Next Article in Journal
Roles of JAK2 in Aging, Inflammation, Hematopoiesis and Malignant Transformation
Previous Article in Journal
Mitochondria and Female Germline Stem Cells—A Mitochondrial DNA Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exosomes and Their Noncoding RNA Cargo Are Emerging as New Modulators for Diabetes Mellitus

1
Center for Regenerative Medicine, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
2
School of Basic Medical Sciences, Qingdao University, Qingdao 266021, China
*
Authors to whom correspondence should be addressed.
Cells 2019, 8(8), 853; https://doi.org/10.3390/cells8080853
Submission received: 2 July 2019 / Revised: 29 July 2019 / Accepted: 6 August 2019 / Published: 8 August 2019

Abstract

:
Diabetes belongs to a group of metabolic disorders characterized by long term high blood glucose levels due to either inadequate production of insulin (Type 1 diabetes, T1DM) or poor response of the recipient cell to insulin (Type 2 diabetes, T2DM). Organ dysfunctions are the main causes of morbidity and mortality due to high glucose levels. Understanding the mechanisms of organ crosstalk may help us improve our basic knowledge and find novel strategies to better treat the disease. Exosomes are part of a newly emerged research area and have attracted a great deal of attention for their capacity to regulate communications between cells. In conditions of diabetes, exosomes play important roles in the pathological processes in both T1DM and T2DM, such as connecting the immune cell response to pancreatic tissue injury, as well as adipocyte stimulation to insulin resistance of skeletal muscle or liver. Furthermore, in recent years, nucleic acids containing exosomes—especially microRNAs (miRNAs) and long noncoding RNAs (lncRNAs)—have been shown to mainly regulate communications between organs in pathological processes of diabetes, including influencing metabolic signals and insulin signals in target tissues, affecting cell viability, and modulating inflammatory pancreatic cells. Moreover, exosome miRNAs show promise in their use as biomarkers or in treatments for diabetes and diabetic complications. Thus, this paper summarizes the recent work on exosomes related to diabetes as well as the roles of exosomal miRNAs and lncRNAs in diabetic pathology and diagnosis in order to help us better understand the exact roles of exosomes in diabetes development.

1. Extracellular Vesicles and Exosomes

Communication between organs is crucial for their normal functioning and pathological physiology [1,2,3]. In addition to the traditional known factors that affect organ communication such as cytokines, hormones, and chemokines, a new family involving extracellular vesicles (EVs) has emerged as a group of important modulators. The generic term “extracellular vesicles” was proposed to define all lipid bilayer-enclosed extracellular structures. EVs can be secreted by almost all cell types, including plant cells, bacteria, and fungi [4,5,6,7,8]. These small vesicles can be released in response to various stimulation, for example, changes in cell physical environments (PH, temperature, irradiation) or cell stress and activation induced by chemical agents [9,10,11,12,13]. Accruing literature reveals four major classes of EVs, which are sorted by size, subcellular origin, and content, named microvesicles, apoptotic bodies, virus-like particles, and exosomes [14,15,16,17,18]. The first three types of EVs are formed by outward budding of the plasma membrane. Exosomes, which are unlike other EVs, range from 30 to 100 nm in size and are formed by an intracellular endocytic trafficking pathway involving fusion of multivesicular late endocytic compartments [multivesicular bodies (MVBs)] with the plasma membrane (Figure 1). The generation of exosomes is initially formed as intraluminal vesicles (ILVs) (Figure 1) [19,20]. When an MVB fuses with the plasma membrane, the intraluminal vesicles (also called exosomes) are released to the extracellular space (Figure 1). The molecular mechanisms of MVB formation include two distinct pathways: the endosomal sorting complex required for transport (ESCRT) dependent pathway, and the ESCRT independent pathway. The ESCRT pathway requires formation of a complex by ESCRT, the sorting protein, Vps4, and the constitutive heat-shock protein, Hsp-70 [21,22,23]. The ESCRT is a family of proteins that associate in successive complexes (ESCRT-0, -I, -II, and -III) at the membranes of MVBs to regulate the formation of ILVs as well as their cargo [4]. The typical tumor susceptibility gene 101 protein (Tsg101) and Alix (encoded by PDCD6IP) for exosome identification belong to the ESCRT complex. The other pathway involves ESCRT-independent pathways. This pathway regulates the assembly of exosomes and requires Hsp70-phospholipid interactions and the activity of acid sphingomyelinase (nSMase), an enzyme that hydrolyzes sphingomyelin without the presence of ESCRTs [24,25] (Figure 1).

2. Exosome Uptake

Exosomes carry diverse cargo molecules, such as signal lipids, functional proteins, and nuclei [26,27,28,29,30], which can mediate intercellular communications by initiating a range of biological responses by acceptor cells. Upon secretion, those exosomes are released into the extracellular space and incorporated with the body fluids. Concurrently, the recipient cells uptake those exosomes through various processes. An exosome can either enter the recipient cell cytosol via phagocytosis/endocytosis or fuse with the recipient plasma membrane and directly release the cargo (Figure 1). During exosome biogenesis, diverse surface proteins are displayed on exosomes and recipient cells, including integrins, lectins/proteoglycans, and T cell immunoglobulin and mucin domain containing protein 4 (Tim4) [31,32]. Integrin plays a crucial role in the recognition of target recipient cells [33,34]. Therefore, exosomes can also interact with the recipient cells via the ligands on the cell surface in a protein–protein interaction, induce internalization, or promote intracellular signaling cascades without being internalized (Figure 1). Exosomes are responsible for a variety of normal physiological processes and abnormal pathological processes, such as the progression of neuronal degeneration [35,36], cancer [37,38], and diabetes.

3. Exosomes and Diabetes

Diabetes is part of a group of metabolic disorders characterized by long-term high blood glucose levels due to either inadequate production of insulin (type 1) or poor response of the recipient cell to insulin (type 2) [39]. According to the International Diabetes Federation (IDF), in 2017, 451 million people (aged 18–99 years) had diabetes worldwide. This number is projected to increase to 693 million by 2045 globally [40]. Thus far, the exact pathology of diabetes is still not fully understood. Studies have shown that the mechanisms underlying diabetes are complex and may involve changes at both the organ and the gene expression levels [41,42,43].

3.1. Exosomes and Type 1 Diabetes

The pathology of type 1 diabetes (T1DM) is known to be related to autoimmune disorders. A complex interaction between pancreatic β-cells and an innate or an adaptive immune system leads to the irreversible destruction of insulin-producing cells in pancreatic islets [44,45,46,47]. With more in-depth research, a relationship between exosomes and type 1 diabetes has emerged in recent years [48]. On one hand, exosomes carry active immune molecules (normally proteins or nuclei) that can activate immune cells, such as T cells and B cells, and induce β-cell apoptosis, thereby contributing to T1DM development [49,50] (Figure 2). On the other hand, researchers found that rat and human pancreatic islets also release intracellular β-cell autoantigens in human T1DM, GAD65, IA-2, and proinsulin in exosomes, which are taken up by and activate dendritic cells [13]. Moreover, insulinoma-derived exosomes stimulate the innate immune response in the Myd88-dependent pathway, which is an inflammatory signaling downstream of members of the Toll-like receptor (TLR) and the interleukin-1 (IL-1) receptor families [51], and exosomes derived from islet mesenchymal stem cell (MSCs) directly activate the T cell response and stimulate the release of interferon gamma (IFN-γ) to induce inflammation, which plays a role in the initiation of autoimmune responses in T1D [52,53] (Figure 2). In addition, some exosomes are promising as effective and practical candidates for T1D therapy. For instance, adipocyte-derived stem cell exosomes exert ameliorative effects on T1DM by multiple pathways, including modulating the immune cell response [54], attenuating podocyte damage [55], and promoting proangiogenic properties [56]. Nonetheless, exosomes can be developed as an effective tool to improve islet transplant by modulating the immune response or as a biomarker of recurrent autoimmunity for islet transplant diagnosis [57,58].

