Next Article in Journal
Raman Multi-Omic Snapshot and Statistical Validation of Structural Differences between Herpes Simplex Type I and Epstein–Barr Viruses
Previous Article in Journal
The Role of Human Endogenous Retrovirus (HERV)-K119 env in THP-1 Monocytic Cell Differentiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exosomes Highlight Future Directions in the Treatment of Acute Kidney Injury

1
Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
2
Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
3
Department of Critical Care Medicine, Center of Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(21), 15568; https://doi.org/10.3390/ijms242115568
Submission received: 20 September 2023 / Revised: 19 October 2023 / Accepted: 23 October 2023 / Published: 25 October 2023
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Acute kidney injury (AKI) is a severe health problem associated with high morbidity and mortality rates. It currently lacks specific therapeutic strategies. This review focuses on the mechanisms underlying the actions of exosomes derived from different cell sources, including red blood cells, macrophages, monocytes, mesenchymal stem cells, and renal tubular cells, in AKI. We also investigate the effects of various exosome contents (such as miRNA, lncRNA, circRNA, mRNA, and proteins) in promoting renal tubular cell regeneration and angiogenesis, regulating autophagy, suppressing inflammatory responses and oxidative stress, and preventing fibrosis to facilitate AKI repair. Moreover, we highlight the interactions between macrophages and renal tubular cells through exosomes, which contribute to the progression of AKI. Additionally, exosomes and their contents show promise as potential biomarkers for diagnosing AKI. The engineering of exosomes has improved their clinical potential by enhancing isolation and enrichment, target delivery to injured renal tissues, and incorporating small molecular modifications for clinical use. However, further research is needed to better understand the specific mechanisms underlying exosome actions, their delivery pathways to renal tubular cells, and the application of multi-omics research in studying AKI.

1. Introduction

Acute kidney injury (AKI) is common and is associated with high rates of mortality and disability. It has been reported that AKI occurs in approximately 20% of hospitalized patients, and in ICU patients, the incidence of AKI can even be as high as 57.3%, with a high mortality rate [1,2]. AKI can result from ischemia-reperfusion injury (IRI), trauma, sepsis, post-major surgery, and exposure to nephrotoxic substances, among others [3,4,5,6]. Due to the complex pathophysiologic mechanisms of AKI, there is currently a lack of specific drugs for its treatment, and some clinical trials have not yielded satisfactory results [5]. Exosomes (Exos), which are widely recognized as promising therapeutic choices for various diseases, hold promise in the development of new treatment strategies for AKI.
Extracellular vesicles (EVs) are a heterogeneous population of membrane-bound vesicles that have not been fully explored in terms of specific types and classifications. Currently, based on their size, biogenic processes, and molecular markers, they can be broadly categorized as follows [7]: microvesicles, exosomes, and apoptotic bodies. However, researchers have also identified some EVs that may represent novel types [8], like tumor-derived tumor bodies versus mitochondrial-derived mitotic vesicles [9]. EVs have been widely applied in regenerative medicine and nanomedicine, participating in the pathophysiology and treatment of various diseases [10,11], including the focus of our discussion, AKI [12].
Exosomes comprise an important type of EV, and they have a diameter ranging from 40 nm to 160 nm, with an average diameter of 100 nm. Exos originate from endosomes and have become an important area of research [13]. Exos were initially believed to be involved in the elimination of cellular waste. However, increasing evidence suggests that exosomes play an important role in intercellular communication. The widely recognized pathways associated with exosome uptake include receptor–ligand binding, membrane fusion, and endocytosis [14].
The contents of exosomes include proteins, lipids, and nucleic acids (such as DNA, mRNA, miRNA, and mitochondrial DNA [15]). Although the synergistic or antagonistic effects of exosome cargos are still not fully understood, numerous studies have confirmed that various cargo components of exosomes are critical to the onset and development of AKI [16].

2. Role of Exosomes of Different Origins in AKI

2.1. Role of Exosomes of MSCs in AKI

Cells of distinct types have been confirmed to secrete exosomes that may be significant in the onset and development of AKI, especially mesenchymal stem cells (MSCs). MSCs are derived from a wide range of sources, including bone marrow, adipose tissue, umbilical cord, human placenta, dental pulp, skin, blood, and urine, as well as from induced pluripotent stem cells (iPSCs) [17].
MSCs have been shown to exert prominent therapeutic effects. Results of a Phase 1 clinical trial revealed that autologous bone marrow-derived MSCs (BMMSCs) increased blood flow and the glomerular filtration rate and reduced inflammation and blood pressure in atherosclerotic renovascular disease [18]. Bone marrow-derived MSCs have been reported to repair cisplatin/glycerol-induced AKI, IRI, and unilateral ureteral obstruction (UUO)-induced renal interstitial fibrosis [19]. However, transplanted MSCs show low survival rates, restricting their therapeutic efficacy. Additionally, the paracrine effects of exosomes in regenerative medicine are of utmost importance. And a meta-analysis has shown that MSC-Exos possess therapeutic effects similar to those of MSCs in AKI [20]. Compared with stem cells, exosomes are easier to extract, store, and transport and they reflect lower immunogenicity and toxicity. They are stable in the circulation, can cross biological barriers, and can be engineered for enhanced efficacy and reduced side effects [21,22,23]. Therefore, the use of MSC-Exos as a replacement for MSCs constitutes a promising area of clinical research.
MSC-Exos have been shown to inhibit renal tubular cell apoptosis, promote tubular proliferation, alleviate inflammatory and oxidative stress, and promote vascular regeneration to repair AKI due to I/R, UUO, or cisplatin [24,25,26,27,28,29]. In addition, exosomes from BMMSCs have been found to reduce kidney fibrosis and inhibit the epithelial–mesenchymal transition in the UUO model. Newly discovered BMMSCs derived from human exfoliated deciduous teeth stem cells show high proliferative potential, self-renewal capability, and low immunogenicity [30]. Secreted exosomes can also alleviate renal injury in cisplatin-induced AKI through anti-apoptotic, anti-inflammatory, and antioxidative mechanisms [31].
Various sources of stem cells also have their unique advantages, such as ease of access, renewability, absence of ethical controversy, and low immunogenicity. Human amniotic epithelial cells (hAECs) have special advantages in clinical translation compared with BMMSCs [32]. To our knowledge, hAEC-Exos achieve protective effects in IRI, cisplatin-induced AKI, and sepsis models in mice [33,34,35]. Human placental mesenchymal stem cells (hPMSCs) are isolated from the amniotic membrane of the placenta, and the exosomes derived from hPMSCs have been shown to ameliorate renal IRI [36]. Adipose-derived mesenchymal stem cell (ADMSC) exosomes are superior to BMMSC exosomes in improving renal function [37]. The potent regenerative function of ADMSCs has attracted attention as a paracrine tool in promoting tubular regeneration in AKI. These cells have been found to alleviate inflammation and oxidative stress, promote tubular proliferation, and alleviate renal injury in sepsis and IRI models [38,39,40,41]. Furthermore, exosomes derived from ADMSCs in retrograde renal injury have been demonstrated to exert a reparative effect. In a feline model of post-renal ureteral obstruction-induced AKI, ADMSC exosome treatment effectively restored the plasma metabolome to a normal physiological steady state [42]. Human umbilical cord mesenchymal stem cell (hucMSC) exosomes regulate autophagy, reduce tubular cell apoptosis and necrosis, stabilize mitochondrial structure, and reduce oxidative stress to alleviate renal damage in cisplatin-induced AKI [43,44,45]. In IRI and UUO models, exosomes from hucMSCs were found to alter the apoptosis transcriptome in proximal tubules [45,46,47]. hucMSC exosomes have also been noted to repair renal tubular cells in an I/R-AKI model in pigs, reducing serum creatinine and blood urea nitrogen levels [48]. iPSCs possess similar characteristics to BMMSCs [49]. The secreted exosomes from iPSCs have similar renal protective properties, effectively safeguarding renal function and integrity, thereby alleviating renal IRI. Additionally, the significant induction of activated extracellular signal-related kinases 1 and 2 (ERK-1 and -2) signaling molecules can be observed [50]. Urine-derived stem cells (USCs) express the renal protective protein Klotho, and their exosomes preferentially localize to the injured kidney area. In both IRI and gentamicin-induced rat models, exosomes derived from USCs exhibit protective effects. USCs improve renal functional impairment through klotho and other similar mechanisms described above [51,52,53] (Table 1). Above all, exosomes of a variety of cellular origins have been found to have a similar regulatory effect in AKI, and in physiological and pathological states, the secretion of exosomes by cells in the body may be regulated in a similar way as hormones.

2.2. Role of Exosomes from Other Sources in AKI

The pathophysiologic processes of AKI do not solely rely on MSCs but also on the participation of blood cells, endothelial cells, and their exosomes. For example, investigators recently uncovered exosomes from red blood cells that showed increased levels of hemoglobin, mediating AKI after cardiopulmonary bypass in animal models, and leading to significant renal damage [60]. Macrophages, as significant inflammatory cells in AKI, have also been shown to secrete exosomes that alleviate endothelial dysfunction and reduce apoptosis and necrosis of renal tubular epithelial cells (TECs) in septic AKI through the neutrophil gelatinase-associated lipocalin-vascular cellular adhesion molecule-1 (NGAL-VCAM1) pathway [61]. Exosomes produced by pericytes may well play a critical role in maintaining the microvascular network [58]. Additionally, transplantation of endothelial progenitor cells (EPCs) significantly reduced renal tissue injury in septic rats [62], and it was shown that exosomes derived from EPCs improved the outcomes of septic mouse models by alleviating inflammation [56,59]. Exosomes from unknown sources in serum also contribute to AKI, as noted in a study in which the application of remote ischemic preconditioning had a protective effect on AKI associated with circulating exosomes [63].
Exosomes from various sources play important roles in different types of AKI, which may be related to the complex systemic effects of AKI (Figure 1). Furthermore, the mechanisms of exosomes primarily involve the following: regulating renal tubular cell proliferation and apoptosis, modulating inflammatory pathways and oxidative stress, inhibiting fibrosis progression, stabilizing endothelial cell function, and promoting angiogenesis—all of which correspond to the pathological mechanisms of AKI. This may suggest that exosomes, from a mechanistic perspective, have the potential to serve as a specific therapeutic direction for AKI.

3. Role of Exosome Content in AKI

To gain more detailed knowledge of the pathophysiologic mechanisms underlying AKI, researchers are conducting in-depth research on exosomes derived from different sources. Therefore, they have shifted their focus toward investigating specific small molecules within exosomes and the corresponding mechanisms by which they regulate AKI. As a result, there has been an increasing amount of research dedicated to the study of the contents of exosomes, particularly various non-coding RNAs. While there have also been some studies on proteins and lipids, their number is relatively limited at present.

