Abstract
Background
Cardiosphere-derived cells (CDCs) confer cardioprotection in acute myocardial infarction (MI) via distinctive macrophage (Mϕ) polarization. Here we demonstrate that CDC-secreted exosomes (CDCexo) recapitulate the cardioprotective effects of CDC therapy known as cellular postconditioning.
Methods
Rats and pigs underwent MI induced by ischemia-reperfusion prior to intracoronary infusion of CDCexo, inert fibroblast exosomes (Fbexo; control), or vehicle. Two days later, infarct size was quantified. Macrophages were isolated from cardiac tissue or bone marrow for downstream analyses. RNA-sequencing was used to determine exosome content and alterations in gene expression profiles in Mϕ.
Results
Administration of CDCexo, but not Fbexo, after reperfusion reduces infarct size in rat and pig models of MI. Furthermore, CDCexo reduce the number of CD68+ Mϕ within infarcted tissue and modify the polarization state of Mϕ so as to mimic that induced by CDCs. CDCexo are enriched in several miRNAs (including miR-146a, miR-181b, and miR-126) relative to Fbexo. Reverse pathway analysis of whole-transcriptome data from CDCexo-primed Mϕ implicated miR-181b as a significant (p=1.3×10−21) candidate mediator of CDC-induced Mϕ polarization and protein kinase C δ (PKCδ) as a downstream target. Otherwise-inert Fbexo loaded selectively with miR-181b alter Mϕ phenotype and confer cardioprotective efficacy in a rat model of MI. Adoptive transfer of PKCδ-suppressed Mϕ recapitulates cardioprotection.
Conclusions
Our data support the hypothesis that exosomal transfer of miR-181b from CDCs into Mϕ reduces PKCδ transcript levels and underlies the cardioprotective effects of CDCs administered after reperfusion.
Keywords: Myocardial infarction, cardioprotection, macrophage, exosome, miRNA
INTRODUCTION
Acute myocardial infarction (MI) elicits a robust innate immune response that mobilizes a large population of neutrophils and macrophages to the myocardium. This response, which plays out over the first few days following injury, occurs in a phasic manner whereby a rapid infiltration of neutrophils is succeeded by a heterogeneous influx of monocytes/macrophages1. The secondary phase of monocyte/macrophage infiltration, which is implicated in the clearance of necrotic cellular debris, deposition of matrix, and maturation of scar tissue, critically determines ultimate infarct size. Depletion of circulating monocytes/macrophages2 or selective depletion of M2-like healing macrophages3 not only exacerbates tissue injury, but also impairs cardiac function post-MI. Although there is macrophage heterogeneity within the myocardium at steady state4 and post-MI5, it is unclear which environmental cues confer the distinct polarization profile(s) observed in vivo6.
Cardiosphere-derived cell (CDC) therapy has been utilized as a strategy to treat ischemic heart disease in both small and large animals, as well as in humans. Despite limited retention within the host myocardial tissue following intracoronary or direct myocardial injection7, the beneficial effects of cell therapy are long-lasting8. Our lab has recently demonstrated that exosomes (nanosized lipid bilayer vesicles; ~30–150nm) are a potent secretory component of CDC therapy following MI9, as confirmed by Barile et al.10. These vesicles are secreted by many cell types throughout the body for local or remote cell-to-cell communication; they contain a specific payload of small RNAs (e.g., miRNA, snoRNA, Y-RNA, etc.)11 and protein (e.g., transcription factors, transmembrane receptors, integrins, etc.)12, and are readily taken up by numerous cell types including macrophages13, fibroblasts14, and endothelial cells15. Here, we extend our recent work on cellular postconditioning by CDCs16–18 by identifying CDC exosomes (CDCexo)19 as key elements of the cellular secretome which confer cardioprotection in MI. We further demonstrate that miR-181b within CDCexo is a critical mediator of macrophage polarization in vitro and cardioprotection in vivo.
METHODS
Experimental Protocol, Animals, & Surgical procedures
All studies were performed at Cedars-Sinai Medical Center in accordance with the Institutional Animal Care and Use Committee (IACUC) guidelines.
Rat
In vivo experimental protocols were performed on 7–10 week old female Wistar-Kyoto (WKY) rats (Charles River Labs, Wilmington, MA). To induce ischemia-reperfusion (IR) injury, rats were provided general anesthesia and then a thoracotomy was performed at the 4th intercostal space to expose the heart and left anterior descending (LAD) coronary artery. A 7–0 silk suture was then used to ligate the LAD, which was subsequently removed after 45 minutes to allow for reperfusion. Twenty minutes later, cells (5×105; in 100µl PBS) or exosomes (35µg, 105µg, or 350µg; in 100µL PBS) were injected into the left ventricular cavity with an aortic cross-clamp, over a period of 20 seconds.
Pig
In vivo experimental protocols were performed on adult female Yucatan mini-pigs. To induce IR injury, pigs were provided general anesthesia and then an angioplasty balloon was threaded into the mid-LAD just after the first diagonal branch and inflated for 90 minutes. Thirty minutes following reperfusion, animals were allocated to receive CDCexo (7.5mg) or placebo (Iscove’s Modified Dulbecco’s Medium, IMDM). To rule out any complications with retention of CDCexo, we performed a direct intra-myocardial injection for delivery. This procedure was performed in an open-chest model (median sternotomy) using a 25G needle. The infarcted area was identified (grey and stiff zone) and ten sequential injections (0.75mg) were administered at the perimeter of the infarct (border zone).
Cardiac Function Measurements
Rat
Transthoracic echocardiography (Vevo770, VisualSonics) was performed on rats at baseline (preischemia) and prior to euthanization (48 hours or 2 weeks). Two-dimensional short and long axes were visualized. Three representative cycles were captured for each animal/time point, and measurements for LV end-systolic dimension, LV end-diastolic dimension, and fractional area of change were obtained and averaged.
Pig
To measure LV function, left ventriculograms were performed before infusion (after MI) and prior to euthanize (48 hours) using a pigtail catheter inserted into the LV. Two incidences were studied: a 45° left anterior oblique projection and a 30° right anterior oblique projection 20° cranial projection. LVEF was determined by averaging those 2 measurements.
Infarct Size Evaluation
Rat
Two days following IR injury, hearts were arrested in diastole following intraventricular injection of 10% KCl. Hearts were then excised, washed in PBS, and cut into serial sections of ~1mm in thickness (from apex to basal edge of infarction). Sections were incubated with TTC (2,3,5-Triphenyl-2H-tetrazolium chloride, 1% solution in PBS) for 20 minutes in the dark, washed with PBS, then imaged and weighed. Infarcts were delineated from viable tissue (white versus red, respectively) and analyzed using ImageJ software as previously reported16. Infarct mass, viable mass, and LV mass were calculated by extrapolating for infarct and non-infarct volumes (based on the areas calculated from both sides of a tissue section) and weight of the tissue. Percentage infarct mass was calculated using (Infarct Mass/Viable Mass) × 100%.
Pig
Two days after infusion, animals underwent lateral thoracotomy. A catheter was inserted into the left appendage and Thioflavin T (50mL, 2%) was injected into the left atrium to assess microvascular obstruction (MVO). Then an angioplasty balloon was inflated at the same position used for the infarct and Gentian violet (50mL, 1.8%) was injected to assess area at risk (AAR). Animals were then euthanized and the heart were explanted and sectioned into 1cm thick slices. TTC staining was performed to determine infarct size (IS). IS, MVO and AAR were then determined as previously described17. AAR was reported as a percent of total LV mass and both IS and MVO were reported as percent of the AAR17.