3.2. Exosomes and Type 2 Diabetes

Type 2 is the main phenotype of diabetes, accounting for more than 90% of the diabetic population [59]. It is characterized by high plasma glucose due to insulin resistance. Excessive accumulation of fat due to obesity in adipose tissue has been considered a major driver in the pathophysiology of insulin resistance [60,61,62]. Recent studies have shown that adipocytes can produce and release exosomes containing bioactive cargo and acting as a mechanism of cell-to-cell communication, which contributes to the incidence of insulin resistance. For example, adipocyte-derived exosomes carrying sonic hedgehog (SHH), a protein known to modulate immunity, induce proinflammatory or M1 polarization of bone marrow-derived macrophages and RAW 264.7 macrophages via the Ptch/PI3K signaling pathway and contribute to insulin resistance [63] (Figure 2). Additionally, exosomes derived from insulin-resistant adipocytes promote plaque burden and plaque vulnerability partly by inducing vasa vasorum angiogenesis in human umbilical vein endothelial cells [64] (Figure 2). In addition to adipocytes, exosomes derived from other cells or organs also contribute to insulin resistance, including bone marrow mesenchymal cells, human placental cells, and cardiomyocytes [65,66,67,68]. Moreover, exosomes are promising as a therapeutic agent for type 2 diabetes. Recent studies have shown that exosomes derived from umbilical cord MSCs relieve β-cell destruction and alleviate type 2 diabetic nephropathy [69,70]. Another investigation also related exosomes to the mechanism underlying the beneficial effects of exercise or bariatric surgery on type 2 diabetes [71,72,73].

4. Exosome Noncoding RNA in Diabetes

Exosomes secreted from different cells participate in the progression of diabetes in distinguishing ways; however, the role of exosomes not only depends on the origin but also is mainly decided by the cargo content. In recent years, exosome nucleic acids, especially noncoding RNA-containing exosomes, have been shown to be involved in a wide range of processes that underlie diabetic progression; those noncoding RNAs have been widely studied and were found to exert their functional roles in remote tissues, which indicated important roles in multiple biological functions through diverse mechanisms, including recruitment of epigenetic modifier proteins, control of mRNA decay and translation, and DNA sequestration of transcription factors [74,75].

4.1. MiRNA in Exosomes and Their Roles in Diabetes

MicroRNAs (miRNAs) are small, noncoding RNA molecules containing approximately 22 nucleotides. miRNAs are found in plants, animals, and some viruses that function in RNA silencing and posttranscriptional regulation of gene expression [76]. Many microRNAs, such as let-7, miR-223, miR-29, miR-103, and miR-107, are known to regulate metabolic disorders (including diabetes) through various molecular pathways, such as modulation of lipid or glucose metabolism, liver gluconeogenesis, insulin secretion, and autophagy [77,78,79,80,81]. MiRNA-carrying exosomes have advantages that work at distant tissues more efficiently. Thus, some exosomal miRNAs are pathological factors in diabetes by targeting key proteins and acting as crucial modulators of insulin sensitivity. In cells, insulin combined with its substrate, insulin receptor substrate-1 (IRS-1), stimulates a cascade signal, including phosphorylation of protein kinase B (PKB/AKT), at serine 473, leading to phosphorylated AKT promoting the translocation of glucose transporter-4 (Glut4) from the cytosol to the membrane for glucose uptake [82]. Katayama and colleagues examined the miRNA expression profile of serum-derived exosome-enriched extracellular vesicles in healthy controls and type 2 diabetic patients. They found that exosome-derived extracellular miR-20b-5p is highly abundant in type 2 diabetic patients, and further in vitro studies showed that exosomal miR-20b-5p targeted AKT-interacting protein (AKTIP), which interacts directly with AKT and modulates AKT activity by enhancing the phosphorylation of AKT regulatory sites and reducing glycogen accumulation in primary human skeletal muscle, resulting in insulin resistance [83]. Consistent with the above findings, another study demonstrated that pancreatic cancer-derived exosomes enter C2C12 myotubes and inhibit insulin and PI3K/Akt signaling, thereby preserving insulin-induced FoxO1 nuclear exclusion and impairing Glut4 trafficking. In this study, microarray methods revealed that certain exosomal microRNAs (miR-450b-3p and miR-151-3p) likely play key roles in this process (Figure 1) [84]. Further, exosome miR-155 derived from adipose tissue macrophages in obesity-induced diabetic mice causes glucose intolerance and insulin resistance by targeting PPARγ, a transcription factor and key regulator of lipid metabolism, when administered to lean mice [85]. Similarly, another elegant experiment on obese mice showed that exosomes from obese mice could induce glucose intolerance in lean mice; in this study, the authors transfected exosomes with the selected miRNA (miR-192, miR-122, miR-27a-3p, and miR-27b-3p; all were demonstrated to be increased in obese mice) and then injected the reconstituted exosomes into lean mice. Surprisingly, the lean mice developed glucose intolerance and insulin resistance as well. The data further showed that exosomal miRNAs induced diabetes in white adipose tissues in lean mice by targeting PPARα [86], which is also a transcription factor and a major regulator of lipid metabolism.
Inadequate pancreatic β-cell numbers is a pathological manifestation of type 1 diabetes. Researchers have found that lymphocyte-derived exosomes miR-142-3p and miR-142-5p from prediabetic mice transferred to pancreatic β-cells trigger the expression of genes involved in chemokine signaling, including Ccl2, Ccl7, and Cxcl10. The induction of these genes may promote the recruitment of immune cells and exacerbate β-cell death during an autoimmune attack, which also contributes to diabetes development [49]. Nonetheless, a recent study demonstrated that bone marrow mesenchymal stem cells derived exosome miR-29b-3p from aged mice mediated insulin resistance in young mice [65], which indicated that exosomal miRNAs could exert their role independently. Thus, exosome cargo with miRNAs is also considered a novel therapeutic agent. Exosome miR-222 was reduced in rabbits with type 1 diabetes; however, MSC-derived exosomes containing miR-222 injected subconjunctivally and intraocularly to these diabetic rabbits could attenuate retinal degeneration [87]. Similarly, intravenous administration of exosome miR-106b and miR-222 derived from bone narrow promoted post-injury β-cell proliferation through Cip/Kip family downregulation in insulin-deficient mice [88]. In addition, MSC-derived exosome miR-146a has anti-inflammatory effects on damaged astrocytes and prevents diabetes-induced cognitive impairment [89].
Consequently, many exosomal miRNAs have been found to be increased in the plasma or the urine of diabetic patients, making them promising circulating biomarkers associated with type 2 diabetes, such as exosomal miR-21-5p, miR-375-3p, miR-133b, miR-342, and miR-30, which are all upregulated in the serum of diabetic subjects [90,91,92], and miR-451-5p, let-7c-5p, miR-362-3p, miR-877-3p, miR-150-5p, and miR-15a-5p, which are upregulated in the urine of diabetic subjects [93,94,95]. It should be noted that, for certain miRNAs, total circulating miRNA levels are distinct from circulating extracellular vesicle miRNA content. For example, a clinical study including 19 children with type 1 diabetes along with 16 healthy controls showed that miR-21-5p derived from serum extracellular vesicles was increased threefold compared with that in nondiabetic individuals, while total serum miR-21-5p was reduced in diabetic participants [92], suggesting that exosomal RNA may be a distinct biomarker from total serum RNA levels. These data are summarized in Table 1.

