring as opposed to noticeable tissue regeneration [14]. Stimulating cardiomyocyte proliferation may, however, provide a means to achieve myocardial regeneration after injury [2, 5, 6, 8, 13].
In this issue of Basic Research in Cardiology, Jung and colleagues propose a new microRNA-based strategy for ischemic heart repair [10]. They initially observed that human-induced pluripotent stem cell-derived cardiomyocytes (iCMs) release large amounts of extracellular vesicles (EVs) when cultured under hypoxic conditions. EVs are membrane-enclosed nanoscale particles that can transfer protein, lipid, RNA, and DNA cargo between cells through the extracellular space [12]. Three microRNAs (miR-20b, miR-92a, and miR-363), originating from a common pre-microRNA transcript, were highly enriched in hypoxic iCM-derived EVs. Treatment with iCM-derived EVs or directly overexpressing the three miRs protected iCMs from hypoxia-induced cell death. Conversely, downregulating the three miRs reduced iCM survival under hypoxic conditions, thus revealing a cell-autonomous protective mechanism [10].
Exploring therapeutic potential, the authors turned to a mouse model of acute MI induced by permanent coronary artery ligation. Injecting EVs or miR-20b-, miR-92a-, and miR-363-mimics encapsulated in a biodegradable polymer into the infarct border zone immediately after MI attenuated left ventricular dilatation and systolic dysfunction as assessed by magnetic resonance imaging (MRI) at 2 and 4 weeks. At 4 weeks, miR-treated mice had developed only very small scars (encompassing ~ 13% of the LV myocardium) compared with control animals (~ 38%). Mechanistically, the authors propose that the EVs, specifically the three miRs, stimulated cardiomyocyte proliferation in the infarct border zone leading to endogenous self-repair of the heart [10]. But is this a plausible explanation?
Supporting their case, the authors found that overexpressing the three miRs in hypoxic iCMs induced mRNA signatures related to cell cycle G1/S transition, DNA replication, and G2/M phase. As evidenced by EdU incorporation and Ki67 and Aurora B expression, these gene expression changes were associated with a modest increase in the number of iCMs progressing through the cell cycle. Using bioinformatic tools and miR-target validation, the authors identified Notch3 as a direct downstream target of the three miRs. Indeed, selectively targeting Notch3 using a small interfering RNA mimicked the effects of miR-overexpression in iCMs. Considering that iCMs provide a suitable platform to study cardiomyocyte proliferation in vitro [4], the authors regrettably do not report the effects of miR-transfection or Notch3 targeting on iCM cell numbers [10].
Quantitation of cell cycle and myofiber marker colocalization via confocal imaging of tissue sections suggested a high rate of cardiomyocyte cell cycle activity in miR-treated infarcted mice [10]. However, this approach is prone to misidentification of cardiomyocyte nuclei when employed in the absence of a cardiomyocyte-restricted nuclear marker or a cell membrane marker [1], as was the case here. Because miR-treatment also appeared to increase cell cycle activity in non-cardiomyocytes, nuclear misidentification would erroneously suggest increased cell cycle activity in the cardiomyocyte compartment of treated animals [18]. It was surprising that miR-mimics would elicit a similar induction of cell cycle activity in in vivo adult cardiomyocytes (which are exceedingly reluctant to proliferate) [3, 17] as compared to iCMs (a cell population which is highly responsive to mitogenic stimuli) [4]. It was also rather surprising that a mitotic marker would exhibit a larger labeling index than an S-phase marker [10], given that S-phase duration is much longer than M-phase duration [16]. Finally, no direct proof of cardiomyocyte cytokinesis is provided in the in vivo studies.
Permanent coronary artery ligation results in transmural necrosis, affecting cardiomyocytes and non-parenchymal cells alike. MRI suggested that the infarcted area was replaced by full-thickness, viable myocardium already 2 weeks after EV delivery. To curtail scar formation within this time frame would require exceptionally high rates of cardiomyocyte proliferation. Notably, miR-mimics were taken up not only by cardiomyocytes, but also vascular cells and fibroblasts in the infarct border zone. Microvessel density in the border zone was ~ 30% greater in miR-treated mice at 4 weeks [10]. Although, with small animal numbers, this potential treatment effect did not reach statistical significance, therapies promoting comparable improvements in border zone capillarization may very well reduce scarring after MI in mice [11, 15, 19]. MiR-mimics may have also reduced cell death after MI as suggested by their effects on hypoxic iCMs in vitro and their impact on the number of caspase 3+ cells in the infarct border zone in vivo [10]. Serial histopathological examinations early after EV or miR-delivery would have been critical to understand the sequence of events and relative contributions of myocardial salvage, improved wound healing, and cardiomyocyte proliferation [5].
While the authors should be congratulated on defining a novel therapeutic approach to infarct repair, more evidence is needed to convince us, beyond a reasonable doubt, that miR-20b-, miR-92a-, and miR-363-mimics mediate their salubrious effects by stimulating myocardial regeneration, as opposed to promoting cardioprotection and wound healing [9, 14, 19].
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.
download
DOWNLOAD
print
DRUCKEN

Unsere Produktempfehlungen

Neuer Inhalt

Print-Titel

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
4.
Zurück zum Zitat Buikema JW, Lee S, Goodyer WR, Maas RG, Chirikian O, Li G, Miao Y, Paige SL, Lee D, Wu H, Paik DT, Rhee S, Tian L, Galdos FX, Puluca N, Beyersdorf B, Hu J, Beck A, Venkamatran S, Swami S, Wijnker P, Schuldt M, Dorsch LM, van Mil A, Red-Horse K, Wu JY, Geisen C, Hesse M, Serpooshan V, Jovinge S, Fleischmann BK, Doevendans PA, van der Velden J, Garcia KC, Wu JC, Sluijter JPG, Wu SM (2020) Wnt activation and reduced cell-cell contact synergistically induce massive expansion of functional human iPSC-derived cardiomyocytes. Cell Stem Cell 27(50–63):e55. https://doi.org/10.1016/j.stem.2020.06.001CrossRef Buikema JW, Lee S, Goodyer WR, Maas RG, Chirikian O, Li G, Miao Y, Paige SL, Lee D, Wu H, Paik DT, Rhee S, Tian L, Galdos FX, Puluca N, Beyersdorf B, Hu J, Beck A, Venkamatran S, Swami S, Wijnker P, Schuldt M, Dorsch LM, van Mil A, Red-Horse K, Wu JY, Geisen C, Hesse M, Serpooshan V, Jovinge S, Fleischmann BK, Doevendans PA, van der Velden J, Garcia KC, Wu JC, Sluijter JPG, Wu SM (2020) Wnt activation and reduced cell-cell contact synergistically induce massive expansion of functional human iPSC-derived cardiomyocytes. Cell Stem Cell 27(50–63):e55. https://​doi.​org/​10.​1016/​j.​stem.​2020.​06.​001CrossRef
10.
Zurück zum Zitat Jung J-H, Ikeda G, Tada Y, von Bornstädt D, Santoso M, Wahlquist C, Rhee S, Jeon Y-J, Yu AC, O’Brien C, Red-Horse K, Appel EA, Mercola M, Woo J, Yang PC (2021) miR-106a-363 cluster in extracellular vesicles promotes endogenous myocardial repair via notch3 pathway in ischemic heart inury. Basic Res Cardiol. https://doi.org/10.1007/s00395-021-00858-8CrossRefPubMed Jung J-H, Ikeda G, Tada Y, von Bornstädt D, Santoso M, Wahlquist C, Rhee S, Jeon Y-J, Yu AC, O’Brien C, Red-Horse K, Appel EA, Mercola M, Woo J, Yang PC (2021) miR-106a-363 cluster in extracellular vesicles promotes endogenous myocardial repair via notch3 pathway in ischemic heart inury. Basic Res Cardiol. https://​doi.​org/​10.​1007/​s00395-021-00858-8CrossRefPubMed
11.
Zurück zum Zitat Korf-Klingebiel M, Reboll MR, Klede S, Brod T, Pich A, Polten F, Napp LC, Bauersachs J, Ganser A, Brinkmann E, Reimann I, Kempf T, Niessen HW, Mizrahi J, Schonfeld HJ, Iglesias A, Bobadilla M, Wang Y, Wollert KC (2015) Myeloid-derived growth factor (C19orf10) mediates cardiac repair following myocardial infarction. Nat Med 21:140–149. https://doi.org/10.1038/nm.3778CrossRefPubMed Korf-Klingebiel M, Reboll MR, Klede S, Brod T, Pich A, Polten F, Napp LC, Bauersachs J, Ganser A, Brinkmann E, Reimann I, Kempf T, Niessen HW, Mizrahi J, Schonfeld HJ, Iglesias A, Bobadilla M, Wang Y, Wollert KC (2015) Myeloid-derived growth factor (C19orf10) mediates cardiac repair following myocardial infarction. Nat Med 21:140–149. https://​doi.​org/​10.​1038/​nm.​3778CrossRefPubMed
15.
Zurück zum Zitat Reboll MR, Korf-Klingebiel M, Klede S, Polten F, Brinkmann E, Reimann I, Schonfeld HJ, Bobadilla M, Faix J, Kensah G, Gruh I, Klintschar M, Gaestel M, Niessen HW, Pich A, Bauersachs J, Gogos JA, Wang Y, Wollert KC (2017) EMC10 (endoplasmic reticulum membrane protein complex subunit 10) is a bone marrow-derived angiogenic growth factor promoting tissue repair after myocardial infarction. Circulation 136:1809–1823. https://doi.org/10.1161/CIRCULATIONAHA.117.029980CrossRefPubMed Reboll MR, Korf-Klingebiel M, Klede S, Polten F, Brinkmann E, Reimann I, Schonfeld HJ, Bobadilla M, Faix J, Kensah G, Gruh I, Klintschar M, Gaestel M, Niessen HW, Pich A, Bauersachs J, Gogos JA, Wang Y, Wollert KC (2017) EMC10 (endoplasmic reticulum membrane protein complex subunit 10) is a bone marrow-derived angiogenic growth factor promoting tissue repair after myocardial infarction. Circulation 136:1809–1823. https://​doi.​org/​10.​1161/​CIRCULATIONAHA.​117.​029980CrossRefPubMed
16.
Zurück zum Zitat Rumyantsev PP (1991) Growth and hyperplasia of cardiac muscle cells. Harwood Academic Publishers, London Rumyantsev PP (1991) Growth and hyperplasia of cardiac muscle cells. Harwood Academic Publishers, London
Metadaten
Titel
Cardioprotection vs. regeneration: the case of extracellular vesicle-derived microRNAs
verfasst von
Kai C. Wollert
Loren J. Field
Publikationsdatum
01.12.2021
Verlag
Springer Berlin Heidelberg
Erschienen in
Basic Research in Cardiology / Ausgabe 1/2021
Print ISSN: 0300-8428
Elektronische ISSN: 1435-1803
DOI
https://doi.org/10.1007/s00395-021-00857-9

Weitere Artikel der Ausgabe 1/2021

Basic Research in Cardiology 1/2021 Zur Ausgabe

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

TAVI versus Klappenchirurgie: Neue Vergleichsstudie sorgt für Erstaunen

21.05.2024 TAVI Nachrichten

Bei schwerer Aortenstenose und obstruktiver KHK empfehlen die Leitlinien derzeit eine chirurgische Kombi-Behandlung aus Klappenersatz plus Bypass-OP. Diese Empfehlung wird allerdings jetzt durch eine aktuelle Studie infrage gestellt – mit überraschender Deutlichkeit.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.