Abstract
Background
Diabetic kidney disease (DKD) is a major complication of diabetes mellitus and a common cause of end-stage kidney disease. The incidence of DKD is rising worldwide and associated with increased morbidity and premature mortality, indicating an urgent need to further explore the underlying pathogenesis and potential biomarkers. Exosomes are nanoscale vesicles secreted by all cell types that play an essential role in cellular homeostasis and intercellular communications by transferring molecular cargoes between different cells.
Summary
Emerging evidence indicates that exosomes are both a crucial signaling mediator and a potential biomarker of DKD. On the one hand, exosomes released by various kidney resident cells facilitate the cell-cell crosstalk as a contributing factor in DKD; on the other hand, exosomes can be detected from urine and blood and have emerged as promising noninvasive biomarkers for DKD.
Key Messages
Herein, we highlight the recent advances in research on the role of exosomes from different kidney resident cells in DKD. We further discuss the potential use of urine exosomes as biomarkers and therapeutic agents.
Keywords: Diabetic kidney disease, Exosomes, Intercellular communication, Biomarker
Introduction
Diabetic kidney disease (DKD) is a progressive kidney disease that occurs as a principal long-term complication of diabetes mellitus (DM). It is one of the most common causes of chronic kidney disease (CKD) and end-stage kidney disease worldwide, accounting for 44.5% of patients requiring dialysis or renal transplantation [1]. Approximately half of the patients with type 2 DM and one-third with type 1 DM develop DKD. In conjunction with the rising incidence of DM, the prevalence of DKD is rapidly increasing, which imposes a huge public health and economic burden on a global level [2, 3]. The natural history of DKD is characterized by microalbuminuria and progressive loss of renal function. Microalbuminuria is currently the best noninvasive marker available for assessing the development and progression of DKD. However, 15–65% of patients with microalbuminuria regress to normoalbuminuria and do not progress to macroalbuminuria or CKD. Moreover, 20–60% of DM patients develop DKD with a decreased glomerular filtration rate but without microalbuminuria [4]. Furthermore, the current prevention and intervention strategies which mainly focus on controlling the hyperglycemia, hypertension, dyslipidemia, and suppressing the renin-angiotensin-aldosterone (RAAS) system do not succeed for all DKD patients since the prevalence of DKD and CKD continues to increase progressively, thus highlighting the urgent need to elucidate the pathogenesis further and thereby identify diagnostic and predictive biomarker and new therapeutic options of DKD.
Thickening of the glomerular basement membrane and tubular basement membrane is the first pathological sign of DKD, accompanied by the proliferation of mesangial cells, podocyte loss, and inflammatory cell infiltration. Kidney resident cells are involved in the whole pathophysiological process of DKD. The cell-cell interactions among kidney resident cells are based on complex intercellular signaling networks. Through cellular communication by secreted factors, exosomes have been recognized as professional information mediators involved in the pathogenesis of DKD. However, the understanding of the role of exosomes in DKD remains limited [5, 6] since the function of exosomes is mainly driven by the transmission of exosomal cargos and the type of cargos depends on the origin cells where exosomes are secreted. In this review, we focus on the role of exosomes derived from different origins, including renal resident cells and other resources such as mesenchymal stromal cells (MSCs), serum and exogenous exosomes by intramuscular injection, and further address how functional urine exosomes are used to diagnose and predict the progression of DKD.
Exosome
Exosome Biogenesis
Exosome is a group of nano-sized extracellular vesicles with a size range of approximately 30–150 nm secreted by cells. It was initially considered as an unconventional way to get rid of the cellular waste that resulted from the maturation of reticulocytes into erythrocytes in 1983 [7] and was subsequently named “exosomes” by Johnstone in 1987 [8]. More than a decade later, Raposo et al. [9] reported that exosomes isolated from Epstein-Barr virus-transformed B lymphocytes could induce T-cell responses and be essential for the adaptive immune response. Research interest in exosomes has grown substantially since then as exosomes were shown to participate in cellular processes, especially in the immune system. Over the years, exosomes have emerged as an essential player in cellular homeostasis and intercellular communication, involved in numerous cellular physiological and pathological processes [10, 11].
Exosome biogenesis is a protein quality control mechanism and can be generated from both the plasma membrane and the endosome membrane [12]. The generation of exosomes begins from endocytosis to form early endosomes by inward budding of the plasma membrane triggered by external stimuli or microbial attacks. Through the endosomal protein sorting and transport device-dependent machinery or endosomal protein sorting and transport device-independent machinery, the early endosomes sprout inward and mature to multivesicular bodies (also called late endosomes) containing interluminal vesicles. Regulated by Rab GTPases, multivesicular bodies are then secreted to the extracellular space through direct budding with the plasma membrane or receptor-ligand binding, or phagocytosis, and the released vesicles are called exosomes [13–16]. After being released from cells, exosomes are then shed into various body fluids and widely distributed in almost all kinds of body fluids such as blood, cerebrospinal fluid, bile, breast milk, synovial fluid, saliva, bile, ascites, amniotic fluid, and pleural effusion, suggesting an irreplaceable role of exosomes in physiological and pathological conditions [17].
Exosomal Cargo and Function
During exosome biogenesis and release, selective cargo loading occurs, and particular cellular constituents are shuttled into exosomes containing various proteins, microRNAs (miRNAs), mRNAs, DNA, lipids, metabolites, cell surface proteins, and many other cellular components [18]. In essence, an exosome contains a mixture of cargos rather than one specific molecule. Thus, the function of exosomes is highly heterogeneous as their various cargos faithfully reflect the cellular states of their origin cells, and the type, quality, and quantity of cargo depend on cell types and the cellular milieu [19]. Due to exosomes’ lipid bilayer membrane structure, cargo within exosomes is protected from degradation and can be successfully transferred to specific recipient cells, mediating cell-cell communication between origin and recipient target cells [11, 20]. Accordingly, exosomes packaged with functional cargo play a vital role in diverse cellular processes and diseases, including DKD [21–24].
The Effect of Exosomes Secreted by Different Cells in DKD
Exosomes transfer autocrine or paracrine signals by cell-cell crosstalk between different kidney resident cells. These involved cells include glomerular endothelial cells (GECs), podocytes, glomerular mesangial cells (GMCs), macrophages, MSCs, and tubular epithelial cells (TECs) and fibroblasts (shown in Fig. 1). The complex functions of exosomes in DKD are summarized as follows and shown in Table 1.
Fig. 1.
Exosome-mediated intercellular communications between renal resident cells in DKD. a Blue arrow: exosomes derived from glomerular endothelial cells (GECs) transferred paracrine signaling to podocytes by transforming growth factor beta 1 (TGF-β1) and glomerular mesangial cells (GMCs) by circRNF169, circSTRN3, and TGF-β1. b Red arrow: exosomes derived from GMCs transferred paracrine signaling to podocytes by TGF-β1 and autocrine signaling to themselves by circ-DLGAP4, angiotensinogen, renin, or circ_0125310. c Green arrow: exosomes derived from mesenchymal stromal cells (MSCs) transferred paracrine signaling to tubular epithelial cells (TECs) by miR-125b and podocytes by miR-215-5p, vascular endothelial growth factor (VEGF), TGF-β1, bone morphogenetic protein 7 (BMP-7), angiogenin, miR-16-5p, and autocrine signaling by miR-let-7a, miR-125a. d Orange arrow: exosomes derived from TECs transferred paracrine signaling to fibroblasts and autocrine signaling by fibulin 1 (FBLN1), miR-483-5p. e Purple arrow: exosomes derived from macrophages transferred paracrine signaling to podocytes by miR-25-3p, GMCs by TGF-β1, and autocrine signaling by inducible nitric oxide synthase (iNOS), interleukin-1β (IL-1β). f Podocytes-derived exosomes transferred autocrine signaling by E74-like ETS transcription factor 3 (Elf3).
