We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Exosomal microRNAs: potential nanotherapeutic targets for diabetic kidney disease

    Lulu Han

    Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China

    Department of Endocrinology, The First Central Hospital of Baoding, Baoding, 071000, China

    ,
    Xiaoning Cai

    Department of Endocrinology, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, China

    &
    Hong Zhou

    *Author for correspondence:

    E-mail Address: zhoubs2013@163.com

    Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China

    Published Online:https://doi.org/10.2217/nnm-2023-0023

    Diabetic kidney disease (DKD) is a primary cause for end-stage renal disease, but no specific therapeutic approaches exist. Exosomal miRNAs, a key functional cargo of nanovesicles, play crucial roles in the pathophysiological processes of DKD. Exosomal miRNAs are involved in cell-to-cell transfer of biological information, mediating nephritic inflammation, oxidative stress, apoptosis, autophagy, epithelial–mesenchymal transition and fibrosis. Circulating exosomal miRNAs derived from urine or serum might function as noninvasive prognostic biomarkers for DKD. Exosomal miRNAs from stem cells have been reported to exert beneficial effects on diabetic kidneys, which suggests that these exosomes might function as potential nanotherapy tools for treating DKD. In this review, we have summarized recent studies based on the association between exosomal miRNAs and DKD.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Zhao L, Zhang J, Lei S et al. Combining glomerular basement membrane and tubular basement membrane assessment improves the prediction of diabetic end-stage renal disease. J. Diabetes 13(7), 572–584 (2021).
    • 2. Sugita E, Hayashi K, Hishikawa A, Itoh H. Epigenetic alterations in podocytes in diabetic nephropathy. Front. Pharmacol. 12, 759299 (2021).
    • 3. Das F, Bera A, Ghosh-Choudhury N et al. High glucose-stimulated enhancer of zeste homolog-2 (EZH2) forces suppression of deptor to cause glomerular mesangial cell pathology. Cell. Signal. 86, 110072 (2021).
    • 4. Vallon V, Thomson SC. Renal function in diabetic disease models: the tubular system in the pathophysiology of the diabetic kidney. Annu. Rev. Physiol. 74, 351–375 (2012).
    • 5. Vartak T, Godson C, Brennan E. Therapeutic potential of pro-resolving mediators in diabetic kidney disease. Adv. Drug Deliv. Rev. 178, 113965 (2021).
    • 6. Thethi T, Kamiyama M, Kobori H. The link between the renin–angiotensin–aldosterone system and renal injury in obesity and the metabolic syndrome. Curr. Hypertens. Rep. 14(2), 160–169 (2012).
    • 7. Magee C, Grieve DJ, Watson CJ, Brazil DP. Diabetic nephropathy: a tangled web to unweave. Cardiovasc. Drugs Ther. 31(5–6), 579–592 (2017).
    • 8. Jung SW, Moon JY. The role of inflammation in diabetic kidney disease. Korean J. Int. Med. 36(4), 753–766 (2021).
    • 9. Samsu N. Diabetic nephropathy: challenges in pathogenesis, diagnosis, and treatment. Biomed. Res. Int. 2021, 1497449 (2021).
    • 10. Lassén E, Daehn IS. Molecular mechanisms in early diabetic kidney disease: glomerular endothelial cell dysfunction. Int. J. Mol. Sci. 21(24), 9456 (2020).
    • 11. Nishad R, Tahaseen V, Kavvuri R et al. Advanced-glycation end-products induce podocyte injury and contribute to proteinuria. Front. Med. 8, 685447 (2021).
    • 12. Zhang PN, Zhou MQ, Guo J et al. Mitochondrial dysfunction and diabetic nephropathy: nontraditional therapeutic opportunities. J. Diabetes Res. 2021, 1010268 (2021).
    • 13. Mostafa DK, Khedr MM, Barakat MK, Abdellatif AA, Elsharkawy AM. Autophagy blockade mechanistically links proton pump inhibitors to worsened diabetic nephropathy and aborts the renoprotection of metformin/enalapril. Life Sci. 265, 118818 (2021).
    • 14. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 367(6478), eaau6977 (2020).
