miR-130a and Tgfβ Content in Extracellular Vesicles Derived from the Serum of Subjects at High Cardiovascular Risk Predicts their In-Vivo Angiogenic Potential

Claudia Cavallari, Federico Figliolini, Marta Tapparo, Massimo Cedrino, Alessandra Trevisan, Lorenza Positello, Pietro Rispoli, Anna Solini, Giuseppe Migliaretti, Giovanni Camussi, Maria Felice Brizzi, Claudia Cavallari, Federico Figliolini, Marta Tapparo, Massimo Cedrino, Alessandra Trevisan, Lorenza Positello, Pietro Rispoli, Anna Solini, Giuseppe Migliaretti, Giovanni Camussi, Maria Felice Brizzi

Abstract

Serum-derived extracellular vesicles (sEV) from healthy donors display in-vivo pro-angiogenic properties. To identify patients that may benefit from autologous sEV administration for pro-angiogenic purposes, sEV angiogenic capability has been evaluated in type 2 diabetic (T2DM) subjects (D), in obese individuals with (OD) and without (O) T2DM, and in subjects with ischemic disease (IC) (9 patients/group). sEV display different angiogenic properties in such cluster of individuals. miRNomic profile and TGFβ content in sEV were evaluated. We found that miR-130a and TGFβ content correlates with sEV in-vitro and in-vivo angiogenic properties, particularly in T2DM patients. Ingenuity Pathway Analysis (IPA) identified a number of genes as among the most significant miR-130a interactors. Gain-of-function experiments recognized homeoboxA5 (HOXA5) as a miR-130a specific target. Finally, ROC curve analyses revealed that sEV ineffectiveness could be predicted (Likelihood Ratio+ (LH+) = 3.3 IC 95% from 2.6 to 3.9) by comparing miR-130a and TGFβ content 'in Series'. We demonstrate that sEV from high cardiovascular risk patients have different angiogenic properties and that miR-130a and TGFβ sEV content predicts 'true ineffective sEVs'. These results provide the rationale for the use of these assays to identify patients that may benefit from autologous sEV administration to boost the angiogenetic process.

Conflict of interest statement

G.C., C.C. and M.F.B. are named as inventors in a related patent application. G.C. is a component of the Scientific Advisory Board of UNICYTE.

