Next Article in Journal
Immunologic and Non-Immunologic Mechanisms Leading to Airway Remodeling in Asthma
Next Article in Special Issue
Impact of Janus Kinase Inhibition with Tofacitinib on Fundamental Processes of Bone Healing
Previous Article in Journal
Low Dose of Penfluridol Inhibits VEGF-Induced Angiogenesis
Previous Article in Special Issue
Immunomodulatory Effects of MSCs in Bone Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Renal Diseases

Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(3), 756; https://doi.org/10.3390/ijms21030756
Submission received: 26 December 2019 / Revised: 21 January 2020 / Accepted: 21 January 2020 / Published: 23 January 2020

Abstract

:
Mesenchymal stem cells (MSCs) have immunomodulatory and regenerative effects in many organs, including the kidney. Emerging evidence has shown that the trophic effects from MSCs are mainly mediated by the paracrine mechanism rather than the direct differentiation of MSCs into injured tissues. These secretomes from MSCs include cytokines, growth factors, chemokines and extracellular vesicles (EVs) containing microRNAs, mRNAs, and proteins. Many research studies have revealed that secretomes from MSCs have potential to ameliorate renal injury in renal disease models, including acute kidney injury and chronic kidney disease through a variety of mechanisms. These trophic mechanisms include immunomodulatory and regenerative effects. In addition, accumulating evidence has uncovered the specific factors and therapeutic mechanisms in MSC-derived EVs. In this article, we summarize the recent advances of immunomodulatory and regenerative effects of EVs from MSCs, especially focusing on the microRNAs.

1. Introduction

Mesenchymal stem cells (MSCs) are multipotent cells with the abilities of self-renewal, regeneration, and multilineage differentiation with respect to tissues such as bone, muscle, and adipocyte [1]. MSCs can be isolated from different kinds of tissues, including bone marrow, adipose tissue, umbilical cord, amniotic fluid, and placenta [1]. MSCs have been shown to ameliorate renal injury in a number of kidney disease models, including acute kidney injury (AKI), chronic kidney disease (CKD), and diabetic nephropathy. MSCs have been shown to directly differentiate and replace the injured area under tissue injury. In addition, MSCs secrete trophic factors such as cytokines, chemokines, growth factors, and extracellular vesicles (EVs) containing proteins, mRNA, and microRNAs (miRNAs). Accumulating evidence has demonstrated that the paracrine action is the dominant mechanism for ameliorating kidney injury. These therapeutic mechanisms are mediated through many biological processes, including anti-apoptosis, anti-inflammation, angiogenesis, and antifibrosis. Among the secretomes from MSCs, EVs have been shown to be the important mediator for renal protection in MSC therapy [2]. Accumulating evidence has revealed the specific miRNAs and proteins in MSC-derived EVs in renal diseases. In this review article, we summarize the current knowledge about therapeutic potential of EVs from MSCs, especially focusing on the immunomodulatory and regenerative effects.

2. Extracellular Vesicles

EVs are microvesicles enclosed by membrane from cells, and they deliver various biological molecules, including miRNAs, mRNA, long non-coding RNAs, tRNAs, circular RNAs, rRNAs, and proteins, into recipient cells [3], and influence their biological functions. EVs have been shown to have key roles in cell–cell communication in health and disease. With the ability to deliver a variety of factors, EVs have been shown to regulate a variety of biological functions, including cell proliferation, anti-apoptosis, antifibrosis, anti-inflammation, and angiogenesis. In fact, EVs from MSCs have been shown to mediate therapeutic effects in renal diseases through immunomodulation and regeneration. EVs can be grouped into several categories, including exosomes, microvesicles, and apoptotic bodies [4]. The diameters of exosomes are between 30 and 100 nm; microvesicles are 50–1000 nm, and apoptotic bodies are 1–5 mm. EVs can also be isolated from urine, amniotic fluid, plasma, and serum, and have been reported to be the biomarkers for specific diseases [5]. EVs have the advantage that they are able to internalize into target cells directly and quickly [6]. Many researchers demonstrated that the treatment of MSC-derived EVs ameliorated renal injury in rodent AKI and CKD models. There was the important problem of heterogeneity because of the different protocols of isolation and characterization. Therefore, the International Society of Extracellular Vesicles (ISEV) was instituted in 2011 to uniform the nomenclatures and methodologies for the isolation and characterization of EVs.

3. Proteins of MSC-Derived EVs

MSCs secrete many varieties of cytokines, chemokines, and growth factors. In addition, MSC-derived EVs also deliver these factors to recipient cells directly. Several studies conducted the proteomic analysis of MSC-derived EVs. For example, Mardpur et al. reported 938 proteins by quantitative proteomic technique [7]. They used the Kyoto Encyclopedia of Genes and Genomics (KEGG) and indicated that enriched proteins in MSC-EVs are involved in extracellular matrix (ECM) receptor interaction, focal adhesion, phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling, actin cytoskeleton, and biosynthesis of antibiotics. They also performed the analyses using Protein Analysis Through Evolutionary Relationships (PARTNER) analysis software, clustered MSC-EVs-proteins into 11 biological processes, and indicated that the cellular and metabolic processes are the highest while immune system response and response to stimulus are also involved. In addition, they revealed that MSC-EVs contain hepatocyte growth factor (HGF), Interleukin (IL)-10, Leukemia inhibitory factor (LIF), C-C motif chemokine ligand (CCL)-2 (also known as monocyte chemotactic protein-1 (MCP-1)), Vascular endothelial growth factor (VEGF)C, CCL-20, C-X-C motif chemokine ligand (CXCL)-2, CXCL-8, and CXCL-16. Among these factors, HGF has been shown to protect from renal injury in many experimental reports. For example, HGF has been shown to inhibit macrophage infiltration by inhibiting CCL-2 in diabetic rat kidney [8]. HGF has also been shown to inhibit glomerulosclerosis through the suppression of transforming growth factor (TGF)-β in diabetic rat kidney [8]. In addition, HGF and TNF-stimulated gene (TSG)-6 from MSCs have been shown to ameliorate inflammation in albumin-induced CKD model via the down-regulation of IL-6, IL-8, TNF-α, CCL-2, and CCL-5, which was partly mediated by the regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling [9]. Kim et al. also reported the proteomic analysis of human bone marrow MSC-EVs by the Database for Annotation, Visualization and Integrated Discovery (DAVID) software and indicated that MSC-EVs contain 730 proteins, and these proteins are mainly involved in cell cycle, proliferation, migration, adhesion (fibronectin, ezrin, Ras GTPase-activating-like protein IQGAP1, and integrin), signaling molecules (mitogen-activated protein kinase (MAPK)1 and CDC42) and morphogenesis [10]. They also indicated these proteins include growth factor receptor (platelet-derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR), and urokinase-type plasminogen activator receptor (uPAR)), Wnt, TGF-β, MAPK, peroxisome proliferator-activated receptor (PPAR), and bone morphogenetic protein (BMP) signaling. This broad spectrum of cellular processes and molecular functions suggest a variety of trophic mechanisms. Eirin et al. also reported the proteomic analysis using porcine adipose tissue-derived MSC-EVs. They indicated enriched proteins in MSC-EVs are involved in angiogenesis, blood coagulation, ECM remodeling, inflammatory response, and apoptosis [11]. They indicated that MSC-EVs contain key proteins for angiogenesis, such as VEGF, angiopoietin-related protein-4, platelet-derived growth factor-C (PDGFC), and WNT7B. Anderson et al. also reported 1927 proteins in MSC-EVs, revealing the modulation of angiogenesis via NF-κB signaling [12]. Lai et al. also performed the proteome analysis of MSC-EVs and indicated the 20S proteasome as the candidate to ameliorate tissue damage [13]. In addition, the specific factor for renal protection has also been reported. Jia et al. demonstrated that 14-3-3ζ in MSC-EVs protected from cisplatin-induced renal injury through the promotion of autophagy by the interaction with autophagy-related gene (ATG)-16L [14]. Taken together, MSC-EVs-derived proteins might have important roles in tissue protection through a variety of mechanisms.

4. mRNAs of MSC-Derived EVs

MSC-EVs contain a variety of mRNAs, and these mRNAs can be transferred into recipient cells. Valadi et al. demonstrated that these transferred mRNA did translate into proteins that can be functional in the recipient cells [15]. In addition, Batagov et al. reported that exosomal mRNAs have a specific structural pattern with the larger fraction in the secreted RNAs as the fragment localizes to the transcript closer to the 3′-end, implying that exosomal RNAs might act as competing RNAs to regulate stability, localization, and mRNA translation [16]. Accumulating evidence indicated the mRNA profile in MSC-EVs. For example, Bruno et al. reported, using reverse transcription (RT)-polymerase chain reaction (PCR) arrays, that these mRNAs might be involved in a variety of biological functions, including immune regulation, cell cycle, actin cytoskeleton regulation, and ECM remodeling [17]. Eirin et al. also characterized the RNA cargos in MSC-EVs, indicating that these mRNAs might be involved in angiogenesis, adipogenesis, transcription, and TGF-β signaling [18]. Importantly, regenerative effects of MSC-EVs treatment in gentamycin and glycerol-induced AKI models were abolished by RNA inactivation with RNase treatment, indicating the important roles of mRNAs and/or miRNAs on renal regeneration [19,20].

5. miRNAs of MSC-Derived EVs

miRNAs are noncoding, single-stranded, small-molecule RNAs. miRNAs silence their target genes through binding to the 3′-UTR. miRNAs in MSC-EVs have been reported to ameliorate renal injury in AKI and CKD rodent models. miRNAs are thought to be one of the essential contributors in MSC-EVs treatment. Indeed, the regenerative effects of MSC-EVs on renal injury were abolished with the knockdown of the ribonuclease III Drosha gene, which is essential for the initial processing step of miRNAs [19], suggesting the dominant roles of miRNAs for renal protection. Several reports indicated that MSC-EVs contain a variety of miRNAs, although the heterogeneity of factors in the different origins of MSCs has also been indicated [21]. The detailed miRNA analysis in MSC-EVs and the possible heterogeneity have previously been reviewed [21]. Previous reports indicated that RNase treatment into MSC-EVs abolished the trophic effects in ischemia/reperfusion (I/R)-induced AKI animal models [17,22], indicating the importance of mRNA and/or miRNAs for renal protection. In addition, accumulating evidence has gradually uncovered the therapeutic mechanisms mediated by specific trophic miRNAs in MSC-EVs for renal immunomodulation and regeneration in AKI and CKD animal models in vivo and in vitro (Table 1). For example, Collino et al. reported the regenerative effects of MSC-EVs-derived miRNAs in glycerol-induced AKI rat model [19]. They indicated that regenerative potential was abolished by the knockdown of the ribonuclease III Drosha gene, which corresponded to the reduced expression of miR-483-5p, miR-191, miR-28-3p, miR-423-5p, miR-744, miR-129-3p, miR-24, and miR-148a family, indicating the important roles of these miRNAs for renal regeneration. In the same line, Zou et al. reported the protective effects of MSC-EVs in I/R-induced AKI rat model and revealed that MSC-EVs promoted cell proliferation and inhibited renal fibrosis through the C-X3-C motif chemokine ligand (CX3CL)1 regulated by miR-15a, miR-15b, and miR-16 [23]. Gu et al. reported that miR-30 in MSC-EVs preserved mitochondrial morphology and inhibited tubular cell apoptosis in I/R-induced AKI rat model [24]. Zhu et al. reported that miR-199a-3p in MSC-EVs inhibited semaphorin3a (Sema3A) expression, which might activate AKT and extracellular signal-regulated kinase (ERK)-signaling pathway, thereby ameliorating cell apoptosis [25]. In addition to the trophic effects in AKI injury models, the renal protection in CKD animal models has been reported. Wang et al. demonstrated that the overexpression of let-7c in MSC-EVs ameliorated renal fibrosis in unilateral ureteral obstruction (UUO) model [26]. In vitro analysis also revealed that MSC-EVs blocked TGF-β-mediated epithelial-mesenchymal transition (EMT) in renal proximal tubular epithelial cells [27,28], and these effects were partly through miR-133b-3p and miR-294 [27]. They also reported that let-7c transferred from MSC-EVs decreased the expression of collagen type IV and matrix metalloproteinase-9 (MMP-9) via inhibition of TGF-β1 signaling in UUO model [26]. In the same line, He et al. reported MSC-EVs treatment ameliorated UUO-induced renal injury via miR-29, miR-30, and miR-210-3p [28]. Grange et al. applied MSC-EVs treatment in type 1 diabetes model and revealed the amelioration of renal fibrosis mediated by miRNA [29]. They suggested that miRNA-EVs might be involved in TGF-β, epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR), ADP-ribosylation factor 6 (ARF6), mammalian target of rapamycin (mTOR), VEGF, p53-apoptosis, ataxia telangiectasia-mutated (ATM), and tumor necrosis factor (TNF) pathway. Zhong et al. reported that miR-451 in MSC-EVs ameliorated diabetic kidney injury through the inhibition of EMT, and the effect relied on the improved cell cycle arrest and the downregulation of P15INK4b (P15) and P19INK4d (P19) [30]. In vitro, Xiang et al. reported that MSC-EVs enhanced autophagy of HK-2 cells and immortalized human proximal tubular cells through miR-145 by inhibiting the PI3K/AKT/mTOR signaling pathway, suggesting a therapeutic effect in renal diseases. These multiple therapeutic mechanisms might be mediated by the combination of several miRNAs in MSC-EVs.

