Ann Pediatr Endocrinol Metab.  2016 Sep;21(3):119-125. 10.6065/apem.2016.21.3.119.

Exosomes as the source of biomarkers of metabolic diseases

Affiliations
  • 1Department of Pharmacology and Medicinal Toxicology Research Center, Inha University School of Medicine, Incheon, Korea. johykang@inha.ac.kr
  • 2Department of Kinesiology, College of Arts and Sports, Inha University School of Medicine, Incheon, Korea.
  • 3Hypoxia-related Disease Research Center, Inha University School of Medicine, Incheon, Korea.

Abstract

Exosomes are extracellular vesicles that contain molecules that regulate the metabolic functions of adjacent or remote cells. Recent in vitro, in vivo and clinical studies support the hypothesis that exosomes released from various cell types play roles in the progression of metabolic disorders including type 2 diabetes. Based on this concept and advances in other diseases, the proteins, mRNA, microRNA and lipids in exosomes isolated from biological fluids have been proposed as biomarkers in metabolic disorders. However, several problems with the development of clinically applicable biomarkers have not been resolved. In this review, the biologic functions of exosomes are briefly introduced, and we discuss the technical and practical pros and cons of different methods of exosome isolation for the identification of exosomal biomarkers of metabolic disorders. Standardization of preanalytical variables and isolation of high-purity exosomes from fully characterized biological fluids will be necessary for the identification of useful exosomal biomarkers that can provide insights into the pathogenic mechanisms of complications of metabolic syndrome and of whole-body metabolism.

Keyword

Exosomes; Diabetes mellitus; Biomarkers; Standardization; Metabolic syndrome X

MeSH Terms

Biomarkers*
Diabetes Mellitus
Exosomes*
Extracellular Vesicles
In Vitro Techniques
Metabolic Diseases*
Metabolic Syndrome X
Metabolism
MicroRNAs
RNA, Messenger
Biomarkers
MicroRNAs
RNA, Messenger

Figure

  • Fig. 1 Biogenesis and secretion of exosomes. Exosomes originate from the endolysosomal pathway and exhibit a homogeneous size and shape. During the maturation of the early endosome, the inward budding of cellular compartments that occurs in the membrane of the endosome produces multivesicular bodies (MVBs). The internal vesicles (intraluminal vesicles; ILVs) within MVBs contain soluble proteins, transmembrane proteins, lipids, receptors, ligands, mRNAs and miRNAs lower inserted box that are responsible for intercellular communication. The internal sorting of these molecules is precisely regulated by several mechanisms including the endosomal sorting complexes required for transport system. MVBs can fuse with lysosomes to recycle the contents of the ILVs or with the plasma membrane to release ILVs as exosomes. The mechanisms that decide between the release of MVBs as exosomes or their allocation to recycling pathways are not fully understood. GPI, glycophosphatidylinositol; TM, transmembrane.

  • Fig. 2 Electron microscopic image of exosomes prepared from cell culture media (CM). After full differentiation of murine myoblast (C2C12 cells) to myocytes, exosomes were isolated from CM by differential ultracentrifugation. (A, B) Small vesicles (white arrows) homogeneously sized <150 nm are clearly identifiable. (C, D) Several aggregated vesicles are also observed when ultracentrifugation is used to prepare exosomes. Arrowheads and asterisk indicate membrane of exosome and exosomal body, respectively.