3.1. Reparative Effects

Non-coding RNAs are abundant components in exosomes and are important in AKI [64]. MicroRNAs (miRNAs), composed of 21–25 nucleotides, induce mRNA degradation or inhibit protein translation by binding to the 3′-untranslated region (UTR) of target mRNAs. In AKI, the levels of miRNAs undergo significant changes, with the upregulation of miR-17-5p, miR-21, and others and the downregulation of miR-20a-5p and miR205, among others [64]. Additionally, depletion of Drosha in MSC-Exos inhibits their regenerative potential; and depletion of miRNAs in exosomes significantly impairs their therapeutic potential in AKI [65]. These findings highlight the important role of miRNAs in exosomes.
Different miRNAs contained in exosomes from various sources have been shown to exert reparative effects in different types of AKI by promoting tubular proliferation and repair, regulating autophagy, suppressing inflammation and oxidative stress, inhibiting fibrosis progression, and promoting angiogenesis. The role of MSC-Exos in AKI has been summarized in previous studies, and this review provides a summary and supplementation of such recent research in Table 2. As described above, exosomes have been shown to play a role in AKI, but most experiments are limited to the animal and cellular levels, only a few involve the analysis of human serum substances, and clinical trials are more rare.

3.2. Damaging Effects

Although numerous studies have shown that miRNAs, especially those derived from MSCs, have positive effects on AKI (including tissue repair and regeneration, restoration of cell cycle arrest, and attenuation of inflammation and fibrosis), increasing evidence suggests that miRNAs contained in exosomes are detrimental in the development of AKI, particularly in the exosomes from TECs, macrophages (Mφ), and the interplay between them.
Macrophage-derived exosomes have been shown to mediate renal glomerular endothelial dysfunction and necroptosis in sepsis-associated AKI [61,72]. They contribute to tubular cell apoptosis and inflammation in cisplatin (Cisp)-induced nephrotoxicity and IRI models [85,86]. Tubular cell-derived exosomes have also been reported to activate fibroblasts [87] through miR-150-5p, promoting renal fibrosis [88]. Proximal TEC-derived exosomes that target mitochondrial transcription factor A (TFAM) mediate Cisp-induced TEC injury [89]. Furthermore, communication between TECs and macrophages through exosomes has been demonstrated to strongly facilitate the progression of AKI. TEC-derived exosomes carrying microRNA-374b-5p regulate macrophage polarization, promoting M1 macrophage activation and thus worsening AKI [90]. Another study revealed that exosomes derived from lipopolysaccharide (LPS)- or hypoxia-inducible factor-1α-stimulated TECs enhanced the activation of M1 macrophages and mediated renal injury in an AKI mouse model [91,92]. Moreover, activated macrophage-derived exosomes are internalized by tubular cells, leading to increased tubular injury [86]; and this may result in a vicious cycle that develops into a cascade of AKI.
In addition to TECs and macrophages, neutrophils have also been found to promote AKI progression through exosomes. In vitro and in vivo experiments have confirmed that neutrophil-derived exosomes containing miR-30d-5p induce M1 macrophage polarization by upregulating NLRP3 inflammasome expression via the nuclear factor κB (NF-κB) signaling pathway, targeting suppressors of cytokine signaling 1, and sirtuin 1 [93] (Table 3).
Due to the complex bidirectional interactions between exosomes and their cargo, the clinical timing of exosome administration becomes crucial. Modulating the content and structure, or modifying the bioactive substances in a precise manner at the appropriate time, can achieve more accurate therapeutic effects while minimizing potential side effects in AKI. However, all of these interventions should be based on a more comprehensive understanding of the physiologic effects of each cargo and a thorough knowledge of the pathological processes of the disease, which requires further investigation. CircRNAs act as regulatory factors of miRNAs and can affect the expression of target genes by binding to miRNAs. They also function as important components of exosomes to regulate various pathological processes. For example, highly stable circ-FANCA is prominently expressed in sepsis-associated AKI patients and LPS-stimulated HK2 cells. Knocking down circ-FANCA delivered through exosomes alleviated HK2 cell damage in LPS-induced septic AKI by targeting the miR-93-5p/OXSR1 axis [96]. Circ_0001818 has also been found to activate the expression of TXNIP, leading to LPS-induced HK2 cell damage [97].
Long non-coding RNAs (lncRNAs) can regulate the stability of miRNAs and also play a crucial role as contents of exosomes. lncRNAs are also involved in AKI, including tubular programmed cell death and inflammation. The expression of lncRNA TUG1 is downregulated in LPS-induced and IRI-induced AKI. At the same time, the upregulation of the TUG1 gene shows a protective effect on IRI-induced kidney injury. Additionally, lncRNA TUG1 from USCs exosomes regulates the stability of ACSL4 mRNA by interacting with SRSF1, thereby regulating ferroptosis and reducing renal IRI [79].
In addition to their functions through non-coding RNAs, exosomes can also exert information exchange by directly transferring mRNA. For example, it has been reported that exosomes isolated from bovine serum albumin-pretreated renal tubular epithelial cells enhanced inflammation and macrophage migration by transferring CCL2 mRNA, thereby enhancing proteinuria-induced kidney injury [81].
Proteins, another major component of exosomes, have been found to wield functions such as adsorption, promoting intercellular information, and receptor–ligand binding in the process of AKI. Previous studies had shown that exosomes derived from hucMSCs carrying 14-3-3ζ had significant therapeutic effects on tissue regeneration in cisplatin-induced AKI [82]. It has also been reported that ASC-EVs target damaged kidneys in a CD44-dependent manner [98]. A single-center prospective cohort study showed that CD26 of urinary exosomes was correlated with the recovery of AKI in the intensive care unit [99]. MSC-derived exosomes overexpressing polyamine oxidase may also be associated with regulating macrophage polarization (the M1 to M2 transition) and mediating the repair process in IRI/AKI [100].
As noted above, most of the current studies focus on miRNAs in AKI, while other non-coding RNAs, mRNAs, and even less-studied components such as mitochondria, proteins, and lipids are largely overlooked. These molecules may also play important roles in AKI, and, therefore, further research is needed in the future.

4. Exosomes and Their Contents Act as Biomarkers for AKI

The current definition of AKI is still limited to parameters such as serum creatinine and urine output measurements, which provide standardized criteria to some extent, but these markers lack sensitivity. Although several new biomarkers have been proposed, their distribution over time was influenced by various factors, and they also had limitations. Exosomes are widely present in bodily fluids, and the contents of exosomes remain relatively stable under extreme conditions, including high RNase activity, low or high pH, long-term room temperature storage, and multiple freeze–thaw cycles [101,102]. Therefore, using exosomes and their contents as potential biomarkers has attracted immense interest.
In recent years, miRNAs derived from exosomes have been explored as biomarkers for AKI. One study showed that miRNA analysis of urinary exosomes can be used to assess the progression of AKI. During injury, miR-16, miR-24, and miR-200c were elevated in urine, while during early recovery, miRs (miR-9a, miR-141, miR-200a, miR-200c, and miR-429) were upregulated together [103]. In the analysis of early biomarkers for LPS-induced AKI in rats, it was found that miR-181a-5p and miR-23b-3p were expressed at higher levels in the LPS-treated group than in the control group [104]. Urinary exosomal microRNA-21 has also been proposed as a biomarker for AKI associated with scrub typhus [105].
In addition to miRNAs, proteins contained in exosomes have been identified as potential biomarkers. One research study showed that urinary exosomes from vancomycin-induced AKI patients exhibited significantly upregulated expression of inflammatory proteins following nephrotoxic injury [106]. An analysis in 2022 revealed urinary exosomal NHE3 in rats with various types of AKI and in 12 patients, suggesting its potential as a diagnostic biomarker for AKI [107]. Furthermore, the combination of uEV-AQP2 and -AQP1 had been suggested to facilitate the estimation of cisplatin-induced decompensated renal injury [108].
Considering that exosomes exist in body fluids, show stability, and are capable of predicting cellular status and reflecting disease development based on their quantity and content, exosomes and their contents have been widely explored as novel non-invasive biomarkers for various diseases. However, routine monitoring methods may be limited by either no detection or low detection efficiency, thus driving the development of innovative detection methods for exosome enrichment and high-efficiency testing to achieve accurate early diagnosis.
In recent years, a rapid isolation system called EXODUS has been developed for exosome detection, enabling fast and efficient separation and detection of exosomes [109]. Another method is surface plasmon-coupled electrochemiluminescence analysis based on plasma nanostructures; this method consists of a polarized electrochemiluminescence sensor for exosomal miRNA detection and has shown significantly improved detection efficiency [110]. Single-exosome counting has also achieved the detection of only 10 enzyme-labeled exosomes per microliter of serum [111]. Furthermore, breakthroughs have been made in the detection limits of individual exosomes using hollow-core, anti-resonant optical fibers. Additionally, microfluidic-based liquid biopsies have been used for exosome separation and detection and have achieved high sensitivity [112]. The continuous development of emerging technologies has improved the detection rate of exosomes, lowered the detection threshold, and achieved high specificity and accuracy, thus promoting the clinical application of exosomes as biomarkers and facilitating early diagnosis of various diseases.

5. Engineering Exosomes

Although there is considerable research indicating the promising application of exosomes and their contents for the treatment of AKI, there are currently no standardized protocols for the large-scale production, efficient targeting, standardized separation and purification methods, and storage conditions for exosomes. This signifies that the quality of exosomes cannot be controlled. And problems associated with the large-scale production of exosomes like production costs and efficiency needed to be solved. Therefore, it is necessary to develop engineered exosomes or utilize modifications on exosomes to achieve their separation and enrichment, and to enhance their targeting ability with respect to injured tissues.

5.1. Isolation and Enrichment

The most commonly used method for exosome isolation is differential ultracentrifugation, which is applied by approximately 80% of researchers worldwide [113]. However, due to the similarities in size characteristics among different types of exosomes, this method can only achieve a crude separation and lacks additional purification. Other methods such as ExtraPEG, immune-affinity chromatography (IAC), and size-exclusion chromatography (SEC) all have advantages and limitations [114,115]. Some studies have suggested that sequentially combining multiple isolation methods can increase exosomal purity, but issues of cost and quantity still require further discussion [116].
Exosome isolation techniques are constantly evolving and show numerous innovations. Tangential flow filtration (TFF) is more suitable for the large-scale production of higher quantities and activity than ultracentrifugation isolation [117]. The ExoSCRT™ technology based on TFF can extract over 4 L of ASC-CM within 4 h, enabling the reproducible production of exosomes with comparable impurity levels that comply with WHO standards [118]. A 3D culture system based on hollow-fiber bioreactors has additionally been implemented to produce MSC-Exos, showing a 19.4-fold improvement in MSC-Exo yield [119].
Exosome enrichment can also be achieved through methods such as transferring target peptide expression plasmids into cells to generate exosomes with target ligands, directing the loading of drugs via electroporation, and creating mixed exosomes through freeze–thaw cycles. Squeezing drug-loaded cells through a series of filters with reduced pore sizes produces biomimetic exosomes [120]. While these novel technologies are still under development, their application in AKI remains limited. Thus, new technologies for exosome isolation and enrichment and their applications to other diseases should also be specified and optimized for AKI.