Table 1.
Exosomes involved intercellular communications in DKD
| Origin | Recipient | Involved molecules | Functions | Relevant mechanism | PMID |
|---|---|---|---|---|---|
| GECs | Podocytes | TGF-β1 | Fibrosis and EMT | Stimulates the EMT and dysfunction of podocytes by releasing exosomes containing TGF-β1 | 28839221 |
| GMCs | circRNF169, circSTRN3 | Inhibited proliferation and promoted EMT | Promoted α-SMA expression | 31497190 | |
| GMCs | TGF-β1 | Promote renal fibrosis | TGF-β1/Smad3 pathway | 27010029, 28659030 | |
| GMCs | GMCs | circ-DLGAP4 | Proliferation and fibrosis | Sponging miR-143 and modulating ERBB3/NF-κB/MMP-2 | 33230102 |
| HMCs | Angiotensinogen, renin | Fibrosis, proliferation | RAS activity | 31000742 | |
| Podocytes | TGF-β1 | Adhesion capacity, apoptosis | TGF-β1/PI3K-Akt signaling pathway | 29378192 | |
| GMCs | circ_0125310 | Promoted cell proliferation and fibrosis | Sponging miR‐422a and targeting the IGF1R/p38 axis | 34854210 | |
| PTECs | Fibroblast | – | Fibrosis, proliferation | – | 32715764 |
| PTECs | FBLN1 | EMT and ECM accumulation | Modulated by miR-1269b | 34977033 | |
| PTECs | miR-483-5p | Promoted fibrosis, ECM deposition | TIMP2 and MAPK1 | 33692334 | |
| Podocytes | – | Elf3 | – | TGF-β1/Smad3 pathway | 31150422 |
| Macrophages | GMCs | TGF-β1 mRNA | Proliferation, fibrosis, inflammation | TGF-β1/Smad3 pathway | 31162940 |
| Macrophages | iNOS and IL-1β | Proliferation, inflammation | NF-κB/p65 signaling pathway | 32388490 | |
| Podocytes | miR-25-3p | Autophagy | Inhibit DUSP | 33107695 | |
| BM-MSCs | Rat TECs | Inflammation, fibrosis, EMT, apoptosis, degeneration | Regulates TGF-β, c-Src/p38-MAPK, ZO-1, TNF-α, ICAM-1, etc. | 27721418 | |
| Human TECs | miR-125b | Autophagy, apoptosis | TRAF6, Akt pathway | 34024846 | |
| Rats | - | Ameliorated fibrosis, autophagy | mTOR pathway | 30467302 | |
| Rats | miR-let-7a | Apoptosis | Downregulation of USP22 | 33347877 | |
| Rats | - | Lower the blood glucose, improve renal function | Inhibits JAK2/STAT3 | 34306382 | |
| adMSCs | Podocytes | miR-215-5p | Metastasis, EMT | Inhibit ZEB2 | 32149094 |
| GMCs | miR-125a | Suppressed fibrosis | Directly bound to HDAC1 and downregulated ET-1 | 33790607 | |
| hUSCs | Podocytes | VEGF, TGF-β1, angiogenin, BMP-7 | Apoptosis, proliferation | Reduces podocytic apoptosis and promotes vascular regeneration and cell survival | 26852014 |
| Podocytes | miR-16-5p | Apoptosis | Suppresses VEGFA expression and podocytic apoptosis | 31568645 | |
| Rats | – | Inflammation, fibrosis | Inhibits inflammation and fibrosis | 32746936 | |
| Muscle | Kidney | miR-23a/27a | Muscle atrophy and renal fibrosis | Akt, FoxO1, PTEN | 29582582 |
| Serum of DKD patients | - | miR-4449 | Regulate the expression of pro-inflammatory cytokines, ROS levels, and pyroptosis | Pyroptosis and oxidative stress | 34732690 |
DKD, diabetic kidney disease; GECs, glomerular endothelial cells; TGF-β1, transforming growth factor beta 1; EMT, epithelial-mesenchymal transition; GMCs, glomerular mesangial cells; RNF169, ring finger protein 169; STRN3, striatin 3; α-SMA, alpha smooth muscle actin; Smad3, SMAD family member 3; DLGAP4, DLG-associated protein 4; ERBB3, Erb-B2 receptor tyrosine kinase 3; NF-κB, nuclear factor kappa-B; MMP-2, matrix metallopeptidase 2; HMCs, human mesangial cells; RAS, reticular activating system; PI3K, phosphatidylinositol-3-kinase; IGF1R, insulin-like growth factor 1 receptor; PTECs, proximal tubular epithelial cells; FBLN1, fibulin 1; ECM, extracellular matrix; TIMP2, TIMP metallopeptidase inhibitor 2; MAPK1, mitogen-activated protein kinase 1; Elf3, E74-like ETS transcription factor 3; iNOS, inducible nitric oxide synthase; IL-1β, interleukin-1β; DUSP, dual-specificity phosphatase; BM-MSCs, bone marrow mesenchymal stem cells; ZO-1, zonula occludens-1; TNF-α, tumor necrosis factor α; ICAM-1, intercellular cell adhesion molecule-1; TRAF6, TNF receptor-associated factor 6; USP22, ubiquitin specific peptidase 22; JAK2/STAT3, Janus kinase 2/signal transducer and activator of transcription 3; adMCS, adipose-derived mesenchymal stem cells; ZEB2, zinc finger E-box binding homeobox 2; HDAC1, histone deacetylase 1; ET-1, endothelin-1; hUSCs, human urine-derived stem cells; BMP-7, bone morphogenetic protein 7; VEGFA, vascular endothelial growth factor A; FoxO1, forkhead box O1; PTEN, phosphatase and tensin homologue; ROS, reactive oxygen species.
