Circulating MicroRNAs in Elderly Type 2 Diabetic Patients

Giuseppina Catanzaro, Zein Mersini Besharat, Martina Chiacchiarini, Luana Abballe, Claudia Sabato, Alessandra Vacca, Paola Borgiani, Francesco Dotta, Manfredi Tesauro, Agnese Po, Elisabetta Ferretti, Giuseppina Catanzaro, Zein Mersini Besharat, Martina Chiacchiarini, Luana Abballe, Claudia Sabato, Alessandra Vacca, Paola Borgiani, Francesco Dotta, Manfredi Tesauro, Agnese Po, Elisabetta Ferretti

Abstract

The circulating microRNAs (miRNAs) associated with type 2 diabetes (T2D) in elderly patients are still being defined. To identify novel miRNA biomarker candidates for monitoring responses to sitagliptin in such patients, we prospectively studied 40 T2D patients (age > 65) with HbA1c levels of 7.5-9.0% on metformin. After collection of baseline blood samples (t0), the dipeptidyl peptidase-IV (DPP-IV) inhibitor (DPP-IVi) sitagliptin was added to the metformin regimen, and patients were followed for 15 months. Patients with HbA1c < 7.5% or HbA1c reduction > 0.5% after 3 and 15 months of therapy were classified as "responders" (group R, n = 34); all others were classified as "nonresponders" (group NR, n = 6). Circulating miRNA profiling was performed on plasma collected in each group before and after 15 months of therapy (t0 and t15). Intra- and intergroup comparison of miRNA profiles pinpointed three miRNAs that correlated with responses to sitagliptin: miR-378, which is a candidate biomarker of resistance to this DPP-IVi, and miR-126-3p and miR-223, which are associated with positive responses to the drug. The translational implications are as immediate as evident, with the possibility to develop noninvasive diagnostic tools to predict drug response and development of chronic complications.

Figures

Figure 1
Figure 1
Experimental design for the identification of circulating miRNAs in T2D patients. The duration of the study was 2 years. The blue horizontal arrow indicates the duration of the different phases of the project. The red vertical arrows indicate the main study periods. In year 1, patients were enrolled and started on metformin + sitagliptin (see Materials and Methods for details). After 3 and 15 months of treatment (t3 and t15), HbA1c values were reassessed and patients were classified as nonresponders (NR) or responders (R). Plasma miRNA levels at baseline (t0) and t15 from groups R and NR were compared as indicated. Comparison of plasma pools: (1) NR-t0 versus R-t0, (2) R-t15 versus R-t0, (3) NR-t15 versus NR-t0, and (4) NR-t15 versus R-t15.
Figure 2
Figure 2
Glycemic control statuses of the patients at baseline and 3 and 15 months after initiation of metformin + sitagliptin. All 40 patients in poor metabolic control were enrolled. HbA1c levels were evaluated after 3 and 15 months from the addition of sitagliptin. On the basis of HbA1c values, patients were divided into responders and nonresponders. All patients showed an initial metabolic response to therapy (t3), whereas after 15 months (t15), 34/40 were responders. Based on this information, patients were divided into five groups: (1) t0 responder samples (R-t0), (2) t0 nonresponder samples (NR-t0), (3) t3 responder samples (R-t3), (4) t15 responder samples (R-t15), and (5) t15 nonresponder samples (NR-t15). miRNA profiling was performed at baseline and after 15 months of sitagliptin addition.
Figure 3
Figure 3
Venn diagram of circulating miRNAs detected in the elderly T2D cohort and in the R and NR subcohorts. Patients were classified as responders (R) or nonresponders (NR) based on their glycemic control status after 15 months of treatment with metformin + sitagliptin (t15). Venn diagrams show the number of miRNAs detected in NR and R plasma samples collected (a) before the start of combined therapy (baseline, t0) and (b) at t15 and (c) at both t0 and t15.
Figure 4
Figure 4
Heat maps showing circulating microRNAs that were differentially expressed in plasma samples from (a) R and NR patients at baseline (t0), prior to the addition of sitagliptin to the maximum-dose metformin regimen; (b) R at t0 and after 15 months of sitagliptin (t15); (c) NR patients at t15 and t0; and (d) R and NR patients at t15.

References

    1. Guay C., Regazzi R. Circulating microRNAs as novel biomarkers for diabetes mellitus. Nature Reviews Endocrinology. 2013;9(9):513–521. doi: 10.1038/nrendo.2013.86.
    1. Pescador N., Pérez-Barba M., Ibarra J. M., Corbatón A., Martínez-Larrad M. T., Serrano-Ríos M. Serum circulating microRNA profiling for identification of potential type 2 diabetes and obesity biomarkers. PLoS One. 2013;8(10, article e77251) doi: 10.1371/journal.pone.0077251.
    1. Kubota T., Kubota N., Kadowaki T. Imbalanced insulin actions in obesity and type 2 diabetes: key mouse models of insulin signaling pathway. Cell Metabolism. 2017;25(4):797–810. doi: 10.1016/j.cmet.2017.03.004.
    1. Zhu H., Leung S. W. Identification of microRNA biomarkers in type 2 diabetes: a meta-analysis of controlled profiling studies. Diabetologia. 2015;58(5):900–911. doi: 10.1007/s00125-015-3510-2.
    1. Mitchell P. S., Parkin R. K., Kroh E. M., et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(30):10513–10518. doi: 10.1073/pnas.0804549105.
    1. Zampetaki A., Kiechl S., Drozdov I., et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circulation Research. 2010;107(6):810–817. doi: 10.1161/CIRCRESAHA.110.226357.
    1. De Smaele E., Ferretti E., Gulino A. MicroRNAs as biomarkers for CNS cancer and other disorders. Brain Research. 2010;1338:100–111. doi: 10.1016/j.brainres.2010.03.103.
    1. Ciccacci C., Morganti R., di Fusco D., et al. Common polymorphisms in MIR146a, MIR128a and MIR27a genes contribute to neuropathy susceptibility in type 2 diabetes. Acta Diabetologica. 2014;51(4):663–671. doi: 10.1007/s00592-014-0582-2.
    1. Barbagallo D., Piro S., Condorelli A. G., et al. miR-296-3p, miR-298-5p and their downstream networks are causally involved in the higher resistance of mammalian pancreatic α cells to cytokine-induced apoptosis as compared to β cells. BMC Genomics. 2013;14(1):p. 62. doi: 10.1186/1471-2164-14-62.
    1. Guay C., Regazzi R. New emerging tasks for microRNAs in the control of β-cell activities. Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 2016;1861(12):2121–2129. doi: 10.1016/j.bbalip.2016.05.003.
    1. Sebastiani G., Po A., Miele E., et al. MicroRNA-124a is hyperexpressed in type 2 diabetic human pancreatic islets and negatively regulates insulin secretion. Acta Diabetologica. 2015;52(3):523–530. doi: 10.1007/s00592-014-0675-y.
    1. Yang Z., Chen H., Si H., et al. Serum miR-23a, a potential biomarker for diagnosis of pre-diabetes and type 2 diabetes. Acta Diabetologica. 2014;51(5):823–831. doi: 10.1007/s00592-014-0617-8.
    1. Butler P. C., Dry S., Elashoff R. GLP-1–based therapy for diabetes: what you do not know can hurt you. Diabetes Care. 2010;33(2):453–455. doi: 10.2337/dc09-1902.
    1. Wang F., He Y., Zhang R., Zeng Q., Zhao X. Combination therapy of metformin plus dipeptidyl peptidase-4 inhibitor versus metformin plus sulfonylurea and their association with a decreased risk of cardiovascular disease in type 2 diabetes mellitus patients. Medicine. 2017;96(36, article e7638) doi: 10.1097/MD.0000000000007638.
    1. de Souza Cazarim M., da Cruz-Cazarim E. L. C., de Oliveira Baldoni A., et al. Cost-effectiveness analysis of different dipeptidyl-peptidase 4 inhibitor drugs for treatment of type 2 diabetes mellitus. Diabetes & Metabolic Syndrome: Clinical Research & Reviews. 2017;11(Supplement 2):S859–S865. doi: 10.1016/j.dsx.2017.07.006.
    1. Mitrakou A. Pathogenesis of hyperglycaemia in type 2 diabetes. Diabetes, Obesity and Metabolism. 2002;4(4):249–254. doi: 10.1046/j.1463-1326.2002.00204.x.
    1. Kim Y.-G., Yoon D., Park S., et al. Dipeptidyl peptidase-4 inhibitors and risk of heart failure in patients with type 2 diabetes mellitus: a population-based cohort study. Circulation: Heart Failure. 2017;10(9, article e003957) doi: 10.1161/CIRCHEARTFAILURE.117.003957.
    1. Monami M., Ahrén B., Dicembrini I., Mannucci E. Dipeptidyl peptidase-4 inhibitors and cardiovascular risk: a meta-analysis of randomized clinical trials. Diabetes, Obesity and Metabolism. 2013;15(2):112–120. doi: 10.1111/dom.12000.
    1. Bansal V. S., Raja C. P., Venkataraman K., Vijayalakshmi M. Genes involved in pancreatic islet cell rejuvenation. The Indian Journal of Medical Research. 2013;137(4):695–703.
    1. Kanasaki K., Shi S., Kanasaki M., et al. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes. 2014;63(6):2120–2131. doi: 10.2337/db13-1029.
    1. Kirschner M. B., Edelman J. J. B., Kao S. C. H., Vallely M. P., van Zandwijk N., Reid G. The impact of hemolysis on cell-free microRNA biomarkers. Frontiers in Genetics. 2013;4:p. 94. doi: 10.3389/fgene.2013.00094.
    1. Catanzaro G., Besharat Z., Garg N., et al. MicroRNAs-proteomic networks characterizing human medulloblastoma-SLCs. Stem Cells International. 2016;2016:10. doi: 10.1155/2016/2683042.2683042
    1. Giannini G., Capalbo C., Ottini L., et al. Clinical classification of BRCA1 DNA missense variants: H1686Q is a novel pathogenic mutation occurring in the ontogenetically invariant THV motif of the N-terminal BRCT domain. Journal of Clinical Oncology. 2008;26(25):4212–4214. doi: 10.1200/JCO.2008.18.2089.
    1. He Y., Ding Y., Liang B., et al. A systematic study of dysregulated microRNA in type 2 diabetes mellitus. International Journal of Molecular Sciences. 2017;18(3):p. 456. doi: 10.3390/ijms18030456.
    1. Zhu H., Shyh-Chang N., Segrè A. V., et al. The Lin28/let-7 axis regulates glucose metabolism. Cell. 2011;147(1):81–94. doi: 10.1016/j.cell.2011.08.033.
    1. Bijkerk R., Duijs J. M. G. J., Khairoun M., et al. Circulating microRNAs associate with diabetic nephropathy and systemic microvascular damage and normalize after simultaneous pancreas–kidney transplantation. American Journal of Transplantation. 2015;15(4):1081–1090. doi: 10.1111/ajt.13072.
    1. Zhao H., Guan J., Lee H. M., et al. Up-regulated pancreatic tissue microRNA-375 associates with human type 2 diabetes through β-cell deficit and islet amyloid deposition. Pancreas. 2010;39(6):843–846. doi: 10.1097/MPA.0b013e3181d12613.
    1. Higuchi C., Nakatsuka A., Eguchi J., et al. Identification of circulating miR-101, miR-375 and miR-802 as biomarkers for type 2 diabetes. Metabolism. 2015;64(4):489–497. doi: 10.1016/j.metabol.2014.12.003.
    1. Wang C., Wan S., Yang T., et al. Increased serum microRNAs are closely associated with the presence of microvascular complications in type 2 diabetes mellitus. Scientific Reports. 2016;6(1, article 20032) doi: 10.1038/srep20032.
    1. Xu L.-l., Shi C.-m., Xu G.-f., et al. TNF-α, IL-6, and leptin increase the expression of miR-378, an adipogenesis-related microRNA in human adipocytes. Cell Biochemistry and Biophysics. 2014;70(2):771–776. doi: 10.1007/s12013-014-9980-x.
    1. Zanutto S., Pizzamiglio S., Ghilotti M., et al. Circulating miR-378 in plasma: a reliable, haemolysis-independent biomarker for colorectal cancer. British Journal of Cancer. 2014;110(4):1001–1007. doi: 10.1038/bjc.2013.819.
    1. Rottiers V., Näär A. M. MicroRNAs in metabolism and metabolic disorders. Nature Reviews Molecular Cell Biology. 2012;13(4):239–250. doi: 10.1038/nrm3313.
    1. Ortega F. J., Mercader J. M., Moreno-Navarrete J. M., et al. Profiling of circulating microRNAs reveals common microRNAs linked to type 2 diabetes that change with insulin sensitization. Diabetes Care. 2014;37(5):1375–1383. doi: 10.2337/dc13-1847.
    1. Liu Y., Gao G., Yang C., et al. The role of circulating microRNA-126 (miR-126): a novel biomarker for screening prediabetes and newly diagnosed type 2 diabetes mellitus. International Journal of Molecular Sciences. 2014;15(6):10567–10577. doi: 10.3390/ijms150610567.
    1. Olivieri F., Spazzafumo L., Bonafè M., et al. miR-21-5p and miR-126a-3p levels in plasma and circulating angiogenic cells: relationship with type 2 diabetes complications. Oncotarget. 2015;6(34):35372–35382. doi: 10.18632/oncotarget.6164.
    1. Zhao Y., Yin Z., Li H., et al. miR-30c protects diabetic nephropathy by suppressing epithelial-to-mesenchymal transition in db/db mice. Aging Cell. 2017;16(2):387–400. doi: 10.1111/acel.12563.
    1. Raut S. K., Singh G. B., Rastogi B., et al. miR-30c and miR-181a synergistically modulate p53–p21 pathway in diabetes induced cardiac hypertrophy. Molecular and Cellular Biochemistry. 2016;417(1-2):191–203. doi: 10.1007/s11010-016-2729-7.
    1. de Graaf C., Donnelly D., Wootten D., et al. Glucagon-like peptide-1 and its class B G protein–coupled receptors: a long march to therapeutic successes. Pharmacological Reviews. 2016;68(4):954–1013. doi: 10.1124/pr.115.011395.
    1. Ding L., Ai D., Wu R., et al. Identification of the differential expression of serum microRNA in type 2 diabetes. Bioscience, Biotechnology, and Biochemistry. 2015;80(3):461–465. doi: 10.1080/09168451.2015.1107460.
    1. Nunez Lopez Y. O., Garufi G., Seyhan A. A. Altered levels of circulating cytokines and microRNAs in lean and obese individuals with prediabetes and type 2 diabetes. Molecular BioSystems. 2017;13(1):106–121. doi: 10.1039/C6MB00596A.
    1. Deiuliis J. A. MicroRNAs as regulators of metabolic disease: pathophysiologic significance and emerging role as biomarkers and therapeutics. International Journal of Obesity. 2016;40(1):88–101. doi: 10.1038/ijo.2015.170.

Source: PubMed

3
Tilaa