In vitro cell culture
Bone marrow-derived Mϕ
Bone marrow was isolated as previously described16. Briefly, bones from rat (femur) or pig (rib) were sterilized with ethanol (70% in H2O) and flushed with PBS (containing 1% FBS, 2mM EDTA) and filtered through a 70µm filter. Red blood cells were lysed with ACK buffer (Invitrogen) then resuspended in IMDM (containing 10% FBS, 10ng/mL m-CSF). The resulting cell suspension was plated and grown for 7 days to obtain Mϕ. Polarization of Mϕ toward M1 (100ng/mL LPS and 50ng/mL IFNγ; Sigma-Aldrich and R&D Systems, respectively), M2 (10ng/mL IL-4 and IL-13; R&D Systems), or MCDCexo (105ug) was performed on the night between days 7 and 8 (~18hrs).
Cardiac Mϕ
Hearts were collected and processed to isolate cardiac Mϕ following ischemic injury, as described16. Briefly, 2 days following MI hearts from animals treated with Fbexo or CDCexo were perfused with PBS and collected. Cardiac tissue was minced and digested, and the resulting suspension filtered (70µm) to generate a single-cell suspension. Mononuclear cells were separated by centrifugation using a density gradient (Histopaque-1083; Sigma), and then resuspended in RPMI (supplemented with 1% FBS) and plated (37°C, 5% CO2). Two hours later, cardiac Mϕ were washed with PBS and used for downstream analyses.
Neonatal Rat Ventricular Myocytes
Cells were isolated and cultured as previously described16. Human Umbilical Vein Endothelial Cells (HUVECs). Cells were purchased (ATCC) and cultured according to the distributor’s recommendations.
Statistics for non-genomic data
Results are expressed as mean ± SEM. Linear regression analysis was performed to test the linear relationship between groups. Each replicate value was considered as an individual point to determine R2. To determine differences between groups at a single time point, data were tested using either two-tailed, unpaired, Student’s t-test or 1-way ANOVA followed by Tukey’s multiple comparisons test (outliers were excluded if Z-score was >3 or <−3). To determine differences between groups at different time points/conditions, data were tested using repeated measures ANOVA followed by Sidak’s multiple hypothesis correction, 2-way ANOVA followed by Holm-Sidak’s multiple hypothesis correction, or multiple t-tests followed by Holm-Sidak’s multiple hypothesis correction (as denoted within the figure legends). To test for linear trend, we performed a 1-way ANOVA followed by a post test for linear trend. All analyses were performed using Prism 5 software (GraphPad) and only differences with a p<0.05 were considered statistically significant.
Refer to the online-only Data Supplement for additional materials and methods.
RESULTS
Characterization of CDC exosomes
CDC exosomes (CDCexo) were isolated from serum-free media conditioned by human CDCs for 5 or 15 days in culture (Supplemental Figure IA). The conditioned media (CM) was then collected, or concentrated by ultrafiltration by centrifugation (UFC, Millipore), to assess particle number and size (Nanoparticle Tracking Analysis, Malvern Nanosight), as well as protein concentration and protein marker expression (Figure 1 and Supplemental Figure IB). The relationship between particle number and protein content was linear in CDCexo from each of two distinct human lines (Figure 1C). Concentration of exosomes by UFC reduced the suspension volume (>10-fold, Figure 1A) without a measurable loss of particles (Supplemental Figure ID). Human dermal fibroblast exosomes (Fbexo; shown to be functionally inert in vivo9) served as a negative control and were isolated and characterized in parallel, under equivalent conditions (Supplemental Figure IC). The mode particle size for CDCexo was ~150nm in diameter, expressing typical exosome markers (including CD63, Alix, and HSP70), as well as the CDC surface marker CD105. Although this size distribution biases higher than what is generally quoted for exosomes on the basis of desiccation-prepared electron microscopy (30–100 nm)20, the observed range is typical of hydrated exosomes measured by dynamic light scattering or nanoparticle tracking analysis20. For uniformity, given the elevated particle number, we performed all subsequent experiments using CDCexo isolated from 15-day conditioned media.
Figure 1. Exosome isolation and characterization.
(A), Nanoparticle tracking analysis revealing particle size distribution and concentration within the conditioned media (CM), and following ultrafiltration by centrifugation (UFC), in human CDC lines (155: 155CDCexo; 220: 220CDCexo). (B), Representative protein immunoblots of exosome (CD63, CD81, Alix, HSP70, and MHCII) and CDC (CD105) markers in human CDC exosomes. (C), Correlation between particle number and protein amount in serially diluted samples of exosomes. Graphs depict mean +/− SEM.
CDCexo administered following reperfusion in a rat model of AMI mimic the cardioprotective effects of CDCs
To examine whether CDCexo could recapitulate the cardioprotective effects observed with CDCs, we delivered CDCexo in vivo. First, we demonstrated that CDCexo were retained within the heart by pre-labeling CDCexo with DiI (Invitrogen), performing a modified version of our in vivo protocol (Figure 2A) by sacrificing rats after 2 hours of reperfusion and examining exosome distribution by fluorescence imaging (Xenogen, Supplemental Figure IIA). Second, in dose-finding studies, rats underwent 45 minutes of ischemia followed by 20 minutes of reperfusion and then received vehicle (PBS), rat allogeneic CDCs (rCDCs), rCDCs with an exosome inhibitor (rCDC + GW4869), or one of several doses of CDCexo (35µg, 105µg, or 350µg). To improve efficacy after intracoronary delivery under aortic cross clamp, exosomes were precipitated with polyethylene glycol prior to delivery (Supplemental Figure IIB). Forty-eight hours later, animals were euthanized and hearts excised for infarct size (IS) determination by TTC staining. Confirming our earlier report16, rCDCs (5×105 cells) reduced IS. Pretreatment of rCDCs with GW4869 abrogated the cardioprotective efficacy of rCDCs, consistent with the idea that exosome secretion is required for rCDC bioactivity. We further tested this idea by applying human CDCexo directly in lieu of cells. CDCexo reduced IS to a similar level as rCDCs at doses of 350µg and 105µg, but not at 35µg (Figure 2B and Supplemental Figure IIC). Based on these data we proceeded to test the potency of CDCexo versus inert Fbexo9, 11. Third, utilizing 350µg protein as our standard dose, we show that CDCexo from two human CDC lines (220 and 155) reduced IS to a similar level as rCDCs, in contrast to Fbexo and PBS controls (Figure 2, C & D). These data were further validated with 3 additional human CDCexo lines (Supplemental Figure IID). The data presented here represent findings of exosomes produced by one primary human CDC line (220, denoted herein as CDCexo); confirmatory findings using exosomes from another human line are found in the supplemental material. Finally, we tested the durability of the cardioprotective effects of CDCexo. When follow-up was extended to 2 weeks (Supplemental Figure IIIA), CDCexo still reduced IS (Supplemental Figure IIIB) and preserved cardiac function (Supplemental Figure III C–D) relative to Fbexo controls. Thus, the functional and structural benefits of CDCexo extend beyond 48 hours, as did the benefits of CDCs16, 18.
Figure 2. Intracoronary infusion of CDC exosomes (CDCexo) in rats reduces ischemic injury when delivered 20 minutes following reperfusion.
(A), Protocol schematic for infusion of exosomes and tissue harvest. (B), Quantification of infarct mass at 48 hrs: dose-responsive effects, as measured by protein concentration, of CDCexo (versus vehicle [PBS], rCDCs, and rCDC + GW4869). rCDC: allogeneic rat CDC; GW4869: exosome inhibitor. n=4–6/group. (C), Representative TTC-stained hearts from animals sacrificed 48hrs after infusion of exosomes from either of two human CDC lines (220CDCexo, 155CDCexo) or fibroblasts (Fbexo; 350µg). (D), Quantification of infarct mass in hearts from groups defined in C (n=5/group, mean +/− SEM). Statistical significance was determined using 1-way ANOVA followed by Tukey’s multiple corrections test. *P<0.05 vs. PBS and Fbexo, respectively.