6. Versatile MSC-EVs-Derived miRNAs Modulate Renal Injury and Homeostasis

In addition to these miRNAs that have directly been shown to be protective in renal diseases, there is accumulating evidence of miRNA being associated with the renal regeneration and also pathophysiology in renal diseases. Among the miRNAs summarized in the previous review paper indicating the miRNA profile from MSC-EVs [21], miR-1246, -23a, -451a, -125b, -199a, -let-7a, -4454/7975, -21, -let-7b, -100, -29a, -144, -29b, -22, -630, -221, -let-7i, -424, -191, -25, -130a, -376a, -27b, -30, -210, -24, -1202, -638, -148a, -532, -378, -let-7f, 486, -10a, -10b, -222, -143, -199b, -218, -135b, -203, -219, -299, -302b, -145, -338, -1260, and -1908, we searched the related papers and summarized the effects of these miRNAs reported in renal diseases (Table 2).
miR-21: miR-21 is one of the most reported miRNAs in renal injury. miR-21 has been reported to silence phosphatase and tensin homolog deleted from chromosome 10 (PTEN) and glycogen synthase kinase (GSK)-3β, thereby reducing NF-κB activity in macrophage [71]. In renal diseases, Song et al. reported that miR-21 ameliorated I/R-induced AKI by inhibiting tubular cell apoptosis and dendritic cell maturation [33]. On the other hand, miR-21 has been shown to correlate with tubulointerstitial injury in kidney biopsies patients in diabetic patients. Indeed, Koling et al. reported that the inhibition of miR-21 ameliorated renal injury in diabetic mice [34]. Xu et al. described the miR-21 as a double-edged sword because of the divergent pathophysiological processes, including inflammation and angiogenesis, and complex roles both on protective and pathological pathways in renal diseases [72]. They indicated that the preconditioning of upregulation of miR-21 contributed to the renal protection in subsequent I/R-induced renal injury through the anti-apoptosis. miR-21 has been shown to be downstream of AKT that mediates its anti-apoptotic effect through the suppression of Fas ligand [73]. Zhu et al. reported that miR-21 inhibited lipopolysaccharide (LPS)-induced NF-κB activation and IL-6 and enhanced IL-10 expression in monocyte cells in lung injury [74]. Conversely, long-term elevation of miR-21 might promote the progression of renal fibrosis [72]. Indeed, miR-21 promoted angiotensin II-induced renal fibrosis by activating the TGF-β1 pathway via suppressing PRARα [37]. Tang et al. reported that the silencing miR-21 inhibited renal interstitial fibrosis via targeting ERK1/2 signaling in mice [36]. miR-21 has also been shown to promote renal fibrosis by targeting PPARα [35]. Furthermore, miR-21 has also been shown to induce renal fibrosis through multiple pathways including AKT, ERK/MAPK, and it has been shown to target Smad7, which is a negative regulator of TGF-β/Smad3 pathway [38,75]. These complex mechanisms of miRNA might result in the effects of the double-edged sword.
miR-199: As described ahead, miR-199a-3p inhibits Sema3A, which activates AKT and ERK signaling, thereby ameliorating cell apoptosis [25]. In the same line, Sun et al. reported that the inhibition of miR-199a-5p suppressed cyst proliferation and induced cell apoptosis in autosomal dominant polycystic kidney disease (ADPKD) tissue and cells by targeting CDKN1C [39]. On the other hand, Yang et al. reported that the inhibition of miR-199a-3p reduced cisplatin-induced cell apoptosis and inhibited caspase-3 activity [40]. mTOR is the target gene of miR-199a [76], regulating cell survival, proliferation, migration, and apoptosis. In addition, miR-199a-3p increased TGF-β1-induced renal fibrosis via silencing of SOCS7 and STAT3 [41]. miR-199a has also been reported to be involved in inflammation via modulating the activation of NF-κB by targeting Klotho in lupus nephritis [42]. Sun et al. reported the increased level of serum miR-199b-5p in diabetic patients [43]. They also indicated that the inhibition of miR-199b ameliorated TGF-β1-induced EMT and renal fibrosis via targeting SIRT1 in diabetic nephropathy [43]. Kang et al. suggested that miR-199b-5p targets klotho, thereby promoting renal tubular injury in diabetic nephropathy [77]. In summary, miR-199 might have dual effects of the double-edged sword in renal diseases similar to miR-21.
let-7 family: the let-7 family has been reported to target collagen I [78,79] as well as being involved in cell cycle, cell proliferation, apoptosis, and immune system. Yan et al. demonstrated that naringenin ameliorated kidney injury through let7a-TGFBR1 signaling in diabetic nephropathy [44]. In addition, the let-7 family has been shown to protect against TGF-β1-induced collagen accumulation, and it is suggested that Col1a2 and Col4a1 might be the targets of the let-7 family [45]. Let-7b has also been reported to regulate renal fibrosis in proximal tubular cells and mesangial cells via regulation of TGFBR1 under renal injury [46]. In addition, let-7i has been reported to target toll-like receptor (TLR)4, thereby inhibiting LPS-induced inflammatory kidney injury [47]. As described ahead, the transfer of let-7c from MSC-EVs ameliorated renal fibrosis in UUO model through the downregulation of Col4a1, MMP-9, TGF-β1, and TGFBR1 [26]. Taken together, the let-7 family might be the promising target for ameliorating renal fibrosis and inflammation in renal diseases.
miR-30: As described ahead, miR-30 preserved mitochondrial morphology and inhibited cell apoptosis in I/R-induced AKI rat model [24]. miR-30c has been shown to target Bnip3L and Hspa5, thereby inhibiting cisplatin-induced tubular epithelial cell apoptosis [48]. In addition, He et al. reported that miR-30a is associated with the inhibition of UUO-induced renal fibrosis under MSC-EVs treatment [28]. miR-30a has been reported to target the snail in hepatocytes [80]. Zhang et al. indicated that miR-30c-5p might decrease I/R-induced renal injury by promoting M1 macrophage toward M2 macrophage and changes in inflammatory cytokines [49]. In addition, miR-30 might protect podocyte by preventing uPAR-ITGB3 signaling through the calcineurin–NFATC pathway [50]. In the same line, Yang et al. reported that TGF-β induced podocyte injury through the downregulation of miR-30 [81]. Angiotensin II has been shown to induce calcineurin signaling and podocyte injury by reducing miR-30, indicating that miR-30 expression might be protective for podocyte [51]. miR-30e has been shown to target Bnip3L and protect against aldosterone-induced podocyte apoptosis and mitochondrial dysfunction [52]. miR-30a has also been shown to inhibit EMT of podocyte through downregulation of NFATc3 [53]. Taken together, miR-30 might have potentials to mediate a variety of trophic effects, including anti-apoptosis, anti-inflammation, and antifibrosis, as well as the protection of podocytes.
miR-29: miR-29a has been reported to target collagen I [78,79] and is known to act as fibrotic regulator in several tissue types [54]. In renal diseases, miR-29a has been reported to protect from diabetic nephropathy via regulation of PPARγ and CB1R [54]. miR-29a has also been shown to modulate DKK1/Wnt/β-catenin signaling, thereby protecting from diabetic glomerular dysfunction [55]. In addition, Wang et al. reported that TGF-β1 reduced the expression of miR29a/b/c family [82]. TGF-β1 inhibited the expression of miR-29 family, thereby promoting fibrosis via increase of ECM component [82]. Hu et al. reported that miR-29b ameliorated angiotensin II-induced EMT of rat tubular epithelial cells through the regulation of PI3K/AKT signaling [56]. In the same line, miR-200, miR-29, and miR-143 have been reported to affect the progression of TGF-β-dependent EMT and fibrosis [83,84]. miR-29 has also been shown to attenuate renal fibrosis in UUO model by downregulating TGF-β signaling [85]. Saito et al. also demonstrated that miR-29b delivery by adeno-associated virus attenuated renal fibrosis in UUO model by suppressing snail expression [57]. In summary, miR-29 might have the potential to block the progression of renal fibrosis through the regulation of TGF-β1/EMT axis.
miR-145: As described ahead, miR-145 might promote autophagy of HK-2 cells by inhibiting the PI3K/AKT/mTOR-signaling pathway. In addition, Liu et al. reported that miR-145 ameliorated TGF-β1-induced EMT in renal proximal tubular cells possibly by inhibiting TGF-β-smad signaling pathway [58]. In the same line, Liu et al., reported that miR-145 overexpression inhibited high-glucose-induced EMT and fibrosis via downregulating ZEB2 [59]. Therefore, miR-145 might be the promising target to protect from kidney injury through the regulation of autophagy and EMT.
miR-210: As described ahead, MSC-EVs might ameliorate UUO-induced renal injury via miR-210-3p [28]. miR-210 has been shown to be upregulated in response to hypoxia-inducible factor (HIF). miR-210 has also been reported to be involved in various pathophysiological pathways such as oxidative stress, cancer, and apoptosis [86]. Liu et al. indicated that miR-210 protected renal tubular cells from apoptosis by targeting HIF-1α [60]. In addition, Liu et al. reported that miR-210 promoted angiogenesis by activation of VEGF pathway in I/R-induced kidney injury [61]. Zhang et al. suggested that MSC-EVs treatment in cisplatin-induced AKI model might ameliorate renal inflammation and cell apoptosis via miR-210/Serpine1 and miR-378/Fos axis [62]. Although further analysis is required to confirm these effects, miR-210 might protect from renal injury in multiple mechanisms.
miR-22: Zhang et al. reported that miR-22 promoted renal fibrosis by targeting PTEN/AKT/mTOR pathway and suppressing autophagy in diabetic nephropathy [63]. Indeed, the inhibition of miR-22 ameliorated kidney fibrosis by targeting BMP-6, BMP-7, and BMP type I receptor [64]. BMP-7 is known to have antifibrotic function through the regulation of EMT and synthesis of ECM [87]. BMP-7 from MSCs has been reported to ameliorate glomerular fibrosis through the inhibition of TGF-β signaling in vivo and in vitro [88]. Therefore, miR-22 might be associated with the progression of renal injury.
miR-125b: miR-125b has been shown to be increased by NF-E2-related factor 2 (Nrf2), which results in the inhibition of aryl hydrocarbon receptor (AhR) repressor and protection from cisplatin-induced renal injury via anti-apoptosis [65]. On the other hand, miR-125b has been shown to contribute to high-glucose-induced reactive oxygen species (ROS) generation and renal tubular cell apoptosis by downregulation of angiotensin-converting-enzyme (ACE) [66]. Further analysis is required to explore the effect of miR-125b in renal diseases.
miR-130a: Liang et al. reported that miR-130a protected LPS-induced glomerular injury by Klotho upregulation [67]. They also indicated that miR-130a activated PI3K/AKT pathway and inhibited Wnt and NF-κB pathway [67]. Liu et al. reported that miR-130a-5p prevented angiotensin II-induced podocyte apoptosis by modulating phospholipase A2 receptor (PLA2R) [68]. In spite of these trophic effects, miR-130a-3p has been reported to target SnoN, thereby promoting renal fibrosis via TGF-β1/Smad pathway, similar to miR-23a [69]. Further analysis is still required to conclude the potential effects of miR-130a in renal diseases.
miR-23a: miR-23a has been shown to be increased in renal tissue in patients with type 2 diabetes mellitus (DM) [70]. Xu et al. reported that miR-23a directly targeted SnoN gene, and miR-23a overexpression promoted high glucose-induced EMT and renal fibrogenesis [70], suggesting the pathophysiological role of miR-23a on the progression of renal fibrosis.
Other possible candidates:
In spite of the limited number of reports in renal diseases, there are several candidate miRNAs associated with renal regeneration and/or pathophysiology.
miR-221: Di et al. reported that miR-221 might target Ets-1, thereby preventing angiotensin II-induced renal fibroblast activation and fibrosis [89].
miR-424: Chen et al. reported that miR-424 expression was increased in I/R renal injury and miR-424 attenuated I/R-induced renal injury through the inhibition of apoptosis via death receptor 6 [90].
miR-191: Qin et al. reported that miR-191-5p reduces sepsis-induced acute kidney injury by targeting oxidative-stress-responsive 1 (OXSR1) in rat model [91].
miR-25: Oh et al. reported that miR-25 expression was downregulated under diabetic condition, which resulted in the increase of NADPH oxidase 4 (NOX4) in early diabetic nephropathy [92].
miR-27b: Zheng et al. reported that miR-27b enhanced puromycin aminonucleoside (PAN)-induced apoptosis and cytoskeleton destruction in podocytes [93].
miR-24: Lorenzen et al. reported that the inhibition of miR-24 in I/R-induced renal injury mice reduced tubular cell apoptosis [94]. On the other hand, Liu et al. reported that miR-24-3p inhibited mesangial cell proliferation and fibrosis through the downregulation of fibroblast growth factor (FGF)-11 [95].
miR-486: Vinas et al. reported that miR-486-5p ameliorated I/R-induced kidney injury via inhibiting PTEN and cell apoptosis [96].
miR-10a: Shan et al. reported that HDAC/miR-10a/CREB1 axis might be associated with renal injury in type 2 DM [97].
miR-218: Yang et al. reported that miR-218 promoted high-glucose-induced apoptosis in podocytes through the regulation of heme oxygenase (HO)-1 [98].
miR-135b: Yang et al. reported that miR-135 family (miR-135a and miR-135b) mediated podocyte injury through the activation of Wnt/β-catenin signaling [99].
miR-203: Liu et al. reported that low miR-203 expression might promote diabetic nephropathy via increasing TLR4 [100].
miR-1908: Xie et al. reported that the transfection of miR-1908 inhibited renal fibrosis via targeting TGF-β1 [101].