4.2. Other Noncoding RNAs in Exosomes and Their Roles in Diabetes

In addition to miRNA, other noncoding RNAs, such as circular RNA (circRNAs), P-element induced Wimpy testis (PIWI)-interacting RNAs (piRNAs), and long noncoding RNA (lncRNAs), are also known to participate in the pathological process of diabetes. CircRNA is a type of noncoding RNA that forms a unique, circular, covalently closed continuous loop. This kind of RNA is resistant to exonuclease-mediated degradation because it does not have 5’ or 3’ ends and is presumably more stable than most linear RNAs. Recent evidence has shown that circRNAs usually regulate the transcription of miRNA-target genes and function as miRNA sponges. Several circRNAs have been implicated in the pathological progression of diabetes, including reducing β-cell proliferation, reducing survival, and affecting insulin secretion [96,97,98]. PiRNAs belong to another class of small RNAs that are 24–31 nucleotides in length. They were first found in germline cells and play a key role in spermatogenesis. PiRNAs interact with PIWI proteins and guide them to silence transposable elements, therefore silencing gene expression, regulating mRNA translation, or sustaining stem cell stability. Recently, researchers found that piRNAs are also expressed in pancreatic islets and participate in the control of β-cell activities [99]. However, although circRNAs and piRNAs are known to exist in exosomes, there is still a lack of evidence showing exosomal circular RNA or piRNA changes in diabetic state.
LncRNA is a kind of nonprotein-coding RNA comprising transcripts longer than 200 nucleotides. LncRNA regulates the expression of genes at epigenetic, transcriptional, and posttranscriptional levels and is similar to miRNA. In recent decades, lncRNAs such as ANRIL, H19, MALAT1, Sox2OT, and MEG3 have been implicated in playing important roles in the pathology of diabetes and all are known to associate with the pathology of type 2 diabetes [100,101,102,103,104]. Although only a few studies related exosomal lncRNAs to diabetes, a recent study did show that, although lncRNA-3134 was nearly unchanged in exosome-free samples, lncRNA-p3134 was increased four-fold in serum exosomes, and the circulating level of lncRNA-p3134 was correlated with fasting blood glucose and HOMA-β levels. In a further study, the authors found that the secretion of lncRNA-p3134 positively regulated glucose-stimulated insulin secretion by promoting key regulators (Pdx-1, MafA, GLUT2, and Tcf7l2) in β-cells, and that the upregulation of lncRNA-p3134 in db/db mice restored insulin synthesis and secretion, suggesting lncRNA-p3134 as a compensatory factor to preserve the β-cell function response to high glucose stimulation [105] (Figure 3).
As exosomes have a double membrane structure, they can protect encapsulated small RNAs from ribonucleases (RNases) in body fluid and have low antigenicity and toxicity [106]. In this case, extracellular vesicles, which include exosomes, are considered ideal carriers for nucleic acid drugs. An extracellular vesicle-mimetic nanovesicle containing lncRNA-H19 is considered an effective treatment that could remarkably accelerate the healing processes of chronic wounds induced by diabetes [107], indicating that noncoding RNA-containing exosomes can be a promising candidate for drug delivery.

5. Limitations and Conclusions

Current research examining the role of exosomes carrying noncoding RNAs in diabetes is still in its infancy, especially that involving lncRNAs, circular RNAs, or piwi-interacting RNAs. Additionally, in the current research, there are still no clear characterizations, and markers are available for exosomes from different cell types, making it difficult to determine the origin of exosomes in circulation [108,109]. In addition, although exosomes as drug vehicles have been investigated widely, there are still some obstacles in developing exosome-based drug delivery systems [110], such as encapsulation efficiency of cargo, selection of appropriate exosome origination cells, drug loading methodology [111], and exosome surface modification [112,113,114]. However, exosomal miRNA and lncRNA have been shown to modulate diabetic progress in various ways, including influencing the metabolic and the insulin signals in target tissues, affecting cell viability, and modulating inflammation in pancreatic cells. With increasing evidence, exosomes definitely appear to have promising and important roles not only in pathogenesis but also in diabetic diagnosis and therapeutics.

Author Contributions

W.C. collected all the data, W.C. and J.W. drafted and wrote the manuscript. All authors have read and approved the final version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China, grant number (81700704, 81622005), the natural science foundation of ShanDong province, grant number (ZR2017BH087).

Conflicts of Interest

The authors confirm that there are no conflicts of interest.