Intramyocardial injection of CDCexo administered following reperfusion in a pig model of AMI mimics the cardioprotective effects of CDCs
To confirm these findings, we validated our work in the porcine model (Figure 3A). Briefly, Yucatan mini-pigs underwent 90 minutes of ischemia followed by 30 minutes of reperfusion before direct open-chest intramyocardial injection of CDCexo (7.5mg) or placebo. Cardiac function was assessed at 48 hours, with subsequent euthanasia and IS determination by TTC. After treatment with CDCexo, LVEF was better preserved (Figure 3B; Supplemental Figure IV, A & B), microvascular occlusion (MVO/AAR) was reduced, and infarct size (IS/AAR) was attenuated relative to placebo (Figure 3, C & D). Furthermore, we noted a reduction in CD68+ macrophage (Mϕ) infiltration into the infarct of CDCexo-treated versus placebo animals (Figure 3, E & F). Together, these in vivo porcine data support the hypothesis that CDCexo are key mediators of CDC-induced cardioprotection, not only in rodents but also in a clinically-relevant large-animal model.
Figure 3. Direct intramyocardial injection of CDCexo in pigs reduces ischemic injury when delivered 30 minutes following reperfusion.
(A), Schematic of protocol. (B), LV ejection fraction (LVEF), measured by echocardiography, immediately following reperfusion (baseline; post-reperfusion, but before treatment) and after 48 hrs. (C), Representative TTC-stained placebo- and CDCexo-treated pigs hearts at 48 hrs. Area-at-risk (AAR; red dotted line), microvascular occlusion (MVO; black dotted line), and infarct size (IS; white dotted line) (D), Quantification of AAR, MVO, and IS in pigs treated with placebo or CDCexo. (E) Representative immunohistochemical stain of CD68+ Mϕ within the infarct region of placebo- and CDCexo-treated pigs. Scale bar: 50µm. (F) Quantification of CD68+ Mϕ described in (E). Graphs depict mean +/− SEM (Placebo: n=5; CDCexo: n=4). Statistical significance was determined using Student’s t-test or repeated measures ANOVA followed by Sidak’s multiple corrections test. *P<0.05.
Implication of uniquely-polarized macrophages as effectors of cardioprotection
The first evidence that inflammation might be involved in cellular postconditioning came from our initial studies in rats16, where we found that CDCs modulate Mϕ toward a cardioprotective phenotype. Mimicking the effects of CDCs16, CDCexo significantly reduced CD68+ Mϕ accumulation within the infarct border zone relative to Fbexo control as measured by immunohistochemistry (Figure 4, A & B; Supplemental Figure II, E & F) and flow cytometry (Figure 4, C–E). Next, we harvested hearts from animals treated with either CDCexo or Fbexo 48 hours after MI then isolated cardiac Mϕ (Figure 4F), as described16. Expression of proinflammatory genes Nos2 and Tnf was significantly attenuated in cardiac Mϕ isolated from CDCexo-treated animals (Figure 4G).
Figure 4. CDCexo effects on Mϕ infiltration and polarization.
(A), Representative confocal images of CD68+ Mϕ within the infarct border zone 48 hrs after reperfusion. (B), Quantification of CD68+ Mϕ in (A) (n=4/group). (C & D), Representative flow cytometry plots showing the gating strategy used to determine CD45+CD11b+CD68+ cardiac Mϕ. (C), Leukocytes were gated (FSC/SSC) and dead cells removed (DAPI−), prior to identification by CD45+ and CD11b+. (D), Representative histograms of CD68+ cells from Fbexo and CDCexo-treated hearts as a result of gating according to (C). Blue line: Negative control. (E), Quantification of flow cytometry data in (D) (n=4/group). (F), Representative image of isolated cardiac Mϕ denoting CD68+ by immunohistochemistry. (G), Gene expression profiles from cardiac Mϕ isolated from infarcted hearts treated with Fbexo or CDCexo (n=3/group). Scale bar: 50µm. Graphs depict mean +/− SEM. Statistical significance was determined using Student’s t-test or multiple t-tests followed by Holm-Sidak’s multiple corrections test. *P<0.05.
In vitro, rat bone marrow (BM)-derived Mϕ exposed to CDCexo (Figure 5A) exhibit a distinctive shift in Mϕ polarization (not observed with Fbexo), including elevation in anti-inflammatory gene expression levels of Arg1, Il4ra, Tgfb1, and Vegfa, without any significant change in Tnf (Figure 5B). These effects occur dose-dependently (Figure 5C and Supplemental Table 1), are consistent among donor lines (Supplemental Figure VA), and are stable following lyophilization and reconstitution (Supplemental Figure VA) or sequential freeze-thaw cycles (Supplemental Figure V, B & C). Pig BM-derived Mϕ exposed to CDCexo exhibited a similar polarization shift (Supplemental Figure VIA). An assay of phagocytosis revealed enhanced uptake of fluorescent beads in the CDCexo-primed Mϕ group relative to M1, M2, or Fbexo-primed Mϕ (Figure 5, D & E; Supplemental Figure VIB), again recapitulating the effects of CDCs16. Together, these data support the notion that CDCexo are crucial in mediating the Mϕ responses underlying cellular postconditioning16.
Figure 5. CDCexo uniquely modulate bone marrow-derived Mϕ polarization.
(A), Schematic of in vitro assay protocol, including bone marrow (BM) harvest, Mϕ differentiation, and treatment. (B), Gene expression changes in response to Fbexo and CDCexo. (C), Dose-response gene expression changes in BM-derived Mϕ 18hrs after exposure to CDCexo. (D), Representative flow cytometry plots and quantification of fluorescent microsphere bead uptake by BM-derived Mϕ following polarization with M1 (LPS & IFNγ), M2 (IL-4 & IL-13), Fbexo, and CDCexo. Blue line: Negative control. (E), Quantification of flow cytometry data in (D). Graphs depict mean +/− SEM with n=3–5/group. Statistical significance was determined using 2-way ANOVA followed by Holm-Sidak’s multiple corrections test, 1-way ANOVA followed by Tukey’s multiple corrections test, or multiple t-tests followed by Holm-Sidak’s multiple corrections test. *P<0.05.
CDCexo have a distinct miRNA signature
Exosomes are intriguing as signaling intermediates with cargo rich in small RNA molecules11, 12. The composition of exosomes is not determined randomly, but rather uniquely regulated by cell source and environmental stressors21. To understand how CDCexo confer Mϕ polarization we performed total RNA-sequencing (RNA-seq) on exosomes isolated from CDCs (CDCexo) and Fbs (Fbexo), then aligned them to a database of both short and long RNA annotations. To identify the most biologically-relevant changes, we performed one set of biological replicates using the Illumina RNA-seq platform and a second using the Ion Torrent platform (Figure 6)22, 23. Globally, the number of reads (and thus the RNA content) mapping to different classes of RNAs were similar between CDCexo and Fbexo; the greatest number of reads mapped to protein coding genes (RefSeq) and tRNAs, while fewer reads mapped to short RNAs (e.g., miRNAs and piRNAs; even fewer mapped to scaRNAs and snoRNAs) (Supplemental Figure VII). However, the largest and most consistent proportional change was observed in the miRNA class, where CDCexo produced 5- and 8-fold greater reads than Fbexo in the Illumina and Ion Torrent libraries, respectively (Figure 6, A & B). Next, we focused on the reads that aligned to miRNA annotations and normalized both sets of libraries using DESeq 2 to identify the miRNAs with consistent changes in expression between CDCexo and Fbexo (Figure 6C). While the two platforms (Illumina and Ion Torrent) had technical differences (Pearson correlation coefficient ~0.6), several miRNAs stood out as having significant differential expression. The miRNAs that were uniquely enriched in Fbexo included miR-199, miR-3676, and miR-3648, while those uniquely enriched in CDCexo included miR-126 and miR-181b; in addition, miR-181a was enriched just below the threshold of significance in CDCexo. Interestingly, miR-181a/b stood out as having large numbers of differentially expressed target genes in CDCexo-treated Mϕ (marked as red in Figure 6C, and described in detail below).
Figure 6. RNA-sequencing analysis of CDCexo and CDCexo-primed Mϕ identify miR-181 as the top miRNA exosome candidate.
(A), Total RNA-sequencing analysis depicting the change between Fbexo and CDCexo in the number of total RNA-seq reads for different RNA classes, shown as a fold-change ratio of Fbexo/CDCexo. RNA categories with a total of less than 1,000 reads aligned to all transcripts are omitted. The libraries sequenced using Illumina (blue) and Ion Torrent (red) sequencing platforms are shown separately, and the RNA classes are ordered by the average fold-change across both platforms. (B), Raw read counts for each of the four samples for the miRNA class (the other classes are shown in Supplemental Figure VII). (C), Volcano plot showing relative expression changes of 677 miRNAs between Fbexo and CDCexo, with normalization and significance testing performed by DEseq2, using the Illumina libraries as one replicate and the Ion Torrent libraries as a second replicate (statistical significance reflects the concordance between the two platforms). Dashed horizontal line: uncorrected p-value of 0.05; Filled points: miRNAs having predicted target genes present in the miRecords database, and further analyzed in D–H; Filled red points and red labels: miRNAs with targets downregulated in CDCexo-polarized Mϕ (detailed in D). (D), The 7 miRNAs with the most significant downregulation of predicted targets in CDCexo-polarized Mϕ, ordered by significance. Top panel: fold-change for all predicted miRNA gene targets; Bottom panel: statistical significance (hypergeometric test, see Supplemental Methods) of the association between predicted miRNA targets and downregulated genes (−log10 p-values); Annotations at the bottom: miRNAs with overexpression in CDCexo compared to Fbexo, from C. (E), Comparison of untreated Mϕ (rn-control) and CDCexo-primed Mϕ (rn-MCDCexo) gene expression to published datasets25, 26. Only genes with homology between rat (rn), human, and mouse (mm) are shown, while those with undetectably low expression in all our Mϕ samples are filtered out. Genes are sorted by differential expression in rn-MCDCexo, from the most downregulated (left, blue) to the most upregulated (right, red). Gene expression values are shown as relative (log-transformed) fold change, relative to other samples in the same study. (F), Venn diagrams highlight the highly significant associations between genes sets of rn-MCDCexo to published miR-181a/b KO (miR-181 KO) thymocytes27; significance values were calculated using a hypergeometric test (see Supplemental Methods). Downregulated genes in our model (+CDCexo) were associated with genes upregulated in miR-181 KO thymocytes (upper left quadrant, p=2.6E-147), and vice-versa (lower right quadrant, p=5.2E-79). These findings were not true for genes regulated in the same direction (upper right and lower left quadrants). (G), Predicted miR-181 target genes from miRecords are shown in the same order as (E), and are concentrated in those genes downregulated rn-MCDCexo and upregulated miR-181 KO thymocytes. (H) Venn diagrams show the strong association of predicted miR-181 targets and those genes downregulated in rn-MCDCexo (p=1.3E-21). Significance values were calculated using a hypergeometric test (see Supplemental Methods). Refer to the Supplemental Methods for sample size and additional methods.
miR-181 as a candidate effector of CDCexo-mediated Mϕ polarization
To investigate the effect of CDCexo miRNAs on Mϕ, we performed RNA-seq on two biological replicates of unprimed (control) or CDCexo-primed (CDCexo) rat BM-derived Mϕ. Then, we used gene annotations from the NextBio functional annotation database (Illumina Inc.)24 to investigate the functions of genes with altered expression following exposure to CDCexo. Specifically, we compared differentially-regulated genes to predicted miRNA targets from the miRecords database (reverse pathway analysis25, 26), and found that target genes for several miRNAs enriched in CDCexo were predominantly downregulated in CDCexo-treated Mϕ (Figure 6D). This downregulation of miRNA target genes was not observed for randomly permuted gene sets of the same size (Supplemental Figure VIIIA). The fact that miRNAs with proportionally higher expression in Fbexo (e.g., miR-30 and let-7) were also downregulated in CDCexo-primed Mϕ may reflect the higher absolute levels of these miRNAs in CDCexo (Figure 6A). Consistent with this interpretation, we found a significant downregulation of predicted targets for all highly-expressed miRNAs examined, whether they had proportionally higher expression in CDCexo or Fbexo (Supplemental Figure VIIIB). However, the three most significant miRNAs (miR-181, miR-27, and miR-19) were all proportionally higher in CDCexo, and thus represented strong candidates for conferring the functional changes observed in Mϕ (Figure 6D). miR-181 also had > 10-fold higher proportional abundance in CDCexo vs. Fbexo (Figure 6C), compared to miR-19 and miR-27 (both had < 2-fold higher abundance). These data pointed to the miR-181 family as a strong candidate for the key regulatory cargo contained in CDCexo.
To further explore transcriptional changes in CDCexo-treated Mϕ, we performed an unbiased comparison of our RNA-seq dataset with two published rodent immune cell RNA-seq datasets (Figure 6E). First, we compared expression changes in our CDCexo-treated cells to a mouse Mϕ RNA-seq dataset23. This revealed a similar pattern of gene expression changes, with our unprimed rat Mϕ resembling the mouse M0 state, and CDCexo-primed rat Mϕ resembling the mouse M2 state (Figure 6E). Since complete concordance was not observed between the two datasets, these findings support the notion16that CDCexo-primed Mϕ are not ordinary M2 Mϕ, but lie somewhere within (or outside) the M1-M2 spectrum. We next compared a mouse miR-181a/b knockout (KO) thymocyte dataset22 with CDCexo-primed Mϕ (Figure 6, E & F). This particular perturbation came up as the most strongly related among thousands of gene perturbation datasets within the NextBio database, and our results demonstrated a very strong degree of concordance (Figure 6E). Overlapping the genes with statistically significant expression changes across two replicates revealed that those downregulated in our model (+CDCexo) were significantly associated (p=2.6E-147) with genes upregulated in miR-181a/b KO thymocytes (Figure 6F, upper left quadrant), and vice-versa (Figure 6F, lower right quadrant, p=5.2E-79). Further, when grouped based on their Gene Ontology (GO) annotation, these genes clustered in biological processes associated with increased translation, metabolism, and oxidative phosphorylation (Supplemental Table 2). Genes downregulated in our model had no statistical association with genes downregulated in miR-181a/b KO thymocytes (Figure 6F, lower left quadrant), nor did the converse (Figure 6F, upper right quadrant). This association is even stronger than the association with predicted miR-181 targets (p=1.3E-21, Figure 6, G & H), further supporting the notion that miR-181 may be a critical factor regulating CDCexo function, and by far the most promising single candidate mediator of the transcriptomic changes observed in CDCexo-primed Mϕ.
miR-181b in exosomes confers cardioprotection
The miR-181 family comprises four known members (miR-181a, −181b, −181c, and −181d), which have been implicated as modulators of myeloid leukemia and stem cell differentiation27. To better understand the functional effects of miR-181, we assessed the gene expression pattern of miR-181 family members in CDCs and CDCexo (Figure 7A; miR-181c and miR-181d were undetectable by qPCR and thus not presented). Levels of miR-181a, but not miR-181b, were higher in human CDCs relative to Fbs (Figure 7A). CDCexo, on the other hand, were highly enriched for miR-181b, but not miR-181a, indicative of highly selective transfer of miRNA cargo into CDCexo (Figure 7A and Supplemental Figure IXA), as has been seen with other miRNAs in other cell types28. The expression levels of miR-181a and miR-181b differed in various isolated cardiac cell types (e.g., cardiomyocytes, endothelial cells, Mϕ; Supplemental Figure IXB) and were not changed within the hearts of untreated animals prior to or following ischemic injury (Figure 7C).