7. Immunomodulatory Effect of EVs from MSCs against Renal Injury

MSCs have been shown to modulate immune cells such as T cells, dendric cells, B cells, natural killer (NK) cells, and regulatory T cells via soluble factors, including indoleamine 2,3 dioxygenase (IDO), nitric oxide (NO), prostaglandin E2 (PGE2), HGF, and TGF-β1, as well as via direct cell–cell contact [102]. In addition, factors in MSC-EVs are capable of modulating immune cells. The modulation of immune system is one of the most important functions of MSC-EVs therapy among all biological processes. For example, Wang et al. reported that MSC-EVs treatment in cisplatin-induced AKI reduced the levels of TNF-α and IL-1β in renal epithelial cells in vitro [103]. In addition, MSC-EVs treatment, like MSCs injection, decreased TNF-α and IL-1β expression in I/R-induced rat renal injury model [104]. In the same line, MSC-EVs treatment prevented gentamicin-induced AKI by downregulating IL-6 and TNF-α as well as up-regulating IL-10 [105]. Wang et al. also reported that MSC-EVs treatment in I/R-induced AKI rat reduced the TNF-α mRNA expression [106]. Eirin et al. reported that MSC-EVs treatment attenuated renal inflammation after renal artery stenosis, and the effect was partly mediated by IL-10 mRNA in MSC-EVs [107]. IL-10 is known to regulate the function of immune cells and determine the M2 macrophage phenotype. M2 macrophage might attenuate inflammation and promote tissue repair. They also demonstrated the decreased expression of TNF-α, IL-6, and IL-1β [107], indicating the complex roles of MSC-EVs on immunomodulation. They also demonstrated that MSC-EVs treatment shifted the renal macrophage phenotype from M1 toward M2. Zou et al. also suggested that NK cell regulatory process is involved in the MSC-EVs effect in I/R-induced AKI [108]. Choi et al. demonstrated that MSC-EVs treatment in UUO model decreased macrophage infiltration in kidney detected by F4/80 staining [109]. Macrophages are the key inflammatory cells involved in kidney inflammation. In addition, He et al. reported that lymphocyte infiltration was reduced with MSC-EVs treatment in 5/6 subtotal nephrectomy mouse model [110]. As described ahead, several specific miRNAs have been reported to possess immunomodulatory effects. For example, miR-21 has been reported to silence PTEN and GSK-3β, thereby reducing NF-κB activity in macrophage [71]. miR-21 inhibited LPS-induced NF-κB activation and IL-6 and enhanced IL-10 expression in monocyte cells [74]. miR-199a has also been reported to be involved in inflammation via modulating the activation of NF-κB by targeting Klotho in Lupus nephritis [42]. miR-210 and miR-378 have also been shown to regulate inflammation by targeting Serpine1 and Fos in cisplatin-induced AKI model [62]. Zou et al. reported that MSC-EVs treatment blocked macrophage infiltration through the downregulation of CX3CL1 by miR-15a, miR-15b, and miR-16 [23]. CX3CL1 has potential as chemo-attraction for macrophage, and thereby the inhibition of CX3CL1 might reduce the accumulation of macrophage in the kidney. As shown ahead, MSC-EVs contain proteins, including HGF, IL-10, LIF, CCL-2, VEGFC, CCL-20, CXCL-2, CXCL-8, CXCL-16, denfensinα1, HERC5, and IFITM2 that are associated with inflammation [7]. CCL2, VEGFC, and CCL20 might bring immune cells nearby and induce immunosuppressive processes. CXCL2, CXCL8, CXCL16, denfensinα1, HERC5, and IFITM2 have been reported to have a role on immune cell chemotaxis and neutrophil degranulation. HGF might inhibit macrophage infiltration by inhibiting CCL-2 in diabetic rat kidney [8] and ameliorate inflammation in albumin-induced CKD model through the downregulation of IL-6, IL-8, TNF-α, CCL-2, and CCL-5 [9]. HGF, IL-10, and TGF-β1 are important factors in suppressing T cell proliferation. TGF-β1 also induces CD4(+)/CD25(+)-regulatory T cells that regulate inflammatory response. NO is also known to have important immunomodulatory effects. NO suppresses T cells through cell cycle arrest and apoptosis. NO also has roles in M1/M2 macrophage fate and cytokine receptor and T cell receptor (TCR) signaling. Gennai et al. reported, using ex vivo perfused human lungs rejected for transplantation, that MSC-EVs increased the NO level in the perfusate [111], suggesting the possibility that MSC-EVs might regulate NO expression and thereby have the potential in immunomodulation under tissue injury. PGE2 is another important factor that affects immunomodulation, especially through the regulation of T cells. PGE2 has been shown to shift Th1 toward Th2/Th17 by inhibiting pro-inflammatory cytokines and inducing Th2 cytokines, including IL-4 and IL-5. Harting et al. demonstrated that MSC-EVs treatment increased the expression of cyclooxygenase-2 (Cox-2) and PGE2, and attenuated pro-inflammatory cytokines [112]. Taken together, MSC-EVs have the strong potential to promote immunomodulation in a variety of mechanisms.
In spite of the trophic effects of MSC-EVs in the point of immunomodulation, MSC-EVs treatment potentially mediates negative immunomodulatory effects [113]. For example, Kilpinen et al. analyzed the secretomes of umbilical-cord-derived MSC-EVs with or without the interferon-gamma (IFN-γ) stimulus and indicated that EVs from MSCs treated with IFN-γ for 24 h contained major histocompatibility Class I (MHCI) and both α and β units of the proteasome complex required for the antigen presentation and activation of T cells [114]. In addition, EVs from MSCs treated with IFN-γ for 48 h also contained major histocompatibility Class II (MHCII). The presence of MHCI and MHCII on the surface of MSC-EVs might trigger the innate immune response. Therefore, MSC-EVs after IFN-γ stimulus might act as immunogenicity and accelerate inflammation. We need to keep in mind the possible negative effects under the MSC-EVs treatment.

8. Regenerative Effect of EVs from MSCs against Renal Injury

In addition to the immunomodulatory effects, MSC-EVs have the potential to promote renal regeneration and protection in a variety of mechanisms (Figure 1).

8.1. Renal Regeneration

In many reports of AKI models induced by I/R and glycerol and cisplatin-induced rodent models, the increase of tubular proliferation was observed under the treatment of MSC-EVs [17,23,105,115,116,117,118,119,120], indicating the regenerative effect of MSC-EVs. Although it is not easy to identify the specific factors for cell proliferation, several factors might be involved. For example, HGF has been reported to enhance mitogenesis in I/R-AKI rat model [121]. mRNA and/or miRNA in MSC-EVs might be involved as well since the enhancement of tubular proliferation with MSC-EVs treatment in glycerol-induced AKI was abolished with RNase treatment. Angiogenesis is another mechanism of renal regeneration in rodent AKI models. MSC-EVs treatment in I/R-induced AKI rat model showed the increase of VEGF expression and capillary vessel density that was abolished with RNase treatment [116]. In addition, MSC-EVs treatment in I/R-induced AKI rat model showed the increase of VEGF and VEGFR2 expression as well as the increase of miR-210, and the overexpression of miR-210 in HUVEC-12 cells increased VEGF and VEGFR2 expression and promoted angiogenesis in vitro, indicating that miR-210 might be involved in MSC-EV-induced angiogenesis by targeting VEGF signaling [61]. The expression of angiogenesis indicators, CD31, von Willebrand factor (vWF), and angiopoietin was upregulated with MCS-EVs treatment in I/R-induced AKI rat model [104]. In addition, in the UUO-induced CKD mouse model, MSC-EVs treatment improved the rarefaction of peritubular capillaries detected by CD31 staining [109]. During renal regeneration after AKI, epithelial de-differentiation has been reported to be an important process that promotes cell survival, migration, and proliferation [122]. HGF, TGF-β1, IGF, and EGF have been reported to be involved in the de-differentiation [122,123]. Indeed, Ju et al. reported that MSC-EVs treatment in I/R-induced AKI rat model promoted tubular epithelial cell de-differentiation via HGF induction that was blocked with RNase treatment, suggesting that HGF induction might be mediated by mRNAs and/or miRNAs [124]. Furthermore, they revealed that the HGF mRNA from MSC-EVs entered the injured tubular cells and was translated into HGF protein, indicating foreign HGF synthesis. In summary, MSC-EVs have a variety of trophic mechanisms for renal regeneration through the regulation of cell proliferation, angiogenesis, and tubular cell de-differentiation.