References

  1. Regev-Rudzki, N.; Wilson, D.W.; Carvalho, T.G.; Sisquella, X.; Coleman, B.M.; Rug, M.; Bursac, D.; Angrisano, F.; Gee, M.; Hill, A.F.; et al. Cell-Cell Communication between Malaria-Infected Red Blood Cells via Exosome-like Vesicles. Cell 2013, 153, 1120–1133. [Google Scholar] [CrossRef] [Green Version]
  2. Roy, S.; Kim, D.; Lim, R. Cell-cell communication in diabetic retinopathy. Vis. Res. 2017, 139, 115–122. [Google Scholar] [CrossRef] [PubMed]
  3. Ohyashiki, J.H.; Umezu, T.; Ohyashiki, K. Extracellular vesicle-mediated cell-cell communication in haematological neoplasms. Philos. Trans. R. Soc. B Biol. Sci. 2018, 373, 20160484. [Google Scholar] [CrossRef] [PubMed]
  4. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  5. Pérez-Bermúdez, P.; Blesa, J.; Soriano, J.M.; Marcilla, A. Extracellular vesicles in food: Experimental evidence of their secretion in grape fruits. Eur. J. Pharm. Sci. 2017, 98, 40–50. [Google Scholar] [CrossRef] [PubMed]
  6. Todorova, D.; Simoncini, S.; Lacroix, R.; Sabatier, F.; Dignat-George, F. Extracellular Vesicles in Angiogenesis. Circ. Res. 2017, 120, 1658–1673. [Google Scholar] [CrossRef] [PubMed]
  7. Merchant, M.L.; Rood, I.M.; Deegens, J.K.J.; Klein, J.B. Isolation and characterization of urinary extracellular vesicles: Implications for biomarker discovery. Nat. Rev. Nephrol. 2017, 13, 731–749. [Google Scholar] [CrossRef] [PubMed]
  8. Choi, J.W.; Um, J.H.; Cho, J.H.; Lee, H.J. Tiny RNAs and their voyage via extracellular vesicles: Secretion of bacterial small RNA and eukaryotic microRNA. Exp. Biol. Med. 2017, 242, 1475–1481. [Google Scholar] [CrossRef]
  9. Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental pH Is a Key Factor for Exosome Traffic in Tumor Cells. J. Biol. Chem. 2009, 284, 34211–34222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Yu, M.; Song, W.; Tian, F.; Dai, Z.; Zhu, Q.; Ahmad, E.; Guo, S.; Zhu, C.; Zhong, H.; Yuan, Y.; et al. Temperature- and rigidity-mediated rapid transport of lipid nanovesicles in hydrogels. Proc. Natl. Acad. Sci. USA 2019, 116, 5362–5369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Jelonek, K.; Widlak, P.; Pietrowska, M. The Influence of Ionizing Radiation on Exosome Composition, Secretion and Intercellular Communication. Protein Pept. Lett. 2016, 23, 656–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kucharzewska, P.; Belting, M. Emerging roles of extracellular vesicles in the adaptive response of tumour cells to microenvironmental stress. J. Extracell. Vesicles 2013, 2, 214074. [Google Scholar] [CrossRef] [PubMed]
  13. Cianciaruso, C.; Phelps, E.A.; Pasquier, M.; Hamelin, R.; Demurtas, D.; Ahmed, M.A.; Piemonti, L.; Hirosue, S.; Swartz, M.A.; De Palma, M.; et al. Primary Human and Rat beta-Cells Release the Intracellular Autoantigens GAD65, IA-2, and Proinsulin in Exosomes Together With Cytokine-Induced Enhancers of Immunity. Diabetes 2017, 66, 460–473. [Google Scholar] [CrossRef] [PubMed]
  14. Van der Pol, E.; Böing, A.N.; Harrison, P.; Sturk, A.; Nieuwland, R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol. Rev. 2012, 64, 676–705. [Google Scholar] [CrossRef] [PubMed]
  15. Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef] [PubMed]
  16. Stahl, P.D.; Raposo, G. Extracellular Vesicles: Exosomes and Microvesicles, Integrators of Homeostasis. Physiology 2019, 34, 169–177. [Google Scholar] [CrossRef] [PubMed]
  17. Hauser, P.; Wang, S.; Didenko, V.V. Apoptotic Bodies: Selective Detection in Extracellular Vesicles. Methods Mol. Biol. 2017, 1554, 193–200. [Google Scholar]
  18. Reiter, K.; Aguilar, P.P.; Wetter, V.; Steppert, P.; Tover, A.; Jungbauer, A. Separation of virus-like particles and extracellular vesicles by flow-through and heparin affinity chromatography. J. Chromatogr. A 2019, 1588, 77–84. [Google Scholar] [CrossRef]
  19. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  20. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  21. Juan, T.; Furthauer, M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin. Cell Dev. Biol. 2018, 74, 66–77. [Google Scholar] [CrossRef] [PubMed]
  22. Adell, M.A.Y.; Migliano, S.M.; Teis, D. ESCRT-III and Vps4: A dynamic multipurpose tool for membrane budding and scission. FEBS J. 2016, 283, 3288–3302. [Google Scholar] [CrossRef]
  23. Dreyer, F.; Baur, A.; Federico, M. Biogenesis and Functions of Exosomes and Extracellular Vesicles. Methods Mol. Biol. 2016, 1448, 201–216. [Google Scholar] [PubMed]
  24. Babst, M. MVB Vesicle Formation: ESCRT-Dependent, ESCRT-Independent and Everything in Between. Curr. Opin. Cell Biol. 2011, 23, 452–457. [Google Scholar] [CrossRef] [PubMed]
  25. Van Niel, G.; Charrin, S.; Simoes, S.; Romao, M.; Rochin, L.; Saftig, P.; Marks, M.S.; Rubinstein, E.; Raposo, G. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev. Cell 2011, 21, 708–721. [Google Scholar] [CrossRef]
  26. Skotland, T.; Sandvig, K.; Llorente, A. Lipids in exosomes: Current knowledge and the way forward. Prog. Lipid Res. 2017, 66, 30–41. [Google Scholar] [CrossRef]
  27. Skotland, T.; Ekroos, K.; Kauhanen, D.; Simolin, H.; Seierstad, T.; Berge, V.; Sandvig, K.; Llorente, A. Molecular lipid species in urinary exosomes as potential prostate cancer biomarkers. Eur. J. Cancer 2017, 70, 122–132. [Google Scholar] [CrossRef]
  28. Li, W.; Li, C.; Zhou, T.; Liu, X.; Liu, X.; Li, X.; Chen, D. Role of exosomal proteins in cancer diagnosis. Mol. Cancer 2017, 16, 145. [Google Scholar] [CrossRef] [PubMed]
  29. Chen, T.; Xi, Q.Y.; Sun, J.J.; Ye, R.S.; Cheng, X.; Sun, R.P.; Wang, S.B.; Shu, G.; Wang, L.N.; Zhu, X.T.; et al. Revelation of mRNAs and proteins in porcine milk exosomes by transcriptomic and proteomic analysis. BMC Vet. Res. 2017, 13, 496. [Google Scholar] [CrossRef]
  30. Shurtleff, M.J.; Yao, J.; Qin, Y.; Nottingham, R.M.; Temoche-Diaz, M.M.; Schekman, R.; Lambowitz, A.M. Broad role for YBX1 in defining the small noncoding RNA composition of exosomes. Proc. Natl. Acad. Sci. USA 2017, 114, E8987–E8995. [Google Scholar] [CrossRef] [Green Version]
  31. Mulcahy, L.A.; Pink, R.C.; Carter, D.R.F. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 1093. [Google Scholar] [CrossRef] [PubMed]
  32. Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes Facilitate Therapeutic Targeting of Oncogenic Kras in Pancreatic Cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef] [PubMed]
  33. Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Mark, M.T.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Christianson, H.C.; Svensson, K.J.; Van Kuppevelt, T.H.; Li, J.P.; Belting, M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc. Natl. Acad. Sci. USA 2013, 110, 17380–17385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Wang, R.; Ding, Q.; Yaqoob, U.; De Assuncao, T.M.; Verma, V.K.; Hirsova, P.; Cao, S.; Mukhopadhyay, D.; Huebert, R.C.; Shah, V.H. Exosome Adherence and Internalization by Hepatic Stellate Cells Triggers Sphingosine 1-Phosphate-dependent Migration. J. Biol. Chem. 2015, 290, 30684–30696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ochieng, J.; Nangami, G.; Sakwe, A.; Rana, T.; Ingram, S.; Goodwin, J.S.; Moye, C.; Lammers, P.; Adunyah, S.E.; Goodwin, J.S. Extracellular histones are the ligands for the uptake of exosomes and hydroxyapatite-nanoparticles by tumor cells via syndecan-4. FEBS Lett. 2018, 592, 3274–3285. [Google Scholar] [CrossRef] [PubMed]