Figures

Figure 1
Figure 1
Nanosight sEV characterisation. (A) Representative images of NTA analysis referred to each group of patients. (B) Dot-plot graph representing NTA size distribution with mean size values for healthy, obese, T2DM, obese/T2DM and ischemic subjects. (C) The histogram reports the number of EVs recovered from the serum of each group of patients. *p < 0.05 obese and ischemic patients vs healthy subjects (One-way ANOVA followed by Multiple Comparison Test). (n = 9 patients/group). H = Healthy subjects; D = Diabetic patients; O = Obese patients; OD = Obese/Diabetic patients; IC = Ischemic patients.
Figure 2
Figure 2
In-vitro and in-vivo angiogenesis in response to sEVs. (A) Dot plot graph reporting the in-vitro proangiogenic activity of sEV recovered from each patient. The number corresponds to each patient per group (see Supplementary Table 3). The dotted line defines the cut-off for effective and ineffective sEV. The light colour corresponds to ineffective sEV per each group. (B) Representative images of vessels formed in response to effective and ineffective sEV. The number refers to patient sEV. (n = 3 each group except for OD. The same sample was used in 3 independent experiments). (C)In-vivo quantitative analysis of vessels counted in 10 sections of Matrigel for each experimental condition. Data represent the mean value of untreated (C) (n = 3) and treated mice with: healthy ineffective sEV (i-sEV), healthy effective sEV (e-sEV); T2DM ineffective sEV (D i-sEV), T2DM effective sEV (D e-sEV); obese ineffective sEV (O i-sEV), obese effective sEV (O e-sEV); obese/T2DM ineffective sEV (OD i-sEV), obese/T2DM effective sEV (OD e-sEV); ischemic ineffective sEV (IC i-sEV), ischemic effective sEV (IC e-sEV). *p < 0.05 healthy e-sEV vs. healthy i-sEV; §p < 0.05 T2DMe-sEV vs. T2DM i-sEV; #p < 0.05 obese e-sEV vs. obese i-sEV; °p < 0.05 obese/T2DM e-sEV vs. obese/T2DM i-sEV; +p < 0.05 ischemic e-sEV vs. ischemic i-sEV ischemic; (One-way ANOVA followed by Multiple Comparison Test). (n = 3 each group except for OD. The same sample was used in 3 independent experiments) ECs (red), erythrocytes (yellow) and Matrigel (light blue) staining in Matrigel plugs. (Original magnification: x200; scale bar: 12 µm).
Figure 3
Figure 3
TGFβ sEV content. (A) Data obtained per patient/group are reported. The upper curve refers to sEV TGFβ content, while the lower to the % of the test of potency. The dotted line defines the cut-off for effective and ineffective sEV. The number corresponds to each patient (n = 3 each group, except for OD. The same sample was used in 3 independent experiments). (B) Min to max columns represent TGFβ content in effective and ineffective sEVs from each group. *p < 0.05 healthy e-sEV vs. healthy i-sEV; #p < 0.05 T2DM e-sEV vs. T2DM i-sEV; §p < 0.05 obese e-sEV vs. obese i-sEV; (One-way ANOVA followed by Multiple Comparison Test). (n = 3, except for OD. The same sample was used in 3 independent experiments).
Figure 4
Figure 4
miR expression and pattern analysis of sEV. (A) miRNome miRNA qPCR profiler array analysis on healthy effective (e-sEV) and ineffective (i-sEV). miRNAs were detected by RT-PCR. Results are reported as Log2−(∆Ct) (Normalized Relative Quantities) values. Dark blues circles correspond to e-sEV, while light red corresponds to i-sEV (n = 3/each). In red and blue, miRNAs upregulated and down regulated, respectively (t-test, p value < 0.05, see Supplementary Table 5). (B) miRNA validation in patient sEV. RT-PCR for miR-126, miR-21, miR-296-3p, miR-210, miR-130a, miR-27a, miR-29a and miR-191 was performed in all patients and healthy donors. Results are reported as 40-Ct. The red circle indicates patients with high miR-210 expression. (C) Gene Ontology analysis. Network analysis of pathways positively correlated with the miRNAs indicated above. Data were obtained from DIANA-mirPath analyses. The significant enrichment of the TGFβ-associated pathway was identified. (D) miR-130a distribution, patient-by-patient, including healthy subjects is reported and compared with the % of the test of potency. (E) Network analysis of pathways positively correlated with miR-130a. Data were obtained from DIANA-mirPath analysis. Only pathways including at least 15 genes were selected.
Figure 5
Figure 5
miR-130a integrated interaction networks, target validation and ROC analysis. (A) Network analysis between miR-130a and mRNA targets. Lines represent interactions between genes and miR-130a predicted using the IPA Software; indirect interactions (dotted lines), direct interactions (continuous lines). Squares include TGFβ and TGFBR. Circles include genes involved in angiogenesis (KDR, EPHB6, ROCK1, HOXA5). (B) Validation of miR-130a target(s). Upper panel RT-PCR relative quantification (RQ): miR-130a relative amount in untreated ECs (1), in ECs transfected with miRNA Scramble (Scr) (2), with mimic-miR-130a (3), or treated with sEV from patients D17 (4), D18 (5), D20 (6), D1 (7), D2 (8), and D4 (9) is reported. (*p < 0.05 samples 3, 4, 5, 6 vs. samples 1, 2, 7, 8, 9). Lower panel: KDR, ROCK1, HOXA5 and β-actin expression was evaluated in the above samples. sEV from patients D17, D18, D20 are enriched in miR-130a, while sEV from D1, D2 and D4 carry low miR-130a level. (C) ROC analysis. miR-130a and TGFβ sEV content from all patients and healthy subjects were analysed. Predictive capacity was evaluated for each of the two measures individually. A table reporting the AUC values, standard errors, p-values and threshold values is also reported.