8.2. Renal Protection

The mechanism regulating cell apoptosis is one of the most important aspects of renal protection. Indeed, the anti-apoptotic pathway has been reported in many studies with MSC-EVs treatment in a variety of rodent renal injury models, including AKI induced by I/R [24,31,33,104,106,116,117,118,120,124,125], cisplatin [48,119,126], gentamicin [105], and glycerol [17], hypoxia-induced renal injury [60], aldosterone-induced renal injury [52], and UUO-induced CKD model [109]. Recent advances have gradually uncovered the specific miRNAs, their targets and signaling pathways involved in the effect of anti-apoptosis (Table 2), including PTEN, AKT, mTOR, dynamin-related protein 1 (DRP1), Sema3A, and ERK signaling pathway. Anti-necrosis with MSC-EVs treatment has also been reported in drug-induced AKI, such as with glycerol, cisplatin, and gentamicin [105,115,119]. Autophagy regulation is another aspect of trophic mechanism with MSC-EVs treatment for renal protection. In cisplatin-induced AKI model, the improvement of autophagy was observed with MSC-EVs treatment [14]. In the study, there was an increase of autophagy marker and LC3II expression in vivo and in vitro with MSC-EVs treatment, and it was associated with 14-3-3ζ expression, which regulates ATG-16L. Another group also indicated that MSC-EVs treatment increased the LC3B expression as well as the ATG-5 and ATG-7 gene expressions in normal rat kidney-52E (NRK-52E) cells through the inhibition of mTOR signaling [103]. In addition, Xiang et al. indicated that MSC-EVs enhanced the expression of LC3II and beclin 1 in HK-2 cells through miR-145 targeting PI3K/AKT/mTOR-signaling pathway [32]. Under pathological conditions, autophagy is induced as the adaptive and protective mechanism for cell survival, thus the regulation of autophagy is an important mechanism for renal protection. Preservation of mitochondria is also an important mechanism for renal protection from AKI. DRP1 is known as the key regulator for mitochondrial fission [127] and is rapidly activated after AKI [128]. Inhibition of DRP1 has been reported to protect the kidney from AKI injury caused by I/R and cisplatin [128,129]. Indeed, MSC-EVs treatment preserved mitochondrial function via transferring miR-30 that inhibited the expression of DRP1 [24]. Improvement of oxidative stress with MSC-EVs treatment is also an important mechanism for renal protection against cisplatin and I/R-induced AKI [104,117,119,125]. Oxidative stress via reactive ROS production induces apoptosis, necrosis, and inflammation in AKI. Zhang et al. reported that MSC-EVs treatment alleviated oxidative stress detected by the reduction of malondialdehyde (MDA) and 8-hydroxy-2′-deoxyguanosine (8-OhdG) as well as the enhancement of Nrf2 and HO-1 in I/R-induced AKI rat model [125]. NOX-2, known as the inducer of ROS production, declined with MSC-EVs treatment in I/R-induced AKI rat model [117]. In addition, Zhou et al. reported that MSC-EVs treatment ameliorated oxidative stress in cisplatin-induced AKI detected by the reduction of 8-OhdG-positive cells, which reduced the cell apoptosis [119]. HGF has also been reported to improve oxidative stress through the suppression of GLUT1 [130]. Taken together, several factors in MSC-EVs might be involved in the regulation of oxidative stress. Renal fibrosis is the pathological process of CKD, which is strongly associated with the progression of renal dysfunction. The improvement of renal fibrosis with MSC-EVs treatment was reported in a variety of renal injury models, including I/R-induced AKI rodent model [23,104,116,117,124], renovascular stenosis-induced pig CKD model [107], UUO-induced renal fibrosis model [26,28,109], diabetes nephropathy mouse model [29,30], and 5/6 subtotal nephrectomy mouse model [110]. In vitro experiment using HK-2 cells, MSC-EVs ameliorated TGF-β1-induced EMT, which was mediated by miR-133b-3p and miR-294 [27]. As described ahead, let-7c from MSC-EVs ameliorated renal fibrosis in UUO model with the downregulation of Col4a1, MMP-9, TGF-β1, and TGFBR1 [26]. In addition, miR-451 in MSC-EVs ameliorated renal fibrosis by targeting P15 and P19, thus inhibiting EMT in diabetic model [30]. MiR-29b in MSC-EVs might target snail, thereby regulating EMT [57]. Taken together, several MSC-EVs-derived miRNAs have the potential to ameliorate renal fibrosis, mainly through the regulation of TGF-β1-EMT axis. In summary, EV-MSCs have a variety of factors involved in many biological processes for renal generation and protection as well as immunomodulation.

9. Conclusions

In this review article, we summarized the recent advances of the immunomodulatory and regenerative effects of MSC-EVs in renal diseases. MSC-EVs treatments have several advantages over MSCs therapy in the point of easy transfer into recipient cells, no risk of tumor formation, and low immunogenicity because of the cell-free sources. Although these cell-free therapies might be promising to treat against renal injury, there are still challenges for clinical use. The heterogeneity of secretomes of MSCs obtained from different tissues might strongly affect the therapeutic potential. In addition, some preconditioning stimuli might affect the quality and quantity of trophic factors in MSC-EVs as well as the engineering or genetic modification for overexpression of specific proteins and miRNAs. Other important challenges are their safety, analysis of MSC cell sources, and isolation method to acquire more effective functions. In conclusion, MSC-EVs therapy might be the promising cell-free therapeutic option for the treatment against renal injury including AKI and CKD. Further exploration is still required for future clinical use for patients with renal diseases.

Author Contributions

K.T., S.K., and J.W. analyzed the literature and wrote and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Joint Research with Kikkoman Co., Ltd. for the publication.

Conflicts of Interest

The authors declare no conflict of interest. The funder had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

MSCsMesenchymal stem cells
AKIAcute kidney injury
CKDChronic kidney disease
EVsExtracellular vesicles
miRNAsMicroRNAs
ISEVThe International Society of Extracellular Vesicles
KEGGThe Kyoto Encyclopedia of Genes and Genomics
ECMExtracellular matrix
PI3KPhosphatidylinositol 3-kinase
AKTProtein kinase B
PARTNERProtein Analysis Through Evolutionary Relationships
HGFHepatocyte growth factor
ILInterleukin
LIFLeukemia inhibitory factor
CCLC-C motif chemokine ligand
MCP-1Monocyte chemotactic protein-1
VEGFVascular endothelial growth factor
CXCLC-X-C motif chemokine ligand
TGFTransforming growth factor
TSGTNF-stimulated gene
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
DAVIDThe Database for Annotation, Visualization and Integrated Discovery
MAPKMitogen-activated protein kinase
PDGFRPlatelet-derived growth factor receptor
EGFREpidermal growth factor receptor
uPARUrokinase-type plasminogen activator receptor
PPARPeroxisome proliferator-activated receptor
BMPBone morphogenetic protein
PDGFCPlatelet-derived growth factor-C
ATGAutophagy related gene
RTReverse transcription
PCRPolymerase chain reaction
I/RIschemia/reperfusion
CX3CLC-X3-C motif chemokine ligand
Sema3ASemaphorin3a
ERKExtracellular signal-regulated kinase
UUOUnilateral ureteral obstruction
EMTEpithelial-mesenchymal transition
MMP-9Matrix metalloproteinase-9
ARF6ADP-ribosylation factor 6
mTORMammalian target of rapamycin
ATMAtaxia telangiectasia-mutated
TNFTumor necrosis factor
P15P15INK4b
P19P19INK4d
PTENPhosphatase and tensin homologue deleted from chromosome 10
GSKGlycogen synthase kinase
LPSLipopolysaccharide
ADPKDAutosomal dominant polycystic kidney disease
TLRToll-like receptor
HIFHypoxia-inducible factor
Nrf2NF-E2-related factor 2
AhRAryl hydrocarbon receptor
ROSReactive oxygen species
ACEAngiotensin-converting-enzyme
PLA2RPhospholipase A2 receptor
DMDiabetes mellitus
OXSR1Oxidative stress responsive 1
NOXNADPH oxidase
PANPuromycin aminonucleoside
FGFFibroblast growth factor
HO-1Heme oxygenase-1
NKNatural killer
IDOIndoleamine 2,3 dioxygenase
NONitric oxide
PGE2Prostaglandin E2
TCRT cell receptor
Cox-2Cyclooxygenase-2
IFN-γinterferon-gamma
MHCImajor histocompatibility Class I
MHCIImajor histocompatibility Class II
vWFVon Willebrand factor
DRP1Dynamic related protein 1
NRK-52ENormal rat kidney-52E
MDAMalondialdehyde
8-OhdG8-hydroxy-2′-deoxyguanosine

