4.7 Article

Extracellular vesicles from human cardiovascular progenitors trigger a reparative immune response in infarcted hearts

Journal

CARDIOVASCULAR RESEARCH
Volume 117, Issue 1, Pages 292-307

Publisher

OXFORD UNIV PRESS
DOI: 10.1093/cvr/cvaa028

Keywords

Acellular therapies; Heart failure; Extracellular vesicles; Immunomodulatory effects

Funding

  1. Institut National de la Santeet de la Recherche Medicale
  2. Assistance Publique Hopitaux de Paris
  3. Universite Paris Descartes
  4. Agence Nationale de la Recherche (LabexRevive) [ANR -10-LABX-73]
  5. Fondation de France [FDF 00066471]
  6. Fondation pour la Recherche Medicale [DEQ20160334910]

Ask authors/readers for more resources

EV-CPC do not elicit an immune response in in vitro human allogeneic models or in immunocompetent animal models. They have the capacity to direct the response of monocyte/macrophages towards resolving inflammation, which enhances their clinical attractiveness as an acellular therapy for cardiac repair.
Aims The cardioprotective effects of human induced pluripotent stem cell-derived cardiovascular progenitor cells (CPC) are largely mediated by the paracrine release of extracellular vesicles (EV). We aimed to assess the immunological behaviour of EV-CPC, which is a prerequisite for their clinical translation. Methods and results Flow cytometry demonstrated that EV-CPC expressed very low levels of immune relevant molecules including HLA Class I, CD80, CD274 (PD-L1), and CD275 (ICOS-L); and moderate levels of ligands of the natural killer (NK) cell activating receptor, NKG2D. In mixed lymphocyte reactions, EV-CPC neither induced nor modulated adaptive allogeneic T cell immune responses. They also failed to induce NK cell degranulation, even at high concentrations. These in vitro effects were confirmed in vivo as repeated injections of EV-CPC did not stimulate production of immunoglobulins or affect the interferon (IFN)-gamma responses from primed splenocytes. In a mouse model of chronic heart failure, intra-myocardial injections of EV-CPC, 3 weeks after myocardial infarction, decreased both the number of cardiac pro-inflammatory Ly6Chigh monocytes and circulating levels of pro-inflammatory cytokines (IL-1 alpha, TNF-alpha, and IFN-gamma). In a model of acute infarction, direct cardiac injection of EV-CPC 2 days after infarction reduced pro-inflammatory macrophages, Ly6Chigh monocytes, and neutrophils in heart tissue as compared to controls. EV-CPC also reduced levels of pro-inflammatory cytokines IL-1 alpha, IL-2, and IL-6, and increased levels of the anti-inflammatory cytokine IL-10. These effects on human macrophages and monocytes were reproduced in vitro; EV-CPC reduced the number of pro-inflammatory monocytes and M1 macrophages, while increasing the number of anti-inflammatory M2 macrophages. Conclusions EV-CPC do not trigger an immune response either in in vitro human allogeneic models or in immunocompetent animal models. The capacity for orienting the response of monocyte/macrophages towards resolution of inflammation strengthens the clinical attractiveness of EV-CPC as an acellular therapy for cardiac repair. [GRAPHICS] .

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available