References

    1. Bastien M, Poirier P, Lemieux I, Després JP. Overview of epidemiology and contribution of obesity to cardiovascular disease. Prog. Cardiovasc. Dis. 2014;56:369–81. doi: 10.1016/j.pcad.2013.10.016.
    1. Deregibus Maria Chiara, Cantaluppi Vincenzo, Calogero Raffaele, Lo Iacono Marco, Tetta Ciro, Biancone Luigi, Bruno Stefania, Bussolati Benedetta, Camussi Giovanni. Endothelial progenitor cell–derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA. Blood. 2007;110(7):2440–2448. doi: 10.1182/blood-2007-03-078709.
    1. Camussi G, Deregibus MC, Cantaluppi V. Role of stem-cell-derived microvesicles in the paracrine action of stem cells. Biochem. Soc. Trans. 2013;41:283–7. doi: 10.1042/BST20120192.
    1. Tetta C, Ghigo E, Silengo L, Deregibus MC, Camussi G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine. 2013;44:11–19. doi: 10.1007/s12020-012-9839-0.
    1. Yuana Y, Sturk A, Nieuwland R. Extracellular vesicles in physiological and pathological conditions. Blood Rev. 2013;27:31–39. doi: 10.1016/j.blre.2012.12.002.
    1. Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J. Clin. Invest. 2016;126:1173–1180. doi: 10.1172/JCI81131.
    1. Shanmuganathan M, Vughs J, Noseda M, Emanueli C. Exosomes: Basic Biology and Technological Advancements Suggesting Their Potential as Ischemic Heart Disease Therapeutics. Front. Physiol. 2018;9:1159. doi: 10.3389/fphys.2018.01159.
    1. Belting M, Christianson HC. Role of exosomes and microvesicles in hypoxia-associated tumour development and cardiovascular disease. Journal of Internal Medicine. 2015;278:251–263. doi: 10.1111/joim.12393.
    1. Faure V, et al. Elevation of circulating endothelial microparticles in patients with chronic renal failure. J. Thromb. Haemost. 2006;4:566–573. doi: 10.1111/j.1538-7836.2005.01780.x.
    1. Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles. 2014;3:24641. doi: 10.3402/jev.v3.24641.
    1. Gidlof O, et al. Platelets activated during myocardial infarction release functional miRNA, which can be taken up by endothelial cells and regulate ICAM1 expression. Blood. 2013;121(3908–17):S1–26.
    1. Pathan M, et al. Vesiclepedia 2019: a compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2018 doi: 10.1093/nar/gky1029.
    1. Catalanotto, C., Cogoni, C. & Zardo, G. MicroRNA in Control of Gene Expression: An Overview of Nuclear Functions. Int. J. Mol. Sci. 17 (2016).
    1. Kondkar AA, Abu-Amero KK. Utility of circulating microRNAs as clinical biomarkers for cardiovascular diseases. Biomed Res. Int. 2015;2015:821823. doi: 10.1155/2015/821823.
    1. Pan Y, et al. Platelet-secreted microRNA-223 promotes endothelial cell apoptosis induced by advanced glycation end products via targeting the insulin-like growth factor 1 receptor. J. Immunol. 2014;192:437–446. doi: 10.4049/jimmunol.1301790.
    1. Hosseinkhani B, Kuypers S, van den Akker NMS, Molin DGM, Michiels L. Extracellular Vesicles Work as a Functional Inflammatory Mediator Between Vascular Endothelial Cells and Immune Cells. Front. Immunol. 2018;9:1789. doi: 10.3389/fimmu.2018.01789.
    1. Zhang H, et al. Serum exosomes mediate delivery of arginase 1 as a novel mechanism for endothelial dysfunction in diabetes. Proc. Natl. Acad. Sci. 2018;115:E6927–E6936. doi: 10.1073/pnas.1721521115.
    1. Togliatto G, et al. PDGF-BB Carried by Endothelial Cell-Derived Extracellular Vesicles Reduces Vascular Smooth Muscle Cell Apoptosis in Diabetes. Diabetes. 2018;67:704–716. doi: 10.2337/db17-0371.
    1. Louise Schacht Revenfeld, A. et al. Diagnostic and Prognostic Potential of Extracellular Vesicles in Peripheral Blood. Clinical Therapeutics36 (2014).
    1. Bei Y, et al. Exercise-induced circulating extracellular vesicles protect against cardiac ischemia–reperfusion injury. Basic Res. Cardiol. 2017;112:38. doi: 10.1007/s00395-017-0628-z.
    1. Cavallari C, et al. Serum-derived extracellular vesicles (EVs) impact on vascular remodeling and prevent muscle damage in acute hind limb ischemia. Sci. Rep. 2017;7:8180. doi: 10.1038/s41598-017-08250-0.
    1. Khurana R, Simons M, Martin JF, Zachary IC. Role of angiogenesis in cardiovascular disease: A critical appraisal. Circulation. 2005;112:1813–1824. doi: 10.1161/CIRCULATIONAHA.105.535294.
    1. Deveza L, Choi J, Yang F. Therapeutic angiogenesis for treating cardiovascular diseases. Theranostics. 2012;2:801–814. doi: 10.7150/thno.4419.
    1. Anand S. A brief primer on microRNAs and their roles in angiogenesis. Vasc. Cell. 2013;5:2. doi: 10.1186/2045-824X-5-2.
    1. Yin K-J, Hamblin M, Chen YE. Angiogenesis-regulating microRNAs and Ischemic Stroke. Curr. Vasc. Pharmacol. 2015;13:352–365. doi: 10.2174/15701611113119990016.
    1. Shalaby SM, El-Shal AS, Shoukry A, Khedr MH, Abdelraheim N. Serum miRNA-499 and miRNA-210: A potential role in early diagnosis of acute coronary syndrome. IUBMB Life. 2016;68:673–682. doi: 10.1002/iub.1529.
    1. Mosch, B., Reissenweber, B., Neuber, C. & Pietzsch, J. Eph receptors and ephrin ligands: Important players in angiogenesis and tumor angiogenesis. J. Oncol. 2010 (2010).
    1. Montalvo J, et al. Rock1 & 2 Perform Overlapping and Unique Roles in Angiogenesis and Angiosarcoma Tumor Progression. Curr Mol Med. 2013;1:205–219. doi: 10.2174/156652413804486296.
    1. Yoshiji H, et al. KDR/Flk-1 is a major regulator of vascular endothelial growth factor–induced tumor development and angiogenesis in murine hepatocellular carcinoma cells. Hepatology. 1999;30:1179–1186. doi: 10.1002/hep.510300509.
    1. Chen Y, Gorski DH. Regulation of angiogenesis through a microRNA (miR-130a) that down-regulates antiangiogenic homeobox genes GAX and HOXA5. Blood. 2008;111:1217–1226. doi: 10.1182/blood-2007-07-104133.
    1. Gai C, et al. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer. 2018;18:439. doi: 10.1186/s12885-018-4364-z.
    1. Einarson TR, Acs A, Ludwig C, Panton UH. Prevalence of cardiovascular disease in type 2 diabetes: a systematic literature review of scientific evidence from across the world in 2007-2017. Cardiovasc. Diabetol. 2018;17:83. doi: 10.1186/s12933-018-0728-6.
    1. Kolluru GK, Bir SC, Kevil CG. Endothelial dysfunction and diabetes: effects on angiogenesis, vascular remodeling, and wound healing. Int. J. Vasc. Med. 2012;2012:918267.
    1. Zeng H, Chen J-X. Microvascular Rarefaction and Heart Failure With Preserved Ejection Fraction. Front. Cardiovasc. Med. 2019;6:1–7. doi: 10.3389/fcvm.2019.00015.
    1. Heusch G, et al. Cardiovascular remodelling in coronary artery disease and heart failure. Lancet. 2014;383:1933–1943. doi: 10.1016/S0140-6736(14)60107-0.