5.2. Targeted Therapy

The improvement of the targeting ability of exosomes to an injured site is currently being conducted to enhance the efficacy of exosomes in AKI treatment. For example, P-selectin has been identified as a biomarker for endothelial cell ischemic AKI. In one study, the authors applied a novel binding method to covalently link evPBP (CDAEWVDVS) with DMPEPEG5000-maleimide (DMPE-PEG-MAL), resulting in the synthesis of DMPEPEG-PBP (DPP). DPP can then be inserted into the membrane of Exos to selectively target the injured kidney. Molecular imaging was thus adopted to quantitatively assess the expression of P-selectin in the kidney, demonstrating a protective effect [121]. Another approach involved the use of peptides or antibodies that specifically bind to target molecules. For instance, the LTH peptide (which binds to Kim1) was conjugated to red blood cell-derived EVs (REVs) using a bifunctional cross-linker that enabled its targeting to the kidney [122]. Furthermore, neutrophil membrane-engineered NEX significantly promoted the targeted enrichment of exosomes in damaged renal tissue, leading to improvements in AKI [123]. Researchers have also discovered that the functionalization of MSC-derived EVs (MSC-EVs) using monocyte membranes through membrane fusion significantly enhanced their targeted capability in damaged tissues [124]. However, these applications are currently focused on cardiac diseases [125], and their usage in AKI has yet to be explored to any great degree. Due to the similarity in pathological damage between AKI and heart disease (i.e., inflammation or mediation by IRI), it is speculated that the therapeutic effects may also be comparable. In addition, various hybridization methods from tumor-therapy modalities could offer valuable insights for targeted exosome therapy in AKI. These methods include the hybridization of EVs with synthetic liposomes, coupling between post-modified diazonium compounds and antibodies that are modified with dibenzocyclooctyne (DBCO) [126], coupling with diacyllipid-DNA aptamers (sgc8) [127], anchoring EVs to superparamagnetic nanoparticles [128], and the induction of electrostatic interactions to facilitate fusion between cationic lipids and exosomes [129].
In summary, the aforementioned methods in tumor or heart disease provide insights into the precise targeted treatment and monitoring of AKI using Exos.

5.3. Exosome Modifications

Exosome modifications can be achieved through gene editing and chemical drug modifications. Exos can serve as carriers to deliver nucleic acids for gene editing, or they can be genetically engineered to incorporate gene sequences that encode specific membrane proteins. However, this technology is limited to currently known gene sequences.
Pre-treatment with small-molecule drugs is a common method to enhance therapeutic effects. Compared with genetic manipulation, small-molecule drugs reflect significant advantages: their effects can be fine-tuned by altering their working concentration, duration, and composition. Researchers have found that resveratrol enhanced the restorative effects of HucMSC exosomes in cisplatin-induced AKI [130]. Similarly, 3,3′-dimethylthiazolyl diphenyl tetrazolium bromide stimulated the self-secretion of Wnt11 in MSC-derived exosomes and promoted wound healing [131]. Supramolecular nanofibers containing arginine-glycine-aspartate (RGD) peptides improved the therapeutic effects of EVs in kidney repair [132], and platelet-rich plasma induced YAP nuclear expression to promote BMMSC proliferation, activating the AKT/Rab27 pathways to facilitate the paracrine secretion of MSC-derived exosomes for glycerol-induced AKI repair [133]. Additionally, hypoxia-induced activation of HIF-1α was also found to increase the loading of miR-21 in exosomes, thereby providing a therapeutic effect against septic/IRI-induced AKI [134].
Enhancing the functionality of exosomes through small-molecule pre-treatment is widely recognized. Although numerous molecules have been evaluated, further exploration is needed to determine the optimal timing and underlying mechanisms of action of small-molecule interventions.

6. Future Directions

6.1. Accelerating Clinical Translation

We have already illustrated the meaningful role of exosomes in AKI, but there are still many challenges to achieving clinical translation. Currently, most experiments are limited to animals or just cellular levels, only a few involve analysis of human serum substances, and clinical trials are even more rare. What is more, one important reason for fewer clinical trials is the shortage of engineering exosomes, lower purity, separation efficiency. So, we would like to find new markers not inferior to the used Kim-1 or P-selectin in AKI and then design specific binding ligands with packaged engineering to implement precise and efficient targeted treatment and monitoring of AKI. On this basis, we would accelerate clinical translation to achieve early diagnosis and treatment of AKI, but at the same time, we cannot ignore potential regulatory hurdles or ethical concerns; although, exosome research involves less ethics than stem cells. It still involves a range of ethical issues, such as informed consent of donors; privacy and personal information confidentiality of donors; exosomes of animal origin; and the participation of ethical review committees and regulatory bodies in exosome research is crucial.
When exosomes are clinically applied, then as drugs, the potential differences in exosome treatment responses of different patients are inevitable. These individual differences can be influenced by a variety of factors, including genetic, epigenetic, and other factors. Some studies have identified genetic variants associated with exosome treatment response, such as mutations in certain genes that may lead to increased resistance or sensitivity of cells to treatment. In addition, epigenetic differences may also lead to different responses to exosome therapy. Furthermore, other factors such as chronic disease, drug use, and immune status may also affect a patient’s response to exosome therapy. And these factors may alter the sensitivity or resistance of cells to treatment. Patients may develop an immune response to treatment, resulting in exosomes being cleared or losing function. More research is needed to understand how these factors affect the effectiveness of exosome treatment.

6.2. Elucidating Pathways of Reaching the Renal Tubules

Tubular injury is a significant aspect of AKI. Many studies have shown that exosomes exert their effects by entering renal tubular epithelial cells; however, further exploration is required to understand how exosomes reach damaged renal tubules. While there is evidence that exosomes can cross the blood–brain barrier, there is limited research on assessing whether exosomes can pass through the glomerular filtration membrane in the kidneys. Concerning the mechanisms by which exosomes reach the renal tubular cells, two hypotheses are proposed. (1) Exosomes enter the primary urine through the glomerular filtration membrane and are then reabsorbed by the renal tubules; and (2) exosomes cannot pass through the glomerular filtration membrane but instead interact with the tubules through a network that involves afferent arterioles, efferent arterioles, and peritubular capillaries. It is also possible that both pathways co-exist simultaneously. The process by which systemically administered exosomes enter the vicinity of the renal tubules thus requires further investigation. We postulate that the development of stable in vivo imaging techniques will facilitate the elucidation of this complex and intricate process.

6.3. Exosomes and Multi-Omics Studies

Current research on exosomes is limited, with analyses focused on miRNAs by matching them with potential target mRNAs to elucidate the function of Exo-associated miRNAs. However, many pathways in the body are intertwined and regulated by multiple factors, making it difficult to fully understand the complexity of diseases using this approach. Research on Exos is still evolving, and multi-omics analysis has become a major area of current work. The combined application of exosomes and multi-omics analysis may well bring us closer to the essence of disease in general and provide a comprehensive understanding of cellular changes. However, as the combined application of Exos and multi-omics analyses in kidney diseases has not yet been explored, we recommend that greater attention be given to this area.

7. Conclusions

This review provides an overview of the mechanisms underlying exosome activity, particularly those derived from blood cells and mesenchymal cells, in AKI. We herein explored the reparative effects of various exosome cargo components in AKI and highlighted the interactions between renal tubular cells and macrophages through exosomes that promote the progression of AKI. Furthermore, this review comprised the clinical potential of exosomes and their contents as biomarkers for the early diagnosis and staging of AKI. We also discussed exosome engineering research in AKI, including isolation and enrichment, targeted therapy, and molecular modifications, as well as research progress in tumor and heart disease. Finally, this review comprised ideas for overcoming the limitations of exosome research in AKI. However, elucidation of the molecular underpinnings of how exosomes reach the renal tubular cells and the combined application of exosomes and multi-omics studies await further investigations in the future.

Author Contributions

All authors were involved in drafting and revising the manuscript. Z.P. and Y.L. considered the direction of this topic and guided each stage of the process; X.Z. was responsible for the writing and tables of the article; J.W. drew the figure and searched the relevant research studies fully; J.Z. and Y.T. translated and finalized the version to be published. All authors vouch for the completeness and accuracy of the data and agreed to submit the manuscript for publication. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (No. 82102273 to Li, No. 81772046 to Peng; No. 82241039 to Peng), the Program of Excellent Doctoral (Postdoctoral) of Zhongnan Hospital of Wuhan University (Grant No. ZNYB2020008 to Prof. Li), and Zhongnan Hospital of Wuhan University Science, Technology and Innovation Seed Fund, Project CXPY2020012 to Prof. Li.