    • 15. He X, Kuang G, Wu Y, Ou C. Emerging roles of exosomal miRNAs in diabetes mellitus. Clin. Transl. Med. 11(6), e468 (2021). • Expounds the critical roles of exosomal miRNAs in diabetes mellitus and its associated complications, which provided us many ideas for this review.
    • 16. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Ann. Rev. Cell Dev. Biol. 30, 255–289 (2014).
    • 17. Noonin C, Thongboonkerd V. Exosome–inflammasome crosstalk and their roles in inflammatory responses. Theranostics 11(9), 4436–4451 (2021).
    • 18. Tafrihi M, Hasheminasab E. miRNAs: biology, biogenesis, their web-based tools, and databases. Microrna 8(1), 4–27 (2019).
    • 19. Zuo Y, Chen L, He X et al. Atorvastatin regulates MALAT1/miR-200c/NRF2 activity to protect against podocyte pyroptosis induced by high glucose. Diabetes Metab. Syndr. Obes. 14, 1631–1645 (2021).
    • 20. He F, Peng F, Xia X et al. miR-135a promotes renal fibrosis in diabetic nephropathy by regulating TRPC1. Diabetologia 57(8), 1726–1736 (2014).
    • 21. Chen YQ, Wang XX, Yao XM et al. MicroRNA-195 promotes apoptosis in mouse podocytes via enhanced caspase activity driven by BCL2 insufficiency. Am. J. Nephrol. 34(6), 549–559 (2011).
    • 22. Lu Y, Liu D, Feng Q, Liu Z. Diabetic nephropathy: perspective on extracellular vesicles. Front. Immunol. 11, 943 (2020).
    • 23. Mohan A, Singh RS, Kumari M et al. Urinary exosomal microRNA-451-5p is a potential early biomarker of diabetic nephropathy in rats. PLOS ONE 11(4), e0154055 (2016).
    • 24. Kim H, Bae YU, Jeon JS et al. The circulating exosomal microRNAs related to albuminuria in patients with diabetic nephropathy. J. Transl. Med. 17(1), 236 (2019). • The authors screened the differently expressed blood exosomal microRNAs of diabetic kidney disease (DKD) patients and analyzed the correlation between exosomal miRNAs and clinical parameters. They provided us potential biomarkers for DKD.
    • 25. Xie Y, Jia Y, Xie C et al. Corrigendum to ‘Urinary exosomal microRNA profiling in incipient type 2 diabetic kidney disease’. J. Diabetes Res. 2018, 5969714 (2018).
    • 26. Prabu P, Rome S, Sathishkumar C et al. MicroRNAs from urinary extracellular vesicles are non-invasive early biomarkers of diabetic nephropathy in type 2 diabetes patients with the ‘Asian Indian phenotype’. Diabetes Metab. 45(3), 276–285 (2019).
    • 27. Wang H, Wang J, Liu T, Leng Y, Yang W. Stem cell-derived exosomal microRNAs: potential therapies in diabetic kidney disease. Biomed. Pharmacother. 164, 114961 (2023).
    • 28. Jin J, Wang Y, Zhao L et al. Exosomal miRNA-215-5p derived from adipose-derived stem cells attenuates epithelial–mesenchymal transition of podocytes by inhibiting ZEB2. Biomed. Res. Int. 2020, 2685305 (2020).
    • 29. Hao Y, Miao J, Liu W et al. Mesenchymal stem cell-derived exosomes carry microRNA-125a to protect against diabetic nephropathy by targeting histone deacetylase 1 and downregulating endothelin-1. Diabetes Metab. Syndr. Obes. 14, 1405–1418 (2021).
    • 30. Duan Y, Chen B, Chen F et al. Exosomal microRNA-16-5p from human urine-derived stem cells ameliorates diabetic nephropathy through protection of podocyte. J. Cell. Mol. Med. 25(23), 10798–10813 (2019). •• Explored the renoprotective effects of stem cell-derived exosomal miRNA on DKD and provided new insights into therapeutic potential for DKD.
    • 31. Cai X, Zou F, Xuan R, Lai X. Exosomes from mesenchymal stem cells expressing microribonucleic acid-125b inhibit the progression of diabetic nephropathy via the tumour necrosis factor receptor-associated factor 6/Akt axis. Endocr. J. 68(7), 817–828 (2021).
    • 32. Akers JC, Gonda D, Kim R, Carter BS, Chen CC. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neurooncol. 113(1), 1–11 (2013).
    • 33. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. Acta Rev. Cancer 1871(2), 455–468 (2019).
    • 34. Lin J, Li J, Huang B et al. Exosomes: novel biomarkers for clinical diagnosis. Sci. World J. 2015, 657086 (2015).
    • 35. Zhang H, Freitas D, Kim HS et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 20(3), 332–343 (2018).