Reference

1. Guescini M, Guidolin D, Vallorani L, Casadei L, Gioacchini AM, Tibollo P, et al. C2C12 myoblasts release micro-vesicles containing mtDNA and proteins involved in signal transduction. Exp Cell Res. 2010; 316:1977–1984. PMID: 20399774.
Article
2. Aoki N, Jin-no S, Nakagawa Y, Asai N, Arakawa E, Tamura N, et al. Identification and characterization of microvesicles secreted by 3T3-L1 adipocytes: redox- and hormone-dependent induction of milk fat globule-epidermal growth factor 8-associated microvesicles. Endocrinology. 2007; 148:3850–3862. PMID: 17478559.
Article
3. Conde-Vancells J, Rodriguez-Suarez E, Embade N, Gil D, Matthiesen R, Valle M, et al. Characterization and comprehensive proteome profiling of exosomes secreted by hepatocytes. J Proteome Res. 2008; 7:5157–5166. PMID: 19367702.
Article
4. Raiborg C, Stenmark H. The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins. Nature. 2009; 458:445–452. PMID: 19325624.
Article
5. Baietti MF, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012; 14:677–685. PMID: 22660413.
Article
6. Villarroya-Beltri C, Baixauli F, Gutiérrez-Vázquez C, Sánchez-Madrid F, Mittelbrunn M. Sorting it out: regulation of exosome loading. Semin Cancer Biol. 2014; 28:3–13. PMID: 24769058.
Article
7. EL Andaloussi S, Mäger I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013; 12:347–357. PMID: 23584393.
Article
8. Simhadri VR, Reiners KS, Hansen HP, Topolar D, Simhadri VL, Nohroudi K, et al. Dendritic cells release HLA-B-associated transcript-3 positive exosomes to regulate natural killer function. PLoS One. 2008; 3:e3377. PMID: 18852879.
Article
9. Chivet M, Hemming F, Pernet-Gallay K, Fraboulet S, Sadoul R. Emerging role of neuronal exosomes in the central nervous system. Front Physiol. 2012; 3:145. PMID: 22654762.
Article
10. Jang YY, Collector MI, Baylin SB, Diehl AM, Sharkis SJ. Hematopoietic stem cells convert into liver cells within days without fusion. Nat Cell Biol. 2004; 6:532–539. PMID: 15133469.
Article
11. Quesenberry PJ, Aliotta JM. Cellular phenotype switching and microvesicles. Adv Drug Deliv Rev. 2010; 62:1141–1148. PMID: 20558219.
Article
12. Lai RC, Chen TS, Lim SK. Mesenchymal stem cell exosome: a novel stem cell-based therapy for cardiovascular disease. Regen Med. 2011; 6:481–492. PMID: 21749206.
Article
13. Timmers L, Lim SK, Hoefer IE, Arslan F, Lai RC, van Oorschot AA, et al. Human mesenchymal stem cell-conditioned medium improves cardiac function following myocardial infarction. Stem Cell Res. 2011; 6:206–214. PMID: 21419744.
Article
14. Camussi G, Deregibus MC, Bruno S, Grange C, Fonsato V, Tetta C. Exosome/microvesicle-mediated epigenetic reprogramming of cells. Am J Cancer Res. 2011; 1:98–110. PMID: 21969178.
15. Rak J, Guha A. Extracellular vesicles: vehicles that spread cancer genes. Bioessays. 2012; 34:489–497. PMID: 22442051.
Article
16. Cai Z, Yang F, Yu L, Yu Z, Jiang L, Wang Q, et al. Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J Immunol. 2012; 188:5954–5961. PMID: 22573809.
Article
17. Bellingham SA, Guo BB, Coleman BM, Hill AF. Exosomes: vehicles for the transfer of toxic proteins associated with neurodegenerative diseases? Front Physiol. 2012; 3:124. PMID: 22563321.
Article
18. Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, et al. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci. 2010; 30:6838–6851. PMID: 20484626.
Article
19. Aswad H, Forterre A, Wiklander OP, Vial G, Danty-Berger E, Jalabert A, et al. Exosomes participate in the alteration of muscle homeostasis during lipid-induced insulin resistance in mice. Diabetologia. 2014; 57:2155–2164. PMID: 25073444.
Article
20. Jayachandran M, Litwiller RD, Lahr BD, Bailey KR, Owen WG, Mulvagh SL, et al. Alterations in platelet function and cell-derived microvesicles in recently menopausal women: relationship to metabolic syndrome and atherogenic risk. J Cardiovasc Transl Res. 2011; 4:811–822. PMID: 21786187.
Article
21. Diamant M, Nieuwland R, Pablo RF, Sturk A, Smit JW, Radder JK. Elevated numbers of tissue-factor exposing microparticles correlate with components of the metabolic syndrome in uncomplicated type 2 diabetes mellitus. Circulation. 2002; 106:2442–2447. PMID: 12417540.