Acknowledgments

I would like to thank my guiders, Peng and Li, for their guidance through each stage of the process, and Gao and Nie for their search work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Levey, A.S.; James, M.T. Acute Kidney Injury. Ann. Intern. Med. 2017, 167, ITC66–ITC80. [Google Scholar] [CrossRef]
  2. Xu, X.; Nie, S.; Liu, Z.; Chen, C.; Xu, G.; Zha, Y.; Qian, J.; Liu, B.; Han, S.; Xu, A.; et al. Epidemiology and Clinical Correlates of AKI in Chinese Hospitalized Adults. Clin. J. Am. Soc. Nephrol. 2015, 10, 1510–1518. [Google Scholar] [CrossRef] [PubMed]
  3. Zarbock, A.; Nadim, M.K.; Pickkers, P.; Gomez, H.; Bell, S.; Joannidis, M.; Kashani, K.; Koyner, J.L.; Pannu, N.; Meersch, M.; et al. Sepsis-associated acute kidney injury: Consensus report of the 28th Acute Disease Quality Initiative workgroup. Nat. Rev. Nephrol. 2023, 19, 401–417. [Google Scholar] [CrossRef]
  4. Messerer, D.A.C.; Halbgebauer, R.; Nilsson, B.; Pavenstädt, H.; Radermacher, P.; Huber-Lang, M. Immunopathophysiology of trauma-related acute kidney injury. Nat. Rev. Nephrol. 2021, 17, 91–111. [Google Scholar] [CrossRef] [PubMed]
  5. Pickkers, P.; Darmon, M.; Hoste, E.; Joannidis, M.; Legrand, M.; Ostermann, M.; Prowle, J.R.; Schneider, A.; Schetz, M. Acute kidney injury in the critically ill: An updated review on pathophysiology and management. Intensive Care Med. 2021, 47, 835–850. [Google Scholar] [CrossRef] [PubMed]
  6. Peerapornratana, S.; Manrique-Caballero, C.L.; Gómez, H.; Kellum, J.A. Acute kidney injury from sepsis: Current concepts, epidemiology, pathophysiology, prevention and treatment. Kidney Int. 2019, 96, 1083–1099. [Google Scholar] [CrossRef]
  7. Grange, C.; Bussolati, B. Extracellular vesicles in kidney disease. Nat. Rev. Nephrol. 2022, 18, 499–513. [Google Scholar] [CrossRef] [PubMed]
  8. Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445. [Google Scholar] [CrossRef]
  9. D’Acunzo, P.; Pérez-González, R.; Kim, Y.; Hargash, T.; Miller, C.; Alldred, M.J.; Erdjument-Bromage, H.; Penikalapati, S.C.; Pawlik, M.; Saito, M.; et al. Mitovesicles are a novel population of extracellular vesicles of mitochondrial origin altered in Down syndrome. Sci. Adv. 2021, 7, eabe5085. [Google Scholar] [CrossRef]
  10. Kim, H.Y.; Kwon, S.; Um, W.; Shin, S.; Kim, C.H.; Park, J.H.; Kim, B.-S. Functional Extracellular Vesicles for Regenerative Medicine. Small 2022, 18, e2106569. [Google Scholar] [CrossRef]
  11. Yang, B.; Chen, Y.; Shi, J. Exosome Biochemistry and Advanced Nanotechnology for Next-Generation Theranostic Platforms. Adv. Mater. 2019, 31, e1802896. [Google Scholar] [CrossRef] [PubMed]
  12. Karpman, D.; Ståhl, A.-L.; Arvidsson, I. Extracellular vesicles in renal disease. Nat. Rev. Nephrol. 2017, 13, 545–562. [Google Scholar] [CrossRef] [PubMed]
  13. Li, S.-R.; Man, Q.-W.; Gao, X.; Lin, H.; Wang, J.; Su, F.-C.; Wang, H.-Q.; Bu, L.-L.; Liu, B.; Chen, G. Tissue-derived extracellular vesicles in cancers and non-cancer diseases: Present and future. J. Extracell. Vesicles 2021, 10, e12175. [Google Scholar] [CrossRef]
  14. Mulcahy, L.A.; Pink, R.C.; Carter, D.R.F. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [PubMed]
  15. Hough, K.P.; Trevor, J.L.; Strenkowski, J.G.; Wang, Y.; Chacko, B.K.; Tousif, S.; Chanda, D.; Steele, C.; Antony, V.B.; Dokland, T.; et al. Exosomal transfer of mitochondria from airway myeloid-derived regulatory cells to T cells. Redox. Biol. 2018, 18, 54–64. [Google Scholar] [CrossRef] [PubMed]
  16. Oh, S.; Kwon, S.-H. Extracellular Vesicles in Acute Kidney Injury and Clinical Applications. Int. J. Mol. Sci. 2021, 22, 8913. [Google Scholar] [CrossRef]
  17. Huang, Y.; Yang, L. Mesenchymal stem cells and extracellular vesicles in therapy against kidney diseases. Stem Cell Res. Ther. 2021, 12, 219. [Google Scholar] [CrossRef]
  18. Abumoawad, A.; Saad, A.; Ferguson, C.M.; Eirin, A.; Herrmann, S.M.; Hickson, L.J.; Goksu, B.B.; Bendel, E.; Misra, S.; Glockner, J.; et al. In a Phase 1a escalating clinical trial, autologous mesenchymal stem cell infusion for renovascular disease increases blood flow and the glomerular filtration rate while reducing inflammatory biomarkers and blood pressure. Kidney Int. 2020, 97, 793–804. [Google Scholar] [CrossRef]
  19. Matsui, F.; Babitz, S.K.; Rhee, A.; Hile, K.L.; Zhang, H.; Meldrum, K.K. Mesenchymal stem cells protect against obstruction-induced renal fibrosis by decreasing STAT3 activation and STAT3-dependent MMP-9 production. Am. J. Physiol. Ren. Physiol. 2017, 312, F25–F32. [Google Scholar] [CrossRef]
  20. Liu, C.; Wang, J.; Hu, J.; Fu, B.; Mao, Z.; Zhang, H.; Cai, G.; Chen, X.; Sun, X. Extracellular vesicles for acute kidney injury in preclinical rodent models: A meta-analysis. Stem Cell Res. Ther. 2020, 11, 11. [Google Scholar] [CrossRef]
  21. Dominguez, J.M.; Dominguez, J.H.; Xie, D.; Kelly, K.J. Human extracellular microvesicles from renal tubules reverse kidney ischemia-reperfusion injury in rats. PLoS ONE 2018, 13, e0202550. [Google Scholar] [CrossRef] [PubMed]
  22. Gurunathan, S.; Kang, M.-H.; Jeyaraj, M.; Qasim, M.; Kim, J.-H. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 2019, 8, 307. [Google Scholar] [CrossRef] [PubMed]
  23. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  24. 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]
  25. Menon, S.; Goldstein, S.L.; Mottes, T.; Fei, L.; Kaddourah, A.; Terrell, T.; Arnold, P.; Bennett, M.R.; Basu, R.K. Urinary biomarker incorporation into the renal angina index early in intensive care unit admission optimizes acute kidney injury prediction in critically ill children: A prospective cohort study. Nephrol. Dial. Transpl. 2016, 31, 586–594. [Google Scholar] [CrossRef]
  26. Heller, F.; Frischmann, S.; Grünbaum, M.; Zidek, W.; Westhoff, T.H. Urinary calprotectin and the distinction between prerenal and intrinsic acute kidney injury. Clin. J. Am. Soc. Nephrol. 2011, 6, 2347–2355. [Google Scholar] [CrossRef]
  27. Wang, S.-Y.; Xu, Y.; Hong, Q.; Chen, X.-M.; Cai, G.-Y. Mesenchymal stem cells ameliorate cisplatin-induced acute kidney injury via let-7b-5p. Cell Tissue Res. 2023, 392, 517–533. [Google Scholar] [CrossRef]
  28. Barreto, E.F.; Rule, A.D.; Murad, M.H.; Kashani, K.B.; Lieske, J.C.; Erwin, P.J.; Steckelberg, J.M.; Gajic, O.; Reid, J.M.; Kane-Gill, S.L. Prediction of the Renal Elimination of Drugs with Cystatin C vs. Creatinine: A Systematic Review. Mayo Clin. Proc. 2019, 94, 500–514. [Google Scholar] [CrossRef]
  29. Basu, R.K.; Kaddourah, A.; Goldstein, S.L. Assessment of a renal angina index for prediction of severe acute kidney injury in critically ill children: A multicentre, multinational, prospective observational study. Lancet Child Adolesc. Health 2018, 2, 112–120. [Google Scholar] [CrossRef]
  30. De Miguel-Gómez, L.; Ferrero, H.; López-Martínez, S.; Campo, H.; López-Pérez, N.; Faus, A.; Hervás, D.; Santamaría, X.; Pellicer, A.; Cervelló, I. Stem cell paracrine actions in tissue regeneration and potential therapeutic effect in human endometrium: A retrospective study. BJOG 2020, 127, 551–560. [Google Scholar] [CrossRef]
  31. Huang, T.-Y.; Chien, M.-S.; Su, W.-T. Therapeutic Potential of Pretreatment with Exosomes Derived from Stem Cells from the Apical Papilla against Cisplatin-Induced Acute Kidney Injury. Int. J. Mol. Sci. 2022, 23, 5721. [Google Scholar] [CrossRef]
  32. Zhang, Q.; Lai, D. Application of human amniotic epithelial cells in regenerative medicine: A systematic review. Stem Cell Res. Ther. 2020, 11, 439. [Google Scholar] [CrossRef] [PubMed]
  33. Ren, Y.; Chen, Y.; Zheng, X.; Wang, H.; Kang, X.; Tang, J.; Qu, L.; Shao, X.; Wang, S.; Li, S.; et al. Human amniotic epithelial cells ameliorate kidney damage in ischemia-reperfusion mouse model of acute kidney injury. Stem Cell Res. Ther. 2020, 11, 410. [Google Scholar] [CrossRef]
  34. Kang, X.; Chen, Y.; Xin, X.; Liu, M.; Ma, Y.; Ren, Y.; Ji, J.; Yu, Q.; Qu, L.; Wang, S.; et al. Human Amniotic Epithelial Cells and Their Derived Exosomes Protect Against Cisplatin-Induced Acute Kidney Injury without Compromising Its Antitumor Activity in Mice. Front. Cell Dev. Biol. 2021, 9, 752053. [Google Scholar] [CrossRef] [PubMed]
  35. Chi, D.; Chen, Y.; Xiang, C.; Yao, W.; Wang, H.; Zheng, X.; Xu, D.; Li, N.; Xie, M.; Wang, S.; et al. Human Amnion Epithelial Cells and Their Derived Exosomes Alleviate Sepsis-Associated Acute Kidney Injury via Mitigating Endothelial Dysfunction. Front. Med. 2022, 9, 829606. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, X.; Wang, N.; Huang, Y.; Li, Y.; Li, G.; Lin, Y.; Atala, A.J.; Hou, J.; Zhao, W. Extracellular vesicles from three dimensional culture of human placental mesenchymal stem cells ameliorated renal ischemia/reperfusion injury. Int. J. Artif. Organs 2022, 45, 181–192. [Google Scholar] [CrossRef]