GEC-Derived Exosomes
GECs are specialized vascular endothelial cells that form the walls of glomerular tuft capillaries. In DKD, GEC dysfunction, including increased permeability, is a key component of diabetes induced organ damage responding with mitochondrial dysfunction and increased reactive oxygen species production [25]. The permeability may allow GEC-derived exosomes to penetrate the glomerular basement membrane to reach podocytes and induce the epithelial-mesenchymal transition (EMT) and dysfunction of podocytes by releasing exosomes containing TGF-β1 [26]. Wu et al. [27] also found that high glucose (HG)-treated GECs secrete more exosomes highly enriched in TGF-β1 mRNA than normal-glucose-treated GECs. They further demonstrated that exosomes derived from HG-treated GECs could promote α-smooth muscle actin (α-SMA) expression and proliferation and extracellular matrix (ECM) protein overproduction in GMCs through the TGF-β1/Smad3 signaling pathway. In contrast, Ling et al. showed that the exosomes derived from HG-treated GECs inhibited GMC proliferation and promoted EMT due to downregulated exosomal circRNF169 and circSTRN3 [28]. The reason underlying the discrepancy between these two studies remains unknown. It is noteworthy that exosomes contain different cargoes under different secretion environments. In addition, the isolation methods affect the quality and quantity of exosomes. Regardless, exosomes are regarded as one of the mechanisms of Tongxinluo in DKD treatment since Tongxinluo was shown to inhibit renal fibrosis by preventing the transfer of TGF-β1 from GECs to GMCs via exosomes in DKD [29].
GMC-Derived Exosomes
GMC is a kind of vital indigenous source of kidney exosomes that might affect the function of the glomerulus in DKD conditions. Exosomes mediate communication between GMCs and podocytes and also GMCs themselves. HG-treated GMCs derived exosomes induced podocyte injury through the TGF-β1-PI3K/AKT pathway. Berberine may be a good candidate to protect against the damage of podocytes by reducing exosomal TGF-β1 and apoptosis and increasing adhesion [30]. In the autocrine pattern, three different studies consistently revealed that exosomes derived from DKD patients, DKD rat models, or HG-treated GMCs could promote proliferation and fibrosis of GMCs by different mechanisms such as an increased level of circ_DLGAP4 sponged miR-143 and regulated the ERBB3/NF-κB/MMP-2 axis [31], higher amounts of RAS components such as angiotensinogen and renin in exosomes [32], and exo-circ_0125310 sponged miR-422a and targeted the IGF1R/p38 axis [33]. These studies indicate that GMC-derived exosomes may promote DKD by inducing podocyte injury, proliferation, and fibrosis of GMCs.
TEC-Derived Exosomes
TECs are the main cells in the kidney and are vulnerable to injury. Accumulated evidence has suggested that tubular injury may contribute to the pathological onset of DKD. We demonstrated a profibrotic effect of HG-treated TEC-derived exosomes on fibroblasts, suggesting a pathogenic role of exosomes in DKD by mediating tubulointerstitial communication and promoting renal fibrosis. Our previous study proved that exosomes derived from TECs could mediate tubulointerstitial communication between tubular cells and fibroblasts, which regulate the proliferation and activation of fibroblasts, contributing to the paracrine signaling mechanism responsible for renal fibrosis in DKD [34]. Liu et al. [35] confirmed the effect of TEC-derived exosomes in promoting renal interstitial fibrosis. They suggested that HNRNPA1-mediated exosomal sorting transported cellular miR-483-5p out of TECs into the urine, thus lessening the restraint of cellular miR-483-5p on MAPK1 and TIMP2 mRNAs and ultimately boosting ECM deposition and the progression of renal interstitial fibrosis in DKD. Moreover, exosomes derived from HG-treated TECs induced EMT in TECs by delivering Fibulin-1, which was modulated by miR-1269b [36]. Together, these studies indicate that TEC-derived exosomes have profibrotic functions in DKD, which could be a novel mechanism in renal fibrosis of DKD.
Podocyte-Derived Exosomes
Podocytes are highly specialized epithelial cells wrapping the glomeruli capillaries to prevent the leakage of proteins into urine [37]. Since these cells normally cannot proliferate, podocyte injury and malfunction lead to proteinuria, accumulation of ECM components, and eventually irreversible glomerulosclerosis. Sakurai et al. [38] reported that AGE induced the secretion of Elf3-containing exosomes from podocytes via the TGF-β-Smad3 signaling pathway. Further study in DKD patients demonstrated that urinary exosomal Elf3 might be an early noninvasive marker for podocyte injury in DKD since its appearance correlated with the existence of irreversible podocyte injury. However, it remains unknown whether podocyte-derived exosomes induce cell-cell communication with other renal resident cells.
Macrophage-Derived Exosomes
Macrophages have essential roles in immune surveillance and the maintenance of kidney homeostasis. During kidney injury, macrophages adapt rapidly to the dynamics of the renal microenvironment [39]. Current evidence suggests a vital communicator role for exosomes derived from macrophages which mediate signaling from macrophages to GMCs or podocytes in DKD. For GMCs, TGF-β1 containing exosomes from macrophages could induce proliferation, fibrosis, and inflammation of GMCs by the TGF-β1/Smad3 pathway [40]. For podocytes, exosomal miR-25-3p from macrophages improved HG-induced podocyte injury by inhibiting DUSP1 expression with consequent autophagy activation [41]. Furthermore, HG-treated-macrophage-derived exosomes contained higher concentrations of IL-1β and iNOS, which activate macrophages by inducing the production of intracellular iNOS and α-SMA and accelerate kidney injury via the NF-κB signaling pathway [42].
MSC-Derived Exosomes
MSCs are a heterogeneous subset of stromal stem cells with multipotent and self-renewing properties and can differentiate into cells of the mesodermal lineage [43]. Recently, MSC-derived exosomes have attracted much attention for their ability to facilitate the renal repair and their potential for DKD therapy [44, 45]. On TECs, MSC-derived exosomes exerted multiple effects, such as anti-inflammation, anti-fibrosis, anti-apoptotic effect, activation of autophagy, and protection of tight junction structure in DKD [46, 47]. On podocytes, MSC-derived exosomes suppressed apoptosis, fibrosis, and proliferation [48–50]. In addition, MSC-derived exosomes play a renal-protective role in DKD by ameliorating fibrosis and inflammation [51–53], inducing autophagy [53], inhibiting apoptosis [54], and lowering blood [55]. The mechanisms by which MSC-derived exosomes exert their therapeutic effects are multifaceted, but in general, mainly by regulating the Akt, mTOR, P38-MAPK, JAK2/STAT3, and TGF-β signaling. Due to the multipotent and self-renewing characteristics of MSCs, their derived exosomes hold an excellent promise for DKD.
Other Resources of Exosomes in Kidneys
Exosomes can be secreted from various cell types into circulation and transported to target cells throughout the body. Gao et al. [56] isolated serum exosomes from DKD patients and found that such exosomes could promote pyroptosis and oxidative stress through the miR-4449/HIC1 pathway. Interestingly, Zhang et al. [57] injected miR-23a/27a in the muscle of DKD mice that could prevent diabetes-induced muscle cachexia and attenuate renal fibrosis lesions via exosome-mediated muscle-kidney crosstalk.
Implication of Urinary Exosomes as Biomarkers of DKD
Current biomarkers for detecting DKD, such as albuminuria and eGFR, are of insufficient sensitivity to detect the early-stage DKD, highlighting the necessity for discovering novel biomarkers. Exosomes can be easily obtained from different body fluids by noninvasive methods and have emerged as a promising tool for analyzing DKD biomarkers, especially for urinary exosomes [58]. In general, plasma exosomes may not pass through the glomerular filtration barrier, and as a result, urinary exosomes mainly come from urogenital system cells. Moreover, the exosomes are protected by their bilayer membrane structure. Thus, urinary exosomes mirror the physiopathological state of the kidney other than serum or circulation [59]. Herein, we summarized the implication of urinary exosomes as biomarkers in DKD (shown in Table 2).