  37. Kulkarni, B.; Kirave, P.; Gondaliya, P.; Jash, K.; Jain, A.; Tekade, R.K.; Kalia, K. Exosomal miRNA in chemoresistance, immune evasion, metastasis and progression of cancer. Drug Discov. Today 2019. [Google Scholar] [CrossRef]
  38. Stefanius, K.; Servage, K.; Santos, M.D.S.; Gray, H.F.; Toombs, J.; Chimalapati, S.; Kim, M.S.; Malladi, V.S.; Brekken, R.; Orth, K. Human pancreatic cancer cell exosomes, but not human normal cell exosomes, act as an initiator in cell transformation. Elife 2019, 8, 40226. [Google Scholar] [CrossRef]
  39. Shi, Y.; Hu, F.B. The global implications of diabetes and cancer. Lancet 2014, 383, 1947–1948. [Google Scholar] [CrossRef]
  40. Cho, N.; Shaw, J.; Karuranga, S.; Huang, Y.; Fernandes, J.D.R.; Ohlrogge, A.; Malanda, B. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res. Clin. Prat. 2018, 138, 271–281. [Google Scholar] [CrossRef]
  41. Naidoo, V.; Naidoo, M.; Ghai, M. Cell and tissue-specific epigenetic changes associated with chronic inflammation in insulin resistance and type 2 diabetes mellitus. Scand. J. Immunol. 2018, 88, e12723. [Google Scholar] [CrossRef] [PubMed]
  42. Yahagi, K.; Kolodgie, F.D.; Lutter, C.; Mori, H.; Romero, M.E.; Finn, A.V.; Virmani, R. Pathology of Human Coronary and Carotid Artery Atherosclerosis and Vascular Calcification in Diabetes Mellitus. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 191–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ashcroft, F.M.; Rorsman, P. Diabetes mellitus and the beta cell: The last ten years. Cell 2012, 148, 1160–1171. [Google Scholar] [CrossRef] [PubMed]
  44. Buzzetti, R.; Zampetti, S.; Maddaloni, E. Adult-onset autoimmune diabetes: Current knowledge and implications for management. Nat. Rev. Endocrinol. 2017, 13, 674–686. [Google Scholar] [CrossRef] [PubMed]
  45. Kakleas, K.; Soldatou, A.; Karachaliou, F.; Karavanaki, K.; Kostas, K.; Alexandra, S.; Feneli, K.; Kyriaki, K. Associated autoimmune diseases in children and adolescents with type 1 diabetes mellitus (T1DM). Autoimmun. Rev. 2015, 14, 781–797. [Google Scholar] [CrossRef]
  46. Bonifacio, E.; Mathieu, C.; Nepom, G.T.; Ziegler, A.G.; Anhalt, H.; Haller, M.J.; Harrison, L.C.; Hebrok, M.; Kushner, J.A.; Norris, J.M.; et al. Rebranding asymptomatic type 1 diabetes: The case for autoimmune beta cell disorder as a pathological and diagnostic entity. Diabetologia 2017, 60, 35–38. [Google Scholar] [CrossRef]
  47. Kahaly, G.J.; Hansen, M.P. Type 1 diabetes associated autoimmunity. Autoimmun. Rev. 2016, 15, 644–648. [Google Scholar] [CrossRef]
  48. Lukić, M.L.; Pejnovic, N.; Lukić, A. New Insight into Early Events in Type 1 Diabetes: Role for Islet Stem Cell Exosomes. Diabetes 2014, 63, 835–837. [Google Scholar] [CrossRef]
  49. Guay, C.; Kruit, J.K.; Rome, S.; Menoud, V.; Mulder, N.L.; Jurdzinski, A.; Mancarella, F.; Sebastiani, G.; Donda, A.; Gonzalez, B.J.; et al. Lymphocyte-Derived Exosomal MicroRNAs Promote Pancreatic beta Cell Death and May Contribute to Type 1 Diabetes Development. Cell Metab. 2019, 29, 348–361. [Google Scholar] [CrossRef]
  50. Dai, Y.D.; Sheng, H.; Dias, P.; Rahman, M.J.; Bashratyan, R.; Regn, D.; Marquardt, K. Autoimmune Responses to Exosomes and Candidate Antigens Contribute to Type 1 Diabetes in Non-Obese Diabetic Mice. Curr. Diabetes Rep. 2017, 17, 130. [Google Scholar] [CrossRef]
  51. Sheng, H.; Hassanali, S.; Nugent, C.; Wen, L.; Hamilton-Williams, E.; Dias, P.; Dai, Y.D. Insulinoma-released exosomes or microparticles are immunostimulatory and can activate autoreactive T cells spontaneously developed in non-obese diabetes mice1. J. Immunol. 2011, 187, 1591–1600. [Google Scholar] [CrossRef] [PubMed]
  52. Klein, L.; Hinterberger, M.; Von Rohrscheidt, J.; Aichinger, M. Autonomous versus dendritic cell-dependent contributions of medullary thymic epithelial cells to central tolerance. Trends Immunol. 2011, 32, 188–193. [Google Scholar] [CrossRef]
  53. Malhotra, D.; Fletcher, A.L.; Astarita, J.; Lukacs-Kornek, V.; Tayalia, P.; Gonzalez, S.F.; Elpek, K.G.; Chang, S.K.; Knoblich, K.; Hemler, M.E.; et al. Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks. Nat. Immunol. 2012, 13, 499–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Nojehdehi, S.; Soudi, S.; Hesampour, A.; Rasouli, S.; Soleimani, M.; Hashemi, S.M. Immunomodulatory effects of mesenchymal stem cell-derived exosomes on experimental type-1 autoimmune diabetes. J. Cell. Biochem. 2018, 119, 9433–9443. [Google Scholar] [CrossRef]
  55. Jin, J.; Shi, Y.; Gong, J.; Zhao, L.; Li, Y.; He, Q.; Huang, H. Exosome secreted from adipose-derived stem cells attenuates diabetic nephropathy by promoting autophagy flux and inhibiting apoptosis in podocyte. Stem Cell Res. Ther. 2019, 10, 95. [Google Scholar] [CrossRef]
  56. Zhu, L.L.; Huang, X.; Yu, W.; Chen, H.; Chen, Y.; Dai, Y.T. Transplantation of adipose tissue-derived stem cell-derived exosomes ameliorates erectile function in diabetic rats. Andrologia 2018, 50, e12871. [Google Scholar] [CrossRef] [PubMed]
  57. Wen, D.; Peng, Y.; Liu, D.; Weizmann, Y.; Mahato, R.I. Mesenchymal stem cell and derived exosome as small RNA carrier and Immunomodulator to improve islet transplantation. J. Control. Release 2016, 238, 166–175. [Google Scholar] [CrossRef] [PubMed]
  58. Korutla, L.; Rickels, M.R.; Hu, R.W.; Freas, A.; Reddy, S.; Habertheuer, A.; Harmon, J.; Korutla, V.; Ram, C.; Naji, A.; et al. Noninvasive diagnosis of recurrent autoimmune type 1 diabetes after islet cell transplantation. Am. J. Transplant. 2019, 19, 1852–1858. [Google Scholar] [CrossRef] [PubMed]
  59. Shah, M.; Vella, A. What is type 2 diabetes? Medicine 2014, 42, 687–691. [Google Scholar] [CrossRef]
  60. Czech, M.P. Insulin action and resistance in obesity and type 2 diabetes. Nat. Med. 2017, 23, 804–814. [Google Scholar] [CrossRef]
  61. Tsai, S.; Clemente-Casares, X.; Revelo, X.S.; Winer, S.; Winer, D.A. Are Obesity-Related Insulin Resistance and Type 2 Diabetes Autoimmune Diseases? Diabetes 2015, 64, 1886–1897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kahn, S.E.; Hull, R.L.; Utzschneider, K.M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 2006, 444, 840–846. [Google Scholar] [CrossRef] [PubMed]
  63. Song, M.; Han, L.; Chen, F.F.; Wang, D.; Wang, F.; Zhang, L.; Wang, Z.H.; Zhong, M.; Tang, M.X.; Zhang, W. Adipocyte-Derived Exosomes Carrying Sonic Hedgehog Mediate M1 Macrophage Polarization-Induced Insulin Resistance via Ptch and PI3K Pathways. Cell. Physiol. Biochem. 2018, 48, 1416–1432. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, F.; Chen, F.F.; Shang, Y.Y.; Li, Y.; Wang, Z.H.; Han, L.; Li, Y.H.; Zhang, L.; Ti, Y.; Zhang, W.; et al. Insulin resistance adipocyte-derived exosomes aggravate atherosclerosis by increasing vasa vasorum angiogenesis in diabetic ApoE mice. Int. J. Cardiol. 2018, 265, 181–187. [Google Scholar] [CrossRef] [PubMed]