  37. Zhang, W.; Zhang, J.; Huang, H. Exosomes from adipose-derived stem cells inhibit inflammation and oxidative stress in LPS-acute kidney injury. Exp. Cell Res. 2022, 420, 113332. [Google Scholar] [CrossRef]
  38. 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]
  39. Gao, F.; Zuo, B.; Wang, Y.; Li, S.; Yang, J.; Sun, D. Protective function of exosomes from adipose tissue-derived mesenchymal stem cells in acute kidney injury through SIRT1 pathway. Life Sci. 2020, 255, 117719. [Google Scholar] [CrossRef]
  40. Zhang, J.B.; Wang, X.Q.; Lu, G.; Huang, H.S.; Xu, S.Y. Adipose-derived mesenchymal stem cells therapy for acute kidney injury induced by ischemia-reperfusion in a rat model. Clin. Exp. Pharmacol. Physiol. 2017, 44, 1232–1240. [Google Scholar] [CrossRef]
  41. Liu, W.; Hu, C.; Zhang, B.; Li, M.; Deng, F.; Zhao, S. Exosomal microRNA-342-5p secreted from adipose-derived mesenchymal stem cells mitigates acute kidney injury in sepsis mice by inhibiting TLR9. Biol. Proced. Online 2023, 25, 10. [Google Scholar] [CrossRef] [PubMed]
  42. Li, W.; Wang, W.; He, X.; Liao, Z.; Aierken, A.; Hua, J.; Wang, Y.; Lu, D.; Zhang, S. Rapid recovery of male cats with postrenal acute kidney injury by treating with allogeneic adipose mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res. Ther. 2022, 13, 379. [Google Scholar] [CrossRef] [PubMed]
  43. 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] [PubMed]
  44. 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]
  45. Viñas, 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]
  46. Cao, J.-Y.; Wang, B.; Tang, T.-T.; Wen, Y.; Li, Z.-L.; Feng, S.-T.; Wu, M.; Liu, D.; Yin, D.; Ma, K.-L.; et al. Exosomal miR-125b-5p deriving from mesenchymal stem cells promotes tubular repair by suppression of p53 in ischemic acute kidney injury. Theranostics 2021, 11, 5248–5266. [Google Scholar] [CrossRef]
  47. Ji, C.; Zhang, J.; Zhu, Y.; Shi, H.; Yin, S.; Sun, F.; Wang, Q.; Zhang, L.; Yan, Y.; Zhang, X.; et al. Exosomes derived from hucMSC attenuate renal fibrosis through CK1δ/β-TRCP-mediated YAP degradation. Cell Death Dis. 2020, 11, 327. [Google Scholar] [CrossRef]
  48. Huang, J.; Cao, H.; Cui, B.; Ma, X.; Gao, L.; Yu, C.; Shen, F.; Yang, X.; Liu, N.; Qiu, A.; et al. Mesenchymal Stem Cells-Derived Exosomes Ameliorate Ischemia/Reperfusion Induced Acute Kidney Injury in a Porcine Model. Front. Cell Dev. Biol. 2022, 10, 899869. [Google Scholar] [CrossRef]
  49. Takebe, T.; Sekine, K.; Enomura, M.; Koike, H.; Kimura, M.; Ogaeri, T.; Zhang, R.-R.; Ueno, Y.; Zheng, Y.-W.; Koike, N.; et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 2013, 499, 481–484. [Google Scholar] [CrossRef]
  50. Lim, S.W.; Kim, K.W.; Kim, B.M.; Shin, Y.J.; Luo, K.; Quan, Y.; Cui, S.; Ko, E.J.; Chung, B.H.; Yang, C.W. Alleviation of renal ischemia/reperfusion injury by exosomes from induced pluripotent stem cell-derived mesenchymal stem cells. Korean J. Intern. Med. 2022, 37, 411–424. [Google Scholar] [CrossRef]
  51. Tian, S.-F.; Jiang, Z.-Z.; Liu, Y.-M.; Niu, X.; Hu, B.; Guo, S.-C.; Wang, N.-S.; Wang, Y. Human urine-derived stem cells contribute to the repair of ischemic acute kidney injury in rats. Mol. Med. Rep. 2017, 16, 5541–5548. [Google Scholar] [CrossRef]
  52. Li, X.; Liao, J.; Su, X.; Li, W.; Bi, Z.; Wang, J.; Su, Q.; Huang, H.; Wei, Y.; Gao, Y.; et al. Human urine-derived stem cells protect against renal ischemia/reperfusion injury in a rat model via exosomal miR-146a-5p which targets IRAK1. Theranostics 2020, 10, 9561–9578. [Google Scholar] [CrossRef]
  53. Chen, Y.; Zhang, C.; Du, Y.; Yang, X.; Liu, M.; Yang, W.; Lei, G.; Wang, G. Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6. Chin. Med. J. 2022, 135, 2467–2477. [Google Scholar] [CrossRef]
  54. Li, L.; Wang, R.; Jia, Y.; Rong, R.; Xu, M.; Zhu, T. Exosomes Derived from Mesenchymal Stem Cells Ameliorate Renal Ischemic-Reperfusion Injury Through Inhibiting Inflammation and Cell Apoptosis. Front. Med. 2019, 6, 269. [Google Scholar] [CrossRef] [PubMed]
  55. Zou, L.; Ma, X.; Wu, B.; Chen, Y.; Xie, D.; Peng, C. Protective effect of bone marrow mesenchymal stem cell-derived exosomes on cardiomyoblast hypoxia-reperfusion injury through the miR-149/let-7c/Faslg axis. Free Radic. Res. 2020, 54, 722–731. [Google Scholar] [CrossRef]
  56. Zhou, Y.; Li, P.; Goodwin, A.J.; Cook, J.A.; Halushka, P.V.; Chang, E.; Fan, H. Exosomes from Endothelial Progenitor Cells Improve the Outcome of a Murine Model of Sepsis. Mol. Ther. 2018, 26, 1375–1384. [Google Scholar] [CrossRef] [PubMed]
  57. Wan, Y.; Yu, Y.; Yu, C.; Luo, J.; Wen, S.; Shen, L.; Wei, G.; Hua, Y. Human umbilical cord mesenchymal stem cell exosomes alleviate acute kidney injury by inhibiting pyroptosis in rats and NRK-52E cells. Ren. Fail. 2023, 45, 2221138. [Google Scholar] [CrossRef] [PubMed]
  58. Tsugawa-Shimizu, Y.; Fujishima, Y.; Kita, S.; Minami, S.; Sakaue, T.-A.; Nakamura, Y.; Okita, T.; Kawachi, Y.; Fukada, S.; Namba-Hamano, T.; et al. Increased vascular permeability and severe renal tubular damage after ischemia-reperfusion injury in mice lacking adiponectin or T-cadherin. Am. J. Physiol. Endocrinol. Metab. 2021, 320, E179–E190. [Google Scholar] [CrossRef]
  59. Zhang, Y.; Huang, H.; Liu, W.; Liu, S.; Wang, X.Y.; Diao, Z.L.; Zhang, A.H.; Guo, W.; Han, X.; Dong, X.; et al. Endothelial progenitor cells-derived exosomal microRNA-21-5p alleviates sepsis-induced acute kidney injury by inhibiting RUNX1 expression. Cell Death Dis. 2021, 12, 335. [Google Scholar] [CrossRef]
  60. Pat, B.; Oh, J.-Y.; Masjoan Juncos, J.X.; Powell, P.C.; Collawn, J.F.; Patel, R.P.; Dell’Italia, L.J. Red blood cell exosome hemoglobin content increases after cardiopulmonary bypass and mediates acute kidney injury in an animal model. J. Thorac. Cardiovasc. Surg. 2022, 164, e289–e308. [Google Scholar] [CrossRef]
  61. Xiang, H.; Xu, Z.; Zhang, C.; Xiong, J. Macrophage-derived exosomes mediate glomerular endothelial cell dysfunction in sepsis-associated acute kidney injury. Cell Biosci. 2023, 13, 46. [Google Scholar] [CrossRef]
  62. Xu, X.; Yang, J.; Li, N.; Wu, R.; Tian, H.; Song, H.; Wang, H. Role of Endothelial Progenitor Cell Transplantation in Rats with Sepsis. Transpl. Proc. 2015, 47, 2991–3001. [Google Scholar] [CrossRef]
  63. Pan, T.; Jia, P.; Chen, N.; Fang, Y.; Liang, Y.; Guo, M.; Ding, X. Delayed Remote Ischemic Preconditioning ConfersRenoprotection against Septic Acute Kidney Injury via Exosomal miR-21. Theranostics 2019, 9, 405–423. [Google Scholar] [CrossRef]
  64. Ren, G.-L.; Zhu, J.; Li, J.; Meng, X.-M. Noncoding RNAs in acute kidney injury. J. Cell Physiol. 2019, 234, 2266–2276. [Google Scholar] [CrossRef] [PubMed]
  65. 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] [PubMed]
  66. Zhang, J.; He, W.; Zheng, D.; He, Q.; Tan, M.; Jin, J. Exosomal-miR-1184 derived from mesenchymal stem cells alleviates cisplatin-associated acute kidney injury. Mol. Med. Rep. 2021, 24, 795. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, R.; Zhu, Y.; Li, Y.; Liu, W.; Yin, L.; Yin, S.; Ji, C.; Hu, Y.; Wang, Q.; Zhou, X.; et al. Human umbilical cord mesenchymal stem cell exosomes alleviate sepsis-associated acute kidney injury via regulating microRNA-146b expression. Biotechnol. Lett. 2020, 42, 669–679. [Google Scholar] [CrossRef] [PubMed]
  68. Viñas, J.L.; Spence, M.; Porter, C.J.; Douvris, A.; Gutsol, A.; Zimpelmann, J.A.; Campbell, P.A.; Burns, K.D. micro-RNA-486-5p protects against kidney ischemic injury and modifies the apoptotic transcriptome in proximal tubules. Kidney Int. 2021, 100, 597–612. [Google Scholar] [CrossRef] [PubMed]
  69. Yu, Y.; Chen, M.; Guo, Q.; Shen, L.; Liu, X.; Pan, J.; Zhang, Y.; Xu, T.; Zhang, D.; Wei, G. Human umbilical cord mesenchymal stem cell exosome-derived miR-874-3p targeting RIPK1/PGAM5 attenuates kidney tubular epithelial cell damage. Cell. Mol. Biol. Lett. 2023, 28, 12. [Google Scholar] [CrossRef]
  70. Zhang, Y.; Wang, J.; Yang, B.; Qiao, R.; Li, A.; Guo, H.; Ding, J.; Li, H.; Ye, H.; Wu, D.; et al. Transfer of MicroRNA-216a-5p From Exosomes Secreted by Human Urine-Derived Stem Cells Reduces Renal Ischemia/Reperfusion Injury. Front. Cell Dev. Biol. 2020, 8, 610587. [Google Scholar] [CrossRef]
  71. Ma, M.; Luo, Q.; Fan, L.; Li, W.; Li, Q.; Meng, Y.; Yun, C.; Wu, H.; Lu, Y.; Cui, S.; et al. The urinary exosomes derived from premature infants attenuate cisplatin-induced acute kidney injury in mice via microRNA-30a-5p/ mitogen-activated protein kinase 8 (MAPK8). Bioengineered 2022, 13, 1650–1665. [Google Scholar] [CrossRef] [PubMed]
  72. Juan, C.-X.; Mao, Y.; Cao, Q.; Chen, Y.; Zhou, L.-B.; Li, S.; Chen, H.; Chen, J.-H.; Zhou, G.-P.; Jin, R. Exosome-mediated pyroptosis of miR-93-TXNIP-NLRP3 leads to functional difference between M1 and M2 macrophages in sepsis-induced acute kidney injury. J. Cell. Mol. Med. 2021, 25, 4786–4799. [Google Scholar] [CrossRef] [PubMed]