    • 36. Bowers EC, Hassanin AAI, Ramos KS. In vitro models of exosome biology and toxicology: new frontiers in biomedical research. Toxicol. In Vitro 64, 104462 (2020).
    • 37. Tkach M, Théry C. Communication by extracellular vesicles: where we are and where we need to go. Cell 164(6), 1226–1232 (2016).
    • 38. Li S, Wu Y, Ding F et al. Engineering macrophage-derived exosomes for targeted chemotherapy of triple-negative breast cancer. Nanoscale 12(19), 10854–10862 (2020).
    • 39. Duan L, Xu L, Xu X et al. Exosome-mediated delivery of gene vectors for gene therapy. Nanoscale 13(3), 1387–1397 (2021).
    • 40. Meldolesi J. Exosomes and ectosomes in intercellular communication. Curr. Biol. 28(8), R435–R444 (2018).
    • 41. Merchant ML, Rood IM, Deegens JKJ, Klein JB. Isolation and characterization of urinary extracellular vesicles: implications for biomarker discovery. Nat. Rev. Nephrol. 13(12), 731–749 (2017).
    • 42. Lakhter AJ, Pratt RE, Moore RE et al. Beta cell extracellular vesicle miR-21-5p cargo is increased in response to inflammatory cytokines and serves as a biomarker of type 1 diabetes. Diabetologia 61(5), 1124–1134 (2018).
    • 43. Chang W, Wang J. Exosomes and their noncoding RNA cargo are emerging as new modulators for diabetes mellitus. Cells 8(8), 853 (2019).
    • 44. Gauthier BR, Cobo-Vuilleumier N, López-Noriega L. Roles of extracellular vesicles associated non-coding RNAs in diabetes mellitus. Front. Endocrinol. 13, 1057407 (2022).
    • 45. Chaput N, Théry C. Exosomes: immune properties and potential clinical implementations. Semin. Immunopathol. 33(5), 419–440 (2011).
    • 46. Cortez MA, Bueso-Ramos C, Ferdin J et al. MicroRNAs in body fluids – the mix of hormones and biomarkers. Nat. Rev. Clin. Oncol. 8(8), 467–477 (2011).
    • 47. Valadi H, Ekström K, Bossios A et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9(6), 654–659 (2007).
    • 48. Guay C, Regazzi R. Circulating microRNAs as novel biomarkers for diabetes mellitus. Nat. Rev. Endocrinol. 9(9), 513–521 (2013).
    • 49. Karolina DS, Tavintharan S, Armugam A et al. Circulating miRNA profiles in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 97(12), E2271–E2276 (2012).
    • 50. Eissa S, Matboli M, Bekhet MM. Clinical verification of a novel urinary microRNA panal: 133b, -342 and -30 as biomarkers for diabetic nephropathy identified by bioinformatics analysis. Biomed. Pharmacother. 83, 92–99 (2016).
    • 51. Jia Y, Guan M, Zheng Z et al. miRNAs in urine extracellular vesicles as predictors of early-stage diabetic nephropathy. J. Diabetes Res. 2016, 7932765 (2016).
    • 52. Park S, Lee K, Park IB et al. The profiles of microRNAs from urinary extracellular vesicles (EVs) prepared by various isolation methods and their correlation with serum EV microRNAs. Diabetes Res. Clin. Pract. 160, 108010 (2020).
    • 53. Zhang W, Zhou X, Zhang H et al. Extracellular vesicles in diagnosis and therapy of kidney diseases. Am. J. Physiol. Renal Physiol. 311(5), F844–F851 (2016).
    • 54. Delić D, Eisele C, Schmid R et al. Urinary exosomal miRNA signature in type II diabetic nephropathy patients. PLOS ONE 11(3), e0150154 (2016).
    • 55. Florijn BW, Duijs J, Levels JH et al. Diabetic nephropathy alters the distribution of circulating angiogenic microRNAs among extracellular vesicles, HDL, and Ago-2. Diabetes 68(12), 2287–2300 (2019).
    • 56. Uil M, Hau CM, Ahdi M et al. Cellular origin and microRNA profiles of circulating extracellular vesicles in different stages of diabetic nephropathy. Clin. Kidney J. 14(1), 358–365 (2021).
    • 57. Su H, Qiao J, Hu J et al. Podocyte-derived extracellular vesicles mediate renal proximal tubule cells dedifferentiation via microRNA-221 in diabetic nephropathy. Mol. Cell. Endocrinol. 518, 111034 (2020).
    • 58. Jin J, Shi Y, Gong J et al. Exosome secreted from adipose-derived stem cells attenuates diabetic nephropathy by promoting autophagy flux and inhibiting apoptosis in podocyte. Stem Cell Res. Ther. 10(1), 95 (2019).