Figure 7. Exosomal transfer of miR-181b reduces infarct size following IR injury.
(A), Human CDCs have elevated expression of miR-181a (but not miR-181b), while CDCexo have enriched miR-181b (but not miR-181a) relative to Fb and Fbexo, respectively (n=3/group). (B), Representative TTC stained hearts 48hrs following IR injury. Fbexo(Scramble): Fbexo with miR-181b scramble; Fbexo(Mimic): Fbexo with miR-181b mimic; CDCexo(Scramble): CDCexo with miR-181b scramble; CDCexo(Antagomir): CDCexo with miR-181b antagomir. (C), Expression of miR-181a and miR-181b in tissue from non-infarcted and infarcted rats, normalized to liver. LV: left ventricle; RV: right ventricle; BZ: border zone; IZ: infarct zone; NZ: normal zone (n=4/group). (D), Quantification of percent infarct mass in (C) (n=4–8/group). (E), Quantification of CD68+ Mϕ in (Supplemental Figure XI). Graphs depict mean +/− SEM. Statistical significance was determined using 2-way ANOVA followed by Holm-Sidak’s multiple corrections tests or 1-way ANOVA followed by Tukey’s multiple corrections test. *P<0.05.
We tested the effects of miR-181b on various cardiac cell types, including endothelial cells and Mϕ, in vitro. Consistent with its known targets29, treatment with miR-181b, but not miR-Scramble, reduced transcript levels for E-selectin, VCAM-1, and ICAM-1 in endothelial cells (Supplemental Figure XA). Interestingly, these effects were reproduced in a similar manner following CDCexo or Fbexo with miR-181b, but not Fbexo, treatment (Supplemental Figure XB). Next, we performed a scratch assay to examine whether miR-181b modulates endothelial migration. While CDCexo enhanced scratch closure in vitro, miR-181b alone had no effect (Supplemental Figure X, C & D).
To further validate the functional efficacy of exosomal miR-181b in vivo, we overexpressed miR-181b in Fbexo (naturally low in miR-181b) or antagonized miR-181b in CDCexo and measured infarct size in our rat model (Figure 2A). Briefly, Fbexo were transfected with miR-181b mimic (Fbexo(Mimic)) (Supplemental Figure IXC) and CDCexo were transfected with an antagomir to miR-181b (CDCexo(Antagomir)); miR-scramble was used as a transfection control in both types of exosomes (Fbexo(Scramble) and CDCexo(Scramble)). After 48 hours of reperfusion, Fbexo(Mimic) and CDCexo(Scramble) reduced infarct size and Mϕ infiltration, although not to the same extent as CDCexo, relative to Fbexo, Fbexo(Scramble), and CDCexo(Antagomir) (Figure 7, B, D, & E; Supplemental Figure XI). Thus, miR-181b reproduces the cardioprotective effects of CDCexo, while blocking miR-181b blunts those effects.
miR-181b inhibits the expression of protein kinase C δ (PKCδ)
To better understand how miR-181b may lead to cardioprotection, we honed in on the target genes for miR-181b within CDCexo-treated Mϕ. Among known miR-181b targets, protein kinase C δ (PKCδ; prkcd) was one of the most significantly downregulated (4.82E-30) (Figure 7F). Consistent with previous reports27, expression levels of prkcd were reduced in Mϕ transfected with miR-181b compared to miR-scramble control (Figure 8A). PKCδ enhances inflammatory gene expression (e.g., TNFα)30, as well as cell differentiation, growth, and death31, and PKCδ inhibitors are known to induce, or enhance, cardioprotection32, 33. We thus hypothesized that PKCδ acts as a downstream effector of CDCexo-mediated cardioprotection, via miR-181b–induced suppression of prkcd in Mϕ. To test this hypothesis, we transfected Mϕ with a specific small interfering RNA (siRNA) which effectively suppressed prkcd (Figure 8B). Given our previous demonstration that adoptive transfer of CDC-conditioned Mϕ mimicked cardioprotection by CDCs, we modified our rat model of IR (Figure 2A) to adoptively transfer Mϕ transfected with either PKCδ siRNA (Mϕ(PKCδ siRNA)) or scramble siRNA (Mϕ(scramble siRNA)). Adoptive transfer of Mϕ(PKCδ siRNA) reduced infarct size (Figure 8, C & D), mimicking the effects of CDCexo (Figure 2 & Figure 7). Thus, our data as well as published work are consistent with the hypothesis that PKCδ inhibition by miR-181b in CDCexo underlies the cardioprotection induced by CDCs (Figure 8E).
Figure 8. Adoptive transfer of Mϕ with reduced levels of PKCδ (prkcd) is cardioprotective.
(A), Mϕ treated with miR-181b mimic have reduced prkcd expression (n=3–4/group). (B), Mϕ treated with PKCδ siRNA (10nM) have reduced prkcd expression (n=3/group). (C), Representative images of TTC stained hearts 48hrs following IR injury and adoptive transfer of Mϕ as indicated: Mϕ(PKCδ siRNA): Mϕ pre-treated with PKCδ siRNA; Mϕ(Scramble): Mϕ pre-treated with scramble siRNA. (D), Quantification of percent infarct mass in (C) (n=5/group). (E), Schematic of our working hypothesis: (i), CDCs secrete exosomes (CDCexo) that are cardioprotective (evidence in Figures 1–3); (ii), CDCexo, rich in miR-181b, alter the polarization state of Mϕ (Figures 4–6); (iii), enrichment of miR-181b within exosomes recapitulates cardioprotection (Figure 7); (iv), downregulation of PKCδ in Mϕ by miR-181b is responsible, at least in part, for cardioprotective response of CDCexo (Figure 8). Graphs depict mean +/− SEM. Statistical significance was determined using Student’s t-test. *P<0.05.
Despite the compelling evidence pinpointing PKCδ as a likely downstream effector, it is clear that exosomes and their miRNAs have protean effects, which may be synergistic; picking a single candidate may oversimplify the actual biology. Supplemental Table 3 lists other miR-181b targets that have been implicated in Mϕ polarization, and which were downregulated in CDCexo-primed Mϕ. Conversely, we observed upregulation (and downregulation in miR-181a/b KO thymocytes; Figure 6F, lower right quadrant) in several genes associated with Mϕ polarization, oxidative phosphorylation, and phagocytosis (Supplemental Table 4). Together, these data point to a dramatic shift in Mϕ polarization by CDCexo that reduces proinflammatory signaling and enhances phagocytosis to promote a cardioprotective response in vivo.