  73. Yu, W.; Zeng, H.; Chen, J.; Fu, S.; Huang, Q.; Xu, Y.; Xu, A.; Lan, H.-Y.; Tang, Y. miR-20a-5p is enriched in hypoxia-derived tubular exosomes and protects against acute tubular injury. Clin. Sci. 2020, 134, 2223–2234. [Google Scholar] [CrossRef]
  74. He, Z.; Wang, H.; Yue, L. Endothelial progenitor cells-secreted extracellular vesicles containing microRNA-93-5p confer protection against sepsis-induced acute kidney injury via the KDM6B/H3K27me3/TNF-α axis. Exp. Cell Res. 2020, 395, 112173. [Google Scholar] [CrossRef] [PubMed]
  75. Li, X.-Y.; Zhang, Y.-Q.; Xu, G.; Li, S.-H.; Li, H. miR-124/MCP-1 signaling pathway modulates the protective effect of itraconazole on acute kidney injury in a mouse model of disseminated candidiasis. Int. J. Mol. Med. 2018, 41, 3468–3476. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, S.; Sun, H.; Kong, W.; Zhang, B. Functional role of microRNA-500a-3P-loaded liposomes in the treatment of cisplatin-induced AKI. IET Nanobiotechnol. 2020, 14, 465–469. [Google Scholar] [CrossRef]
  77. Jiang, L.; Liu, X.-Q.; Ma, Q.; Yang, Q.; Gao, L.; Li, H.-D.; Wang, J.-N.; Wei, B.; Wen, J.; Li, J.; et al. hsa-miR-500a-3P alleviates kidney injury by targeting MLKL-mediated necroptosis in renal epithelial cells. FASEB J. 2019, 33, 3523–3535. [Google Scholar] [CrossRef]
  78. Shi, L.; Zhang, Y.; Xia, Y.; Li, C.; Song, Z.; Zhu, J. MiR-150-5p protects against septic acute kidney injury via repressing the MEKK3/JNK pathway. Cell. Signal. 2021, 86, 110101. [Google Scholar] [CrossRef]
  79. Sun, Z.; Wu, J.; Bi, Q.; Wang, W. Exosomal lncRNA TUG1 derived from human urine-derived stem cells attenuates renal ischemia/reperfusion injury by interacting with SRSF1 to regulate ASCL4-mediated ferroptosis. Stem Cell Res. Ther. 2022, 13, 297. [Google Scholar] [CrossRef]
  80. Chen, L.; Xu, J.-Y.; Tan, H.-B. LncRNA TUG1 regulates the development of ischemia-reperfusion mediated acute kidney injury through miR-494-3p/E-cadherin axis. J. Inflamm. 2021, 18, 12. [Google Scholar] [CrossRef]
  81. Shen, B.; Liu, J.; Zhang, F.; Wang, Y.; Qin, Y.; Zhou, Z.; Qiu, J.; Fan, Y. CCR2 Positive Exosome Released by Mesenchymal Stem Cells Suppresses Macrophage Functions and Alleviates Ischemia/Reperfusion-Induced Renal Injury. Stem Cells Int. 2016, 2016, 1240301. [Google Scholar] [CrossRef] [PubMed]
  82. 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 14-3-3ζ enhanced autophagy via modulation of ATG16L in preventing cisplatin-induced acute kidney injury. Am. J. Transl. Res. 2018, 10, 101–113. [Google Scholar] [PubMed]
  83. Du, J.; Sun, Q.; Wang, Z.; Wang, F.; Chen, F.; Wang, H.; Shang, G.; Chen, X.; Ding, S.; Li, C.; et al. Tubular epithelial cells derived-exosomes containing CD26 protects mice against renal ischemia/reperfusion injury by maintaining proliferation and dissipating inflammation. Biochem. Biophys. Res. Commun. 2021, 553, 134–140. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, C.; George, S.K.; Wu, R.; Thakker, P.U.; Abolbashari, M.; Kim, T.-H.; Ko, I.K.; Zhang, Y.; Sun, Y.; Jackson, J.; et al. Reno-protection of Urine-derived Stem Cells in A Chronic Kidney Disease Rat Model Induced by Renal Ischemia and Nephrotoxicity. Int. J. Biol. Sci. 2020, 16, 435–446. [Google Scholar] [CrossRef] [PubMed]
  85. Yuan, L.; Yang, J.; Liu, F.; Li, L.; Liu, J.; Chen, Y.; Cheng, J.; Lu, Y.; Yuan, Y. Macrophage-derived exosomal miR-195a-5p impairs tubular epithelial cells mitochondria in acute kidney injury mice. FASEB J. 2023, 37, e22691. [Google Scholar] [CrossRef]
  86. Zhang, Z.; Chen, H.; Zhou, L.; Li, C.; Lu, G.; Wang, L. Macrophage-derived exosomal miRNA-155 promotes tubular injury in ischemia-induced acute kidney injury. Int. J. Mol. Med. 2022, 50, 116. [Google Scholar] [CrossRef]
  87. Guan, H.; Peng, R.; Mao, L.; Fang, F.; Xu, B.; Chen, M. Injured tubular epithelial cells activate fibroblasts to promote kidney fibrosis through miR-150-containing exosomes. Exp. Cell Res. 2020, 392, 112007. [Google Scholar] [CrossRef]
  88. Zhou, X.; Zhao, S.; Li, W.; Ruan, Y.; Yuan, R.; Ning, J.; Jiang, K.; Xie, J.; Yao, X.; Li, H.; et al. Tubular cell-derived exosomal miR-150-5p contributes to renal fibrosis following unilateral ischemia-reperfusion injury by activating fibroblast in vitro and in vivo. Int. J. Biol. Sci. 2021, 17, 4021–4033. [Google Scholar] [CrossRef]
  89. Guo, Y.; Ni, J.; Chen, S.; Bai, M.; Lin, J.; Ding, G.; Zhang, Y.; Sun, P.; Jia, Z.; Huang, S.; et al. MicroRNA-709 Mediates Acute Tubular Injury through Effects on Mitochondrial Function. J. Am. Soc. Nephrol. 2018, 29, 449–461. [Google Scholar] [CrossRef]
  90. Ding, C.; Zheng, J.; Wang, B.; Li, Y.; Xiang, H.; Dou, M.; Qiao, Y.; Tian, P.; Ding, X.; Xue, W. Exosomal MicroRNA-374b-5p From Tubular Epithelial Cells Promoted M1 Macrophages Activation and Worsened Renal Ischemia/Reperfusion Injury. Front. Cell Dev. Biol. 2020, 8, 587693. [Google Scholar] [CrossRef]
  91. Lv, L.-L.; Feng, Y.; Wu, M.; Wang, B.; Li, Z.-L.; Zhong, X.; Wu, W.-J.; Chen, J.; Ni, H.-F.; Tang, T.-T.; et al. Exosomal miRNA-19b-3p of tubular epithelial cells promotes M1 macrophage activation in kidney injury. Cell Death Differ. 2020, 27, 210–226. [Google Scholar] [CrossRef]
  92. Li, Z.-L.; Lv, L.-L.; Tang, T.-T.; Wang, B.; Feng, Y.; Zhou, L.-T.; Cao, J.-Y.; Tang, R.-N.; Wu, M.; Liu, H.; et al. HIF-1α inducing exosomal microRNA-23a expression mediates the cross-talk between tubular epithelial cells and macrophages in tubulointerstitial inflammation. Kidney Int. 2019, 95, 388–404. [Google Scholar] [CrossRef] [PubMed]
  93. Jiao, Y.; Zhang, T.; Zhang, C.; Ji, H.; Tong, X.; Xia, R.; Wang, W.; Ma, Z.; Shi, X. Exosomal miR-30d-5p of neutrophils induces M1 macrophage polarization and primes macrophage pyroptosis in sepsis-related acute lung injury. Crit. Care 2021, 25, 356. [Google Scholar] [CrossRef] [PubMed]
  94. Yin, Q.; Zhao, Y.-J.; Ni, W.-J.; Tang, T.-T.; Wang, Y.; Cao, J.-Y.; Yin, D.; Wen, Y.; Li, Z.-L.; Zhang, Y.-L.; et al. MiR-155 deficiency protects renal tubular epithelial cells from telomeric and genomic DNA damage in cisplatin-induced acute kidney injury. Theranostics 2022, 12, 4753–4766. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, X.-B.; Chen, X.; Li, D.-J.; Qi, G.-N.; Dai, Y.-Q.; Gu, J.; Chen, M.-Q.; Hu, S.; Liu, Z.-Y.; Yang, Z.-M. Inhibition of miR-155 Ameliorates Acute Kidney Injury by Apoptosis Involving the Regulation on TCF4/Wnt/β-Catenin Pathway. Nephron 2019, 143, 135–147. [Google Scholar] [CrossRef] [PubMed]
  96. Li, H.; Zhang, X.; Wang, P.; Zhou, X.; Liang, H.; Li, C. Knockdown of circ-FANCA alleviates LPS-induced HK2 cell injury via targeting miR-93-5p/OXSR1 axis in septic acute kidney injury. Diabetol. Metab. Syndr. 2021, 13, 7. [Google Scholar] [CrossRef] [PubMed]
  97. Kuang, F.; Wang, B.; You, T.; Liu, Y.; Li, P.; Wang, J. Circ_0001818 targets miR-136-5p to increase lipopolysaccharide-induced HK2 cell injuries by activating TXNIP/NLRP3 inflammasome pathway. Shock 2023, 60, 110–120. [Google Scholar] [CrossRef]
  98. Herrera, M.B.; Bussolati, B.; Bruno, S.; Morando, L.; Mauriello-Romanazzi, G.; Sanavio, F.; Stamenkovic, I.; Biancone, L.; Camussi, G. Exogenous mesenchymal stem cells localize to the kidney by means of CD44 following acute tubular injury. Kidney Int. 2007, 72, 430–441. [Google Scholar] [CrossRef] [PubMed]
  99. Du, J.; Li, Y.; Sun, Q.; Wang, Z.; Wang, F.; Chen, F.; Wang, H.; Liu, Y.; Zhou, H.; Shang, G.; et al. Urinary exosomal CD26 is associated with recovery from acute kidney injury in intensive care units: A prospective cohort study. Clin. Chem. Lab. Med. 2021, 59, 1535–1546. [Google Scholar] [CrossRef]
  100. Xie, X.; Yang, X.; Wu, J.; Tang, S.; Yang, L.; Fei, X.; Wang, M. Exosome from indoleamine 2,3-dioxygenase-overexpressing bone marrow mesenchymal stem cells accelerates repair process of ischemia/reperfusion-induced acute kidney injury by regulating macrophages polarization. Stem Cell Res. Ther. 2022, 13, 367. [Google Scholar] [CrossRef] [PubMed]
  101. Succar, L.; Pianta, T.J.; Davidson, T.; Pickering, J.W.; Endre, Z.H. Subclinical chronic kidney disease modifies the diagnosis of experimental acute kidney injury. Kidney Int. 2017, 92, 680–692. [Google Scholar] [CrossRef]
  102. Yun, S.J.; Jeong, P.; Kim, W.-T.; Kim, T.H.; Lee, Y.-S.; Song, P.H.; Choi, Y.-H.; Kim, I.Y.; Moon, S.-K.; Kim, W.-J. Cell-free microRNAs in urine as diagnostic and prognostic biomarkers of bladder cancer. Int. J. Oncol. 2012, 41, 1871–1878. [Google Scholar] [CrossRef]
  103. Sonoda, H.; Lee, B.R.; Park, K.-H.; Nihalani, D.; Yoon, J.-H.; Ikeda, M.; Kwon, S.-H. miRNA profiling of urinary exosomes to assess the progression of acute kidney injury. Sci. Rep. 2019, 9, 4692. [Google Scholar] [CrossRef] [PubMed]
  104. Da-Silva, C.C.S.; Anauate, A.C.; Guirao, T.P.; Novaes, A.D.S.; Maquigussa, E.; Boim, M.A. Analysis of exosome-derived microRNAs as early biomarkers of lipopolysaccharide-induced acute kidney injury in rats. Front. Physiol. 2022, 13, 944864. [Google Scholar] [CrossRef] [PubMed]