References

  1. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [Green Version]
  2. Borgovan, T.; Crawford, L.; Nwizu, C.; Quesenberry, P. Stem cells and extracellular vesicles: biological regulators of physiology and disease. Am. J. Physiol. Cell Physiol. 2019, 317, C155–C166. [Google Scholar] [CrossRef]
  3. Lee, Y.; El Andaloussi, S.; Wood, M.J. Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 2012, 21, R125–R134. [Google Scholar] [CrossRef] [Green Version]
  4. Rad, F.; Pourfathollah, A.A.; Yari, F.; Mohammadi, S.; Kheirandish, M. Microvesicles preparation from mesenchymal stem cells. Med. J. Islam. Repub. Iran 2016, 30, 398. [Google Scholar]
  5. Keller, S.; Ridinger, J.; Rupp, A.K.; Janssen, J.W.; Altevogt, P. Body fluid derived exosomes as a novel template for clinical diagnostics. J. Transl. Med. 2011, 9, 86. [Google Scholar] [CrossRef] [Green Version]
  6. Kooijmans, S.A.A.; Schiffelers, R.M.; Zarovni, N.; Vago, R. Modulation of tissue tropism and biological activity of exosomes and other extracellular vesicles: New nanotools for cancer treatment. Pharmacol. Res. 2016, 111, 487–500. [Google Scholar] [CrossRef]
  7. Mardpour, S.; Hamidieh, A.A.; Taleahmad, S.; Sharifzad, F.; Taghikhani, A.; Baharvand, H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J. Cell Physiol. 2019, 234, 8249–8258. [Google Scholar] [CrossRef] [PubMed]
  8. Lv, S.S.; Liu, G.; Wang, J.P.; Wang, W.W.; Cheng, J.; Sun, A.L.; Liu, H.Y.; Nie, H.B.; Su, M.R.; Guan, G.J. Mesenchymal stem cells transplantation ameliorates glomerular injury in streptozotocin-induced diabetic nephropathy in rats via inhibiting macrophage infiltration. Int. Immunopharmacol. 2013, 17, 275–282. [Google Scholar] [CrossRef] [PubMed]
  9. Wu, H.J.; Yiu, W.H.; Li, R.X.; Wong, D.W.; Leung, J.C.; Chan, L.Y.; Zhang, Y.; Lian, Q.; Lin, M.; Tse, H.F.; et al. Mesenchymal stem cells modulate albumin-induced renal tubular inflammation and fibrosis. PLoS ONE 2014, 9, e90883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kim, H.S.; Choi, D.Y.; Yun, S.J.; Choi, S.M.; Kang, J.W.; Jung, J.W.; Hwang, D.; Kim, K.P.; Kim, D.W. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J. Proteome Res. 2012, 11, 839–849. [Google Scholar] [CrossRef] [PubMed]
  11. Eirin, A.; Zhu, X.Y.; Puranik, A.S.; Woollard, J.R.; Tang, H.; Dasari, S.; Lerman, A.; van Wijnen, A.J.; Lerman, L.O. Comparative proteomic analysis of extracellular vesicles isolated from porcine adipose tissue-derived mesenchymal stem/stromal cells. Sci. Rep. 2016, 6, 36120. [Google Scholar] [CrossRef] [PubMed]
  12. Anderson, J.D.; Johansson, H.J.; Graham, C.S.; Vesterlund, M.; Pham, M.T.; Bramlett, C.S.; Montgomery, E.N.; Mellema, M.S.; Bardini, R.L.; Contreras, Z.; et al. Comprehensive Proteomic Analysis of Mesenchymal Stem Cell Exosomes Reveals Modulation of Angiogenesis via Nuclear Factor-KappaB Signaling. Stem. Cells 2016, 34, 601–613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lai, R.C.; Tan, S.S.; Teh, B.J.; Sze, S.K.; Arslan, F.; de Kleijn, D.P.; Choo, A.; Lim, S.K. Proteolytic Potential of the MSC Exosome Proteome: Implications for an Exosome-Mediated Delivery of Therapeutic Proteasome. Int. J. Proteom. 2012, 2012, 971907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jia, H.; Liu, W.; Zhang, B.; Wang, J.; Wu, P.; Tandra, N.; Liang, Z.; Ji, C.; Yin, L.; Hu, X.; et al. HucMSC exosomes-delivered 14-3-3zeta enhanced autophagy via modulation of ATG16L in preventing cisplatin-induced acute kidney injury. Am. J. Transl. Res. 2018, 10, 101–113. [Google Scholar] [PubMed]
  15. Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Batagov, A.O.; Kurochkin, I.V. Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3’-untranslated regions. Biol. Direct. 2013, 8, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Bruno, S.; Grange, C.; Deregibus, M.C.; Calogero, R.A.; Saviozzi, S.; Collino, F.; Morando, L.; Busca, A.; Falda, M.; Bussolati, B.; et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J. Am. Soc. Nephrol. 2009, 20, 1053–1067. [Google Scholar] [CrossRef] [Green Version]
  18. Eirin, A.; Riester, S.M.; Zhu, X.Y.; Tang, H.; Evans, J.M.; O’Brien, D.; van Wijnen, A.J.; Lerman, L.O. MicroRNA and mRNA cargo of extracellular vesicles from porcine adipose tissue-derived mesenchymal stem cells. Gene 2014, 551, 55–64. [Google Scholar] [CrossRef] [Green Version]
  19. Collino, F.; Bruno, S.; Incarnato, D.; Dettori, D.; Neri, F.; Provero, P.; Pomatto, M.; Oliviero, S.; Tetta, C.; Quesenberry, P.J.; et al. AKI Recovery Induced by Mesenchymal Stromal Cell-Derived Extracellular Vesicles Carrying MicroRNAs. J. Am. Soc. Nephrol. 2015, 26, 2349–2360. [Google Scholar] [CrossRef]
  20. Murphy, D.J.; Seltzer, B.L.; Yesalis, C.E. Comparison of two methodologies to measure agricultural occupational fatalities. Am. J. Public. Health 1990, 80, 198–200. [Google Scholar] [CrossRef]
  21. Tsuji, K.; Kitamura, S.; Wada, J. Secretomes from Mesenchymal Stem Cells against Acute Kidney Injury: Possible Heterogeneity. Stem. Cells Int. 2018, 2018, 8693137. [Google Scholar] [CrossRef] [PubMed]
  22. Deregibus, M.C.; Cantaluppi, V.; Calogero, R.; Lo Iacono, M.; Tetta, C.; Biancone, L.; Bruno, S.; Bussolati, B.; Camussi, G. Endothelial progenitor cell derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA. Blood 2007, 110, 2440–2448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zou, X.; Zhang, G.; Cheng, Z.; Yin, D.; Du, T.; Ju, G.; Miao, S.; Liu, G.; Lu, M.; Zhu, Y. Microvesicles derived from human Wharton’s Jelly mesenchymal stromal cells ameliorate renal ischemia-reperfusion injury in rats by suppressing CX3CL1. Stem. Cell Res. Ther. 2014, 5, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Gu, D.; Zou, X.; Ju, G.; Zhang, G.; Bao, E.; Zhu, Y. Mesenchymal Stromal Cells Derived Extracellular Vesicles Ameliorate Acute Renal Ischemia Reperfusion Injury by Inhibition of Mitochondrial Fission through miR-30. Stem. Cells Int. 2016, 2016, 2093940. [Google Scholar] [CrossRef] [Green Version]
  25. Zhu, G.; Pei, L.; Lin, F.; Yin, H.; Li, X.; He, W.; Liu, N.; Gou, X. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury via transferring miR-199a-3p. J. Cell Physiol. 2019, 234, 23736–23749. [Google Scholar] [CrossRef]
  26. Wang, B.; Yao, K.; Huuskes, B.M.; Shen, H.H.; Zhuang, J.; Godson, C.; Brennan, E.P.; Wilkinson-Berka, J.L.; Wise, A.F.; Ricardo, S.D. Mesenchymal Stem Cells Deliver Exogenous MicroRNA-let7c via Exosomes to Attenuate Renal Fibrosis. Mol. Ther. 2016, 24, 1290–1301. [Google Scholar] [CrossRef] [Green Version]
  27. Wang, Y.; Fu, B.; Sun, X.; Li, D.; Huang, Q.; Zhao, W.; Chen, X. Differentially expressed microRNAs in bone marrow mesenchymal stem cell-derived microvesicles in young and older rats and their effect on tumor growth factor-beta1-mediated epithelial-mesenchymal transition in HK2 cells. Stem. Cell Res. Ther. 2015, 6, 185. [Google Scholar] [CrossRef] [Green Version]
  28. He, J.; Wang, Y.; Lu, X.; Zhu, B.; Pei, X.; Wu, J.; Zhao, W. Micro-vesicles derived from bone marrow stem cells protect the kidney both in vivo and in vitro by microRNA-dependent repairing. Nephrology 2015, 20, 591–600. [Google Scholar] [CrossRef]
  29. Grange, C.; Tritta, S.; Tapparo, M.; Cedrino, M.; Tetta, C.; Camussi, G.; Brizzi, M.F. Stem cell-derived extracellular vesicles inhibit and revert fibrosis progression in a mouse model of diabetic nephropathy. Sci. Rep. 2019, 9, 4468. [Google Scholar] [CrossRef] [Green Version]
  30. Zhong, L.; Liao, G.; Wang, X.; Li, L.; Zhang, J.; Chen, Y.; Liu, J.; Liu, S.; Wei, L.; Zhang, W.; et al. Mesenchymal stem cells-microvesicle-miR-451a ameliorate early diabetic kidney injury by negative regulation of P15 and P19. Exp. Biol. Med. Maywood 2019. [Google Scholar] [CrossRef]
  31. Yuan, X.; Wang, X.; Chen, C.; Zhou, J.; Han, M. Bone mesenchymal stem cells ameliorate ischemia/reperfusion-induced damage in renal epithelial cells via microRNA-223. Stem. Cell Res. Ther. 2017, 8, 146. [Google Scholar] [CrossRef] [PubMed]
  32. Xiang, J.; Jiang, T.; Zhang, W.; Xie, W.; Tang, X.; Zhang, J. Human umbilical cord-derived mesenchymal stem cells enhanced HK-2 cell autophagy through MicroRNA-145 by inhibiting the PI3K/AKT/mTOR signaling pathway. Exp. Cell Res. 2019, 378, 198–205. [Google Scholar] [CrossRef] [PubMed]
  33. Song, N.; Zhang, T.; Xu, X.; Lu, Z.; Yu, X.; Fang, Y.; Hu, J.; Jia, P.; Teng, J.; Ding, X. miR-21 Protects Against Ischemia/Reperfusion-Induced Acute Kidney Injury by Preventing Epithelial Cell Apoptosis and Inhibiting Dendritic Cell Maturation. Front. Physiol. 2018, 9, 790. [Google Scholar] [CrossRef] [PubMed]
  34. Chau, B.N.; Xin, C.; Hartner, J.; Ren, S.; Castano, A.P.; Linn, G.; Li, J.; Tran, P.T.; Kaimal, V.; Huang, X.; et al. MicroRNA-21 promotes fibrosis of the kidney by silencing metabolic pathways. Sci. Transl. Med. 2012, 4, 121ra118. [Google Scholar] [CrossRef] [Green Version]
  35. Tang, C.R.; Luo, S.G.; Lin, X.; Wang, J.; Liu, Y. Silenced miR-21 inhibits renal interstitial fibrosis via targeting ERK1/2 signaling pathway in mice. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 110–116. [Google Scholar] [CrossRef]
  36. Lyu, H.; Li, X.; Wu, Q.; Hao, L. Overexpression of microRNA-21 mediates Ang II-induced renal fibrosis by activating the TGF-beta1/Smad3 pathway via suppressing PPARalpha. J. Pharmacol. Sci. 2019, 141, 70–78. [Google Scholar] [CrossRef]
  37. Zhong, X.; Chung, A.C.; Chen, H.Y.; Dong, Y.; Meng, X.M.; Li, R.; Yang, W.; Hou, F.F.; Lan, H.Y. miR-21 is a key therapeutic target for renal injury in a mouse model of type 2 diabetes. Diabetologia 2013, 56, 663–674. [Google Scholar] [CrossRef]
  38. Sun, L.; Zhu, J.; Wu, M.; Sun, H.; Zhou, C.; Fu, L.; Xu, C.; Mei, C. Inhibition of MiR-199a-5p reduced cell proliferation in autosomal dominant polycystic kidney disease through targeting CDKN1C. Med. Sci. Monit. 2015, 21, 195–200. [Google Scholar] [CrossRef] [Green Version]