    1. Gili M, Orsello A, Gallo S, Brizzi MF. Diabetes-associated macrovascular complications: cell-based therapy a new tool? Endocrine. 2013;44:557–575. doi: 10.1007/s12020-013-9936-8.
    1. Cohn JN, Ferrari R, Sharpe N. Cardiac remodeling—concepts and clinical implications: a consensus paper from an international forum on cardiac remodeling. J. Am. Coll. Cardiol. 2000;35:569–582. doi: 10.1016/S0735-1097(99)00630-0.
    1. Kourembanas S. Exosomes: Vehicles of Intercellular Signaling, Biomarkers, and Vectors of Cell Therapy. Annu. Rev. Physiol. 2015;77:13–27. doi: 10.1146/annurev-physiol-021014-071641.
    1. Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J. Control. Release. 2015;219:396–405. doi: 10.1016/j.jconrel.2015.07.030.
    1. Oh S-J, et al. VEGF and VEGF-C: Specific Induction of Angiogenesis and Lymphangiogenesis in the Differentiated Avian Chorioallantoic Membrane. Dev. Biol. 1997;188:96–109. doi: 10.1006/dbio.1997.8639.
    1. Wu H, et al. Epsin deficiency promotes lymphangiogenesis through regulation of VEGFR3 degradation in diabetes. J. Clin. Invest. 2018;128:4025–4043. doi: 10.1172/JCI96063.
    1. Kim, A., Shah, A. S. & Nakamura, T. Extracellular Vesicles: A Potential Novel Regulator of Obesity and Its Associated Complications. Child. (Basel, Switzerland)5 (2018).
    1. Togliatto G, et al. Obesity reduces the pro-angiogenic potential of adipose tissue stem cell-derived extracellular vesicles (EVs) by impairing miR-126 content: impact on clinical applications. Int. J. Obes. 2015;40:102. doi: 10.1038/ijo.2015.123.
    1. Chiva-Blanch G, et al. Microparticle Shedding by Erythrocytes, Monocytes and Vascular Smooth Muscular. Cells Is Reduced by Aspirin in Diabetic Patients. Rev. Esp. Cardiol. (Engl. Ed). 2016;69:672–680. doi: 10.1016/j.recesp.2015.12.034.
    1. Sabatier F, et al. Type 1 And Type 2 Diabetic Patients Display Different Patterns of Cellular Microparticles. Diabetes. 2002;51:2840–2845. doi: 10.2337/diabetes.51.9.2840.
    1. Chen J, et al. Proangiogenic compositions of microvesicles derived from human umbilical cord mesenchymal stem cells. PLoS One. 2014;9:e115316. doi: 10.1371/journal.pone.0115316.
    1. Zhang B, et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/beta-catenin pathway. Stem Cells Transl. Med. 2015;4:513–522. doi: 10.5966/sctm.2014-0267.
    1. Chiva-Blanch G, et al. Monocyte-derived circulating microparticles (CD14(+), CD14(+)/CD11b(+) and CD14(+)/CD142(+)) are related to long-term prognosis for cardiovascular mortality in STEMI patients. Int. J. Cardiol. 2017;227:876–881. doi: 10.1016/j.ijcard.2016.11.302.
    1. Devlin C, Greco S, Martelli F, Ivan M. miR-210: More than a silent player in hypoxia. IUBMB Life. 2011;63:94–100.
    1. Eguchi A, et al. Circulating adipocyte-derived extracellular vesicles are novel markers of metabolic stress. J. Mol. Med. (Berl). 2016;94:1241–1253. doi: 10.1007/s00109-016-1446-8.
    1. Murakami T, et al. Impact of weight reduction on production of platelet-derived microparticles and fibrinolytic parameters in obesity. Thromb. Res. 2007;119:45–53. doi: 10.1016/j.thromres.2005.12.013.
    1. Pardo F, Villalobos-Labra R, Sobrevia B, Toledo F, Sobrevia L. Extracellular vesicles in obesity and diabetes mellitus. Mol. Aspects Med. 2018;60:81–91. doi: 10.1016/j.mam.2017.11.010.