  105. Yun, C.Y.; Lim, J.H.; Oh, J.H.; Cho, A.Y.; Lee, K.Y.; Sun, I.O. Urinary Exosomal microRNA-21 as a Marker for Scrub Typhus-Associated Acute Kidney Injury. Genet. Test. Mol. Biomark. 2021, 25, 140–144. [Google Scholar] [CrossRef]
  106. Awdishu, L.; Le, A.; Amato, J.; Jani, V.; Bal, S.; Mills, R.H.; Carrillo-Terrazas, M.; Gonzalez, D.J.; Tolwani, A.; Acharya, A.; et al. Urinary Exosomes Identify Inflammatory Pathways in Vancomycin Associated Acute Kidney Injury. Int. J. Mol. Sci. 2021, 22, 2784. [Google Scholar] [CrossRef] [PubMed]
  107. Yu, Y.; Ren, Z.; Xie, A.; Jia, Y.; Xue, Y.; Wang, P.; Ji, D.; Wang, X. Assessment of Urinary Exosomal NHE3 as a Biomarker of Acute Kidney Injury. Diagnostics 2022, 12, 2634. [Google Scholar] [CrossRef] [PubMed]
  108. Sonoda, H.; Oshikawa-Hori, S.; Ikeda, M. An Early Decrease in Release of Aquaporin-2 in Urinary Extracellular Vesicles After Cisplatin Treatment in Rats. Cells 2019, 8, 139. [Google Scholar] [CrossRef] [PubMed]
  109. Chen, Y.; Zhu, Q.; Cheng, L.; Wang, Y.; Li, M.; Yang, Q.; Hu, L.; Lou, D.; Li, J.; Dong, X.; et al. Exosome detection via the ultrafast-isolation system: EXODUS. Nat. Methods 2021, 18, 212–218. [Google Scholar] [CrossRef]
  110. Rayamajhi, S.; Nguyen, T.D.T.; Marasini, R.; Aryal, S. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery. Acta Biomater. 2019, 94, 482–494. [Google Scholar] [CrossRef]
  111. Liu, C.; Xu, X.; Li, B.; Situ, B.; Pan, W.; Hu, Y.; An, T.; Yao, S.; Zheng, L. Single-Exosome-Counting Immunoassays for Cancer Diagnostics. Nano Lett. 2018, 18, 4226–4232. [Google Scholar] [CrossRef] [PubMed]
  112. Zhao, L.; Wang, H.; Fu, J.; Wu, X.; Liang, X.-Y.; Liu, X.-Y.; Wu, X.; Cao, L.-L.; Xu, Z.-Y.; Dong, M. Microfluidic-based exosome isolation and highly sensitive aptamer exosome membrane protein detection for lung cancer diagnosis. Biosens. Bioelectron. 2022, 214, 114487. [Google Scholar] [CrossRef]
  113. Gardiner, C.; Di Vizio, D.; Sahoo, S.; Théry, C.; Witwer, K.W.; Wauben, M.; Hill, A.F. Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. J. Extracell. Vesicles 2016, 5, 32945. [Google Scholar] [CrossRef]
  114. Rider, M.A.; Hurwitz, S.N.; Meckes, D.G. ExtraPEG: A Polyethylene Glycol-Based Method for Enrichment of Extracellular Vesicles. Sci. Rep. 2016, 6, 23978. [Google Scholar] [CrossRef] [PubMed]
  115. Zarovni, N.; Corrado, A.; Guazzi, P.; Zocco, D.; Lari, E.; Radano, G.; Muhhina, J.; Fondelli, C.; Gavrilova, J.; Chiesi, A. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods 2015, 87, 46–58. [Google Scholar] [CrossRef]
  116. Ludwig, A.-K.; De Miroschedji, K.; Doeppner, T.R.; Börger, V.; Ruesing, J.; Rebmann, V.; Durst, S.; Jansen, S.; Bremer, M.; Behrmann, E.; et al. Precipitation with polyethylene glycol followed by washing and pelleting by ultracentrifugation enriches extracellular vesicles from tissue culture supernatants in small and large scales. J. Extracell. Vesicles 2018, 7, 1528109. [Google Scholar] [CrossRef]
  117. Busatto, S.; Vilanilam, G.; Ticer, T.; Lin, W.-L.; Dickson, D.W.; Shapiro, S.; Bergese, P.; Wolfram, J. Tangential Flow Filtration for Highly Efficient Concentration of Extracellular Vesicles from Large Volumes of Fluid. Cells 2018, 7, 273. [Google Scholar] [CrossRef]
  118. Lee, J.H.; Ha, D.H.; Go, H.-K.; Youn, J.; Kim, H.-K.; Jin, R.C.; Miller, R.B.; Kim, D.-H.; Cho, B.S.; Yi, Y.W. Reproducible Large-Scale Isolation of Exosomes from Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells and Their Application in Acute Kidney Injury. Int. J. Mol. Sci. 2020, 21, 4774. [Google Scholar] [CrossRef]
  119. Cao, J.; Wang, B.; Tang, T.; Lv, L.; Ding, Z.; Li, Z.; Hu, R.; Wei, Q.; Shen, A.; Fu, Y.; et al. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res. Ther. 2020, 11, 206. [Google Scholar] [CrossRef]
  120. He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome Theranostics: Biology and Translational Medicine. Theranostics 2018, 8, 237–255. [Google Scholar] [CrossRef]
  121. Zhang, K.; Li, R.; Chen, X.; Yan, H.; Li, H.; Zhao, X.; Huang, H.; Chen, S.; Liu, Y.; Wang, K.; et al. Renal Endothelial Cell-Targeted Extracellular Vesicles Protect the Kidney from Ischemic Injury. Adv. Sci. 2023, 10, e2204626. [Google Scholar] [CrossRef] [PubMed]
  122. Tang, T.-T.; Wang, B.; Li, Z.-L.; Wen, Y.; Feng, S.-T.; Wu, M.; Liu, D.; Cao, J.-Y.; Yin, Q.; Yin, D.; et al. Kim-1 Targeted Extracellular Vesicles: A New Therapeutic Platform for RNAi to Treat AKI. J. Am. Soc. Nephrol. 2021, 32, 2467–2483. [Google Scholar] [CrossRef] [PubMed]
  123. Wu, P.; Tang, Y.; Jin, C.; Wang, M.; Li, L.; Liu, Z.; Shi, H.; Sun, Z.; Hou, X.; Chen, W.; et al. Neutrophil membrane engineered HucMSC sEVs alleviate cisplatin-induced AKI by enhancing cellular uptake and targeting. J. Nanobiotechnol. 2022, 20, 353. [Google Scholar] [CrossRef] [PubMed]
  124. Hu, S.; Wang, X.; Li, Z.; Zhu, D.; Cores, J.; Wang, Z.; Li, J.; Mei, X.; Cheng, X.; Su, T.; et al. Platelet membrane and stem cell exosome hybrid enhances cellular uptake and targeting to heart injury. Nano Today 2021, 39, 101210. [Google Scholar] [CrossRef] [PubMed]
  125. Li, Q.; Huang, Z.; Wang, Q.; Gao, J.; Chen, J.; Tan, H.; Li, S.; Wang, Z.; Weng, X.; Yang, H.; et al. Targeted immunomodulation therapy for cardiac repair by platelet membrane engineering extracellular vesicles via hitching peripheral monocytes. Biomaterials 2022, 284, 121529. [Google Scholar] [CrossRef]
  126. Kim, M.S.; Haney, M.J.; Zhao, Y.; Yuan, D.; Deygen, I.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: In vitro and in vivo evaluations. Nanomedicine 2018, 14, 195–204. [Google Scholar] [CrossRef]
  127. Zou, J.; Shi, M.; Liu, X.; Jin, C.; Xing, X.; Qiu, L.; Tan, W. Aptamer-Functionalized Exosomes: Elucidating the Cellular Uptake Mechanism and the Potential for Cancer-Targeted Chemotherapy. Anal. Chem. 2019, 91, 2425–2430. [Google Scholar] [CrossRef]
  128. Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef]
  129. Piffoux, M.; Silva, A.K.A.; Wilhelm, C.; Gazeau, F.; Tareste, D. Modification of Extracellular Vesicles by Fusion with Liposomes for the Design of Personalized Biogenic Drug Delivery Systems. ACS Nano 2018, 12, 6830–6842. [Google Scholar] [CrossRef]
  130. Fazekas, B.; Griffin, M.D. Mesenchymal stromal cell-based therapies for acute kidney injury: Progress in the last decade. Kidney Int. 2020, 97, 1130–1140. [Google Scholar] [CrossRef]
  131. Shi, H.; Xu, X.; Zhang, B.; Xu, J.; Pan, Z.; Gong, A.; Zhang, X.; Li, R.; Sun, Y.; Yan, Y.; et al. 3,3’-Diindolylmethane stimulates exosomal Wnt11 autocrine signaling in human umbilical cord mesenchymal stem cells to enhance wound healing. Theranostics 2017, 7, 1674–1688. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, C.; Shang, Y.; Chen, X.; Midgley, A.C.; Wang, Z.; Zhu, D.; Wu, J.; Chen, P.; Wu, L.; Wang, X.; et al. Supramolecular Nanofibers Containing Arginine-Glycine-Aspartate (RGD) Peptides Boost Therapeutic Efficacy of Extracellular Vesicles in Kidney Repair. ACS Nano 2020, 14, 12133–12147. [Google Scholar] [CrossRef] [PubMed]
  133. Ji, C.; Zhang, J.; Zhou, Z.; Shi, H.; Liu, W.; Sun, F.; Zhang, C.; Zhang, L.; Sun, Z.; Qian, H. Platelet-rich plasma promotes MSCs exosomes paracrine to repair acute kidney injury via AKT/Rab27 pathway. Am. J. Transl. Res. 2021, 13, 1445–1457. [Google Scholar] [PubMed]
  134. Ding, Z.-Y.; Tang, T.-T.; Li, Z.-L.; Cao, J.-Y.; Lv, L.-L.; Wen, Y.; Wang, B.; Liu, B.-C. Therapeutic Effect of Extracellular Vesicles Derived from HIF Prolyl Hydroxylase Domain Enzyme Inhibitor-Treated Cells on Renal Ischemia/Reperfusion Injury. Kidney Dis. 2022, 8, 206–216. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Exosomes from various sources including red blood cells, macrophages, monocytes, and mesenchymal stem cells in promoting renal tubular cell regeneration and angiogenesis, regulating autophagy, suppressing inflammatory responses and oxidative stress, and preventing fibrosis to facilitate AKI repair through their contents (such as miRNA, lncRNA, circRNA, mRNA, and proteins).
Figure 1. Exosomes from various sources including red blood cells, macrophages, monocytes, and mesenchymal stem cells in promoting renal tubular cell regeneration and angiogenesis, regulating autophagy, suppressing inflammatory responses and oxidative stress, and preventing fibrosis to facilitate AKI repair through their contents (such as miRNA, lncRNA, circRNA, mRNA, and proteins).
Ijms 24 15568 g001
Table 1. Mechanisms of exosomes of different origins in AKI.
Table 1. Mechanisms of exosomes of different origins in AKI.
Exo SourceModelMechanismPathways/MarkersReference
BMMSCIRI
Rats
Mice
HK-2 (H/R)
Anti-apoptotic
Reduces oxidative stress
Anti-inflammatory
Promote angiogenesis
Inhibits endoplasmic reticulum stress
IL-6, TNF-α, NF-κB IFN-γ ↓
caspase-9, cleaved caspase-3, Bax, and Bcl-2 ↓
BIP
miR-149/let-7c/Faslg
[24,54,55]
BMMSCUUO
Rats
Inhibits fibrosis
Epithelial mesenchymal transformation
Anti-inflammatory
Anti-apoptotic
STAT3 ↓
MMP-9P
Colla1,α-SMA ↓
PCNA ↓
[19]
BMMSCCisp
Mice
Anti-apoptotic
Reduces DNA damage
Inhibiting p53[27]
SCAPCisp
NRK-52E
Anti-apoptotic
Reduces oxidative stress
Anti-inflammatory
NF-κβ, IL-1β, p53 ↓ Bcl-2 ↑ Bax, CASP8, CASP9, and CASP3 ↓[31]
hAECIRI mice
HK-2 (H/R)
Anti-apoptotic
Anti-inflammatory
Promote angiogenesis
extracellular matrix
growth factor
cytokine production,
immunomodulation
[33]
hAECSepsis
LPS-treated HUVECs CLP mice
Anti-inflammatory
Maintained endothelial cell adhesion junction
Mitigating endothelial dysfunction
phosphor-p65, p65, VCAM-1, ZO-1[35]
hAECCisp
mice
Anti-inflammatoryTNF-α/MAPK and caspase signaling ↓[34]
ADMSCIRIAnti-apoptotic
Anti-inflammatory
Reduces oxidative stress, DNA damage
Promote angiogenesis
Inhibits fibrosis
Relieves tubular damage
TNF-α/NF-κB/IL-1β/MIF/PAI-1/Cox-2 ↓
NOX-1/NOX-2/oxidized protein ↓ Bax/caspase-3/PARP ↓ Smad3/TGF-β ↓
[38]
ADMSCSepsis
CLP mice
LPS HK-2
Anti-apoptotic
Anti-inflammatory
Promote angiogenesis
Autophagy
SIRT1
TLR9
[37,39,41]
ADMSCPostrenal AKI
Cats
Metabolomecarnitine, melibiose, D-Glucosamine, cytidine, dihydroorotic acid, stachyose ↑[42]
USCsPlasma from AKI patients
IRI rats
IRI mice
HK-2 (H/R)
Anti-inflammatory
Inhibits fibrosis
Reduces oxidative stress
TRAF6
IRAK1
NF-κB p65
IL10 and TGFβ1 ↑
IFN-γ and IL-1β ↓
[51,52,53]
IPSCIRI
HK-2 (H/R)
Reduce renal damageERK 1/2 signaling[50]
HuMSCCisp NRK-52E cellsPromotes TEC proliferation
Anti-apoptotic
Anti-inflammatory
Reduces oxidative stress
Promotes mitochondrial fusion
Autophagy
fibronectin, α-SMA, GSDMD, caspase-1, IL-1β and NLRP3 ↓[56]
HuMSCUUO ratsInhibits fibrosis CK1δ/β-TRCP inhibited YAP activity[47]
HuMSCIRI
rats
Anti-inflammatory
Inhibits fibrosis
Reduces oxidative stress
[45,46,57]
MacrophagesSepsis
CLP mice
Relieves endothelial cell dysfunction
Reduces tubular cell apoptosis, pyroptosis
HMGB, VCAM1 ↓[56]
PericytesIRI miceReduces tubular cell damage
Maintain capillary stability
Cadherin
APN
[58]
EPCSepsis
CLP rats
Anti-inflammatoryRUNX1 ↓[59]
Abbreviations: BMMSC, bone marrow mesenchymal stem cell; ADMSC, adipose-derived mesenchymal stem cell; SCAP, apical papillary stem cell; USCs, urine-derived mesenchymal stem cells; IPSC, induced pluripotent stem cell; hAEC, human amniotic mesenchymal stem cell; EPC, endothelial progenitor cell; VCAM-1, vascular cell adhesion molecule-1; VCAM1, vascular cell adhesion molecule 1; APN, Adiponectin; ↓, Decreased expression; ↑, increased expression.
Table 2. Reparative role of exosomal contents in AKI.
Table 2. Reparative role of exosomal contents in AKI.
ContentExo SourceModelMechanismPathwaysReference
miR-199a-3pBMMSCIRI mice
HK-2 (H/R)
Anti-apoptotic
Reduces oxidative stress
Sema3A ↓—ERK, AKT ↑[24]
miR-let-7b-5pBMMSCCisp miceAnti-apoptotic
Reduces DNA damage
p53 ↓—DNA damage and apoptosis pathway activity ↓[27]
miR-1184BMMSCHK-2 by cispAnti-apoptotic
Anti-inflammatory
Break the G1 block
Targeting FOXO4, p27 Kip1 and CDK2[66]
miR-146bHucMSCLPS HK-2
CLP mice
Anti-inflammatoryIRAK1 ↓
NF-κB ↓
[67]
miR-486-5pHucMSCIRI micePromotes proliferation angiogenesis PTEN ↓—Akt phosphorylation[45]
miR-486-5pHucMSCIRI miceAnti-apoptotic
Anti-inflammatory
protein kinase B ↑
TNF pathway ↓
phosphatase and tensin homolog↓
[68]
miR-125b-5pHucMSCIRI mice
HK-2 (H/R)
Break the G2/M block
Anti-apoptotic
P53 ↓—CDK1 and Cyclin B1 ↑
modulation of Bcl-2 and Bax
accumulated in proximal tubules by virtue of the VLA-4 and LFA-1
[46]
miR-874-3pHucMSCUUO mice
HK-2 by cisp
Reduce necrosis
Promotes mitochondrial fusion
RIPK1
PGAM5 ↓—dephosphorylation of the S637 site of the Drp1 gene
[69]
miR-342-5pADMSCSepsis
patients with sepsis-associated AKI
CLP mice
HK-2 by LPS
Enhanced autophagy
Anti-inflammatory
Reduced BUN and SCr levels
TLR9 ↓—autophagy ↑
BUN, SCr, ↓
[41]
miR-216a-5pUSCIRI rats
HK-2 (H/R)
Promote proliferation
Angiogenesis
PTEN ↓—Akt phosphorylation[70]
miR-146a-5pUSCIRI rats
HK-2 (H/R)
Anti-inflammatory
Anti-apoptotic
Target 3′UTR of IRAK1—NF-κB ↓[52]
miR-30a-5pUSCpCisp mice
HK-2 by cisp
Anti-apoptotic
Anti-inflammatory
MAPK8 ↓[71]
miR-93-5pMacrophage (M2 > M1)CLP mice
TCMK-1 cells by LPS
Renal epithelial cell pyroptosis
Anti-inflammatory
Anti-apoptotic
TXNIP—pyroptosis in renal epithelial cell[72]
miR-590-3pTECAKI patients after cardiac surgery
HK-2 (H/R)
Autophagy miR-590-3p was highly enriched in the plasma exosomes of young AKI patients after cardiac surgery
Beclin-1 and LC3II ↑
[53]
miR-20a-5pTEC HHK-2 (H/R)
IRI mice
Promotes TEC proliferation,
Promotes mitochondrial fusion,
Attenuating necrosis
macrophages infiltration ↓[73]
miR-21-5pECFCCLP miceAnti-inflammatory
Promotes proliferation angiogenesis
RUNX1 ↓[60]
miR-486-5pECFCIRI micePromotes proliferation angiogenesisPTEN ↓—Akt phosphorylation[45]
miR-93-5pEPCHK-2 by LPS
CLP mice
Anti-apoptotic
Anti-inflammatory
Regulating KDM6BH/3K27me3/TNF-α axis[74]
miR-124UnknownCIS miceAnti-inflammatoryMCP-1 ↑[75]
miR500a3pUnknownHK-2 by cispReduce necrosisRIPK3 and MLKL ↓[76,77]
miR-150-5pUnknownHK-2 by LPS
LPS mice
Anti-apoptotic
Anti-inflammatory and oxidative stress
MEKK3/JNK pathway[78]
lncRNA TUG1USCHK-2 (H/R)
IRI mice
Reduce FerroptosisACSL4 ↑—TUG1’s repression ↓
miR-494-3p—E-cadherin and TUG1
[79]
[80]
CCR2BMMSCIRI miceInhibits macrophage recruitment activationUnknown[81]
14-3-3zHucMSCNRK-52E by cisp
Cisp Rats
AutophagyATG16L↑[82]
CD26TECIRI miceAnti-inflammatory
Promotes TEC proliferation
CXCR4, SDF1 ↓[83]
KlothoUSCAKI
rats
Anti-apoptotic
Anti-inflammatory
Promotes proliferation angiogenesis
fibrosis, monocyte infiltration ↓
SOD1 ↑
[84]
Abbreviations: BMMSC, bone marrow mesenchymal stem cell; ADMSC, adipose-derived mesenchymal stem cell; EPC, endothelial progenitor cell; Sema3A, semaphorin 3A; AKT, activated protein kinase B; ERK, extracellular-signal-regulated kinase; FOXO4, forkhead box O4; IRAK1, interleukin (IL)-1 receptor-associated kinase; PTEN, Phosphatase and tensin homolog; PGAM5, phosphoglycerate mutase 5; RIPK1, Receptor-interacting protein kinase 1; MAPK8, mitogen-activated protein kinase 8; LC3II, microtubule-associated protein 1 light chain 3 beta; RUNX1, runt-related transcription factor 1; SDF1, stromal derived factor-1; SOD1, superoxide dismutase 1; TEC H, hypoxic tubular epithelial cell; ECFC, endothelial colony-forming cell; CIS, Candida infection with sepsis; USCp, premature USC; ↓, Decreased expression; ↑, increased expression.
Table 3. Damaging role of exosomal contents in AKI.
Table 3. Damaging role of exosomal contents in AKI.
ContentExo SourceModelMechanismPathwaysReference
miR-195a-5pMacrophageCisp micePromote TEC apoptosis and damage TEC mitochondriaunknown[85]
miR-155MacrophageIRI/cisp mice
HK-2 (cisp)
NRK-52E (H/R)
Promote TEC damage, promote inflammation
limiting the telomeric dysfunction and the genomic DNA damage
TRF1, CDK12 ↓
TCF4/Wnt/β-Catenin
[86,94,95]
miR-19b-3pLPS-TECSepsis
LPS mice
Promotes inflammation, macrophage 1 polarizationNF-κB/SOCS-1[91]
miR-150-5pTECIRI
UIRI rats
NRK-52E (H/R)
Activates fibroblasts and promotes fibrosis suppressor of cytokine signaling 1 to activate fibroblast ↓[88]
miR374b-5pTECIRI micePromote Macrophage 1 polarizationTransferring miR-374b-5p[90]
miR-709PTCCisp mice
human AKI kidney
Promote TEC apoptosis and mitochondria damage TFAM ↓
miR-709 in PTCs of patients ↑
[89]
miR-23aTEC HUUO/IRI
mice and TEC
Promotes inflammation
Macrophage activation
suppression of the ubiquitin editor A20[92]
miR-30d-5pNeutrophils
mice
SepsisPromotes inflammation, macrophage 1 polarization
Promotes pyroptosis of macrophages
NLRP3—NF-κB ↑
SOCS-1 and SIRT1 ↓
[93]
Circ-FANCASepsis patient
LPS-HK2
Sepsis
LPS HK-2
Promotes inflammation, apoptosis
Inhibit proliferation (G0/G1 arrest)
sponging OXSR1[96]
Circ0001818Unknown
(Serum)
LPS-HK2Promote LPS-HK2 damagemiR-136-5p—TXNIP[97]
Abbreviations: TEC H, hypoxic tubular epithelial cell; PTCs, proximal tubular cells; TFAM, mitochondrial transcription factor A; LPS, Lipopolysaccharides, ↓, Decreased expression; ↑, increased expression.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, X.; Wang, J.; Zhang, J.; Tan, Y.; Li, Y.; Peng, Z. Exosomes Highlight Future Directions in the Treatment of Acute Kidney Injury. Int. J. Mol. Sci. 2023, 24, 15568. https://doi.org/10.3390/ijms242115568

AMA Style

Zhang X, Wang J, Zhang J, Tan Y, Li Y, Peng Z. Exosomes Highlight Future Directions in the Treatment of Acute Kidney Injury. International Journal of Molecular Sciences. 2023; 24(21):15568. https://doi.org/10.3390/ijms242115568

Chicago/Turabian Style

Zhang, Xiaoyu, Jing Wang, Jing Zhang, Yuwei Tan, Yiming Li, and Zhiyong Peng. 2023. "Exosomes Highlight Future Directions in the Treatment of Acute Kidney Injury" International Journal of Molecular Sciences 24, no. 21: 15568. https://doi.org/10.3390/ijms242115568

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