DISCUSSION
Myocardial IR injury leads to necrosis and apoptosis of the underlying myocardium (myocytes, endothelial cells, etc.)34. The acute inflammatory response that follows IR injury is a physiologically archetypal sequence of events that begins with the mobilization and infiltration of neutrophils and follows with Mϕ influx35. Several studies have highlighted the importance of Mϕ in models of myocardial tissue injury, including Mϕ depletion studies in MI2, 16, 36, regenerative models of MI4, or apical myocardial resection37. It is also becoming clearer that Mϕ heterogeneity (differences in polarization state in a spectrum typically defined by M1 and M2 states at the extremes38) is a nuanced process that is highly dependent upon microenvironmental cues (protein-dependent38, extracellular vesicles39, or pH/ion imbalance40).
Over the past decade, cell therapy has demonstrated some success in the treatment of ischemic disease, despite limited cell retention41. Our group has recently demonstrated in pigs17 and rats16 the existence of a novel cardioprotective process known as cellular postconditioning: CDCs delivered within 30 minutes of reperfusion dramatically reduce infarct size. Macrophages are required for cellular postconditioning, and appear to work by adopting a unique highly-phagocytic polarization state (non-M1, non-M2) in response to CDCs16. We have also shown that CDC exosomes (CDCexo) increase viable mass, promote angiogenesis, and reduce scar size in established MI9, 14.
Exosomes are intriguing paracrine signals that can shuttle payloads from cell to cell12. Not surprisingly, exosomes are extremely diverse in their contents and vary greatly among cell types. Their characteristic traits are reflected in their vesicular composition (lipid, protein, small RNA, etc.), heterogeneity (totality of loaded and unloaded vesicles), and concentration (number of particles secreted) in vivo and in vitro21, which are further modulated in time and space by environmental cues42. Our lab9, 14 and at least 2 others10, 43 have demonstrated the bioactivity of exosomes derived from cardiac stem cell populations.
Here, we demonstrate that CDCexo are a unique population of exosomes that have a distinct protein expression pattern (CD63, HSP70, and CD105) and miR profile (including high expression of miR-181b). In contrast to exosomes secreted by cardiac fibroblasts, CDCexo do not exacerbate tissue injury44; instead, they confer cardioprotection when delivered following reperfusion by localizing to the ischemic tissue, reducing infarct size, reducing the total number of CD68+ Mϕ, and shifting the polarization state of Mϕ toward a distinct phenotype. Interestingly, CDCexo appear to specifically home to the site of injury within infarcted myocardium. Localization to this area of interest may confer a local restructuring of the microenvironment and polarize recruited monocytes/Mϕ toward a reparative phenotype. Although the mechanisms for CDCexo localization remain to be defined, homing signals, such as angiotensin II type I receptor (targets exosomes to cardiomyocytes, skeletal muscle, and mesenteric resistance arterioles)45, may be responsible for exosome retention at the site of injury. Furthermore, CDCexo enhance the endogenous phagocytic capacity of Mϕ, a characteristic trait that may enhance clearance of necrotic cell debris and alleviate excessive proinflammatory stress within the infarcted heart35 (a salutary process known as efferocytosis46).
This work further expands our understanding of how CDCs confer cardioprotection and Mϕ polarization16. By reverse pathway analysis on transcriptomic data from both CDCexo (cargo) and CDCexo-primed Mϕ (recipient), we were able to pinpoint one key RNA component through which CDCexo confer these functional benefits. Probing our own sequencing data against publicly-available gene expression and miRNA target prediction databases, we identified miR-181b as our top candidate; this miRNA species displayed the highest relative abundance within CDCexo and the strongest correlation in its downregulation of target genes in CDCexo-primed Mϕ. miR-181b has been implicated in the attenuation of NF-κB signaling in endothelial cells29, 47 and suppression of myeloid differentiation in acute myeloid leukemia27, consistent with our findings that CDCexo-primed Mϕ reduce proinflammatory gene expression and polarize Mϕ away from an M1 phenotype. Furthermore, the observed suppression of the miR-181b target PKCδ and concomitant increases in genes involved in translation and oxidative phosphorylation hint that CDCexo shift the metabolic profile of Mϕ48 toward a cardioprotective phenotype. Adding a miR-181b mimic to otherwise-inert Fbexo qualitatively reproduces CDCexo cardioprotection, while blocking miR-181b in CDCexo blunts cardioprotection. Although the evidence points to the functional efficacy of miR-181b in reducing infarct size and Mϕ polarization, the lack of complete congruence with our CDCexo data is not surprising. CDCexo contain a multitude of small RNAs and proteins, not only miR-181b. Many of the other components undoubtedly are bioactive, exerting the overall functional benefits as an ensemble.
Our data provide compelling evidence for the role of CDCexo in polarizing Mϕ to a phenotype that improves the healing response following injury. Specifically, we demonstrate that miR-181b is a key component within CDCexo that confers cardioprotection by targeting PKCδ within Mϕ to induce a distinctive polarization state. These data add to the list of cardioprotective/cardioregenerative properties previously attributed to CDCexo (anti-fibrotic effects; reduction of cardiomyocyte death and hypertrophy; myocardial regeneration; improved cardiac function; and angiogenesis)9, 11, 14. Although we define here miR-181b as one important component within CDCexo, the remaining composition (other noncoding RNAs, as well as proteins and lipids) of CDCexo requires further examination49 to unravel the details of synthesis, release, homing, pathway regulation, and cellular uptake of CDCexo in vitro and in vivo. Likewise, while we have implicated PKCδ as one downstream contributor to CDCexo-mediated cardioprotection, other plausible effectors remain to be investigated. Such redundancy would be consistent with the notion that exosomes contain a plethora of bioactive molecules which can target multiple signaling pathways synergistically50.
Supplementary Material
CLINICAL PERSPECTIVE.
What Is New?
Here we discover that exosomes mediate the cardioprotective effects of cardiosphere-derived cells (CDCs) administered after reperfusion in rats with acute myocardial infarction.
Treatment with either CDCs or their secreted exosomes reduces infarct size and improves functional recovery.
A specific microRNA species within CDC-derived exosomes, acting on macrophages, is implicated as a key mediator of the cardioprotective benefits.
What Are the Clinical Implications?
Allogeneic CDCs are already in advanced clinical testing for reduction of chronic scar when administered months after myocardial infarction (ALLSTAR trial; ClinicalTrials.gov Identifier NCT01458405).
The present findings give new reason to test the idea that allogeneic CDCs may be efficacious in preventing scar formation and improving cardiac function when given in the early reperfusion period adjunctive to percutaneous coronary intervention.
Acknowledgments
We thank Jackie Valle, Elijah Kravets, Carolina Castillo, Baiming Sun, Antonio Echavez, and Xiaoyu Da for technical assistance.
Sources of Funding
This work was supported by the Board of Governors of the Cedars-Sinai Medical Center. General laboratory support was provided by grants to EM from the National Institutes of Health (R01HL124074) and the Department of Defense (CSR205330).
Footnotes
Disclosures
EM owns founder’s equity in, and serves as unpaid advisor to, Capricor Inc. GdC is a paid consultant for Capricor. Capricor neither provided funding for this work nor did the company have approval rights over the manuscript. The other authors declare no competing financial interests.