  65. Su, T.; Xiao, Y.; Xiao, Y.; Guo, Q.; Li, C.; Huang, Y.; Deng, Q.; Wen, J.; Zhou, F.; Luo, X.H. Bone Marrow Mesenchymal Stem Cells-Derived Exosomal MiR-29b-3p Regulates Aging-Associated Insulin Resistance. ACS Nano 2019, 13, 2450–2462. [Google Scholar] [CrossRef] [PubMed]
  66. Nair, S.; Jayabalan, N.; Guanzon, D.; Palma, C.; Scholz-Romero, K.; Elfeky, O.; Zuñiga, F.; Ormazabal, V.; Diaz, E.; Rice, G.E.; et al. Human placental exosomes in gestational diabetes mellitus carry a specific set of miRNAs associated with skeletal muscle insulin sensitivity. Clin. Sci. 2018, 132, 2451–2467. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, X.; Gu, H.; Huang, W.; Peng, J.; Li, Y.; Yang, L.; Qin, D.; Essandoh, K.; Wang, Y.; Peng, T.; et al. Hsp20-Mediated Activation of Exosome Biogenesis in Cardiomyocytes Improves Cardiac Function and Angiogenesis in Diabetic Mice. Diabetes 2016, 65, 3111–3128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Tang, S.; Luo, F.; Feng, Y.M.; Wei, X.; Miao, H.; Lu, Y.B.; Tang, Y.; Ding, D.F.; Jin, J.F.; Zhu, Q. Neutral Ceramidase Secreted Via Exosome Protects Against Palmitate-Induced Apoptosis in INS-1 Cells. Exp. Clin. Endocrinol. Diabetes 2017, 125, 130–135. [Google Scholar] [CrossRef]
  69. Sun, Y.; Shi, H.; Yin, S.; Ji, C.; Zhang, X.; Zhang, B.; Wu, P.; Shi, Y.; Mao, F.; Yan, Y.; et al. Human Mesenchymal Stem Cell Derived Exosomes Alleviate Type 2 Diabetes Mellitus by Reversing Peripheral Insulin Resistance and Relieving beta-Cell Destruction. ACS Nano 2018, 12, 7613–7628. [Google Scholar] [CrossRef]
  70. Nagaishi, K.; Mizue, Y.; Chikenji, T.; Otani, M.; Nakano, M.; Saijo, Y.; Tsuchida, H.; Ishioka, S.; Nishikawa, A.; Saito, T.; et al. Umbilical cord extracts improve diabetic abnormalities in bone marrow-derived mesenchymal stem cells and increase their therapeutic effects on diabetic nephropathy. Sci. Rep. 2017, 7, 8484. [Google Scholar] [CrossRef]
  71. Li, G.; Liu, H.; Ma, C.; Chen, Y.; Wang, J.; Yang, Y. Exosomes are the novel players involved in the beneficial effects of exercise on type 2 diabetes. J. Cell. Physiol. 2019, 234, 14896–14905. [Google Scholar] [CrossRef] [PubMed]
  72. Safdar, A.; Saleem, A.; Tarnopolsky, M.A. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat. Rev. Endocrinol. 2016, 12, 504–517. [Google Scholar] [CrossRef] [PubMed]
  73. Witczak, J.K.; Min, T.; Prior, S.L.; Stephens, J.W.; James, P.; Rees, A. Bariatric Surgery Is Accompanied by Changes in Extracellular Vesicle-Associated and Plasma Fatty Acid Binding Protein 4. Obes. Surg. 2018, 28, 767–774. [Google Scholar] [CrossRef] [PubMed]
  74. Chang, W. Non-coding RNAs and Berberine: A new mechanism of its anti-diabetic activities. Eur. J. Pharmacol. 2017, 795, 8–12. [Google Scholar] [CrossRef] [PubMed]
  75. Wong, W.K.; Sørensen, A.E.; Joglekar, M.V.; Hardikar, A.A.; Dalgaard, L.T. Non-Coding RNA in Pancreas and beta-Cell Development. Noncoding RNA 2018, 4, E41. [Google Scholar] [PubMed]
  76. Vishnoi, A.; Rani, S. MiRNA Biogenesis and Regulation of Diseases: An Overview. Methods Mol. Biol. 2017, 1509, 1–10. [Google Scholar]
  77. Brennan, E.; Wang, B.; McClelland, A.; Mohan, M.; Marai, M.; Beuscart, O.; Derouiche, S.; Gray, S.; Pickering, R.; Tikellis, C.; et al. Protective Effect of let-7 miRNA Family in Regulating Inflammation in Diabetes-Associated Atherosclerosis. Diabetes 2017, 66, 2266–2277. [Google Scholar] [CrossRef] [Green Version]
  78. Jiang, L.Q.; Franck, N.; Egan, B.; Sjögren, R.J.O.; Katayama, M.; Duque-Guimaraes, D.; Arner, P.; Zierath, J.R.; Krook, A. Autocrine role of interleukin-13 on skeletal muscle glucose metabolism in type 2 diabetic patients involves microRNA let-7. Am. J. Physiol. Metab. 2013, 305, E1359–E1366. [Google Scholar] [CrossRef] [Green Version]
  79. Vickers, K.C.; Landstreet, S.R.; Levin, M.G.; Shoucri, B.M.; Toth, C.L.; Taylor, R.C.; Palmisano, B.T.; Tabet, F.; Cui, H.L.; Rye, K.A.; et al. MicroRNA-223 coordinates cholesterol homeostasis. Proc. Natl. Acad. Sci. USA 2014, 111, 14518–14523. [Google Scholar] [CrossRef] [Green Version]
  80. Massart, J.; Sjögren, R.J.; Lundell, L.S.; Mudry, J.M.; Franck, N.; O’Gorman, D.J.; Egan, B.; Zierath, J.R.; Krook, A. Altered miR-29 Expression in Type 2 Diabetes Influences Glucose and Lipid Metabolism in Skeletal Muscle. Diabetes 2017, 66, 1807–1818. [Google Scholar] [CrossRef] [Green Version]
  81. Foley, N.H.; O’Neill, L.A. miR-107: A Toll-like receptor-regulated miRNA dysregulated in obesity and type II diabetes. J. Leukoc. Biol. 2012, 92, 521–527. [Google Scholar] [CrossRef] [PubMed]
  82. Nicholson, K.M.; Anderson, N.G. The protein kinase B/Akt signalling pathway in human malignancy. Cell. Signal. 2002, 14, 381–395. [Google Scholar] [CrossRef]
  83. Katayama, M.; Wiklander, O.P.; Fritz, T.; Caidahl, K.; Andaloussi, S.E.; Zierath, J.R.; Krook, A. Circulating Exosomal miR-20b-5p is Elevated in Type 2 Diabetes and Could Impair Insulin Action in Human Skeletal Muscle. Diabetes 2018, 68, 515–526. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, L.; Zhang, B.; Zheng, W.; Kang, M.; Chen, Q.; Qin, W.; Li, C.; Zhang, Y.; Shao, Y.; Wu, Y. Exosomes derived from pancreatic cancer cells induce insulin resistance in C2C12 myotube cells through the PI3K/Akt/FoxO1 pathway. Sci. Rep. 2017, 7, 5384. [Google Scholar] [CrossRef] [PubMed]
  85. Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell 2017, 171, 372–384. [Google Scholar] [CrossRef] [PubMed]
  86. Castaño, C.; Kalko, S.; Novials, A.; Párrizas, M. Obesity-associated exosomal miRNAs modulate glucose and lipid metabolism in mice. Proc. Natl. Acad. Sci. USA 2018, 115, 12158–12163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Safwat, A.; Sabry, D.; Ragiae, A.; Amer, E.; Mahmoud, R.; Shamardan, R. Adipose mesenchymal stem cells–derived exosomes attenuate retina degeneration of streptozotocin-induced diabetes in rabbits. J. Circ. Biomark. 2018, 7, 1849454418807827. [Google Scholar] [CrossRef] [PubMed]
  88. Tsukita, S.; Yamada, T.; Takahashi, K.; Munakata, Y.; Hosaka, S.; Takahashi, H.; Gao, J.; Shirai, Y.; Kodama, S.; Asai, Y.; et al. MicroRNAs 106b and 222 Improve Hyperglycemia in a Mouse Model of Insulin-Deficient Diabetes via Pancreatic beta-Cell Proliferation. EBioMedicine 2017, 15, 163–172. [Google Scholar] [CrossRef]
  89. Kubota, K.; Nakano, M.; Kobayashi, E.; Mizue, Y.; Chikenji, T.; Otani, M.; Nagaishi, K.; Fujimiya, M. An enriched environment prevents diabetes-induced cognitive impairment in rats by enhancing exosomal miR-146a secretion from endogenous bone marrow-derived mesenchymal stem cells. PLoS ONE 2018, 13, e0204252. [Google Scholar] [CrossRef]
  90. Eissa, S.; Matboli, M.; Bekhet, M.M. Clinical verification of a novel urinary microRNA panal: 133b, 342 and 30 as biomarkers for diabetic nephropathy identified by bioinformatics analysis. Biomed. Pharmacother. 2016, 83, 92–99. [Google Scholar] [CrossRef]
  91. Fu, Q.; Jiang, H.; Yang, T. Injury Factors Alter miRNAs Profiles of Exosomes Derived from Islets and Circulation. Diabetes 2018, 67, 3986–3999. [Google Scholar] [CrossRef]
