Analysis of Plasma MicroRNAs as Predictors and Biomarkers of Aging and Frailty in Humans

Iryna Rusanova, María E Diaz-Casado, Marisol Fernández-Ortiz, Paula Aranda-Martínez, Ana Guerra-Librero, Francisco J García-García, Germaine Escames, Leocadio Mañas, Darío Acuña-Castroviejo, Iryna Rusanova, María E Diaz-Casado, Marisol Fernández-Ortiz, Paula Aranda-Martínez, Ana Guerra-Librero, Francisco J García-García, Germaine Escames, Leocadio Mañas, Darío Acuña-Castroviejo

Abstract

Although circulating microRNAs (miRNAs) can modulate gene expression and affect immune system response, little is known about their participation in age-associated frailty syndrome and sarcopenia. The aim of this study was to determine miRNAs as possible biomarkers of age and frailty and their correlation with oxidative and inflammatory state in human blood. Three inflammation-related miRNAs (miR-21, miR-146a, and miR-223) and one miRNA related with the control of melatonin synthesis (miR-483) were analyzed. Twenty-two healthy adults, 34 aged robust, and 40 aged fragile patients were selected for this study. The expression of plasma miRNAs was assessed by RT-qPCR; plasma cytokines (IL-6, IL-8, IL-10, and TNFα) were analyzed by commercial kits, and plasma advanced oxidation protein products (AOPP) and lipid oxidation (LPO) were spectrophotometrically measured. Fragile subjects had higher miR-21 levels than control subjects, whereas miR-223 and miR-483 levels increased at a similar extend in both aged groups. All cytokines measured increased in aged groups compared with controls, without differences between robust and fragile subjects. The fragile group had a TNFα/IL-10 ratio significantly higher than robust and control groups. Aged groups also had higher AOPP and LPO levels than controls. Women presented higher AOPP and LPO levels and increased expression of miR-483 compared with men. Positive correlations between miR-21 and AOPP and between miR-483 and IL-8 were detected. The expression of miR-21 and the TNFα/IL-10 ratio were correlated positively with the presence of frailty, which suggests that these markers can be considered as possible biomarkers for age-related frailty.

Figures

Figure 1
Figure 1
miRNA levels in control and aged groups (a)–(d). Relative expression of these miRNAs was calculated using the 2−ΔΔCt method. The expression levels were normalized against U6 snRNA and against the average of the control group (Ct, general normalization). Data are presented as means ± SEM. ∗P < 0.05, ∗∗P < 0.01, and ∗∗∗P < 0.001 versus the control group. Regression and correlation analysis with relative expression of miRNAs, calculated using analysis of the Spearman correlation coefficient. Only those miRNAs that had significant correlation are shown (e) and (f). C: control group; R: robust group; F: fragile group.
Figure 2
Figure 2
Plasma levels of IL-6, IL-8, TNFα, and IL-10 in control and aged groups of subjects ((a)–(d)). TNFα/IL-10 ratio in the three studied groups (e). Data are presented as means ± SEM. ∗P < 0.05, ∗∗P < 0.01 and ∗∗∗P < 0.001 versus the control group. #P < 0.05 versus the robust group.
Figure 3
Figure 3
Lipid peroxidation (LPO) and advanced oxidation protein product (AOPP) levels in plasma of subjects of three studied groups ((a) and (b)). LPO and AOPP levels in plasma were also classified according to gender ((c) and (d)). Data are presented as means ± SEM. ∗P < 0.05, ∗∗P < 0.01, and ∗∗∗P < 0.001 versus the control group. Correlation analysis between miR-21 relative expression and AOPP levels (e), calculated using analysis of the Spearman correlation coefficient. C: control group; R: robust group; F: fragile group.

References

    1. Beard J. R., Officer A., de Carvalho I. A., et al. The world report on ageing and health: a policy framework for healthy ageing. Lancet. 2016;387(10033):2145–2154. doi: 10.1016/S0140-6736(15)00516-4.
    1. European Commission Directorate-General for Economic and Financial Affairs. The 2015 Ageing Report Economic and budgetary projections for the 28 EU Member States (2013-2060) Brussels: European Economy; 2015.
    1. Rosenberg I. H. Sarcopenia: origins and clinical relevance. The Journal of Nutrition. 1997;127(5):990S–991S. doi: 10.1093/jn/127.5.990S.
    1. Cruz-Jentoft A., Baeyens J., Bauer J., et al. Sarcopenia: European consensus on definition and diagnosis: report of the European Working Group on Sarcopenia in Older People. Age and Ageing. 2010;39(4):412–423. doi: 10.1093/ageing/afq034.
    1. Xue Q. L. The frailty syndrome: definition and natural history. Clinics in Geriatric Medicine. 2011;27(1):1–15. doi: 10.1016/j.cger.2010.08.009.
    1. Montero-Fernández N., Serra-Rexach J. A. Role of exercise on sarcopenia in the elderly. European Journal of Physical and Rehabilitation Medicine. 2013;49(1):131–143.
    1. Bandeen-Roche K., Seplaki C. L., Huang J., et al. Frailty in older adults: a nationally representative profile in the United States. The Journals of Gerontology: Series A. 2015;70(11):1427–1434. doi: 10.1093/gerona/glv133.
    1. Ambros V. The functions of animal microRNAs. Nature. 2004;431(7006):350–355. doi: 10.1038/nature02871.
    1. Winter J., Jung S., Keller S., Gregory R. I., Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nature Cell Biology. 2009;11(3):228–234. doi: 10.1038/ncb0309-228.
    1. Weilner S., Grillari-Voglauer R., Redl H., Grillari J., Nau T. The role of microRNAs in cellular senescence and age-related conditions of cartilage and bone. Acta Orthopaedica. 2015;86(1):92–99. doi: 10.3109/17453674.2014.957079.
    1. Li Y. T., Chen S. Y., Wang C. R., et al. Brief report: amelioration of collagen-induced arthritis in mice by lentivirus-mediated silencing of microRNA-223. Arthritis and Rheumatism. 2012;64(10):3240–3245. doi: 10.1002/art.34550.
    1. Fan J., Kou X., Yang Y., Chen N. MicroRNA-regulated proinflammatory cytokines in sarcopenia. Mediators of Inflammation. 2016;2016:9. doi: 10.1155/2016/1438686.1438686
    1. Martin-Cano F. E., Camello-Almaraz C., Acuña-Castroviejo D., Pozo M. J., Camello P. J. Age-related changes in mitochondrial function of mouse colonic smooth muscle: beneficial effects of melatonin. Journal of Pineal Research. 2014;56(2):163–174. doi: 10.1111/jpi.12109.
    1. Garcia-Garcia F. J., Gutierrez Avila G., Alfaro-Acha A., et al. The prevalence of frailty syndrome in an older population from Spain. The Toledo study for healthy aging. The Journal of Nutrition, Health & Aging. 2011;15(10):852–856. doi: 10.1007/s12603-011-0075-8.
    1. Fried L. P., Tangen C. M., Walston J., et al. Frailty in older adults: evidence for a phenotype. The Journals of Gerontology: Series A. 2001;56(3):M146–M157. doi: 10.1093/gerona/56.3.M146.
    1. Lauretani F., Russo C. R., Bandinelli S., et al. Age-associated changes in skeletal muscles and their effect on mobility: an operational diagnosis of sarcopenia. Journal of Applied Physiology. 2003;95(5):1851–1860. doi: 10.1152/japplphysiol.00246.2003.
    1. Ottenbacher K. J., Branch L. G., Ray L., Gonzales V. A., Peek M. K., Hinman M. R. The reliability of upper- and lower-extremity strength testing in a community survey of older adults. Archives of Physical Medicine and Rehabilitation. 2002;83(10):1423–1427. doi: 10.1053/apmr.2002.34619.
    1. Orme J. G., Reis J., Herz E. J. Factorial and discriminant validity of the center for epidemiological studies depression (CES-D) scale. Journal of Clinical Psychology. 1986;42(1):28–33. doi: 10.1002/1097-4679(198601)42:1<28::AID-JCLP2270420104>;2-T.
    1. Guralnik J. M., Ferrucci L., Pieper C. F., et al. Lower extremity function and subsequent disability: consistency across studies, predictive models, and value of gait speed alone compared with the short physical performance battery. The Journals of Gerontology: Series A. 2000;55(4):M221–M231. doi: 10.1093/gerona/55.4.M221.
    1. Washburn R. A., Smith K. W., Jette A. M., Janney C. A. The physical activity scale for the elderly (PASE): development and evaluation. Journal of Clinical Epidemiology. 1993;46(2):153–162. doi: 10.1016/0895-4356(93)90053-4.
    1. Chen X., Liang H., Guan D., et al. A combination of Let-7d, Let-7g and Let-7i serves as a stable reference for normalization of serum microRNAs. PLoS One. 2013;8(11, article e79652) doi: 10.1371/journal.pone.0079652.
    1. Zile M. R., Mehurg S. M., Arroyo J. E., Stroud R. E., DeSantis S. M., Spinale F. G. Relationship between the temporal profile of plasma microRNA and left ventricular remodeling in patients after myocardial infarction. Circulation. Cardiovascular Genetics. 2011;4(6):614–619. doi: 10.1161/CIRCGENETICS.111.959841.
    1. Witko-Sarsat V., Friedlander M., Capeillère-Blandin C., et al. Advanced oxidation protein products as a novel marker of oxidative stress in uremia. Kidney International. 1996;49(5):1304–1313. doi: 10.1038/ki.1996.186.
    1. Esterbauer H., Cheeseman K. H. [42] Determination of aldehydic lipid peroxidation products: malonaldehyde and 4-hydroxynonenal. Methods in Enzymology. 1990;186:407–421. doi: 10.1016/0076-6879(90)86134-H.
    1. Olivieri F., Spazzafumo L., Santini G., et al. Age-related differences in the expression of circulating microRNAs: miR-21 as a new circulating marker of inflammaging. Mechanisms of Ageing and Development. 2012;133(11-12):675–685. doi: 10.1016/j.mad.2012.09.004.
    1. Fabbri M., Paone A., Calore F., Galli R., Croce C. M. A new role for microRNAs, as ligands of Toll-like receptors. RNA Biology. 2014;10(2):169–174. doi: 10.4161/rna.23144.
    1. Olivieri F., Capri M., Bonafè M., et al. Circulating miRNAs and miRNA shuttles as biomarkers: Perspective trajectories of healthy and unhealthy aging. Mechanisms of Ageing and Development. 2017;165(Part B):162–170. doi: 10.1016/j.mad.2012.09.004.
    1. Xu T., Zhou Q., Che L., et al. Circulating miR-21, miR-378, and miR-940 increase in response to an acute exhaustive exercise in chronic heart failure patients. Oncotarget. 2016;7(11):12414–12425. doi: 10.18632/oncotarget.6966.
    1. Li T., Morgan M. J., Choksi S., Zhang Y., Kim Y. S., Liu Z. G. MicroRNAs modulate the noncanonical transcription factor NF-κB pathway by regulating expression of the kinase IKKα during macrophage differentiation. Nature Immunology. 2010;11(9):799–805. doi: 10.1038/ni.1918.
    1. Bhaumik D., Scott G. K., Schokrpur S., et al. MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL–8. Aging. 2009;1(4):402–411. doi: 10.18632/aging.100042.
    1. Holmberg J., Alajbegovic A., Gawlik K. I., Elowsson L., Durbeej M. Laminin α2 chain-deficiency is associated with microRNA deregulation in skeletal muscle and plasma. Frontiers in Aging Neuroscience. 2014;6:p. 155. doi: 10.3389/fnagi.2014.00155.
    1. Li C. W., Wang W. H., Chen B. S. Investigating the specific core genetic-and-epigenetic networks of cellular mechanisms involved in human aging in peripheral blood mononuclear cells. Oncotarget. 2016;7(8):8556–8579. doi: 10.18632/oncotarget.7388.
    1. Clokie S. J. H., Lau P., Kim H. H., Coon S. L., Klein D. C. MicroRNAs in the pineal gland: miR-483 regulates melatonin synthesis by targeting arylalkylamine N-acetyltransferase. The Journal of Biological Chemistry. 2012;287(30):25312–25324. doi: 10.1074/jbc.M112.356733.
    1. Wang D. T., Yin Y., Yang Y. J., et al. Resveratrol prevents TNF-α-induced muscle atrophy via regulation of Akt/mTOR/FoxO1 signaling in C2C12 myotubes. International Immunopharmacology. 2014;19(2):206–213. doi: 10.1016/j.intimp.2014.02.002.
    1. Jung H. J., Lee K.-P., Milholland B., et al. Comprehensive miRNA profiling of skeletal muscle and serum in induced and normal mouse muscle atrophy during aging. The Journals of Gerontology: Series A. 2017;72(11):1483–1491. doi: 10.1093/gerona/glx025.

Source: PubMed

3
订阅