    1. Sridurongrit S, Larsson J, Schwartz R, Ruiz-Lozano P, Kaartinen V. Signaling via the Tgf-beta type I receptor Alk5 in heart development. Dev. Biol. 2008;322:208–218. doi: 10.1016/j.ydbio.2008.07.038.
    1. Pohlers D, et al. TGF-β and fibrosis in different organs - molecular pathway imprints. Biochim. Biophys. Acta - Mol. Basis Dis. 2009;1792:746–756. doi: 10.1016/j.bbadis.2009.06.004.
    1. Evrard SM, et al. The profibrotic cytokine transforming growth factor-β1 increases endothelial progenitor cell angiogenic properties. J. Thromb. Haemost. 2012;10:670–679. doi: 10.1111/j.1538-7836.2012.04644.x.
    1. Guduric-Fuchs J, et al. Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types. BMC Genomics. 2012;13:357. doi: 10.1186/1471-2164-13-357.
    1. Guay C, Regazzi R. Circulating microRNAs as novel biomarkers for diabetes mellitus. Nat. Rev. Endocrinol. 2013;9:513–521. doi: 10.1038/nrendo.2013.86.
    1. Lee EK, et al. miR-130 Suppresses Adipogenesis by Inhibiting Peroxisome Proliferator-Activated Receptor Expression. Mol. Cell. Biol. 2011;31:626–638. doi: 10.1128/MCB.00894-10.
    1. Motawi TK, Shaker OG, Ismail MF, Sayed NH. Peroxisome Proliferator-Activated Receptor Gamma in Obesity and Colorectal Cancer: The Role of Epigenetics. Sci. Rep. 2017;7:1–8. doi: 10.1038/s41598-017-11180-6.
    1. Lopatina T, et al. Platelet-derived growth factor regulates the secretion of extracellular vesicles by adipose mesenchymal stem cells and enhances their angiogenic potential. Cell Commun. Signal. 2014;12:26. doi: 10.1186/1478-811X-12-26.
    1. Couffinhal T, et al. Mouse model of angiogenesis. Am. J. Pathol. 1998;152:1667–1679.
    1. Collino F, et al. Exosome and Microvesicle-Enriched Fractions Isolated from Mesenchymal Stem Cells by Gradient Separation Showed Different Molecular Signatures and Functions on Renal Tubular Epithelial Cells. Stem Cell Rev. 2017;13:226–243. doi: 10.1007/s12015-016-9713-1.
    1. Cavallari C, et al. Online Hemodiafiltration Inhibits Inflammation-Related Endothelial Dysfunction and Vascular Calcification of Uremic Patients Modulating miR-223 Expression in Plasma Extracellular Vesicles. J. Immunol. 2019;202:2372–2383. doi: 10.4049/jimmunol.1800747.
    1. Grund B, Sabin C. Analysis of biomarker data: logs, odds ratios, and receiver operating characteristic curves. Curr. Opin. HIV AIDS. 2010;5:473–479. doi: 10.1097/COH.0b013e32833ed742.
    1. Hanley JA, McNeil BJ. The meaning and use of the area under a receiver operating characteristic (ROC) curve. Radiology. 1982;143:29–36. doi: 10.1148/radiology.143.1.7063747.
    1. Zweig MH, Campbell G. Receiver-operating characteristic (ROC) plots: a fundamental evaluation tool in clinical medicine. Clin. Chem. 1993;39:561–577. doi: 10.1093/clinchem/39.4.561.
    1. Altman, D. G. Practical statistics for medical research. (Chapman & Halls, 1990).
    1. Harper R, Reeves B. Reporting of precision of estimates for diagnostic accuracy: a review. BMJ. 1999;318:1322–1323. doi: 10.1136/bmj.318.7194.1322.
    1. Nam J-M. Confidence Limits for the Ratio of Two Binomial Proportions Based on Likelihood Scores: Non-Iterative Method. Biometrical J. 1995;37:375–379. doi: 10.1002/bimj.4710370311.

Source: PubMed

3
Suscribir