References
- 1.Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007;204:3037–3047. doi: 10.1084/jem.20070885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH, van Luyn MJ. Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice. Am J Pathol. 2007;170:818–829. doi: 10.2353/ajpath.2007.060547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ma Y, Halade GV, Zhang J, Ramirez TA, Levin D, Voorhees A, Jin YF, Han HC, Manicone AM, Lindsey ML. Matrix metalloproteinase-28 deletion exacerbates cardiac dysfunction and rupture after myocardial infarction in mice by inhibiting M2 macrophage activation. Circ Res. 2013;112:675–688. doi: 10.1161/CIRCRESAHA.111.300502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Aurora AB, Porrello ER, Tan W, Mahmoud AI, Hill JA, Bassel-Duby R, Sadek HA, Olson EN. Macrophages are required for neonatal heart regeneration. J Clin Invest. 2014;124:1382–1392. doi: 10.1172/JCI72181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Nahrendorf M, Swirski FK. Monocyte and macrophage heterogeneity in the heart. Circ Res. 2013;112:1624–1633. doi: 10.1161/CIRCRESAHA.113.300890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Okabe Y, Medzhitov R. Tissue-specific signals control reversible program of localization and functional polarization of macrophages. Cell. 2014;157:832–844. doi: 10.1016/j.cell.2014.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Terrovitis JV, Smith RR, Marban E. Assessment and optimization of cell engraftment after transplantation into the heart. Circ Res. 2010;106:479–494. doi: 10.1161/CIRCRESAHA.109.208991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Malliaras K, Ibrahim A, Tseliou E, Liu W, Sun B, Middleton RC, Seinfeld J, Wang L, Sharifi BG, Marban E. Stimulation of endogenous cardioblasts by exogenous cell therapy after myocardial infarction. EMBO Mol Med. 2014;6:760–777. doi: 10.1002/emmm.201303626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ibrahim AGE, Cheng K, Marbán E. Exosomes as Critical Agents of Cardiac Regeneration Triggered by Cell Therapy. Stem Cell Reports. 2014;2:606–619. doi: 10.1016/j.stemcr.2014.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Barile L, Lionetti V, Cervio E, Matteucci M, Gherghiceanu M, Popescu LM, Torre T, Siclari F, Moccetti T, Vassalli G. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res. 2014;103:530–541. doi: 10.1093/cvr/cvu167. [DOI] [PubMed] [Google Scholar]
- 11.Cambier L, de Couto G, Ibrahim A, Echavez AK, Valle J, Liu W, Kreke M, Smith RR, Marban L, Marban E. Y RNA fragment in extracellular vesicles confers cardioprotection via modulation of IL-10 expression and secretion. EMBO Mol Med. 2017;9:337–352. doi: 10.15252/emmm.201606924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–579. doi: 10.1038/nri855. [DOI] [PubMed] [Google Scholar]
- 13.Imai T, Takahashi Y, Nishikawa M, Kato K, Morishita M, Yamashita T, Matsumoto A, Charoenviriyakul C, Takakura Y. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J Extracell Vesicles. 2015;4:26238. doi: 10.3402/jev.v4.26238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tseliou E, Fouad J, Reich H, Slipczuk L, de Couto G, Aminzadeh M, Middleton R, Valle J, Weixin L, Marban E. Fibroblasts Rendered Antifibrotic, Antiapoptotic, and Angiogenic by Priming With Cardiosphere-Derived Extracellular Membrane Vesicles. J Am Coll Cardiol. 2015;66:599–611. doi: 10.1016/j.jacc.2015.05.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Paggetti J, Haderk F, Seiffert M, Janji B, Distler U, Ammerlaan W, Kim YJ, Adam J, Lichter P, Solary E, Berchem G, Moussay E. Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts. Blood. 2015;126:1106–1117. doi: 10.1182/blood-2014-12-618025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.de Couto G, Liu W, Tseliou E, Sun B, Makkar N, Kanazawa H, Arditi M, Marban E. Macrophages mediate cardioprotective cellular postconditioning in acute myocardial infarction. J Clin Invest. 2015;125:3147–3162. doi: 10.1172/JCI81321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kanazawa H, Tseliou E, Malliaras K, Yee K, Dawkins JF, De Couto G, Smith RR, Kreke M, Seinfeld J, Middleton RC, Gallet R, Cheng K, Luthringer D, Valle I, Chowdhury S, Fukuda K, Makkar RR, Marban L, Marban E. Cellular Post-Conditioning: Allogeneic Cardiosphere-Derived Cells Reduce Infarct Size and Attenuate Microvascular Obstruction When Administered After Reperfusion in Pigs With Acute Myocardial Infarction. Circ Heart Fail. 2015;8:322–332. doi: 10.1161/CIRCHEARTFAILURE.114.001484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kanazawa H, Tseliou E, Dawkins JF, De Couto G, Gallet R, Malliaras K, Yee K, Kreke M, Valle I, Smith RR, Middleton RC, Ho CS, Dharmakumar R, Li D, Makkar RR, Fukuda K, Marban L, Marban E. Durable Benefits of Cellular Postconditioning: Long-Term Effects of Allogeneic Cardiosphere-Derived Cells Infused After Reperfusion in Pigs with Acute Myocardial Infarction. J Am Heart Assoc. 2016;5:e002796. doi: 10.1161/JAHA.115.002796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gallet R, Dawkins J, Valle J, Simsolo E, de Couto G, Middleton R, Tseliou E, Luthringer D, Kreke M, Smith RR, Marban L, Ghaleh B, Marban E. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2016:ehw240. doi: 10.1093/eurheartj/ehw240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chernyshev VS, Rachamadugu R, Tseng YH, Belnap DM, Jia Y, Branch KJ, Butterfield AE, Pease LF, 3rd, Bernard PS, Skliar M. Size and shape characterization of hydrated and desiccated exosomes. Anal Bioanal Chem. 2015;407:3285–3301. doi: 10.1007/s00216-015-8535-3. [DOI] [PubMed] [Google Scholar]
- 21.Yanez-Mo M, Siljander PR, Andreu Z, Zavec AB, Borras FE, Buzas EI, Buzas K, Casal E, Cappello F, Carvalho J, Colas E, Cordeiro-da Silva A, Fais S, Falcon-Perez JM, Ghobrial IM, Giebel B, Gimona M, Graner M, Gursel I, Gursel M, Heegaard NH, Hendrix A, Kierulf P, Kokubun K, Kosanovic M, Kralj-Iglic V, Kramer-Albers EM, Laitinen S, Lasser C, Lener T, Ligeti E, Line A, Lipps G, Llorente A, Lotvall J, Mancek-Keber M, Marcilla A, Mittelbrunn M, Nazarenko I, Nolte-’t Hoen EN, Nyman TA, O’Driscoll L, Olivan M, Oliveira C, Pallinger E, Del Portillo HA, Reventos J, Rigau M, Rohde E, Sammar M, Sanchez-Madrid F, Santarem N, Schallmoser K, Ostenfeld MS, Stoorvogel W, Stukelj R, Van der Grein SG, Vasconcelos MH, Wauben MH, De Wever O. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066. doi: 10.3402/jev.v4.27066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Henao-Mejia J, Williams A, Goff LA, Staron M, Licona-Limon P, Kaech SM, Nakayama M, Rinn JL, Flavell RA. The microRNA miR-181 is a critical cellular metabolic rheostat essential for NKT cell ontogenesis and lymphocyte development and homeostasis. Immunity. 2013;38:984–997. doi: 10.1016/j.immuni.2013.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Martinez FO, Helming L, Milde R, Varin A, Melgert BN, Draijer C, Thomas B, Fabbri M, Crawshaw A, Ho LP, Ten Hacken NH, Cobos Jimenez V, Kootstra NA, Hamann J, Greaves DR, Locati M, Mantovani A, Gordon S. Genetic programs expressed in resting and IL-4 alternatively activated mouse and human macrophages: similarities and differences. Blood. 2013;121:e57–e69. doi: 10.1182/blood-2012-06-436212. [DOI] [PubMed] [Google Scholar]
- 24.Kupershmidt I, Su QJ, Grewal A, Sundaresh S, Halperin I, Flynn J, Shekar M, Wang H, Park J, Cui W, Wall GD, Wisotzkey R, Alag S, Akhtari S, Ronaghi M. Ontology-based meta-analysis of global collections of high-throughput public data. PLoS One. 2010;5:e13066. doi: 10.1371/journal.pone.0013066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Arora A, Simpson DA. Individual mRNA expression profiles reveal the effects of specific microRNAs. Genome Biol. 2008;9:R82. doi: 10.1186/gb-2008-9-5-r82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Xiao F, Zuo Z, Cai G, Kang S, Gao X, Li T. miRecords: an integrated resource for microRNA-target interactions. Nucleic Acids Res. 2009;37:D105–D110. doi: 10.1093/nar/gkn851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Su R, Lin HS, Zhang XH, Yin XL, Ning HM, Liu B, Zhai PF, Gong JN, Shen C, Song L, Chen J, Wang F, Zhao HL, Ma YN, Yu J, Zhang JW. MiR-181 family: regulators of myeloid differentiation and acute myeloid leukemia as well as potential therapeutic targets. Oncogene. 2015;34:3226–3239. doi: 10.1038/onc.2014.274. [DOI] [PubMed] [Google Scholar]
- 28.Mittelbrunn M, Gutierrez-Vazquez C, Villarroya-Beltri C, Gonzalez S, Sanchez-Cabo F, Gonzalez MA, Bernad A, Sanchez-Madrid F. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun. 2011;2:282. doi: 10.1038/ncomms1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sun X, Icli B, Wara AK, Belkin N, He S, Kobzik L, Hunninghake GM, Vera MP, Registry M, Blackwell TS, Baron RM, Feinberg MW. MicroRNA-181b regulates NF-kappaB-mediated vascular inflammation. J Clin Invest. 2012;122:1973–1990. doi: 10.1172/JCI61495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kontny E, Kurowska M, Szczepanska K, Maslinski W. Rottlerin, a PKC isozyme-selective inhibitor, affects signaling events and cytokine production in human monocytes. J Leukoc Biol. 2000;67:249–258. doi: 10.1002/jlb.67.2.249. [DOI] [PubMed] [Google Scholar]
- 31.Duquesnes N, Lezoualc’h F, Crozatier B. PKC-delta and PKC-epsilon: foes of the same family or strangers? J Mol Cell Cardiol. 2011;51:665–673. doi: 10.1016/j.yjmcc.2011.07.013. [DOI] [PubMed] [Google Scholar]
- 32.Sivaraman V, Hausenloy DJ, Kolvekar S, Hayward M, Yap J, Lawrence D, Di Salvo C, Yellon DM. The divergent roles of protein kinase C epsilon and delta in simulated ischaemia-reperfusion injury in human myocardium. J Mol Cell Cardiol. 2009;46:758–764. doi: 10.1016/j.yjmcc.2009.02.013. [DOI] [PubMed] [Google Scholar]
- 33.Yogalingam G, Hwang S, Ferreira JC, Mochly-Rosen D. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) phosphorylation by protein kinase Cdelta (PKCdelta) inhibits mitochondria elimination by lysosomal-like structures following ischemia and reoxygenation-induced injury. J Biol Chem. 2013;288:18947–18960. doi: 10.1074/jbc.M113.466870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Reimer KA, Lowe JE, Rasmussen MM, Jennings RB. The wavefront phenomenon of ischemic cell death 1 Myocardial infarct size vs duration of coronary occlusion in dogs. Circulation. 1977;56:786–794. doi: 10.1161/01.cir.56.5.786. [DOI] [PubMed] [Google Scholar]
- 35.Frangogiannis NG. The inflammatory response in myocardial injury, repair, and remodelling. Nat Rev Cardiol. 2014;11:255–265. doi: 10.1038/nrcardio.2014.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Frantz S, Hofmann U, Fraccarollo D, Schafer A, Kranepuhl S, Hagedorn I, Nieswandt B, Nahrendorf M, Wagner H, Bayer B, Pachel C, Schon MP, Kneitz S, Bobinger T, Weidemann F, Ertl G, Bauersachs J. Monocytes/macrophages prevent healing defects and left ventricular thrombus formation after myocardial infarction. FASEB J. 2013;27:871–881. doi: 10.1096/fj.12-214049. [DOI] [PubMed] [Google Scholar]
- 37.Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN, Sadek HA. Transient regenerative potential of the neonatal mouse heart. Science. 2011;331:1078–1080. doi: 10.1126/science.1200708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol. 2011;11:750–761. doi: 10.1038/nri3088. [DOI] [PubMed] [Google Scholar]
- 39.Ismail N, Wang Y, Dakhlallah D, Moldovan L, Agarwal K, Batte K, Shah P, Wisler J, Eubank TD, Tridandapani S, Paulaitis ME, Piper MG, Marsh CB. Macrophage microvesicles induce macrophage differentiation and miR-223 transfer. Blood. 2013;121:984–995. doi: 10.1182/blood-2011-08-374793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, Cline GW, Phillips AJ, Medzhitov R. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014;513:559–563. doi: 10.1038/nature13490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jansen Of Lorkeers SJ, Eding JE, Vesterinen HM, van der Spoel TI, Sena ES, Duckers HJ, Doevendans PA, Macleod MR, Chamuleau SA. Similar effect of autologous and allogeneic cell therapy for ischemic heart disease: systematic review and meta-analysis of large animal studies. Circ Res. 2015;116:80–86. doi: 10.1161/CIRCRESAHA.116.304872. [DOI] [PubMed] [Google Scholar]
- 42.Simons M, Raposo G. Exosomes--vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21:575–581. doi: 10.1016/j.ceb.2009.03.007. [DOI] [PubMed] [Google Scholar]
- 43.Gray WD, French KM, Ghosh-Choudhary S, Maxwell JT, Brown ME, Platt MO, Searles CD, Davis ME. Identification of therapeutic covariant microRNA clusters in hypoxia-treated cardiac progenitor cell exosomes using systems biology. Circ Res. 2015;116:255–263. doi: 10.1161/CIRCRESAHA.116.304360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, Just A, Remke J, Zimmer K, Zeug A, Ponimaskin E, Schmiedl A, Yin X, Mayr M, Halder R, Fischer A, Engelhardt S, Wei Y, Schober A, Fiedler J, Thum T. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest. 2014;124:2136–2146. doi: 10.1172/JCI70577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pironti G, Strachan RT, Abraham D, Mon-Wei Yu S, Chen M, Chen W, Hanada K, Mao L, Watson LJ, Rockman HA. Circulating Exosomes Induced by Cardiac Pressure Overload Contain Functional Angiotensin II Type 1 Receptors. Circulation. 2015;131:2120–2130. doi: 10.1161/CIRCULATIONAHA.115.015687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, Bratton DL, Oldenborg PA, Michalak M, Henson PM. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005;123:321–334. doi: 10.1016/j.cell.2005.08.032. [DOI] [PubMed] [Google Scholar]
- 47.Sun X, He S, Wara AK, Icli B, Shvartz E, Tesmenitsky Y, Belkin N, Li D, Blackwell TS, Sukhova GK, Croce K, Feinberg MW. Systemic delivery of microRNA-181b inhibits nuclear factor-kappaB activation, vascular inflammation, and atherosclerosis in apolipoprotein E-deficient mice. Circ Res. 2014;114:32–40. doi: 10.1161/CIRCRESAHA.113.302089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25:771–784. doi: 10.1038/cr.2015.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Choi DS, Kim DK, Kim YK, Gho YS. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 2013;13:1554–1571. doi: 10.1002/pmic.201200329. [DOI] [PubMed] [Google Scholar]
- 50.Vyas N, Dhawan J. Exosomes: mobile platforms for targeted and synergistic signaling across cell boundaries. Cell Mol Life Sci. 2016 doi: 10.1007/s00018-016-2413-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.