  39. Yang, A.; Liu, F.; Guan, B.; Luo, Z.; Lin, J.; Fang, W.; Liu, L.; Zuo, W. p53 induces miR-199a-3p to suppress mechanistic target of rapamycin activation in cisplatin-induced acute kidney injury. J. Cell Biochem. 2019, 120, 17625–17634. [Google Scholar] [CrossRef]
  40. Yang, R.; Xu, X.; Li, H.; Chen, J.; Xiang, X.; Dong, Z.; Zhang, D. p53 induces miR199a-3p to suppress SOCS7 for STAT3 activation and renal fibrosis in UUO. Sci. Rep. 2017, 7, 43409. [Google Scholar] [CrossRef] [Green Version]
  41. Ye, H.; Su, B.; Ni, H.; Li, L.; Chen, X.; You, X.; Zhang, H. microRNA-199a may be involved in the pathogenesis of lupus nephritis via modulating the activation of NF-kappaB by targeting Klotho. Mol. Immunol. 2018, 103, 235–242. [Google Scholar] [CrossRef] [PubMed]
  42. Sun, Z.; Ma, Y.; Chen, F.; Wang, S.; Chen, B.; Shi, J. miR-133b and miR-199b knockdown attenuate TGF-beta1-induced epithelial to mesenchymal transition and renal fibrosis by targeting SIRT1 in diabetic nephropathy. Eur. J. Pharmacol. 2018, 837, 96–104. [Google Scholar] [CrossRef] [PubMed]
  43. Yan, N.; Wen, L.; Peng, R.; Li, H.; Liu, H.; Peng, H.; Sun, Y.; Wu, T.; Chen, L.; Duan, Q.; et al. Naringenin Ameliorated Kidney Injury through Let-7a/TGFBR1 Signaling in Diabetic Nephropathy. J. Diabetes Res. 2016, 2016, 8738760. [Google Scholar] [CrossRef] [PubMed]
  44. Park, J.T.; Kato, M.; Lanting, L.; Castro, N.; Nam, B.Y.; Wang, M.; Kang, S.W.; Natarajan, R. Repression of let-7 by transforming growth factor-beta1-induced Lin28 upregulates collagen expression in glomerular mesangial cells under diabetic conditions. Am. J. Physiol. Renal. Physiol. 2014, 307, F1390–F1403. [Google Scholar] [CrossRef] [Green Version]
  45. Wang, B.; Jha, J.C.; Hagiwara, S.; McClelland, A.D.; Jandeleit-Dahm, K.; Thomas, M.C.; Cooper, M.E.; Kantharidis, P. Transforming growth factor-beta1-mediated renal fibrosis is dependent on the regulation of transforming growth factor receptor 1 expression by let-7b. Kidney Int. 2014, 85, 352–361. [Google Scholar] [CrossRef] [Green Version]
  46. Qi, M.; Yin, L.; Xu, L.; Tao, X.; Qi, Y.; Han, X.; Wang, C.; Xu, Y.; Sun, H.; Liu, K.; et al. Dioscin alleviates lipopolysaccharide-induced inflammatory kidney injury via the microRNA let-7i/TLR4/MyD88 signaling pathway. Pharmacol. Res. 2016, 111, 509–522. [Google Scholar] [CrossRef]
  47. Du, B.; Dai, X.M.; Li, S.; Qi, G.L.; Cao, G.X.; Zhong, Y.; Yin, P.D.; Yang, X.S. MiR-30c regulates cisplatin-induced apoptosis of renal tubular epithelial cells by targeting Bnip3L and Hspa5. Cell Death Dis. 2017, 8, e2987. [Google Scholar] [CrossRef] [Green Version]
  48. Zhang, C.; Yu, S.; Zheng, B.; Liu, D.; Wan, F.; Ma, Y.; Wang, J.; Gao, Z.; Shan, Z. miR-30c-5p Reduces Renal Ischemia-Reperfusion Involving Macrophage. Med. Sci. Monit. 2019, 25, 4362–4369. [Google Scholar] [CrossRef]
  49. Lang, Y.; Zhao, Y.; Zheng, C.; Lu, Y.; Wu, J.; Zhu, X.; Zhang, M.; Yang, F.; Xu, X.; Shi, S.; et al. MiR-30 family prevents uPAR-ITGB3 signaling activation through calcineurin-NFATC pathway to protect podocytes. Cell Death Dis. 2019, 10, 401. [Google Scholar] [CrossRef]
  50. Zhao, Y.; Wu, J.; Zhang, M.; Zhou, M.; Xu, F.; Zhu, X.; Zhou, X.; Lang, Y.; Yang, F.; Yun, S.; et al. Angiotensin II induces calcium/calcineurin signaling and podocyte injury by downregulating microRNA-30 family members. J. Mol. Med. (Berl.) 2017, 95, 887–898. [Google Scholar] [CrossRef]
  51. Guo, Y.; Deng, X.; Chen, S.; Yang, L.; Ni, J.; Wang, R.; Lin, J.; Bai, M.; Jia, Z.; Huang, S.; et al. MicroRNA-30e targets BNIP3L to protect against aldosterone-induced podocyte apoptosis and mitochondrial dysfunction. Am. J. Physiol. Renal. Physiol. 2017, 312, F589–F598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Peng, R.; Zhou, L.; Zhou, Y.; Zhao, Y.; Li, Q.; Ni, D.; Hu, Y.; Long, Y.; Liu, J.; Lyu, Z.; et al. MiR-30a Inhibits the Epithelial--Mesenchymal Transition of Podocytes through Downregulation of NFATc3. Int. J. Mol. Sci. 2015, 16, 24032–24047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Tung, C.W.; Ho, C.; Hsu, Y.C.; Huang, S.C.; Shih, Y.H.; Lin, C.L. MicroRNA-29a Attenuates Diabetic Glomerular Injury through Modulating Cannabinoid Receptor 1 Signaling. Molecules 2019, 24, 264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hsu, Y.C.; Chang, P.J.; Ho, C.; Huang, Y.T.; Shih, Y.H.; Wang, C.J.; Lin, C.L. Protective effects of miR-29a on diabetic glomerular dysfunction by modulation of DKK1/Wnt/beta-catenin signaling. Sci. Rep. 2016, 6, 30575. [Google Scholar] [CrossRef] [Green Version]
  55. Hu, H.; Hu, S.; Xu, S.; Gao, Y.; Zeng, F.; Shui, H. miR-29b regulates Ang II-induced EMT of rat renal tubular epithelial cells via targeting PI3K/AKT signaling pathway. Int. J. Mol. Med. 2018, 42, 453–460. [Google Scholar] [CrossRef] [Green Version]
  56. Saito, S.; Ohno, S.I.; Harada, Y.; Oikawa, K.; Fujita, K.; Mineo, S.; Gondo, A.; Kanno, Y.; Kuroda, M. rAAV6-mediated miR-29b delivery suppresses renal fibrosis. Clin. Exp. Nephrol. 2019, 23, 1345–1356. [Google Scholar] [CrossRef]
  57. Liu, H.; He, X.J.; Li, G.J.; Ding, Q.X.; Liang, W.X.; Fan, J. Effects of microRNA-145 on epithelial-mesenchymal transition of TGF-beta1-induced human renal proximal tubular epithelial cells. Zhongguo Dang Dai Er Ke Za Zhi 2017, 19, 712–718. [Google Scholar]
  58. Liu, B.; Qiang, L.; Wang, G.D.; Duan, Q.; Liu, J. LncRNA MALAT1 facilities high glucose induced endothelial to mesenchymal transition and fibrosis via targeting miR-145/ZEB2 axis. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 3478–3486. [Google Scholar] [CrossRef]
  59. Liu, L.L.; Li, D.; He, Y.L.; Zhou, Y.Z.; Gong, S.H.; Wu, L.Y.; Zhao, Y.Q.; Huang, X.; Zhao, T.; Xu, L.; et al. miR-210 protects renal cell against hypoxia-induced apoptosis by targeting HIF-1 alpha. Mol. Med. 2017, 23, 258–271. [Google Scholar] [CrossRef] [Green Version]
  60. Liu, F.; Lou, Y.L.; Wu, J.; Ruan, Q.F.; Xie, A.; Guo, F.; Cui, S.P.; Deng, Z.F.; Wang, Y. Upregulation of microRNA-210 regulates renal angiogenesis mediated by activation of VEGF signaling pathway under ischemia/perfusion injury in vivo and in vitro. Kidney Blood Press Res. 2012, 35, 182–191. [Google Scholar] [CrossRef]
  61. Zhang, C.; Ma, P.; Zhao, Z.; Jiang, N.; Lian, D.; Huo, P.; Yang, H. miRNAmRNA regulatory network analysis of mesenchymal stem cell treatment in cisplatininduced acute kidney injury identifies roles for miR210/Serpine1 and miR378/Fos in regulating inflammation. Mol. Med. Rep. 2019, 20, 1509–1522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Zhang, Y.; Zhao, S.; Wu, D.; Liu, X.; Shi, M.; Wang, Y.; Zhang, F.; Ding, J.; Xiao, Y.; Guo, B. MicroRNA-22 Promotes Renal Tubulointerstitial Fibrosis by Targeting PTEN and Suppressing Autophagy in Diabetic Nephropathy. J. Diabetes Res. 2018, 2018, 4728645. [Google Scholar] [CrossRef] [PubMed]
  63. Long, J.; Badal, S.S.; Wang, Y.; Chang, B.H.; Rodriguez, A.; Danesh, F.R. MicroRNA-22 is a master regulator of bone morphogenetic protein-7/6 homeostasis in the kidney. J. Biol. Chem. 2013, 288, 36202–36214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Joo, M.S.; Lee, C.G.; Koo, J.H.; Kim, S.G. miR-125b transcriptionally increased by Nrf2 inhibits AhR repressor, which protects kidney from cisplatin-induced injury. Cell Death Dis. 2013, 4, e899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Huang, Y.F.; Zhang, Y.; Liu, C.X.; Huang, J.; Ding, G.H. microRNA-125b contributes to high glucose-induced reactive oxygen species generation and apoptosis in HK-2 renal tubular epithelial cells by targeting angiotensin-converting enzyme 2. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 4055–4062. [Google Scholar]
  66. Liang, H.; Yang, K.; Xin, M.; Liu, X.; Zhao, L.; Liu, B.; Wang, J. MiR-130a protects against lipopolysaccharide-induced glomerular cell injury by upregulation of Klotho. Pharmazie 2017, 72, 468–474. [Google Scholar] [CrossRef]
  67. Liu, D.; Liu, F.; Wang, X.; Qiao, Y.; Pan, S.; Yang, Y.; Hu, Y.; Zhang, Y.; Tian, F.; Liu, Z. MiR-130a-5p prevents angiotensin II-induced podocyte apoptosis by modulating M-type phospholipase A2 receptor. Cell Cycle 2018, 17, 2484–2495. [Google Scholar] [CrossRef] [Green Version]
  68. Ai, K.; Zhu, X.; Kang, Y.; Li, H.; Zhang, L. miR-130a-3p inhibition protects against renal fibrosis in vitro via the TGF-beta1/Smad pathway by targeting SnoN. Exp. Mol. Pathol. 2019, 112, 104358. [Google Scholar] [CrossRef]
  69. Xu, H.; Sun, F.; Li, X.; Sun, L. Down-regulation of miR-23a inhibits high glucose-induced EMT and renal fibrogenesis by up-regulation of SnoN. Hum. Cell 2018, 31, 22–32. [Google Scholar] [CrossRef] [Green Version]
  70. Das, A.; Ganesh, K.; Khanna, S.; Sen, C.K.; Roy, S. Engulfment of apoptotic cells by macrophages: a role of microRNA-21 in the resolution of wound inflammation. J. Immunol. 2014, 192, 1120–1129. [Google Scholar] [CrossRef] [Green Version]
  71. Kolling, M.; Kaucsar, T.; Schauerte, C.; Hubner, A.; Dettling, A.; Park, J.K.; Busch, M.; Wulff, X.; Meier, M.; Scherf, K.; et al. Therapeutic miR-21 Silencing Ameliorates Diabetic Kidney Disease in Mice. Mol. Ther. 2017, 25, 165–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Xu, X.; Kriegel, A.J.; Jiao, X.; Liu, H.; Bai, X.; Olson, J.; Liang, M.; Ding, X. miR-21 in ischemia/reperfusion injury: a double-edged sword? Physiol. Genom. 2014, 46, 789–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Sayed, D.; He, M.; Hong, C.; Gao, S.; Rane, S.; Yang, Z.; Abdellatif, M. MicroRNA-21 is a downstream effector of AKT that mediates its antiapoptotic effects via suppression of Fas ligand. J. Biol. Chem. 2010, 285, 20281–20290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Wynn, T.A. Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J. Clin. Investig. 2007, 117, 524–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Loboda, A.; Sobczak, M.; Jozkowicz, A.; Dulak, J. TGF-beta1/Smads and miR-21 in Renal Fibrosis and Inflammation. Mediat. Inflamm. 2016, 2016, 8319283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Li, Q.; Xia, X.; Ji, J.; Ma, J.; Tao, L.; Mo, L.; Chen, W. MiR-199a-3p enhances cisplatin sensitivity of cholangiocarcinoma cells by inhibiting mTOR signaling pathway and expression of MDR1. Oncotarget 2017, 8, 33621–33630. [Google Scholar] [CrossRef] [Green Version]