Table 2.
Urinary exosomal biomarkers for DKD
| Exosomal | Molecules | Species | Change | Actions | PMID |
|---|---|---|---|---|---|
| Proteins | WT1 | Human | ↑ | Predict proteinuria and decreased renal function, especially for GFR <60 mL/min/1.73 m2 | 23544132 |
| Plasmin, prostasin, and urokinase | Human | ↑ | Correlated directly with urine albumin | 25609736 | |
| Elf3 | Human | ↑ | Elf3 was detectable only in DKD patients. Irreversible injuries in podocytes were predicted | 31150422 | |
| glcUMOD | Human | ↑ | Forms AGEs | 28592554 | |
| AMBP, MLL3 | Human | ↑ | - | 24211404 | |
| FBLN1 | Human | ↑ | Correlated with the severity of tubular injury | 34977033 | |
| AQP5, AQP2 | Human | ↑ | Positive correlation between uAQP5 and the histological class of DN | 28246612 | |
| CD63 | Human | ↑ | Therapeutic indicator | 28116765 | |
| Regucalcin, SMP30 | Rat | ↓ | - | 26072307 | |
| VDAC1 | Human | ↓ | - | 24211404 | |
| miRNAs | let-7c-5p | Human | ↑ | Correlated with renal function, level of eGFR, and disease progression | 29739042 |
| miR-19b-3p | Mice | ↑ | Correlated with the severity of albuminuria, tubulointerstitial inflammation, and SOCS-1 level | 31097789 | |
| miR-188-5p | Human | ↑ | Regulates HG-induced EMT in HK-2 cells via the PTEN/PI3K/Akt pathway | 32340338 | |
| miR-150-3p, miR-760, miR-3677-3p, miR-548ah-3p, miR-548p, miR-320e, miR-23c | Human | ↑ | - | 32340338 | |
| miR-21-5p, let-7e-5p, miR-23b-3p | Human | ↑ | Associated with renal function, renal sclerosis, fibrosis, detection of DKD, and poor renal function | 31358876 | |
| miR-4534 | Human | ↑ | Involved in the FoxO/BNIP3 signaling pathway, correlated with proteinuria levels in DKD. | 32982978 | |
| miR-516b-5p | Human | ↑ | Correlated with proteinuria levels | 32982978 | |
| miR-1269b | Human | ↑ | Correlated with tubular injury | 34977033 | |
| miR-451 | Human, Rat | ↑ | Correlated with renal failure | 32322216 | |
| miR-451-5p, miR-16 | Rat | ↑ | Predict albuminuria at an earlier time point | 27101382 | |
| miR-320c, miR-6068 | Human | ↑ | Graded correlation with UACR. | 26930277 | |
| miR-1234-5p, miR-6133, miR-4270, miR-4739, miR-371b-5p, miR-638, miR-572, miR-1227-5p, miR-6126, miR-1915-5p, miR-4778-5p, miR-2861 | Human | ↑ | - | 26930277 | |
| miR-133b, miR-342, miR-30a | Human | ↑ | Early marker before the onset of albuminuria; associated with renal function and proteinuria levels | 27470555 | |
| miR-362-3p, miR-877-3p, miR-150-5p, miR-638, miR-133b | Human | ↑ | Early biomarker for DKD. | 29038788 | |
| miR-145, miR-130a | Human | ↑ | Early biomarker for DKD. | 24223694 | |
| miR-15b, miR-34a, and miR-636 | Human | ↑ | Positive correlated with renal function and proteinuria levels | 27475263 | |
| miR-15b-5p | Human, mice | ↑ | Predicted kidney injury in DKD | 31991106 | |
| miR-15a-5p | Human | ↓ | Mediated fibrosis | 29038788 | |
| miR29c-5p, miR-15b-5p | Human | ↓ | Predicted the development of DKD | 29739042 | |
| miR-30b-5p, miR-125b-5p | Human | ↓ | Detected DKD and poor renal function | 31358876 | |
| miR-133a-3p, miR-153-3p | Human | ↓ | Early biomarker for DKD | 32340338 | |
| miR-155, miR-424 | Human | ↓ | Early biomarker for DKD | 24223694 | |
| mRNA | WT1 | Human | ↑ | Reflected damage of diabetic glomeruli, predicted the decline in eGFR | 30282862 |
WT1, Wilms tumor 1; GFR, glomerular filtration rate; Elf3, E74-like ETS transcription factor 3; DKD, diabetic kidney disease; glcUMOD, glycated uromodulin; AGEs, advanced glycation end products; AMBP, α-microglobulin/bikunin precursor; MLL3, histone-lysine N-methyltransferase; FBLN1, Fibulin-1; AQP, aquaporins; uAQP5, urine aquaporins 5; SMP30, senescence marker protein-30; VDAC1, voltage-dependent anion-selective channel protein 1; SOCS1, suppressor of cytokine signaling 1; EMT, epithelial-mesenchymal transition; PTEN, phosphatase and tensin homologue; PI3K, phosphatidylinositol-3-kinase; FoxO, forkhead box O; BNIP3, BCL2 interacting protein 3; UACR, urine albumin-to-creatinine ratio.
Urinary Exosomal Proteins
Several urinary exosomal proteins have been implicated in DKD detection. The first proteomic study of human urinary exosomes for DKD patients suggested increased AMBP and MLL3 and decreased VDAC1 levels as biomarkers for diagnosing and pathological follow-up in DKD [60]. However, urinary exosomal WT1 was the first protein detected in DKD patients. It was associated with a significant increase in urine protein-to-creatinine ratio, albumin-to-creatinine ratio, serum creatinine, and a decline in eGFR [61]. WT1, shed by renal epithelial cells, has generally been regarded as a biomarker for podocyte injury. Another study showed that increased plasmin, prostasin, and urokinase directly correlated with proteinuria levels [62]. Elf3, a predictor of irreversible podocyte injuries, was also suggested as a biomarker of DKD with high specificity which is only detectable in urinary exosomes of DKD patients rather than DM patients or healthy controls [38]. Increased FBLN1 was associated with the severity of tubular injury [36], while AQP5 and AQP2 were associated with the histological class or degree of DKD [63]. Urinary glcUMOD concentration can predict DKD status, particularly in patients with CKD1-3a stages aged <65 years and with a urine glcUMOD concentration ≥9,000 arbitrary units [64]. In another study, urinary exosomal CD63 was higher in normoalbuminuria patients than microalbuminuria patients, and it might be a therapeutic indicator in DKD, especially after α-lipoic acid treatment [65]. By contrast, regucalcin and SMP30 were strongly downregulated in both DKD kidney tissue and urinary exosomes, suggesting their potential for early diagnosis and progression monitoring of DKD [66].