  92. Lakhter, A.J.; Pratt, R.E.; Moore, R.E.; Doucette, K.K.; Maier, B.F.; DiMeglio, L.A.; Sims, E.K. 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 2018, 61, 1124–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Li, W.; Yang, S.; Qiao, R.; Zhang, J. Potential Value of Urinary Exosome-Derived let-7c-5p in the Diagnosis and Progression of Type II Diabetic Nephropathy. Clin. Lab. 2018, 64, 709–718. [Google Scholar] [CrossRef] [PubMed]
  94. Mohan, A.; Singh, R.S.; Kumari, M.; Garg, D.; Upadhyay, A.; Ecelbarger, C.M.; Tripathy, S.; Tiwari, S. Urinary Exosomal microRNA-451-5p Is a Potential Early Biomarker of Diabetic Nephropathy in Rats. PLoS ONE 2016, 11, e0154055. [Google Scholar] [CrossRef] [PubMed]
  95. Xie, Y.; Jia, Y.; Cuihua, X.; Hu, F.; Xue, M.; Xue, Y. Urinary Exosomal MicroRNA Profiling in Incipient Type 2 Diabetic Kidney Disease. J. Diabetes Res. 2017, 2017, 1–10. [Google Scholar] [CrossRef] [PubMed]
  96. Fang, Y.; Wang, X.; Li, W.; Han, J.; Jin, J.; Su, F.; Zhang, J.; Huang, W.; Xiao, F.; Pan, Q.; et al. Screening of circular RNAs and validation of circANKRD36 associated with inflammation in patients with type 2 diabetes mellitus. Int. J. Mol. Med. 2018, 42, 1865–1874. [Google Scholar] [CrossRef] [PubMed]
  97. Stoll, L.; Sobel, J.; Rodriguez-Trejo, A.; Guay, C.; Lee, K.; Venø, M.T.; Kjems, J.; Laybutt, D.R.; Regazzi, R. Circular RNAs as novel regulators of beta-cell functions in normal and disease conditions. Mol. Metab. 2018, 9, 69–83. [Google Scholar] [CrossRef] [PubMed]
  98. Xu, H.; Guo, S.; Li, W.; Yu, P. The circular RNA Cdr1as, via miR-7 and its targets, regulates insulin transcription and secretion in islet cells. Sci. Rep. 2015, 5, 12453. [Google Scholar] [CrossRef] [PubMed]
  99. Henaoui, I.S.; Jacovetti, C.; Mollet, I.G.; Guay, C.; Sobel, J.; Eliasson, L.; Regazzi, R. PIWI-interacting RNAs as novel regulators of pancreatic beta cell function. Diabetologia 2017, 60, 1977–1986. [Google Scholar] [CrossRef] [PubMed]
  100. Kong, Y.; Hsieh, C.H.; Alonso, L.C. ANRIL: A lncRNA at the CDKN2A/B Locus with Roles in Cancer and Metabolic Disease. Front. Endocrinol. 2018, 9, 405. [Google Scholar] [CrossRef]
  101. Gao, Y.; Wu, F.; Zhou, J.; Yan, L.; Jurczak, M.J.; Lee, H.Y.; Yang, L.; Mueller, M.; Zhou, X.B.; Dandolo, L.; et al. The H19/let-7 double-negative feedback loop contributes to glucose metabolism in muscle cells. Nucleic Acids Res. 2014, 42, 13799–13811. [Google Scholar] [CrossRef] [PubMed]
  102. Hu, M.; Wang, R.; Li, X.; Fan, M.; Lin, J.; Zhen, J.; Chen, L.; Lv, Z. LncRNA MALAT1 is dysregulated in diabetic nephropathy and involved in high glucose-induced podocyte injury via its interplay with beta-catenin. J. Cell. Mol. Med. 2017, 21, 2732–2747. [Google Scholar] [CrossRef] [PubMed]
  103. Li, C.P.; Wang, S.H.; Wang, W.Q.; Song, S.G.; Liu, X.M. Long Noncoding RNA-Sox2OT Knockdown Alleviates Diabetes Mellitus-Induced Retinal Ganglion Cell (RGC) injury. Cell. Mol. Neurobiol. 2017, 37, 361–369. [Google Scholar] [CrossRef] [PubMed]
  104. Kameswaran, V.; Bramswig, N.C.; McKenna, L.B.; Penn, M.; Schug, J.; Hand, N.J.; Chen, Y.; Choi, I.; Vourekas, A.; Won, K.J.; et al. Epigenetic regulation of the DLK1-MEG3 microRNA cluster in human type 2 diabetic islets. Cell Metab. 2014, 19, 135–145. [Google Scholar] [CrossRef] [PubMed]
  105. Ruan, Y.; Lin, N.; Ma, Q.; Chen, R.; Zhang, Z.; Wen, W.; Chen, H.; Sun, J. Circulating LncRNAs Analysis in Patients with Type 2 Diabetes Reveals Novel Genes Influencing Glucose Metabolism and Islet beta-Cell Function. Cell. Physiol. Biochem. 2018, 46, 335–350. [Google Scholar] [CrossRef] [PubMed]
  106. Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208. [Google Scholar] [CrossRef] [Green Version]
  107. Tao, S.C.; Rui, B.Y.; Wang, Q.Y.; Zhou, D.; Zhang, Y.; Guo, S.C. Extracellular vesicle-mimetic nanovesicles transport LncRNA-H19 as competing endogenous RNA for the treatment of diabetic wounds. Drug Deliv. 2018, 25, 241–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Yamashita, T.; Takahashi, Y.; Nishikawa, M.; Takakura, Y. Effect of exosome isolation methods on physicochemical properties of exosomes and clearance of exosomes from the blood circulation. Eur. J. Pharm. Biopharm. 2016, 98, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Li, P.; Kaslan, M.; Lee, S.H.; Yao, J.; Gao, Z. Progress in Exosome Isolation Techniques. Theranostics 2017, 7, 789–804. [Google Scholar] [CrossRef]
  110. Fitts, C.A.; Ji, N.; Li, Y.; Tan, C. Exploiting Exosomes in Cancer Liquid Biopsies and Drug Delivery. Adv. Healthc. Mater. 2019, 8, e1801268. [Google Scholar] [CrossRef]
  111. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef] [PubMed]
  112. Wiklander, O.P.B.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tamura, R.; Uemoto, S.; Tabata, Y. Augmented liver targeting of exosomes by surface modification with cationized pullulan. Acta Biomater. 2017, 57, 274–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Fang, X.; Duan, Y.; Adkins, G.B.; Pan, S.; Wang, H.; Liu, Y.; Zhong, W. Highly Efficient Exosome Isolation and Protein Analysis by an Integrated Nanomaterial-Based Platform. Anal. Chem. 2018, 90, 2787–2795. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Exosome biogenesis and regulation. Schematic representation of exosome biogenesis and uptake. Exosomes are initially formed as intraluminal vesicles by multivesicular bodies (MVBs). MVB formation has two mechanisms. The endosomal sorting complex required for transport (ESCRT)-dependent pathway requires a complex constituent of ESCRT, Vps4 (vacuolar protein sorting-associated protein), and Hsp70 (heat-shock protein). The typical proteins Tsg101 and Alix for exosome identification belong to the ESCRT complex and represent ESCRT. The other pathway involves ESCRT-independent pathways, which are dependent on Hsp70 binding phospholipids to assemble MVBs. After secretion, the recipient cells mainly uptake those exosomes in three ways: (1) endocytosis, by which exosomes enter into the recipient cell cytosol directly; (2) fusion, by which the exosome cargo is mainly released to the recipient cytosol; or (3) ligands, by which exosomes interact with the recipient cells either by inducing internalization or eliciting intracellular signaling cascades without being internalized.
Figure 1. Exosome biogenesis and regulation. Schematic representation of exosome biogenesis and uptake. Exosomes are initially formed as intraluminal vesicles by multivesicular bodies (MVBs). MVB formation has two mechanisms. The endosomal sorting complex required for transport (ESCRT)-dependent pathway requires a complex constituent of ESCRT, Vps4 (vacuolar protein sorting-associated protein), and Hsp70 (heat-shock protein). The typical proteins Tsg101 and Alix for exosome identification belong to the ESCRT complex and represent ESCRT. The other pathway involves ESCRT-independent pathways, which are dependent on Hsp70 binding phospholipids to assemble MVBs. After secretion, the recipient cells mainly uptake those exosomes in three ways: (1) endocytosis, by which exosomes enter into the recipient cell cytosol directly; (2) fusion, by which the exosome cargo is mainly released to the recipient cytosol; or (3) ligands, by which exosomes interact with the recipient cells either by inducing internalization or eliciting intracellular signaling cascades without being internalized.