Article
22. D'Alessandra Y, Devanna P, Limana F, Straino S, Di Carlo A, Brambilla PG, et al. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur Heart J. 2010; 31:2765–2773. PMID: 20534597.
23. Feng B, Chen Y, Luo Y, Chen M, Li X, Ni Y. Circulating level of microparticles and their correlation with arterial elasticity and endothelium-dependent dilation in patients with type 2 diabetes mellitus. Atherosclerosis. 2010; 208:264–269. PMID: 19674745.
Article
24. Kim SJ, Moon GJ, Cho YH, Kang HY, Hyung NK, Kim D, et al. Circulating mesenchymal stem cells microparticles in patients with cerebrovascular disease. PLoS One. 2012; 7:e37036. PMID: 22615882.
Article
25. Ogata M, Inanami O, Nakajima M, Nakajima T, Hiraoka W, Kuwabara M. Ca(2+)-dependent and caspase-3-independent apoptosis caused by damage in Golgi apparatus due to 2,4,5,7-tetrabromorhodamine 123 bromide-induced photodynamic effects. Photochem Photobiol. 2003; 78:241–247. PMID: 14556310.
Article
26. Safdar A, Saleem A, Tarnopolsky MA. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat Rev Endocrinol. 2016; 12:504–517. PMID: 27230949.
Article
27. Jalabert A, Vial G, Guay C, Wiklander OP, Nordin JZ, Aswad H, et al. Exosome-like vesicles released from lipid-induced insulin-resistant muscles modulate gene expression and proliferation of beta recipient cells in mice. Diabetologia. 2016; 59:1049–1058. PMID: 26852333.
Article
28. Lee JE, Moon PG, Lee IK, Baek MC. Proteomic analysis of extracellular vesicles released by adipocytes of Otsuka Long-Evans Tokushima Fatty (OLETF) Rats. Protein J. 2015; 34:220–235. PMID: 25998041.
Article
29. Santovito D, De Nardis V, Marcantonio P, Mandolini C, Paganelli C, Vitale E, et al. Plasma exosome microRNA profiling unravels a new potential modulator of adiponectin pathway in diabetes: effect of glycemic control. J Clin Endocrinol Metab. 2014; 99:E1681–E1685. PMID: 24937531.
Article
30. Deng ZB, Poliakov A, Hardy RW, Clements R, Liu C, Liu Y, et al. Adipose tissue exosome-like vesicles mediate activation of macrophage-induced insulin resistance. Diabetes. 2009; 58:2498–2505. PMID: 19675137.
Article
31. Jansen F, Yang X, Hoelscher M, Cattelan A, Schmitz T, Proebsting S, et al. Endothelial microparticle-mediated transfer of MicroRNA-126 promotes vascular endothelial cell repair via SPRED1 and is abrogated in glucose-damaged endothelial microparticles. Circulation. 2013; 128:2026–2038. PMID: 24014835.
Article
32. Wang X, Huang W, Liu G, Cai W, Millard RW, Wang Y, et al. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells. J Mol Cell Cardiol. 2014; 74:139–150. PMID: 24825548.
Article
33. O'Neill S, Bohl M, Gregersen S, Hermansen K, O'Driscoll L. Blood-based biomarkers for metabolic syndrome. Trends Endocrinol Metab. 2016; 27:363–374. PMID: 27150849.
34. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001; 69:89–95. PMID: 11240971.
35. Lawson C, Vicencio JM, Yellon DM, Davidson SM. Microvesicles and exosomes: new players in metabolic and cardiovascular disease. J Endocrinol. 2016; 228:R57–R71. PMID: 26743452.
Article
36. Koeck ES, Iordanskaia T, Sevilla S, Ferrante SC, Hubal MJ, Freishtat RJ, et al. Adipocyte exosomes induce transforming growth factor beta pathway dysregulation in hepatocytes: a novel paradigm for obesity-related liver disease. J Surg Res. 2014; 192:268–275. PMID: 25086727.
Article
37. Nicklas BJ, Penninx BW, Cesari M, Kritchevsky SB, Newman AB, Kanaya AM, et al. Association of visceral adipose tissue with incident myocardial infarction in older men and women: the Health, Aging and Body Composition Study. Am J Epidemiol. 2004; 160:741–749. PMID: 15466496.
Article
38. Kanhai DA, Kappelle LJ, van der Graaf Y, Uiterwaal CS, Visseren FL. SMART Study Group. The risk of general and abdominal adiposity in the occurrence of new vascular events and mortality in patients with various manifestations of vascular disease. Int J Obes (Lond). 2012; 36:695–702. PMID: 21712810.
Article