    • 59. Rayego-Mateos S, Morgado-Pascual JL, Opazo-Ríos L et al. Pathogenic pathways and therapeutic approaches targeting inflammation in diabetic nephropathy. Int. J. Mol. Sci. 21(11), 3798 (2020).
    • 60. Ding X, Jing N, Shen A et al. miR-21-5p in macrophage-derived extracellular vesicles affects podocyte pyroptosis in diabetic nephropathy by regulating A20. J. Endocrinol. Invest. 44(6), 1175–1184 (2021).
    • 61. Lee SE, Jang JE, Kim HS et al. Mesenchymal stem cells prevent the progression of diabetic nephropathy by improving mitochondrial function in tubular epithelial cells. Exp. Mol. Med. 51(7), 1–14 (2019).
    • 62. Duan Y, Luo Q, Wang Y et al. Adipose mesenchymal stem cell-derived extracellular vesicles containing microRNA-26a-5p target TLR4 and protect against diabetic nephropathy. J. Biol. Chem. 295(37), 12868–12884 (2020).
    • 63. Lv J, Hao YN, Wang XP, Lu WH, Xie LY, Niu D. Bone marrow mesenchymal stem cell-derived exosomal miR-30e-5p ameliorates high-glucose induced renal proximal tubular cell pyroptosis by inhibiting ELAVL1. Ren. Fail. 45(1), 2177082 (2023).
    • 64. Gao C, Wang B, Chen Q et al. Serum exosomes from diabetic kidney disease patients promote pyroptosis and oxidative stress through the miR-4449/HIC1 pathway. Nutr. Diabetes 11(1), 33 (2021).
    • 65. Huang Y, Li R, Zhang L et al. Extracellular vesicles from high glucose-treated podocytes induce apoptosis of proximal tubular epithelial cells. Front. Physiol. 11, 579296 (2020).
    • 66. Zhang S, Wu J, Zhu X et al. A novel approach to identify the mechanism of miR-145-5p toxicity to podocytes based on the essential genes targeting analysis. Mol. Ther. Nucl. Acids 26, 749–759 (2021). •• Confirmed that extracellular vesicles could act as a stable transporter for miRNAs to perform their regulatory functions. Provides novel thoughts for the future development of nanotechnology-based drug-delivery systems.
    • 67. Tsai Y, Kuo M, Hung W et al. High glucose induces mesangial cell apoptosis through miR-15b-5p and promotes diabetic nephropathy by extracellular vesicle delivery. Mol. Ther. 28(3), 963–974 (2020). •• Verified the important role of exosomal miRNA in the pathogenesis of DKD in vivo and in vitro.
    • 68. Jiang ZZ, Liu YM, Niu X et al. Exosomes secreted by human urine-derived stem cells could prevent kidney complications from type I diabetes in rats. Stem Cell. Res. Ther. 7, 24 (2016).
    • 69. Nagaishi K, Mizue Y, Chikenji T et al. Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes. Sci. Rep. 6, 34842 (2016).
    • 70. Liu T, Jin Q, Yang L et al. Regulation of autophagy by natural polyphenols in the treatment of diabetic kidney disease: therapeutic potential and mechanism. Front. Endocrinol. 14, 1142276 (2023).
    • 71. Shapouri-Moghaddam A, Mohammadian S, Vazini H et al. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 233(9), 6425–6440 (2018).
    • 72. Huang H, Liu H, Tang J et al. M2 macrophage-derived exosomal miR-25-3p improves high glucose-induced podocytes injury through activation autophagy via inhibiting DUSP1 expression. IUBMB Life 72(12), 2651–2662 (2020).
    • 73. Hu R, Li X, Peng C et al. miR-196b-5p-enriched extracellular vesicles from tubular epithelial cells mediated aldosterone-induced renal fibrosis in mice with diabetes. BMJ Open Diabetes Res. Care 8(1), e001101 (2020).
    • 74. Jin J, Qian F, Zheng D et al. Mesenchymal stem cells attenuate renal fibrosis via exosomes-mediated delivery of microRNA let-7i-5p antagomir. Int. J. Nanomed. 16, 3565–3578 (2021).
    • 75. Liu D, Liu F, Li Z et al. HNRNPA1-mediated exosomal sorting of miR-483-5p out of renal tubular epithelial cells promotes the progression of diabetic nephropathy-induced renal interstitial fibrosis. Cell Death Dis. 12(3), 255 (2021).
    • 76. Zhong L, Liao G, Wang X et al. Mesenchymal stem cells-microvesicle-miR-451a ameliorate early diabetic kidney injury by negative regulation of P15 and P19. Exp. Biol. Med. 243(15–16), 1233–1242 (2018).