Plasma-derived exosome characterization reveals a distinct microRNA signature in long duration Type 1 diabetes

Marta Garcia-Contreras, Sanket H Shah, Alejandro Tamayo, Paul D Robbins, Ronald B Golberg, Armando J Mendez, Camillo Ricordi, Marta Garcia-Contreras, Sanket H Shah, Alejandro Tamayo, Paul D Robbins, Ronald B Golberg, Armando J Mendez, Camillo Ricordi

Abstract

Type 1 diabetes mellitus (T1DM) results from an autoimmune attack against the insulin-producing ß cells which leads to chronic hyperglycemia. Exosomes are lipid vesicles derived from cellular multivesicular bodies that are enriched in specific miRNAs, potentially providing a disease-specific diagnostic signature. To assess the value of exosome miRNAs as biomarkers for T1DM, miRNA expression in plasma-derived exosomes was measured. Nanoparticle tracking analysis and transmission electron microscopy confirmed the presence of plasma-derived exosomes (EXOs) isolated by differential centrifugation. Total RNA extracted from plasma-derived EXOs of 12 T1DM and 12 control subjects was hybridized onto Nanostring human v2 miRNA microarray array and expression data were analyzed on nSolver analysis software. We found 7 different miRNAs (1 up-regulated and 6 down-regulated), that were differentially expressed in T1DM. The selected candidate miRNAs were validated by qRT-PCR analysis of cohorts of 24 T1DM and 24 control subjects. Most of the deregulated miRNAs are involved in progression of T1DM. These findings highlight the potential of EXOs miRNA profiling in the diagnosis as well as new insights into the molecular mechanisms involved in T1DM.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Work flow of study design and sample processing. Plasma from T1DM (n = 36) and control subjects (n = 36) was collected. Exosomes were isolated by ultracentrifugation and characterized by TEM and NTA analysis. Total exosome RNA was isolated and use for the miRNA microarray analysis (discovery set) or for the qRT-PCR validation (validation set) (A). Plasma exosomes were analyzed under electron microscopy which displayed the same morphology in T1DM and control subjects (B,C). Particle and Size distribution of exosomes analyzed by the Nanoparticle tracking analysis of T1DM and control subjects (D,E). Exosomal RNAs determined by the Agilent RNA Pico Chip. Exosomal RNA samples contained no detectable 18S and 28S rRNAs (G). Validation of selected exosome protein expression by flow cytometry (control read peak) (F). Small RNA Densitometry traces profiles were used to quantify and compare the relative abundance of various small RNAs in T1DM (H) and Control subjects (I).
Figure 2
Figure 2
Differential miRNA content in T1DM and control subjects. Heatmap of the per-row normalized expression levels of selected miRNAs differentially expressed in T1DM and control subjects plasma-derived exosomes (A). Scatter plot of proteins enriched in Control or T1D subjects, n = 12 per group, using P < 0.05 (multiple t-test) and array threshold as cutoffs. Noted in the legend are Control subjects overexpressed miRNAs (green), T1D overexpressed miRNAs (red) (B). Correlation plot of exosome miRNAs, indicating in red and green those miRNAs expressed differentially T1DM and control subject’s exosomes (C).
Figure 3
Figure 3
Differential microarray miRNA expression in T1DM and control subjects. Individual changes in the miRNA plasma exosome levels of the one up-regulated and six down-regulated miRNAs in T1DM patients (n = 12) and control subjects (n = 12). The signal intensities were normalized to the total signal intensity of the microarray. The horizontal lines indicate the mean normalized signal intensity for each group. Statistically significant differences were determined by Student’s t-test.
Figure 4
Figure 4
Box-and-whisker plots of the expression levels of the seven selected miRNAs in an independent set of T1DM (n = 24) and control subjects (n = 24). The comparative cycle threshold (Ct) method was used to quantify the levels of exosome miRNAs in T1DM and control subjects. The relative ratio was calculated using the ΔCt method. The Ct value of miR-631 was used as an internal standard. (*p-value 

Figure 5

Effect of plasma-derived exosomes from…

Figure 5

Effect of plasma-derived exosomes from T1D and controls on insulin secretion of perifused…

Figure 5
Effect of plasma-derived exosomes from T1D and controls on insulin secretion of perifused islets. Groups of 350 IEQ islets each, were stimulated every minute as shown in (A) for low (3 mM) → high (11 mM) → low (3 mM) glucose challenge followed by a KCl step. Pancreatic islet perifusion, normalized iInsulin secretion data to islet volume (IEQ). Data are expressed as ng insulin/islet equivalents (IEQs) per minute (A,B) or area under the curve (AUC) for each secretagogue (CE). Statistical analysis was performed by Student’s t-test.
Figure 5
Figure 5
Effect of plasma-derived exosomes from T1D and controls on insulin secretion of perifused islets. Groups of 350 IEQ islets each, were stimulated every minute as shown in (A) for low (3 mM) → high (11 mM) → low (3 mM) glucose challenge followed by a KCl step. Pancreatic islet perifusion, normalized iInsulin secretion data to islet volume (IEQ). Data are expressed as ng insulin/islet equivalents (IEQs) per minute (A,B) or area under the curve (AUC) for each secretagogue (CE). Statistical analysis was performed by Student’s t-test.

References

    1. Herold KC, Vignali DA, Cooke A, Bluestone JA. Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol. 2013;13:243–256. doi: 10.1038/nri3422.
    1. Winter WE, Schatz DA. Autoimmune markers in diabetes. Clin Chem. 2011;57:168–175. doi: 10.1373/clinchem.2010.148205.
    1. Orban T, et al. Pancreatic islet autoantibodies as predictors of type 1 diabetes in the Diabetes Prevention Trial-Type 1. Diabetes care. 2009;32:2269–2274. doi: 10.2337/dc09-0934.
    1. Planas R, et al. Gene expression profiles for the human pancreas and purified islets in type 1 diabetes: new findings at clinical onset and in long-standing diabetes. Clinical and experimental immunology. 2010;159:23–44. doi: 10.1111/j.1365-2249.2009.04053.x.
    1. Eliasson L, Esguerra JL. Role of non-coding RNAs in pancreatic beta-cell development and physiology. Acta physiologica. 2014;211:273–284. doi: 10.1111/apha.12285.
    1. Poy MN, et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature. 2004;432:226–230. doi: 10.1038/nature03076.
    1. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–579.
    1. Valadi H, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature cell biology. 2007;9:654–659. doi: 10.1038/ncb1596.
    1. Cheng, L., Sharples, R. A., Scicluna, B. J. & Hill, A. F. Exosomes provide a protective and enriched source of miRNA for biomarker profiling compared to intracellular and cell-free blood. Journal of extracellular vesicles3, doi:10.3402/jev.v3.23743 (2014).
    1. Lakhter AJ, Sims EK. Minireview: Emerging Roles for Extracellular Vesicles in Diabetes and Related Metabolic Disorders. Mol Endocrinol. 2015;29:1535–1548. doi: 10.1210/me.2015-1206.
    1. Skog J, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature cell biology. 2008;10:1470–1476. doi: 10.1038/ncb1800.
    1. Ferrante SC, et al. Adipocyte-derived exosomal miRNAs: a novel mechanism for obesity-related disease. Pediatr Res. 2015;77:447–454. doi: 10.1038/pr.2014.202.
    1. Shi R, et al. Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget. 2015;6:26971–26981. doi: 10.18632/oncotarget.4699.
    1. Liang B, et al. Characterization and proteomic analysis of ovarian cancer-derived exosomes. J Proteomics. 2013;80:171–182. doi: 10.1016/j.jprot.2012.12.029.
    1. Sodar BW, et al. Low-density lipoprotein mimics blood plasma-derived exosomes and microvesicles during isolation and detection. Sci Rep. 2016;6:24316. doi: 10.1038/srep24316.
    1. Yang J, Wei F, Schafer C, Wong DT. Detection of tumor cell-specific mRNA and protein in exosome-like microvesicles from blood and saliva. PloS one. 2014;9:e110641. doi: 10.1371/journal.pone.0110641.
    1. Guay C, Menoud V, Rome S, Regazzi R. Horizontal transfer of exosomal microRNAs transduce apoptotic signals between pancreatic beta-cells. Cell Commun Signal. 2015;13:17. doi: 10.1186/s12964-015-0097-7.
    1. Figliolini F, et al. Isolation, characterization and potential role in beta cell-endothelium cross-talk of extracellular vesicles released from human pancreatic islets. PLoS One. 2014;9:e102521. doi: 10.1371/journal.pone.0102521.
    1. Gould, S. J. & Raposo, G. As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles2, doi:10.3402/jev.v2i0.20389 (2013).
    1. Li Y, et al. Identification of endogenous controls for analyzing serum exosomal miRNA in patients with hepatitis B or hepatocellular carcinoma. Disease markers. 2015;2015:893594.
    1. Chatterjee A, et al. A cross comparison of technologies for the detection of microRNAs in clinical FFPE samples of hepatoblastoma patients. Scientific reports. 2015;5:10438. doi: 10.1038/srep10438.
    1. Nielsen LB, et al. Circulating levels of microRNA from children with newly diagnosed type 1 diabetes and healthy controls: evidence that miR-25 associates to residual beta-cell function and glycaemic control during disease progression. Experimental diabetes research. 2012;2012:896362.
    1. Setyowati Karolina D, Sepramaniam S, Tan HZ, Armugam A, Jeyaseelan K. miR-25 and miR-92a regulate insulin I biosynthesis in rats. RNA biology. 2013;10:1365–1378. doi: 10.4161/rna.25557.
    1. Bork-Jensen J, et al. Glucose tolerance is associated with differential expression of microRNAs in skeletal muscle: results from studies of twins with and without type 2 diabetes. Diabetologia. 2015;58:363–373. doi: 10.1007/s00125-014-3434-2.
    1. Hoekstra M, van der Sluis RJ, Kuiper J, Van Berkel TJ. Nonalcoholic fatty liver disease is associated with an altered hepatocyte microRNA profile in LDL receptor knockout mice. The Journal of nutritional biochemistry. 2012;23:622–628. doi: 10.1016/j.jnutbio.2011.03.005.
    1. Knezevic I, et al. A novel cardiomyocyte-enriched microRNA, miR-378, targets insulin-like growth factor 1 receptor: implications in postnatal cardiac remodeling and cell survival. The Journal of biological chemistry. 2012;287:12913–12926. doi: 10.1074/jbc.M111.331751.
    1. Gerin I, et al. Roles for miRNA-378/378* in adipocyte gene expression and lipogenesis. American journal of physiology. Endocrinology and metabolism. 2010;299:E198–206.
    1. Ishida M, et al. MicroRNA-378 regulates adiponectin expression in adipose tissue: a new plausible mechanism. PloS one. 2014;9:e111537. doi: 10.1371/journal.pone.0111537.
    1. Congcong Yin, M. W., Jai, Li., Ruicong, She., Li, Zhou & Qingsheng, Mi. Serum miRNAs as potential biomarkers for early prediction of type 1 diabetes. The FASEB Journal vol. 30 no. 1 Supplement 307.3 (2016).
    1. Bijkerk R, et al. Circulating microRNAs associate with diabetic nephropathy and systemic microvascular damage and normalize after simultaneous pancreas-kidney transplantation. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2015;15:1081–1090. doi: 10.1111/ajt.13072.
    1. Baldeon Rojas L, et al. Study on inflammation-related genes and microRNAs, with special emphasis on the vascular repair factor HGF and miR-574-3p, in monocytes and serum of patients with T2D. Diabetology & metabolic syndrome. 2016;8:6. doi: 10.1186/s13098-015-0113-5.
    1. Marchand, L. et al. miRNA-375 a Sensor of Glucotoxicity Is Altered in the Serum of Children with Newly Diagnosed Type 1 Diabetes. Journal of Diabetes Research, doi:10.1155/2016/1869082 (2016).
    1. Del Prato S, Marchetti P, Bonadonna RC. Phasic insulin release and metabolic regulation in type 2 diabetes. Diabetes. 2002;51(Suppl 1):S109–116. doi: 10.2337/diabetes.51.2007.S109.
    1. Buchwald P, Cechin SR, Weaver JD, Stabler CL. Experimental evaluation and computational modeling of the effects of encapsulation on the time-profile of glucose-stimulated insulin release of pancreatic islets. Biomedical engineering online. 2015;14:28. doi: 10.1186/s12938-015-0021-9.

Source: PubMed

3
订阅