  77. Kang, W.L.; Xu, G.S. Atrasentan increased the expression of klotho by mediating miR-199b-5p and prevented renal tubular injury in diabetic nephropathy. Sci. Rep. 2016, 6, 19979. [Google Scholar] [CrossRef]
  78. Balakrishnan, I.; Yang, X.; Brown, J.; Ramakrishnan, A.; Torok-Storb, B.; Kabos, P.; Hesselberth, J.R.; Pillai, M.M. Genome-wide analysis of miRNA-mRNA interactions in marrow stromal cells. Stem. Cells 2014, 32, 662–673. [Google Scholar] [CrossRef] [Green Version]
  79. Schug, J.; McKenna, L.B.; Walton, G.; Hand, N.; Mukherjee, S.; Essuman, K.; Shi, Z.; Gao, Y.; Markley, K.; Nakagawa, M.; et al. Dynamic recruitment of microRNAs to their mRNA targets in the regenerating liver. BMC Genom. 2013, 14, 264. [Google Scholar] [CrossRef] [Green Version]
  80. Zhang, J.; Zhang, H.; Liu, J.; Tu, X.; Zang, Y.; Zhu, J.; Chen, J.; Dong, L.; Zhang, J. miR-30 inhibits TGF-beta1-induced epithelial-to-mesenchymal transition in hepatocyte by targeting Snail1. Biochem. Biophys. Res. Commun. 2012, 417, 1100–1105. [Google Scholar] [CrossRef]
  81. Yang, Q.; Sun, M.; Chen, Y.; Lu, Y.; Ye, Y.; Song, H.; Xu, X.; Shi, S.; Wang, J. Triptolide protects podocytes from TGF-beta-induced injury by preventing miR-30 downregulation. Am. J. Transl. Res. 2017, 9, 5150–5159. [Google Scholar] [PubMed]
  82. Wang, B.; Komers, R.; Carew, R.; Winbanks, C.E.; Xu, B.; Herman-Edelstein, M.; Koh, P.; Thomas, M.; Jandeleit-Dahm, K.; Gregorevic, P.; et al. Suppression of microRNA-29 expression by TGF-beta1 promotes collagen expression and renal fibrosis. J. Am. Soc. Nephrol. 2012, 23, 252–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Cantaluppi, V.; Gatti, S.; Medica, D.; Figliolini, F.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Biancone, L.; Tetta, C.; Camussi, G. Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia-reperfusion injury by microRNA-dependent reprogramming of resident renal cells. Kidney Int. 2012, 82, 412–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chandrasekaran, K.; Karolina, D.S.; Sepramaniam, S.; Armugam, A.; Wintour, E.M.; Bertram, J.F.; Jeyaseelan, K. Role of microRNAs in kidney homeostasis and disease. Kidney Int. 2012, 81, 617–627. [Google Scholar] [CrossRef] [Green Version]
  85. Wang, H.; Wang, B.; Zhang, A.; Hassounah, F.; Seow, Y.; Wood, M.; Ma, F.; Klein, J.D.; Price, S.R.; Wang, X.H. Exosome-Mediated miR-29 Transfer Reduces Muscle Atrophy and Kidney Fibrosis in Mice. Mol. Ther. 2019, 27, 571–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Gan, L.; Liu, Z.; Wei, M.; Chen, Y.; Yang, X.; Chen, L.; Xiao, X. MiR-210 and miR-155 as potential diagnostic markers for pre-eclampsia pregnancies. Medicine 2017, 96, e7515. [Google Scholar] [CrossRef] [PubMed]
  87. Hills, C.E.; Squires, P.E. The role of TGF-beta and epithelial-to mesenchymal transition in diabetic nephropathy. Cytokine Growth Factor Rev. 2011, 22, 131–139. [Google Scholar] [CrossRef] [Green Version]
  88. Lv, S.; Liu, G.; Sun, A.; Wang, J.; Cheng, J.; Wang, W.; Liu, X.; Nie, H.; Guan, G. Mesenchymal stem cells ameliorate diabetic glomerular fibrosis in vivo and in vitro by inhibiting TGF-beta signalling via secretion of bone morphogenetic protein 7. Diab. Vasc. Dis. Res. 2014, 11, 251–261. [Google Scholar] [CrossRef]
  89. Di, J.; Jiang, L.; Zhou, Y.; Cao, H.; Fang, L.; Wen, P.; Li, X.; Dai, C.; Yang, J. Ets-1 targeted by microrna-221 regulates angiotensin II-induced renal fibroblast activation and fibrosis. Cell Physiol. Biochem. 2014, 34, 1063–1074. [Google Scholar] [CrossRef]
  90. Chen, S.; Yao, Y.; Lin, F.; Bian, F.; Zhu, C.; Jiang, G. MiR-424 is over-expressed and attenuates ischemia-reperfusion kidney injury via p53 and death receptor 6 pathway. Am. J. Transl. Res. 2019, 11, 1965–1979. [Google Scholar]
  91. Qin, Y.; Wang, G.; Peng, Z. MicroRNA-191-5p diminished sepsis-induced acute kidney injury through targeting oxidative stress responsive 1 in rat models. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Oh, H.J.; Kato, M.; Deshpande, S.; Zhang, E.; Das, S.; Lanting, L.; Wang, M.; Natarajan, R. Inhibition of the processing of miR-25 by HIPK2-Phosphorylated-MeCP2 induces NOX4 in early diabetic nephropathy. Sci. Rep. 2016, 6, 38789. [Google Scholar] [CrossRef] [PubMed]
  93. Zheng, Z.; Hu, H.; Tong, Y.; Hu, Z.; Cao, S.; Shan, C.; Lin, W.; Yin, Y.; Li, Z. MiR-27b regulates podocyte survival through targeting adenosine receptor 2B in podocytes from non-human primate. Cell Death Dis. 2018, 9, 1133. [Google Scholar] [CrossRef] [PubMed]
  94. Lorenzen, J.M.; Kaucsar, T.; Schauerte, C.; Schmitt, R.; Rong, S.; Hubner, A.; Scherf, K.; Fiedler, J.; Martino, F.; Kumarswamy, R.; et al. MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J. Am. Soc. Nephrol. 2014, 25, 2717–2729. [Google Scholar] [CrossRef] [Green Version]
  95. Liu, H.; Wang, X.; Wang, Z.Y.; Li, L. Circ_0080425 inhibits cell proliferation and fibrosis in diabetic nephropathy via sponging miR-24-3p and targeting fibroblast growth factor 11. J. Cell Physiol. 2019. [Google Scholar] [CrossRef]
  96. Vinas, J.L.; Burger, D.; Zimpelmann, J.; Haneef, R.; Knoll, W.; Campbell, P.; Gutsol, A.; Carter, A.; Allan, D.S.; Burns, K.D. Transfer of microRNA-486-5p from human endothelial colony forming cell-derived exosomes reduces ischemic kidney injury. Kidney Int. 2016, 90, 1238–1250. [Google Scholar] [CrossRef] [Green Version]
  97. Shan, Q.; Zheng, G.; Zhu, A.; Cao, L.; Lu, J.; Wu, D.; Zhang, Z.; Fan, S.; Sun, C.; Hu, B.; et al. Epigenetic modification of miR-10a regulates renal damage by targeting CREB1 in type 2 diabetes mellitus. Toxicol. Appl. Pharmacol. 2016, 306, 134–143. [Google Scholar] [CrossRef]
  98. Yang, H.; Wang, Q.; Li, S. MicroRNA-218 promotes high glucose-induced apoptosis in podocytes by targeting heme oxygenase-1. Biochem. Biophys. Res. Commun. 2016, 471, 582–588. [Google Scholar] [CrossRef]
  99. Yang, X.; Wang, X.; Nie, F.; Liu, T.; Yu, X.; Wang, H.; Li, Q.; Peng, R.; Mao, Z.; Zhou, Q.; et al. miR-135 family members mediate podocyte injury through the activation of Wnt/beta-catenin signaling. Int. J. Mol. Med. 2015, 36, 669–677. [Google Scholar] [CrossRef] [Green Version]
  100. Liu, Z.M.; Zheng, H.Y.; Chen, L.H.; Li, Y.L.; Wang, Q.; Liao, C.F.; Li, X.W. Low expression of miR-203 promoted diabetic nephropathy via increasing TLR4. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 5627–5634. [Google Scholar] [CrossRef]
  101. Xie, F.; Li, X.; Wei, C.; Gou, L.; Dang, Y.; Shan, Z. [Transfected miR-1908 inhibits renal fibrosis via targeting transforming growth factor beta 1]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2015, 31, 1682–1685. [Google Scholar] [PubMed]
  102. Gao, F.; Chiu, S.M.; Motan, D.A.; Zhang, Z.; Chen, L.; Ji, H.L.; Tse, H.F.; Fu, Q.L.; Lian, Q. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016, 7, e2062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Wang, B.; Jia, H.; Zhang, B.; Wang, J.; Ji, C.; Zhu, X.; Yan, Y.; Yin, L.; Yu, J.; Qian, H.; et al. Pre-incubation with hucMSC-exosomes prevents cisplatin-induced nephrotoxicity by activating autophagy. Stem. Cell Res. Ther. 2017, 8, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lin, K.C.; Yip, H.K.; Shao, P.L.; Wu, S.C.; Chen, K.H.; Chen, Y.T.; Yang, C.C.; Sun, C.K.; Kao, G.S.; Chen, S.Y.; et al. Combination of adipose-derived mesenchymal stem cells (ADMSC) and ADMSC-derived exosomes for protecting kidney from acute ischemia-reperfusion injury. Int. J. Cardiol. 2016, 216, 173–185. [Google Scholar] [CrossRef] [PubMed]
  105. Reis, L.A.; Borges, F.T.; Simoes, M.J.; Borges, A.A.; Sinigaglia-Coimbra, R.; Schor, N. Bone marrow-derived mesenchymal stem cells repaired but did not prevent gentamicin-induced acute kidney injury through paracrine effects in rats. PLoS ONE 2012, 7, e44092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wang, R.; Lin, M.; Li, L.; Li, L.; Qi, G.; Rong, R.; Xu, M.; Zhu, T. [Bone marrow mesenchymal stem cell-derived exosome protects kidney against ischemia reperfusion injury in rats]. Zhonghua Yi Xue Za Zhi 2014, 94, 3298–3303. [Google Scholar] [PubMed]
  107. Eirin, A.; Zhu, X.Y.; Puranik, A.S.; Tang, H.; McGurren, K.A.; van Wijnen, A.J.; Lerman, A.; Lerman, L.O. Mesenchymal stem cell-derived extracellular vesicles attenuate kidney inflammation. Kidney Int. 2017, 92, 114–124. [Google Scholar] [CrossRef]
  108. Zou, X.; Gu, D.; Zhang, G.; Zhong, L.; Cheng, Z.; Liu, G.; Zhu, Y. NK Cell Regulatory Property is Involved in the Protective Role of MSC-Derived Extracellular Vesicles in Renal Ischemic Reperfusion Injury. Hum. Gene. Ther. 2016, 27, 926–935. [Google Scholar] [CrossRef]
  109. Choi, H.Y.; Lee, H.G.; Kim, B.S.; Ahn, S.H.; Jung, A.; Lee, M.; Lee, J.E.; Kim, H.J.; Ha, S.K.; Park, H.C. Mesenchymal stem cell-derived microparticles ameliorate peritubular capillary rarefaction via inhibition of endothelial-mesenchymal transition and decrease tubulointerstitial fibrosis in unilateral ureteral obstruction. Stem. Cell Res. Ther. 2015, 6, 18. [Google Scholar] [CrossRef] [Green Version]
  110. He, J.; Wang, Y.; Sun, S.; Yu, M.; Wang, C.; Pei, X.; Zhu, B.; Wu, J.; Zhao, W. Bone marrow stem cells-derived microvesicles protect against renal injury in the mouse remnant kidney model. Nephrology 2012, 17, 493–500. [Google Scholar] [CrossRef]
  111. Gennai, S.; Monsel, A.; Hao, Q.; Park, J.; Matthay, M.A.; Lee, J.W. Microvesicles Derived From Human Mesenchymal Stem Cells Restore Alveolar Fluid Clearance in Human Lungs Rejected for Transplantation. Am. J. Transpl. 2015, 15, 2404–2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Harting, M.T.; Srivastava, A.K.; Zhaorigetu, S.; Bair, H.; Prabhakara, K.S.; Toledano Furman, N.E.; Vykoukal, J.V.; Ruppert, K.A.; Cox, C.S., Jr.; Olson, S.D. Inflammation-Stimulated Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Inflammation. Stem. Cells 2018, 36, 79–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Veceric-Haler, Z.; Cerar, A.; Perse, M. (Mesenchymal) Stem Cell-Based Therapy in Cisplatin-Induced Acute Kidney Injury Animal Model: Risk of Immunogenicity and Tumorigenicity. Stem. Cells Int. 2017, 2017, 7304643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kilpinen, L.; Impola, U.; Sankkila, L.; Ritamo, I.; Aatonen, M.; Kilpinen, S.; Tuimala, J.; Valmu, L.; Levijoki, J.; Finckenberg, P.; et al. Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning. J. Extracell Vesicles 2013, 2. [Google Scholar] [CrossRef]