Cells 08 00853 g001
Figure 2. Exosomes regulate diabetic pathological process. Schematic representation of exosomes participating in the pathologies of type 1 diabetes (T1D) and type 2 diabetes (T2D). (a) Immune cell-derived exosomes can activate immune cells and induce β-cell apoptosis and cell death. (b) In certain models, pancreatic islets also release intracellular β-cell autoantigens in exosomes in turn to favor the immune response. (c) Adipocyte-derived exosomes carrying sonic hedgehog (SHH), a protein known to modulate immunity, induce proinflammatory or M1 polarization of bone marrow-derived macrophages and induce insulin resistance in the main insulin-sensitive organs (skeletal muscle, liver, adipose).
Figure 2. Exosomes regulate diabetic pathological process. Schematic representation of exosomes participating in the pathologies of type 1 diabetes (T1D) and type 2 diabetes (T2D). (a) Immune cell-derived exosomes can activate immune cells and induce β-cell apoptosis and cell death. (b) In certain models, pancreatic islets also release intracellular β-cell autoantigens in exosomes in turn to favor the immune response. (c) Adipocyte-derived exosomes carrying sonic hedgehog (SHH), a protein known to modulate immunity, induce proinflammatory or M1 polarization of bone marrow-derived macrophages and induce insulin resistance in the main insulin-sensitive organs (skeletal muscle, liver, adipose).
Cells 08 00853 g002
Figure 3. The molecular mechanisms by which exosomes containing miRNA and long noncoding RNA (lncRNA) regulate communication between organs. In the diabetic state, miRNA- and lncRNA-carrying exosomes exert their biological function by targeting different tissues. (1) Exosome lncRNA-p3134 was downregulated in a diabetic model, and lncRNA-p3134 could positively regulate glucose-stimulated insulin secretion by promoting key regulators (Pdx-1, MafA, GLUT2, and Tcf7l2) in β-cells. In this case, downregulation of lncRNA-p3134 contributed to the pathology of diabetes. (2) Exosomal miRNA (miR-20b-5p, miR-155, miR-450b-3p, miR151-3p, and miR-29b-3p) was upregulated in the diabetic model and targeted skeletal muscles via various insulin signaling regulatory proteins (PPAR, AKT, GLUT4, and FOXO) and then contributed to the pathology of diabetes. (3) Exosomal miRNA (miR-122, miR-192, miR-27a/b, miR-155, miR-29b-3p) was upregulated in diabetic models and targeted adipocytes via PPAR proteins and then contributed to the pathology of diabetes. (4) Exosomal miRNA (miR-142-3p and miR-142-5p) was upregulated in diabetic models and targeted pancreatic cells via upregulated cytokines, which then contributed to the pathology of diabetes. (5) Other exosomal miRNAs (miR-106, miR-146a, miR-222, and miR-486) are promising therapeutic agents to protect pancreatic cells or treat diabetic complications by targeting certain organs, such as astrocytes, retinal tissue, and renal cells.
Figure 3. The molecular mechanisms by which exosomes containing miRNA and long noncoding RNA (lncRNA) regulate communication between organs. In the diabetic state, miRNA- and lncRNA-carrying exosomes exert their biological function by targeting different tissues. (1) Exosome lncRNA-p3134 was downregulated in a diabetic model, and lncRNA-p3134 could positively regulate glucose-stimulated insulin secretion by promoting key regulators (Pdx-1, MafA, GLUT2, and Tcf7l2) in β-cells. In this case, downregulation of lncRNA-p3134 contributed to the pathology of diabetes. (2) Exosomal miRNA (miR-20b-5p, miR-155, miR-450b-3p, miR151-3p, and miR-29b-3p) was upregulated in the diabetic model and targeted skeletal muscles via various insulin signaling regulatory proteins (PPAR, AKT, GLUT4, and FOXO) and then contributed to the pathology of diabetes. (3) Exosomal miRNA (miR-122, miR-192, miR-27a/b, miR-155, miR-29b-3p) was upregulated in diabetic models and targeted adipocytes via PPAR proteins and then contributed to the pathology of diabetes. (4) Exosomal miRNA (miR-142-3p and miR-142-5p) was upregulated in diabetic models and targeted pancreatic cells via upregulated cytokines, which then contributed to the pathology of diabetes. (5) Other exosomal miRNAs (miR-106, miR-146a, miR-222, and miR-486) are promising therapeutic agents to protect pancreatic cells or treat diabetic complications by targeting certain organs, such as astrocytes, retinal tissue, and renal cells.
Cells 08 00853 g003
Table 1. Current studies of exosomal microRNAs (miRNA) in diabetes.
Table 1. Current studies of exosomal microRNAs (miRNA) in diabetes.
Exosome MiRNA NameMain Indication for DiabetesExpression in Diabetic StatueExosome Extracted FromEffected CellsExperimental ModelReference
MiR-20b-5pPathological factorupregulateSerumPrimary human skeletal muscle cellsT2D patientsKatayama, M et al. 2018. [83]
MiR-122,miR-192,miR-27a-3p, miR-27b-3pPathological factorupregulateSerumWhite adipose tissueDiet-induced obesity miceCastano, C et al. 2018 [86]
MiR-142-3p, miR-142-5p, miR-155Pathological factorupregulateLymphocytePancreatic β cellsNon-obese diabetic miceGuay, C et al. 2018 [49]
MiR-155Pathological factorupregulateAdipose tissue macrophageLiver, adipose tissue, and muscleObese miceYing, W et al. 2017 [85]
MiR-29b-3pPathological factorupregulateBone marrow mesenchymal stem cellsAdipocytes, C2C12 myocytes, and primary cultured hepatocytesAging-related insulin resistance miceSun, T et al. 2019 [65]
MiR-486Therapy agentupregulateAdipose derived stem cellsPodocyte cellsSpontaneous diabetic miceJin, J et al. 2019 [55]
MiR-222Therapy agentdownregulateAdipose tissue mesenchymal stem cellsRetinal tissueT1D rabbitSafwat, A et al. 2018 [87]
MiR-146aTherapy agentdownregulateBone narrow stem /stromal cellAstrocytesT1D miceKubota, K et al. 2018 [89]
MiR-106b, miR-222Therapy agentdownregulateBone narrow stem cellPancreatic β cellsT1D miceTsukita, S et al. 2017 [88]
MiR-21-5pBiomarkerupregulateSerumNRT1D patientsLakhter, A et al. 2018 [92]
MiR-375-3pBiomarkerupregulateSerumNRNew-onset T1D and T2D patientsFu, Q et al. 2018. [91]
MiR-133b, miR-342, miR-30BiomarkerupregulateSerumNRT2D patientsEissa, S et al. 2017 [90]
MiR-451-5pBiomarkerupregulateUrinaryNRT1D ratsMohan, A et al. 2016 [94]
Let-7c-5pBiomarkerupregulateUrinaryNRT2D patients with kidney diseaseLi, W et al. 2018 [93]
MiR-362-3p, miR-877-3p, miR-150-5p, miR-15a-5pBiomarkerupregulateUrinaryNRT2D patients with kidney diseaseXie, Y et al. 2017 [95]
T1D, type 1 diabetes; T2D, type 2 diabetes; NR, not reported.

Share and Cite

MDPI and ACS Style

Chang, W.; Wang, J. Exosomes and Their Noncoding RNA Cargo Are Emerging as New Modulators for Diabetes Mellitus. Cells 2019, 8, 853. https://doi.org/10.3390/cells8080853

AMA Style

Chang W, Wang J. Exosomes and Their Noncoding RNA Cargo Are Emerging as New Modulators for Diabetes Mellitus. Cells. 2019; 8(8):853. https://doi.org/10.3390/cells8080853

Chicago/Turabian Style

Chang, Wenguang, and Jianxun Wang. 2019. "Exosomes and Their Noncoding RNA Cargo Are Emerging as New Modulators for Diabetes Mellitus" Cells 8, no. 8: 853. https://doi.org/10.3390/cells8080853

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop