دورية أكاديمية

Evaluation and manipulation of tissue and cellular distribution of cardiac progenitor cell-derived extracellular vesicles

التفاصيل البيبلوغرافية
العنوان: Evaluation and manipulation of tissue and cellular distribution of cardiac progenitor cell-derived extracellular vesicles
المؤلفون: Marieke T. Roefs, Wolf Heusermann, Maike A. D. Brans, Christian Snijders Blok, Zhiyong Lei, Pieter Vader, Joost P. G. Sluijter
المصدر: Frontiers in Pharmacology, Vol 13 (2022)
بيانات النشر: Frontiers Media S.A., 2022.
سنة النشر: 2022
المجموعة: LCC:Therapeutics. Pharmacology
مصطلحات موضوعية: extracellular vesicle, exosome, heart, biodistribution, targeting, endothelial cell, Therapeutics. Pharmacology, RM1-950
الوصف: Cardiac progenitor cell-derived extracellular vesicles (CPC-EVs) have been successfully applied via different delivery routes for treating post-myocardial infarction injury in several preclinical models. Hence, understanding the in vivo fate of CPC-EVs after systemic or local, i.e. myocardial, delivery is of utmost importance for the further therapeutic application of CPC-EVs in cardiac repair. Here, we studied the tissue- and cell distribution and retention of CPC-EVs after intramyocardial and intravenous injection in mice by employing different EV labeling and imaging techniques. In contrast to progenitor cells, CPC-EVs demonstrated no immediate flush-out from the heart upon intramyocardial injection and displayed limited distribution to other organs over time, as determined by near-infrared imaging in living animals. By employing CUBIC tissue clearing and light-sheet fluorescent microscopy, we observed CPC-EV migration in the interstitial space of the myocardium shortly after EV injection. Moreover, we demonstrated co-localization with cTnI and CD31-positive cells, suggesting their interaction with various cell types present in the heart. On the contrary, after intravenous injection, most EVs accumulated in the liver. To potentiate such a potential systemic cardiac delivery route, targeting the cardiac endothelium could provide openings for directed CPC-EV therapy. We therefore evaluated whether decorating EVs with targeting peptides (TPs) RGD-4C or CRPPR connected to Lamp2b could enhance EV delivery to endothelial cells. Expression of both TPs enhanced CPC-EV uptake under in vitro continuous flow, but did not affect uptake under static cell culture conditions. Together, these data demonstrate that the route of administration influences CPC-EV biodistribution pattern and suggest that specific TPs could be used to target CPC-EVs to the cardiac endothelium. These insights might lead to a better application of CPC-EV therapeutics in the heart.
نوع الوثيقة: article
وصف الملف: electronic resource
اللغة: English
تدمد: 1663-9812
Relation: https://www.frontiersin.org/articles/10.3389/fphar.2022.1052091/full; https://doaj.org/toc/1663-9812
DOI: 10.3389/fphar.2022.1052091
URL الوصول: https://doaj.org/article/c30c5196cdb4453789c74e97f785554a
رقم الأكسشن: edsdoj.30c5196cdb4453789c74e97f785554a
قاعدة البيانات: Directory of Open Access Journals
الوصف
تدمد:16639812
DOI:10.3389/fphar.2022.1052091