Urinary Exosomal miRNAs
By modulating gene expression, miRNAs influence various cellular and physiological processes, including proliferation, differentiation, cell death, and organ development. There is an increasing body of evidence that miRNAs are crucial regulators and biomarkers of DKD. Li et al. [67] showed an upregulation of let-7c-5p in urinary exosomes of type II diabetes, which was associated with the decline of renal function and DKD progression. In contrast, miR29c-5p and miR-15b-5p were downregulated, which could also predict the progression of DKD. Lv et al. [68] revealed that a high level of miR-19b-3p was associated with the severity of albuminuria, tubulointerstitial inflammation, and SOCS-1 levels. Both miR-4534 and miR-516b-5p were upregulated and correlated with proteinuria levels in DKD, an observation probably related to the involvement of miR-4534 in the FoxO/BNIP3 signaling pathway [69]. Increased miR-21-5p, let-7e-5p, miR-23b-3 and decreased miR-30b-5p, miR-125b-5p were associated with renal function, renal sclerosis, fibrosis, which could detect DKD and reduced renal function [70]. Furthermore, miR-1269b was correlated with tubular injury, and miR-451 was correlated with renal failure, while an increased level of miR-451-5p or miR-16 might predict albuminuria at an earlier time point [36, 71, 72].
Lee et al. conducted the next-generation sequencing to analyze miRNAs in urinary exosomes of DKD and found that miR-188-5p, miR-150-3p, miR-760, miR-3677-3p, miR-548ah-3p, miR-548p, miR-320e, and miR-23c were significantly upregulated in DKD patients, while miR-133a-3p and miR-153-3p were downregulated. Functionally, they demonstrated that miR-188-5p promoted EMT in HG-treated HK-2 cells through the PTEN/PI3K/Akt pathway [73]. Delic and colleagues [74] analyzed urinary exosomes of type II DKD patients by Agilent’s miRNA microarrays, showing the upregulation of 14 miRNAs (miR-320c, miR-6068, miR-1234-5p, miR-6133, miR-4270, miR-4739, miR-371b-5p, miR-638, miR-572, miR-1227-5p, miR-6126, miR-1915-5p, miR-4778-5p, and miR-2861) and downregulation of miR-30d-5p and miR-30e-5p. Further investigation showed the correlation of miR-320c and miR-6068 with UACR levels in these patients, but the function of these miRNAs was not explored.
Other studies also suggested specific urinary exosomal miRNAs as early biomarkers for DKD, including upregulated miR-133b, miR-342, miR-30a, miR-362-3p, miR-877-3p, miR-150-5p, miR-638, miR-133b, miR-145, miR-130a and downregulated miR-155, miR-424, miR-133a-3p, miR-153-3p [73, 75–77]. Moreover, some miRNAs were revealed to be correlated with renal function and proteinuria levels, such as miR-15b, miR-34a, miR-636, miR-15b-5p, miR-30b-5p, and miR-125b-5p [70, 78, 79].
Besides the protein level of WT1, its mRNA could also predict renal functional decline and damage of diabetic glomeruli in DKD patients [80]. Recently, one study compared the differences of mRNAs, miRNAs, lncRNAs, and circRNAs profiles in exosomes derived from HG-treated human TECs for 48 h and revealed differential expression with 169 lncRNAs, 885 mRNAs, 3 circRNAs, and 152 miRNAs; however, no validation experiments were proceeded [81]. Therefore, urinary exosomal proteins and miRNAs may be novel noninvasive biomarkers for DKD. Further clinical trials should investigate their biomarker efficacy and test the possible combination use of multiple biomarkers for early detection of DKD.
Biogenesis of Exosomes in DKD
Compared with the research on the cargo and function of exosomes, very little is known about the biogenesis or particle number of exosomes in DKD. Nonetheless, we recently reported a significant decrease in exosome secretion in DKD models, including both Akita diabetic mice and HG-treated renal tubular cells [34]. Mechanistically, FOXO1 was shown to be phosphorylated and inactivated in DKD, leading to the downregulation of RAB27B, a key regulator of exosome secretion [82]. Consistently, Hassanpour et al. [83] showed that chronic exposure of human endothelial progenitor cells to diabetic sera suppressed exosome secretion or biogenesis. However, there are studies showing a higher particle number of exosomes in DKD models. For example, the numbers of exosomes released from GECs treated with HG for 24 h were 1.5–3 times higher than the normal-glucose-treated group [26–28]. The cause of the discrepancy between these studies is unclear, but it may be related to the experimental conditions, including the cell and tissue types and the duration of diabetic exposure. In this regard, longer than 1-week exposure of cells to the diabetic condition [83] yielded results that are consistent with in vivo model of weeks of diabetes [34, 82]. Of note, DKD is a chronic progressive disease. Thus, an in vitro model with a longer diabetic exposure time may better reflect the natural course of DKD in human patients.
Conclusion
Exosomes mediate communication by autocrine or paracrine mechanisms between different kidney resident cells. Recent studies have demonstrated the changes of exosomes in DKD, including both exosomal cargos and numbers. Functionally, exosomes have been implicated in the pathogenesis and development of DKD. Despite these progresses, important questions mainly open in this field. For example, since an exosome contains multiple cargo molecules, it is interesting and meaningful to clarify the role of specific exosomal cargos in DKD, which may lead to the identification of novel therapeutic targets for DKD. In addition, how do cells control the content and number of cargos inside exosomes and what influences the binding of exosomes with specific kidney cell types in DKD? Addressing this question is critical to understanding the function of exosomes in DKD. Also, we have just begun to understand how kidney cells control the number of exosomes or exosome secretion in DKD. As eluded above, there may be a biphasic change in exosome secretion after diabetic exposure. In the early time, the exosome section may be increased, but chronic exposure to diabetic conditions leads to a significant decrease in exosome secretion. Certainly, exosome secretion can be regulated by RAB27B and FOXO1 [82], but it also depends on cell types and cellular conditions.
Strikingly, urinary exosomes showed a particular advantage as biomarkers for DKD. miRNAs and proteins in urinary exosomes are the most studied molecules that are indicative of renal function and disease progression in DKD. However, it is noteworthy that some of them might not be specific for DKD but general CKD. Furthermore, most of these studies did not compare the efficacy of biomarker candidates with albuminuria except for WT1 protein [61]. Further studies need to validate the clinical applicability of these potential biomarkers. Moreover, further research is encouraged to test the diagnostic potential or prognostic role of other exosomal components in DKD, besides miRNAs and proteins.
Conflict of Interest Statement
The authors have no conflicts of interest to declare.
Funding Sources
This work was partly supported by the National Natural Science Foundation of China (82200763), Hunan Health Commission (202202063532), Natural Science Foundation of Hunan province (2022JJ40226), Natural Science Foundation of Changsha City (kq2202435), and PhD. Foundation of Hunan Provincial People’s Hospital (BSJJ202112).
Author Contributions
Conceptualization: J.W. and Z.D.; original draft preparation: J.W.; review and editing: J.W. M.Z., Y.Y., Y.L., P.F., and Z.D.
Funding Statement
This work was partly supported by the National Natural Science Foundation of China (82200763), Hunan Health Commission (202202063532), Natural Science Foundation of Hunan province (2022JJ40226), Natural Science Foundation of Changsha City (kq2202435), and PhD. Foundation of Hunan Provincial People’s Hospital (BSJJ202112).
References
- 1. CfDCa Prevention . Chronic kidney disease in the United States, 2021. Atlanta, GA: US Department of Health and Human Services; 2021 March 4. p. 2021. [Google Scholar]
- 2. GBD Chronic Kidney Disease Collaboration . Global, regional, and national burden of chronic kidney disease, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2020;395(10225):709–33. 10.1016/S0140-6736(20)30045-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Yang C, Gao B, Zhao X, Su Z, Sun X, Wang HY, et al. Executive summary for China kidney disease network (CK-NET) 2016 annual data report. Kidney Int. 2020;98(6):1419–23. 10.1016/j.kint.2020.09.003. [DOI] [PubMed] [Google Scholar]
- 4. Reutens AT. Epidemiology of diabetic kidney disease. Med Clin North Am. 2013;97(1):1–18. 10.1016/j.mcna.2012.10.001. [DOI] [PubMed] [Google Scholar]
- 5. Lu Y, Liu D, Feng Q, Liu Z. Diabetic nephropathy: perspective on extracellular vesicles. Front Immunol. 2020;11:943. 10.3389/fimmu.2020.00943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Chen J, Zhang Q, Liu D, Liu Z. Exosomes: advances, development and potential therapeutic strategies in diabetic nephropathy. Metabolism. 2021;122:154834. 10.1016/j.metabol.2021.154834. [DOI] [PubMed] [Google Scholar]
- 7. Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell. 1983;33(3):967–78. 10.1016/0092-8674(83)90040-5. [DOI] [PubMed] [Google Scholar]
- 8. Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262(19):9412–20. 10.1016/s0021-9258(18)48095-7. [DOI] [PubMed] [Google Scholar]
- 9. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183(3):1161–72. 10.1084/jem.183.3.1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977. 10.1126/science.aau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164(6):1226–32. 10.1016/j.cell.2016.01.043. [DOI] [PubMed] [Google Scholar]
- 12. Pegtel DM, Gould SJ. Annu Rev Biochem. 2019;88:487–514. 10.1146/annurev-biochem-013118-111902. [DOI] [PubMed] [Google Scholar]
- 13. Kowal J, Tkach M, Thery C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–25. 10.1016/j.ceb.2014.05.004. [DOI] [PubMed] [Google Scholar]
- 14. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79. 10.1038/nri855. [DOI] [PubMed] [Google Scholar]
- 15. Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, et al. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 2013;126(Pt 24):5553–65. 10.1242/jcs.128868. [DOI] [PubMed] [Google Scholar]
- 16. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 2010;12(1):19–30; sup pp 1-13. 10.1038/ncb2000. [DOI] [PubMed] [Google Scholar]
- 17. Lin J, Li J, Huang B, Liu J, Chen X, Chen XM, et al. Exosomes: novel biomarkers for clinical diagnosis. ScientificWorldJournal. 2015;2015:657086. 10.1155/2015/657086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Willms E, Johansson HJ, Mager I, Lee Y, Blomberg KEM, Sadik M, et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci Rep. 2016;6:22519. 10.1038/srep22519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193–208. 10.1007/s00018-017-2595-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113(8):E968–77. 10.1073/pnas.1521230113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Simons M, Raposo G. Exosomes – vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21(4):575–81. 10.1016/j.ceb.2009.03.007. [DOI] [PubMed] [Google Scholar]
- 22. Mathieu M, Martin-Jaular L, Lavieu G, Thery C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17. 10.1038/s41556-018-0250-9. [DOI] [PubMed] [Google Scholar]
- 23. Lv LL, Feng Y, Tang TT, Liu BC. New insight into the role of extracellular vesicles in kidney disease. J Cell Mol Med. 2019;23(2):731–9. 10.1111/jcmm.14101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Zhang W, Zhou X, Zhang H, Yao Q, Liu Y, Dong Z. Extracellular vesicles in diagnosis and therapy of kidney diseases. Am J Physiol Renal Physiol. 2016;311(5):F844–51. 10.1152/ajprenal.00429.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Ramnath RD, Satchell SC. Glomerular endothelial cells: assessment of barrier properties in vitro. Methods Mol Biol. 2020;2067:145–51. 10.1007/978-1-4939-9841-8_11. [DOI] [PubMed] [Google Scholar]
- 26. Wu X, Gao Y, Xu L, Dang W, Yan H, Zou D, et al. Exosomes from high glucose-treated glomerular endothelial cells trigger the epithelial-mesenchymal transition and dysfunction of podocytes. Sci Rep. 2017;7(1):9371. 10.1038/s41598-017-09907-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Wu XM, Gao YB, Cui FQ, Zhang N. Exosomes from high glucose-treated glomerular endothelial cells activate mesangial cells to promote renal fibrosis. Biol Open. 2016;5(4):484–91. 10.1242/bio.015990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Ling L, Tan Z, Zhang C, Gui S, Cui Y, Hu Y, et al. CircRNAs in exosomes from high glucose-treated glomerular endothelial cells activate mesangial cells. Am J Transl Res. 2019;11(8):4667–82. [PMC free article] [PubMed] [Google Scholar]
- 29. Wu XM, Gao YB, Xu LP, Zou DW, Zhu ZY, Wang XL, et al. Tongxinluo inhibits renal fibrosis in diabetic nephropathy: involvement of the suppression of intercellular transfer of TGF-[formula: see text]1-containing exosomes from GECs to GMCs. Am J Chin Med. 2017;45(5):1075–92. 10.1142/S0192415X17500586. [DOI] [PubMed] [Google Scholar]
- 30. Wang YY, Tang LQ, Wei W. Berberine attenuates podocytes injury caused by exosomes derived from high glucose-induced mesangial cells through TGFβ1-PI3K/AKT pathway. Eur J Pharmacol. 2018;824:185–92. 10.1016/j.ejphar.2018.01.034. [DOI] [PubMed] [Google Scholar]
- 31. Bai S, Xiong X, Tang B, Ji T, Li X, Qu X, et al. Exosomal circ_DLGAP4 promotes diabetic kidney disease progression by sponging miR-143 and targeting ERBB3/NF-κB/MMP-2 axis. Cell Death Dis. 2020;11(11):1008. 10.1038/s41419-020-03169-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. da Silva Novaes A, Borges FT, Maquigussa E, Varela VA, Dias MVS, Boim MA. Influence of high glucose on mesangial cell-derived exosome composition, secretion and cell communication. Sci Rep. 2019;9(1):6270. 10.1038/s41598-019-42746-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Zhu Y, Zha F, Tang B, Ji TT, Li XY, Feng L, et al. Exosomal hsa_circ_0125310 promotes cell proliferation and fibrosis in diabetic nephropathy via sponging miR-422a and targeting the IGF1R/p38 axis. J Cell Mol Med. 2022;26(1):151–62. 10.1111/jcmm.17065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Wen J, Ma Z, Livingston MJ, Zhang W, Yuan Y, Guo C, et al. Decreased secretion and profibrotic activity of tubular exosomes in diabetic kidney disease. Am J Physiol Renal Physiol. 2020;319(4):F664–73. 10.1152/ajprenal.00292.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Liu D, Liu F, Li Z, Pan S, Xie J, Zhao Z, et al. HNRNPA1-mediated exosomal sorting of miR-483-5p out of renal tubular epithelial cells promotes the progression of diabetic nephropathy-induced renal interstitial fibrosis. Cell Death Dis. 2021;12(3):255. 10.1038/s41419-021-03460-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Tsai Y-C, Hung W-W, Chang W-A, Wu P-H, Wu L-Y, Lee S-C, et al. Autocrine exosomal fibulin-1 as a target of MiR-1269b induces epithelial-mesenchymal transition in proximal tubule in diabetic nephropathy.Front Cell Dev Biol. 2021;9:789716. 10.3389/fcell.2021.789716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Scott RP, Quaggin SE. Review series: the cell biology of renal filtration. J Cell Biol. 2015;209(2):199–210. 10.1083/jcb.201410017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Sakurai A, Ono H, Ochi A, Matsuura M, Yoshimoto S, Kishi S, et al. Involvement of Elf3 on Smad3 activation-dependent injuries in podocytes and excretion of urinary exosome in diabetic nephropathy. PLoS One. 2019;14(5):e0216788. 10.1371/journal.pone.0216788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Tang PMK, Nikolic-Paterson DJ, Lan HY. Macrophages: versatile players in renal inflammation and fibrosis. Nat Rev Nephrol. 2019;15(3):144–58. 10.1038/s41581-019-0110-2. [DOI] [PubMed] [Google Scholar]
- 40. Zhu Q-J, Zhu M, Xu X-X, Meng X-M, Wu Y-G. Exosomes from high glucose-treated macrophages activate glomerular mesangial cells via TGF-β1/Smad3 pathway in vivo and in vitro. FASEB J. 2019;33(8):9279–90. 10.1096/fj.201802427RRR. [DOI] [PubMed] [Google Scholar]
- 41. Huang H, Liu H, Tang J, Xu W, Gan H, Fan Q, et al. M2 macrophage-derived exosomal miR-25-3p improves high glucose-induced podocytes injury through activation autophagy via inhibiting DUSP1 expression. IUBMB Life. 2020;72(12):2651–62. 10.1002/iub.2393. [DOI] [PubMed] [Google Scholar]
- 42. Zhu M, Sun X, Qi X, Xia L, Wu Y. Exosomes from high glucose-treated macrophages activate macrophages andinduce inflammatory responses via NF-κB signaling pathway in vitro and in vivo. Int Immunopharmacol. 2020;84:106551. 10.1016/j.intimp.2020.106551. [DOI] [PubMed] [Google Scholar]
- 43. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8(9):726–36. 10.1038/nri2395. [DOI] [PubMed] [Google Scholar]
- 44. Hickson LJ, Eirin A, Conley SM, Taner T, Bian X, Saad A, et al. Diabetic kidney disease alters the transcriptome and function of human adipose-derived mesenchymal stromal cells but maintains immunomodulatory and paracrine activities important for renal repair. Diabetes. 2021;70(7):1561–74. 10.2337/db19-1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Abdel Aziz MT, Wassef MAA, Ahmed HH, Rashed L, Mahfouz S, Aly MI, et al. The role of bone marrow derived-mesenchymal stem cells in attenuation of kidney function in rats with diabetic nephropathy. Diabetol Metab Syndr. 2014;6(1):34. 10.1186/1758-5996-6-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Nagaishi K, Mizue Y, Chikenji T, Otani M, Nakano M, Konari N, et al. Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes. Sci Rep. 2016;6:34842. 10.1038/srep34842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Cai X, Zou F, Xuan R, Lai XY. Exosomes from mesenchymal stem cells expressing microribonucleic acid-125b inhibit the progression of diabetic nephropathy via the tumour necrosis factor receptor-associated factor 6/Akt axis. Endocr J. 2021;68(7):817–28. 10.1507/endocrj.EJ20-0619. [DOI] [PubMed] [Google Scholar]
- 48. Jin J, Wang Y, Zhao L, Zou W, Tan M, He Q. Exosomal miRNA-215-5p derived from adipose-derived stem cells attenuates epithelial-mesenchymal transition of podocytes by inhibiting ZEB2. Biomed Res Int. 2020;2020:2685305. 10.1155/2020/2685305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Jiang ZZ, Liu YM, Niu X, Yin JY, Hu B, Guo SC, et al. Exosomes secreted by human urine-derived stem cells could prevent kidney complications from type I diabetes in rats. Stem Cell Res Ther. 2016;7:24. 10.1186/s13287-016-0287-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Duan YR, Chen BP, Chen F, Yang SX, Zhu CY, Ma YL, et al. Exosomal microRNA-16-5p from human urine-derived stem cells ameliorates diabetic nephropathy through protection of podocyte. J Cell Mol Med. 2021;25(23):10798–813. 10.1111/jcmm.14558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Xiang E, Han B, Zhang Q, Rao W, Wang Z, Chang C, et al. Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Res Ther. 2020;11(1):336. 10.1186/s13287-020-01852-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Hao Y, Miao J, Liu W, Cai K, Huang X, Peng L. Mesenchymal stem cell-derived exosomes carry MicroRNA-125a to protect against diabetic nephropathy by targeting histone deacetylase 1 and downregulating endothelin-1. Diabetes Metab Syndr Obes. 2021;14:1405–18. 10.2147/DMSO.S286191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Ebrahim N, Ahmed IA, Hussien NI, Dessouky AA, Farid AS, Elshazly AM, et al. Mesenchymal stem cell-derived exosomes ameliorated diabetic nephropathy by autophagy induction through the mTOR signaling pathway. Cells. 2018;7(12):226. 10.3390/cells7120226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Mao R, Shen J, Hu X. BMSCs-derived exosomal microRNA-let-7a plays a protective role in diabetic nephropathy via inhibition of USP22 expression. Life Sci. 2021;268:118937. 10.1016/j.lfs.2020.118937. [DOI] [PubMed] [Google Scholar]
- 55. Wang S, Bao L, Fu W, Deng L, Ran J. Protective effect of exosomes derived from bone marrow mesenchymal stem cells on rats with diabetic nephropathy and its possible mechanism. Am J Transl Res. 2021;13(6):6423–30. [PMC free article] [PubMed] [Google Scholar]
- 56. Gao C, Wang B, Chen Q, Wang M, Fei X, Zhao N. Serum exosomes from diabetic kidney disease patients promote pyroptosis and oxidative stress through the miR-4449/HIC1 pathway. Nutr Diabetes. 2021;11(1):33. 10.1038/s41387-021-00175-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Zhang A, Li M, Wang B, Klein JD, Price SR, Wang XH. miRNA-23a/27a attenuates muscle atrophy and renal fibrosis through muscle-kidney crosstalk. J Cachexia Sarcopenia Muscle. 2018;9(4):755–70. 10.1002/jcsm.12296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Lin CH, Chang YC, Chuang LM. Early detection of diabetic kidney disease: present limitations and future perspectives. World J Diabetes. 2016;7(14):290–301. 10.4239/wjd.v7.i14.290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Gudehithlu KP, Garcia-Gomez I, Vernik J, Brecklin C, Kraus M, Cimbaluk DJ, et al. In diabetic kidney disease urinary exosomes better represent kidney specific protein alterations than whole urine. Am J Nephrol. 2015;42(6):418–24. 10.1159/000443539.In [DOI] [PubMed] [Google Scholar]
- 60. Zubiri I, Posada-Ayala M, Sanz-Maroto A, Calvo E, Martin-Lorenzo M, Gonzalez-Calero L, et al. Diabetic nephropathy induces changes in the proteome of human urinary exosomes as revealed by label-free comparative analysis. J Proteomics. 2014;96:92–102. 10.1016/j.jprot.2013.10.037. [DOI] [PubMed] [Google Scholar]
- 61. Kalani A, Mohan A, Godbole MM, Bhatia E, Gupta A, Sharma RK, et al. Wilm’s tumor-1 protein levels in urinary exosomes from diabetic patients with or without proteinuria. PLoS One. 2013;8(3):e60177. 10.1371/journal.pone.0060177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Andersen H, Friis UG, Hansen PBL, Svenningsen P, Henriksen JE, Jensen BL. Diabetic nephropathy is associated with increased urine excretion of proteases plasmin, prostasin and urokinase and activation of amiloride-sensitive current in collecting duct cells. Nephrol Dial Transplant. 2015;30(5):781–9. 10.1093/ndt/gfu402. [DOI] [PubMed] [Google Scholar]
- 63. Rossi L, Nicoletti MC, Carmosino M, Mastrofrancesco L, Di Franco A, Indrio F, et al. Urinary excretion of kidney aquaporins as possible diagnostic biomarker of diabetic nephropathy. J Diabetes Res. 2017;2017:4360357. 10.1155/2017/4360357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Chang CC, Chen CY, Huang CH, Wu CL, Wu HM, Chiu PF, et al. Urinary glycated uromodulin in diabetic kidney disease. Clin Sci. 2017;131(15):1815–29. 10.1042/CS20160978. [DOI] [PubMed] [Google Scholar]
- 65. Sun H, Yao W, Tang Y, Zhuang W, Wu D, Huang S, et al. Urinary exosomes as a novel biomarker for evaluation of alpha-lipoic acid’s protective effect in early diabetic nephropathy. J Clin Lab Anal. 2017;31(6):e22129. 10.1002/jcla.22129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Zubiri I, Posada-Ayala M, Benito-Martin A, Maroto AS, Martin-Lorenzo M, Cannata-Ortiz P, et al. Kidney tissue proteomics reveals regucalcin downregulation in response to diabetic nephropathy with reflection in urinary exosomes. Transl Res. 2015;166(5):474–84.e4. 10.1016/j.trsl.2015.05.007. [DOI] [PubMed] [Google Scholar]
- 67. 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(5):709–18. 10.7754/Clin.Lab.2018.171031. [DOI] [PubMed] [Google Scholar]
- 68. Lv LL, Feng Y, Wu M, Wang B, Li ZL, Zhong X, et al. Exosomal miRNA-19b-3p of tubular epithelial cells promotes M1 macrophage activation in kidney injury. Cell Death Differ. 2020;27(1):210–26. 10.1038/s41418-019-0349-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Zhao Y, Shen A, Guo F, Song Y, Jing N, Ding X, et al. Urinary exosomal MiRNA-4534 as a novel diagnostic biomarker for diabetic kidney disease. Front Endocrinol. 2020;11:590. 10.3389/fendo.2020.00590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Zang J, Maxwell AP, Simpson DA, McKay GJ. Differential expression of urinary exosomal MicroRNAs miR-21-5p and miR-30b-5p in individuals with diabetic kidney disease. Sci Rep. 2019;9(1):10900. 10.1038/s41598-019-47504-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Mohan A, Singh RS, Kumari M, Garg D, Upadhyay A, Ecelbarger CM, et al. Urinary exosomal microRNA-451-5p is a potential early biomarker of diabetic nephropathy in rats. PLoS One. 2016;11(4):e0154055. 10.1371/journal.pone.0154055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Kumari M, Mohan A, Ecelbarger CM, Gupta A, Prasad N, Tiwari S. miR-451 loaded exosomes are released by the renal cells in response to injury and associated with reduced kidney function in human. Front Physiol. 2020;11:234. 10.3389/fphys.2020.00234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Lee WC, Li LC, Ng HY, Lin PT, Chiou TTY, Kuo WH, et al. Urinary exosomal MicroRNA signatures in nephrotic, biopsy-proven diabetic nephropathy. J Clin Med. 2020;9(4):1220. 10.3390/jcm9041220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Delić D, Eisele C, Schmid R, Baum P, Wiech F, Gerl M, et al. Urinary exosomal miRNA signature in type II diabetic nephropathy patients. PLoS One. 2016;11(3):e0150154. 10.1371/journal.pone.0150154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Eissa S, Matboli M, Bekhet MM. Clinical verification of a novel urinary microRNA panal: 133b, -342 and -30 as biomarkers for diabetic nephropathy identified by bioinformatics analysis. Biomed Pharmacother. 2016;83:92–9. 10.1016/j.biopha.2016.06.018. [DOI] [PubMed] [Google Scholar]
- 76. 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;2017:6978984. 10.1155/2017/6978984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, et al. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One. 2013;8(11):e73798. 10.1371/journal.pone.0073798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Tsai YC, Kuo MC, Hung WW, Wu LY, Wu PH, Chang WA, et al. High glucose induces mesangial cell apoptosis through miR-15b-5p and promotes diabetic nephropathy by extracellular vesicle delivery. Mol Ther. 2020;28(3):963–74. 10.1016/j.ymthe.2020.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Eissa S, Matboli M, Aboushahba R, Bekhet MM, Soliman Y. Urinary exosomal microRNA panel unravels novel biomarkers for diagnosis of type 2 diabetic kidney disease. J Diabetes Complications. 2016;30(8):1585–92. 10.1016/j.jdiacomp.2016.07.012. [DOI] [PubMed] [Google Scholar]
- 80. Abe H, Sakurai A, Ono H, Hayashi S, Yoshimoto S, Ochi A, et al. Urinary exosomal mRNA of WT1 as diagnostic and prognostic biomarker for diabetic nephropathy. J Med Invest. 2018;65(3.44):208–15. 10.2152/jmi.65.208. [DOI] [PubMed] [Google Scholar]
- 81. Zhou S, Fang J, Hu M, Pan S, Liu D, Xing G, et al. Determining the influence of high glucose on exosomal lncRNAs, mRNAs, circRNAs and miRNAs derived from human renal tubular epithelial cells. Aging. 2021;13(6):8467–80. 10.18632/aging.202656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Zeng M, Wen J, Ma Z, Xiao L, Liu Y, Kwon S, et al. FOXO1-Mediated downregulation of RAB27B leads to decreased exosome secretion in diabetic kidneys. Diabetes. 2021;70(7):1536–48. 10.2337/db20-1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Hassanpour M, Cheraghi O, Brazvan B, Hiradfar A, Aghamohammadzadeh N, Rahbarghazi R, et al. Chronic exposure of human endothelial progenitor cells to diabetic condition abolished the regulated kinetics activity of exosomes. Iran J Pharm Res. 2018;17(3):1068–80. [PMC free article] [PubMed] [Google Scholar]