  115. Bruno, S.; Tapparo, M.; Collino, F.; Chiabotto, G.; Deregibus, M.C.; Soares Lindoso, R.; Neri, F.; Kholia, S.; Giunti, S.; Wen, S.; et al. Renal Regenerative Potential of Different Extracellular Vesicle Populations Derived from Bone Marrow Mesenchymal Stromal Cells. Tissue Eng. Part A 2017, 23, 1262–1273. [Google Scholar] [CrossRef]
  116. Zou, X.; Gu, D.; Xing, X.; Cheng, Z.; Gong, D.; Zhang, G.; Zhu, Y. Human mesenchymal stromal cell-derived extracellular vesicles alleviate renal ischemic reperfusion injury and enhance angiogenesis in rats. Am. J. Transl. Res. 2016, 8, 4289–4299. [Google Scholar]
  117. Zhang, G.; Zou, X.; Miao, S.; Chen, J.; Du, T.; Zhong, L.; Ju, G.; Liu, G.; Zhu, Y. The anti-oxidative role of micro-vesicles derived from human Wharton-Jelly mesenchymal stromal cells through NOX2/gp91(phox) suppression in alleviating renal ischemia-reperfusion injury in rats. PLoS ONE 2014, 9, e92129. [Google Scholar] [CrossRef]
  118. Choi, H.Y.; Moon, S.J.; Ratliff, B.B.; Ahn, S.H.; Jung, A.; Lee, M.; Lee, S.; Lim, B.J.; Kim, B.S.; Plotkin, M.D.; et al. Microparticles from kidney-derived mesenchymal stem cells act as carriers of proangiogenic signals and contribute to recovery from acute kidney injury. PLoS ONE 2014, 9, e87853. [Google Scholar] [CrossRef] [Green Version]
  119. Zhou, Y.; Xu, H.; Xu, W.; Wang, B.; Wu, H.; Tao, Y.; Zhang, B.; Wang, M.; Mao, F.; Yan, Y.; et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem. Cell Res. Ther. 2013, 4, 34. [Google Scholar] [CrossRef] [Green Version]
  120. Gatti, S.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Cantaluppi, V.; Tetta, C.; Camussi, G. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol. Dial. Transpl. 2011, 26, 1474–1483. [Google Scholar] [CrossRef] [Green Version]
  121. Miller, S.B.; Martin, D.R.; Kissane, J.; Hammerman, M.R. Hepatocyte growth factor accelerates recovery from acute ischemic renal injury in rats. Am. J. Physiol. 1994, 266, F129–F134. [Google Scholar] [CrossRef] [PubMed]
  122. Ishibe, S.; Cantley, L.G. Epithelial-mesenchymal-epithelial cycling in kidney repair. Curr. Opin. Nephrol. Hypertens 2008, 17, 379–385. [Google Scholar] [CrossRef] [PubMed]
  123. Gobe, G.C.; Johnson, D.W. Distal tubular epithelial cells of the kidney: Potential support for proximal tubular cell survival after renal injury. Int. J. Biochem. Cell Biol. 2007, 39, 1551–1561. [Google Scholar] [CrossRef]
  124. Ju, G.Q.; Cheng, J.; Zhong, L.; Wu, S.; Zou, X.Y.; Zhang, G.Y.; Gu, D.; Miao, S.; Zhu, Y.J.; Sun, J.; et al. Microvesicles derived from human umbilical cord mesenchymal stem cells facilitate tubular epithelial cell dedifferentiation and growth via hepatocyte growth factor induction. PLoS ONE 2015, 10, e0121534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Zhang, G.; Zou, X.; Huang, Y.; Wang, F.; Miao, S.; Liu, G.; Chen, M.; Zhu, Y. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Protect Against Acute Kidney Injury Through Anti-Oxidation by Enhancing Nrf2/ARE Activation in Rats. Kidney Blood Press. Res. 2016, 41, 119–128. [Google Scholar] [CrossRef] [PubMed]
  126. Bruno, S.; Grange, C.; Collino, F.; Deregibus, M.C.; Cantaluppi, V.; Biancone, L.; Tetta, C.; Camussi, G. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS ONE 2012, 7, e33115. [Google Scholar] [CrossRef] [PubMed]
  127. Liesa, M.; Palacin, M.; Zorzano, A. Mitochondrial dynamics in mammalian health and disease. Physiol. Rev. 2009, 89, 799–845. [Google Scholar] [CrossRef] [Green Version]
  128. Zhan, M.; Brooks, C.; Liu, F.; Sun, L.; Dong, Z. Mitochondrial dynamics: regulatory mechanisms and emerging role in renal pathophysiology. Kidney Int. 2013, 83, 568–581. [Google Scholar] [CrossRef] [Green Version]
  129. Brooks, C.; Wei, Q.; Cho, S.G.; Dong, Z. Regulation of mitochondrial dynamics in acute kidney injury in cell culture and rodent models. J. Clin. Investig. 2009, 119, 1275–1285. [Google Scholar] [CrossRef]
  130. Lv, S.; Cheng, J.; Sun, A.; Li, J.; Wang, W.; Guan, G.; Liu, G.; Su, M. Mesenchymal stem cells transplantation ameliorates glomerular injury in streptozotocin-induced diabetic nephropathy in rats via inhibiting oxidative stress. Diabetes Res. Clin. Pract. 2014, 104, 143–154. [Google Scholar] [CrossRef]
Figure 1. Schema of the effects of mesenchymal stem cell extracellular vesicles (MSC-EVs) in renal injury. Arrows: promotion of the processes; T-bars: inhibition of the processes.
Figure 1. Schema of the effects of mesenchymal stem cell extracellular vesicles (MSC-EVs) in renal injury. Arrows: promotion of the processes; T-bars: inhibition of the processes.
Ijms 21 00756 g001
Table 1. The study of miRNA therapy from MSC-EVs in renal diseases.
Table 1. The study of miRNA therapy from MSC-EVs in renal diseases.
AnimalModelMiRNAsTarget Protein/Gene/SignalFunctionRef.
RatGlycerolmiRNAsmultipleanti-inflammation[19]
MouseI/RmiR-223NLRP3anti-apoptosis[31]
RatI/RmiR-15a/-15b/-16CX3CL1antifibrosis/anti-inflammation[23]
RatI/RmiR-30DRP1anti-apoptosis/preserve mitochondria[24]
HK-2 cell-miR-145PI3K/AKT/mTORpromote autophagy[32]
HK-2 cell-miR-133b-3p/-294TGF-β1/EMTantifibrosis[27]
MouseUUOmiRNAsEMTantifibrosis[28]
MouseUUOlet-7c-antifibrosis[26]
MouseDiabetesmiR-451aP15/P19/EMTantifibrosis[30]
MouseDiabetesmiRNAs-antifibrosis[29]
I/R: Ischemia/reperfusion; UUO: Unilateral ureteral obstruction; CX3CL1: C-X3-C motif chemokine ligand 1; DRP1: Dynamic related protein 1; PI3K: Phosphatidylinositol 3-kinase; AKT: Protein kinase B; mTOR: Mammalian target of rapamycin; TGF-β1: Transforming growth factor -β1; EMT: Epithelial-mesenchymal transition; P15: P15INK4b; P19: P19INK4d.
Table 2. MiRNAs in MSC-EVs in renal regeneration and pathophysiology.
Table 2. MiRNAs in MSC-EVs in renal regeneration and pathophysiology.
MiRNARenal InjuryTarget Protein/Gene/SignalFunctionReference
miR-21I/RPTEN/AKT/mTOR/HIFanti-apoptosis-
PDCD4/NF-κBinhibit dendritic cell maturation[33]
DiabetesCdc25/Cdk6promote DKD:mesangial expansion, renal fibrosis, macrophage[34]
UUOPRARαpromote renal fibrosis[35]
UUOERK1/ERK2promote renal fibrosis[36]
Ang-IIPRARα/TGF-β1promote renal fibrosis[37]
DiabetesSmad7/TGF-β/NF-κBpromote renal fibrosis/inflammation[38]
miR-199
miR-199aI/RSema3A/AKT/ERKanti-apoptosis[25]
PKDCDKN1Cinhibition of cyst growth[39]
CisplatinCaspase-3promote apoptosis[40]
TGF-β1SOCS7/STAT3promote renal fibrosis[41]
LupusKlotho/NF-κBinflammation[42]
miR-199bDiabetesSIRT1promote renal fibrosis[43]
Let-7 familyDiabetesTGFBR1ameliorate DKD[44]
TGF-β1COL1a2/COL4a1improve renal injury[45]
TGF-β1TGFBR1antifibrosis[46]
LPSTLR4reduce inflammatory injury[47]
UUOMMP9/TGF-β1/TGFBR1antifibrosis[26]
miR-30I/RDRP1anti-apoptosis/preserve mitochondria[24]
CisplatinBnip3L/Hspa5anti-apoptosis[48]
I/RM1-M2 macrophage transitionanti-inflammation[49]
UUOEMTantifibrosis[28]
miR-30-KDuPAR-ITGB3protect podocytophaty[50]
Ang-IICalcium/calcineurin signalingprotect podocytophaty[51]
AldosteroneBnip3anti-apoptosis/preserve mitochondria[52]
AdriamycinNFATc3/EMTprotect podocytopathy[53]
miR-29
miR-29aDiabetesPPARγ/CB1Rameliorate DKD[54]
DiabetesDKK1/Wnt/bcateninameliorate DKD[55]
miR-29bAng-IIPI3K/AKTreduce EMT[56]
UUOSnailantifibrosis[57]
miR-145-PI3K/AKT/mTORpromote autophagy[32]
TGF-β1TGF-β/Smad signalingantifibrosis[58]
High-glucoseZEB2/EMTantifibrosis[59]
miR-210UUOEMTantifibrosis[28]
HypoxiaHIF-1αanti-apoptosis[60]
I/RVEGF pathwaypromote angiogenesis[61]
CisplatinSerpine1inflammatory regulation[62]
miR-22DiabetesPTEN/AKT/mTORpromote fibrosis/suppress autophagy[63]
UUOBMP-6/BMP-7 signalingpromote fibrosis[64]
miR-125bCisplatinAhR receptoranti-apoptosis[65]
High-glucoseACE/ROSpromote apoptosis[66]
miR-130aLPSKlotho/PI3K/AKT/Wnt/NF-κBprotect glomerular injury[67]
Ang-IIPLA2Ranti-apoptosis[68]
TGF-β1SnoN/TGF-β1/Smadpromote fibrosis[69]
miR-23aHigh-glucoseEMTpromote fibrosis[70]
miR-15a/15b/16I/RCX3CL1antifibrosis/anti-inflammation[23]
I/R: ischemia/reperfusion; UUO: unilateral ureteral obstruction; Ang-II: Angiotensin-II; PKD: olycystic kidney disease; TGF-β1: transforming growth factor-β1; KD: knockdown; LPS: lipopolysaccharide; PTEN: phosphatase and tensin homolog deleted from chromosome 10; AKT: protein kinase B; mTOR: mammalian target of rapamycin; HIF: hypoxia-inducible factor; NF-κB: nuclear factor-κb; PRARα: peroxisome proliferator-activated receptorα; ERK: extracellular signal-regulated kinase; Sema3A: semaphorin3a; TLR4: toll-like receptor4; MMP9: matrix metalloproteinase9; DRP1: dynamic related protein 1; EMT: epithelial-mesenchymal transition; VEGF: vascular endothelial growth factor; BMP: bone morphogenetic protein; AhR: aryl hydrocarbon receptor; ACE: angiotensin-converting-enzyme; ROS: reactive oxygen species; PLA2R: phospholipase A2 receptor; CX3CL1: C-X-C motif chemokine ligand1; DKD: diabetic kidney disease.

Share and Cite

MDPI and ACS Style

Tsuji, K.; Kitamura, S.; Wada, J. Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Renal Diseases. Int. J. Mol. Sci. 2020, 21, 756. https://doi.org/10.3390/ijms21030756

AMA Style

Tsuji K, Kitamura S, Wada J. Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Renal Diseases. International Journal of Molecular Sciences. 2020; 21(3):756. https://doi.org/10.3390/ijms21030756

Chicago/Turabian Style

Tsuji, Kenji, Shinji Kitamura, and Jun Wada. 2020. "Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Renal Diseases" International Journal of Molecular Sciences 21, no. 3: 756. https://doi.org/10.3390/ijms21030756

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