39. Livshits MA, Khomyakova E, Evtushenko EG, Lazarev VN, Kulemin NA, Semina SE, et al. Corrigendum: Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci Rep. 2016; 6:21447. PMID: 27022711.
40. Cocucci E, Racchetti G, Podini P, Meldolesi J. Enlargeosome traffic: exocytosis triggered by various signals is followed by endocytosis, membrane shedding or both. Traffic. 2007; 8:742–757. PMID: 17488290.
Article
41. Eken C, Gasser O, Zenhaeusern G, Oehri I, Hess C, Schifferli JA. Polymorphonuclear neutrophil-derived ectosomes interfere with the maturation of monocyte-derived dendritic cells. J Immunol. 2008; 180:817–824. PMID: 18178820.
Article
42. Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014; 3:26913. PMID: 25536934.
Article
43. Linares R, Tan S, Gounou C, Arraud N, Brisson AR. High-speed centrifugation induces aggregation of extracellular vesicles. J Extracell Vesicles. 2015; 4:29509. PMID: 26700615.
Article
44. Van Deun J, Mestdagh P, Sormunen R, Cocquyt V, Vermaelen K, Vandesompele J, et al. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J Extracell Vesicles. 2014; 3:24858.
Article
45. Kalra H, Adda CG, Liem M, Ang CS, Mechler A, Simpson RJ, et al. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics. 2013; 13:3354–3364. PMID: 24115447.
Article
46. Hong CS, Funk S, Muller L, Boyiadzis M, Whiteside TL. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J Extracell Vesicles. 2016; 5:29289. PMID: 27018366.
Article
47. Fiandaca MS, Kapogiannis D, Mapstone M, Boxer A, Eitan E, Schwartz JB, et al. Identification of preclinical Alzheimer's disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement. 2015; 11:600–607.e1. PMID: 25130657.
Article
48. Shi M, Liu C, Cook TJ, Bullock KM, Zhao Y, Ginghina C, et al. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson's disease. Acta Neuropathol. 2014; 128:639–650. PMID: 24997849.
Article
49. Lobb RJ, Becker M, Wen SW, Wong CS, Wiegmans AP, Leimgruber A, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015; 4:27031. PMID: 26194179.
Article
50. Agouni A, Ducluzeau PH, Benameur T, Faure S, Sladkova M, Duluc L, et al. Microparticles from patients with metabolic syndrome induce vascular hypo-reactivity via Fas/Fasligand pathway in mice. PLoS One. 2011; 6:e27809. PMID: 22110764.
Article
51. Agouni A, Lagrue-Lak-Hal AH, Ducluzeau PH, Mostefai HA, Draunet-Busson C, Leftheriotis G, et al. Endothelial dysfunction caused by circulating microparticles from patients with metabolic syndrome. Am J Pathol. 2008; 173:1210–1219. PMID: 18772329.
Article
52. Amabile N, Cheng S, Renard JM, Larson MG, Ghorbani A, McCabe E, et al. Association of circulating endothelial microparticles with cardiometabolic risk factors in the Framingham Heart Study. Eur Heart J. 2014; 35:2972–2979. PMID: 24742886.
Article
53. Chen Y, Feng B, Li X, Ni Y, Luo Y. Plasma endothelial microparticles and their correlation with the presence of hypertension and arterial stiffness in patients with type 2 diabetes. J Clin Hypertens (Greenwich). 2012; 14:455–460. PMID: 22747618.
Article
54. Kranendonk ME, de Kleijn DP, Kalkhoven E, Kanhai DA, Uiterwaal CS, van der Graaf Y, et al. Extracellular vesicle markers in relation to obesity and metabolic complications in patients with manifest cardiovascular disease. Cardiovasc Diabetol. 2014; 13:37. PMID: 24498934.
Article
55. Burger D, Thibodeau JF, Holterman CE, Burns KD, Touyz RM, Kennedy CR. Urinary podocyte microparticles identify prealbuminuric diabetic glomerular injury. J Am Soc Nephrol. 2014; 25:1401–1407. PMID: 24676640.
Article
56. Sun AL, Deng JT, Guan GJ, Chen SH, Liu YT, Cheng J, et al. Dipeptidyl peptidase-IV is a potential molecular biomarker in diabetic kidney disease. Diab Vasc Dis Res. 2012; 9:301–308. PMID: 22388283.
Article
57. Zubiri I, Posada-Ayala M, Sanz-Maroto A, Calvo E, Martin-Lorenzo M, Gonzalez-Calero L, et al. Diabetic nephropathy induces changes in the proteome of human urinary exosomes as revealed by label-free comparative analysis. J Proteomics. 2014; 96:92–102. PMID: 24211404.
Article
58. Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013; 2:20360.
Article
Full Text Links
  • APEM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr