
Efficacy of exosomes in acute kidney injury treatment and the associated mechanism (Review)
- Authors:
- Published online on: March 26, 2025 https://doi.org/10.3892/mmr.2025.13503
- Article Number: 137
-
Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Acute kidney injury (AKI) involves rapid deterioration of kidney function, typically occurring over a period of hours to days, and is defined by an increase in serum creatinine levels, a decrease in urine output or both. AKI occurs in 10–15% of hospitalized patients and >50% of patients in intensive care units. Since AKI is a highly heterogeneous disease, its complex pathophysiological mechanisms are not completely understood (1). Currently, no effective treatment or prevention method is available for AKI (1,2). AKI can be caused by several drugs and toxins, such as cisplatin-induced AKI (Cis-AKI) and vancomycin-induced AKI (V-AKI). In addition, sepsis-induced AKI (S-AKI) and ischemia-reperfusion (I/R)-induced AKI (I/R-AKI) are also commonly reported (3–5). The main pathogenic mechanisms of AKI are oxidative stress and cytotoxicity caused by the accumulation of reactive oxygen species (ROS) and toxins due to dysfunction of enzyme-promoted and non-enzyme-promoted antioxidant defense systems, resulting in renal tissue damage and renal dysfunction (6–9).
Extracellular vesicles (EVs) are a heterogeneous group of membrane-bound vesicles and EVs can be divided into micro-vesicles, exosomes and apoptotic bodies according to their size, biological processes and molecular markers (10). Exosomes are an important type of EV, with a size of 40–200 nm and an average diameter of 100 nm (10,11). Exosomes originate from endosomes and are secreted and released by a variety of cells, including mesenchymal stem cells (MSCs) and neutrophils (11). Increasing evidence suggests that exosomes serve an important role in intercellular communication, and the pathways related to exosome uptake include receptor-ligand binding, membrane fusion and endocytosis (10–13). For example, proteins, microRNAs (miRNAs/miRs), circular RNAs, lipids, DNA and long non-coding RNAs (lncRNAs) are delivered to target cells to regulate cell functions (10–15). Although the potential mechanisms underlying the targeted regulation of exosomes are diverse and complex, several studies have revealed the regulatory role of exosome-supported biomolecules in renal I/R injury (12,16,17). However, further investigation of the effects of exosomes on AKI progression is necessary. Exosomes secreted by cells and substances from different sources differ in terms of their efficacy and mechanism of action on AKI, which are discussed and summarized in the present review.
Therapeutic effect of exosomes on AKI
Effects of exosomes derived from MSCs on AKI
Previous studies have revealed that MSCs have the capacity for self-renewal, differentiation, immune regulation and nutritional support, and are essential in regenerative medicine because of their ability to create a microenvironment conducive to the repair of damaged tissues (17,18). Previously, a phase 1a clinical trial demonstrated that MSCs can increase renal blood flow and the glomerular filtration rate, and reduce inflammation in the kidneys after stenosis (18–20). The therapeutic effect of MSCs is generally considered to be mediated by the secretion of various factors, including cytokines, cell growth factors and exosomes (21). Exosome-rich components are recognized as key active therapeutic ingredients for MSC-based therapies. Compared with adult stem cell therapy, exosome therapy has certain advantages, including increased consistency, enhanced potency, greater scalability of manufacturing and a wide range of sources (22,23). MSCs can be isolated from a variety of tissues, including the bone marrow, fat, umbilical cord and placenta, and extracted from substances such as dental pulp, skin, blood and urine (10,24,25).
Exosomes from different types of MSCs can promote tubular repair, alleviate tubular epithelial cell damage, inhibit inflammation, oxidative stress and apoptosis, and alleviate renal I/R injury (14,18,26–30). Previous studies have shown that the transfusion of human umbilical cord-derived MSC (huMSC) exosomal miR-874-3p into a mouse model of Cis-AKI and transfection into Cis-HK-2 reduced the activation of necroptosis, and promoted mitochondrial homeostasis and the repair of tubular epithelial cell injury by targeting the receptor-interacting serine/threonine protein kinase 1 (RIPK1)/PGAM5 pathway (27,29). Adipose-derived stem cell-derived exosomal CIRCVMA21 and miR-342-5p alleviated lipopolysaccharide (LPS)-stimulated HK-2 cell damage, inhibited oxidative stress and inflammation, increased renal function, and restored normal tissue morphology by targeting miR-16-5P and inhibiting toll-like receptor 9 (3,31). In addition, physiological homeostasis of the plasma metabolome could be maintained in a male cat model of postrenal AKI, effectively improving renal function of the cats (3,25,28,31,32). Furthermore, the exosomes miR-199a-3p and miR-199a-5p from bone marrow mesenchymal stem cells also have effects on AKI. First, miR-199a-3p inhibits apoptosis and inflammation in I/R mouse models by activating the AKT/ERK pathway. Secondly, miR-199a-5p targeted immunoglobulin protein, inhibited apoptosis and alleviated endoplasmic reticulum stress in I/R mouse models (12,14).
In addition, human urine-derived stem cell (hUSC) exosomes carrying miR-146a-5p and miR-216a-5p also play a role. First, miR-146a-5p inhibits inflammation and apoptosis and promotes renal tubular cell proliferation by targeting interleukin-1 receptor-associated kinase-1. Second, miR-216a-5p targets the PTEN/Akt signaling pathway, inhibits inflammation and oxidative stress, and reduces I/R-AKI in mice (33,34). Furthermore, klotho, a reno-protective protein expressed by hUSCs, migrates to the site of kidney injury and carries out a protective role through a similar mechanism (35). In addition, ferroptosis is a unique mode of cell death induced by iron-mediated lipid peroxidation accumulation and plasma membrane rupture. Exosomes derived from hUSCs carry the lncRNA taurine upregulated 1 and regulate achaete-scute homolog 4-mediated iron death by interacting with serine/arginine-rich splicing factor 1 to inhibit tubular cell epithelial cell damage and alleviate kidney injury (36,37). Iron overload promotes M1 macrophage activation and subsequent inflammation (38). Therefore, future work should combine anti-ferroptosis and anti-inflammatory therapy to potentially increase the therapeutic efficacy of the treatment for inflammation. In combination with immunization and other methods, the application of MSC-exosome-mediated ferroptosis in the treatment of various diseases is expected to increase (36,38–41). Future studies may explore whether exosomes serve roles in other forms of cell death, such as copper-induced cell death, in the occurrence and development of disease (42).
Although studies have shown that exosomes derived from MSCs serve a positive role in the development of AKI (3,12,14,27,31,33,34,43), evidence from animal model studies is limited. This is partly because of ethical constraints; thus, the participation of regulatory bodies, such as ethical review committees, in exosome research is key. In addition, due to the shortcomings of MSCs, such as insufficient clinical validation, unstable sources and quality, and the complex physiological structure and mechanism of the human body, when MSCs are used as drugs, it is necessary to consider whether resistance or immune reactions may occur in the process, which may prevent initial therapeutic effects and exacerbate damage (10,40,41,44) (Fig. 1; Table I).
Efficacy of exosomes derived from other cells in AKI treatment and the associated mechanisms
In addition to MSCs, exosomes secreted by other cells and substances have therapeutic effects on AKI. Exosomes from the urine of premature infants were effective against Cis-AKI (45). Exosomal miR-30a-5p serves a role by activating MAPK8. It reduces the expression of cisplatin-induced cleaved caspase-3, monocyte chemoattractant protein-1 (MCP-1) and Bax, and increases Bcl-2 expression (45). Therefore, miR-30a-5p inhibits cell inflammation and apoptosis, and serves a protective role in the kidney. In V-AKI, human urinary exosomes may serve as biomarkers for drug-induced kidney injury, and exhibit potential as biomarkers, targets and therapeutics for immunomodulatory compounds (45,46).
Exosomal miR-486-5p from endothelial colony-forming cells is effective against I/R-AKI. Exosomal miR-486-5p serves an important role by targeting the PTEN/Akt pathway to alleviate renal tubular epithelial cell (TEC) inflammation and inhibit fibrosis (47,48). In mouse models of CLP, endothelial progenitor cell-derived exosomal miR-21-5p improved kidney function by inhibiting runt-related transcription factor 1 expression, apoptosis and inflammatory responses (49).
Human amniotic epithelial cells (hAECs) are non-MSCs that are isolated from the placental amniotic epithelial membrane. hAECs respond to immunomodulatory therapy, express anti-inflammatory proteins, including IL-10 and IL-12, and have the advantages of abundant sources, large quantities and genetic stability (50). In mice subjected to cecal ligation and puncture, hAEC-derived exosomes inhibited vascular cell adhesion factor 1 through immunomodulation, thereby controlling cellular inflammation and reducing fluorescein isothiocyanate-dextran leakage. Therefore, the integrity of the endothelial monolayer and glomerular endothelial cells was protected, and the functional damage to endothelial cells induced by s-AKI was alleviated (51,52).
TECs are typically the site of injury caused by hypoxia, inflammation and toxins, and TEC injury serves a role in promoting disease progression. miR-19b-3p in exosomes from TECs activate the NF-κB signaling pathway in macrophages by targeting suppressor of cytokine-signaling-1. Increased p65 and phosphorylated-p65 protein levels increase the expression levels of MCP-1 and TNF-α, regulate the activation mechanism of M1 macrophages and promote the progression of renal tubulointerstitial inflammation facilitated by macrophages. Exosomal miR-19b-3p levels are positively associated with the severity of proteinuria (53–55). This finding is the opposite of the protective effect of mesenchymal stem cell exosomes on the kidneys reported in previous studies (33,53), and it is hypothesized that the inflammatory response in AKI may be caused by the exosome-mediated inflammatory pathway through the promotion of TEC macrophage activation. Therefore, miR-16b-3p is a potential target for the treatment of AKI (53,54).
The study also revealed that exosomes from the plasma of patients undergoing heart surgery were effective in mouse models of I/R. Exosomal miR-590-3p reduced I/R-induced oxidative stress by targeting RIPK1. Furthermore, LC3 II and Beclin-1 protein expression levels were decreased, p62 protein expression was increased, and TNF receptor-associated factor 6 could be targeted to regulate autophagy and alleviate LPS-induced AKI and podocyte apoptosis, which had a protective effect on the kidneys (55).
The therapeutic effect of cellular exosomes on S-AKI has also been investigated. Remote ischemic preconditioning (rIPC) also has a protective effect. rIPC is a beneficial stimulator triggered by transient I/R in remote tissues (2). Previous studies have revealed that rIPC has a beneficial effect on a variety of organs (56–59). After rIPC, the kidney promotes the expression of miR-21, which inhibits PTEN expression and NF-κB activity. Therefore, inhibition of HIF-1α abrogates renoprotection induced by rIPC in mice (2,56) (Fig. 2; Table II).
Novel advances in exosome therapy
Engineering exosomes
Several studies have shown that MSCs and other cell- and substance-derived exosomes have positive roles in the treatment and prevention of AKI (3,31,33,34,53,55,57). However, these exosomes are deficient in the treatment of AKI, and their therapeutic effects are limited. At present, no standard system exists for the mass production, separation and storage of exosomes. Therefore, the engineering of exosomes is necessary. This process can enhance the targeting of injured sites and treatment specificity or improve resistance to body clearance. The engineering of exosomes involves the modification of natural exosomes using biological and chemical engineering techniques, and the modification of endogenous and exogenous loads through bioengineering refers to the introduction of targeting motifs by genetically fusing membrane-bound proteins (60–62). Chemical engineering refers to the addition of substances such as antibodies, proteins and small molecules through lipid chemical reactions, membrane-bound protein chemical reactions or lipid-lipid interactions, amplifying the advantages and specific functions of various nano-delivery systems (55,60). To date, engineered exosomes have been studied for the treatment of a variety of diseases, such as Cis-AKI. huMSC-derived exosomes exhibit a low targeting ability and therapeutic specificity for tissue damage repair. Therefore, neutrophil cell membrane-engineered huMSC-sEVs (NEXs) have been developed to enhance the specificity of targeting huMSC-sEVs. NEXs effectively target the site of kidney injury, considerably reduce ROS levels, and strengthen anti-inflammatory, antioxidative stress and antiapoptotic effects (43). Exosome engineering provides a more accurate and personalized treatment outlook for AKI (10,43,62,63).
Smart nano-exosomes
Smart nano-exosomes are nano-cellular double vesicles that are present in the endosomal region of the majority of eukaryotes and the cytoplasm of several types of bacteria (64). Smart nano-exosomes are formed when multivesicular bodies are secreted together with fused plasma membranes through the exocytosis of intracellular vesicles (65). Nanomaterials with considerable pharmacokinetic, bioavailability and biodistribution properties, such as low toxicity and immunogenicity, penetrate other cells without being targeted by the immune system or toxic reactions, and enable more efficient penetration and delivery of cargoes carrying molecules (66). Thus, these materials have the potential to further elucidate complex biological responses, and represent a promising tool for the diagnosis and treatment of multiple diseases (64–66). Currently, nanoscale single-cell gene manipulation is an effective method to screen for intelligent exosomes that can be directly modified by stem cell modification to increase the ‘intelligent’ properties of exosomes (67–69). Nano-exosomes have been investigated for the treatment of viral diseases, cancer and cardiovascular diseases (70,71), but there is a gap in AKI research. Previous studies have shown that intelligent nano-exosomes are a rich source of potential biomarkers, and that their secretion into extracellular regions provides convenient conditions for the examination of body fluids such as urine and blood (72,73). Therefore, studies should explore whether they can be used as novel biomarkers for the diagnosis of AKI (68–73).
Applications of nanotechnology in AKI
Gene therapy and stem cell therapy use can improve the effectiveness of AKI treatment, but early use of stem cell therapy for AKI treatment requires research to confirm its long-term safety and assess the risk of developing fibrosis (74). Therefore, due to the rapid development of nanotechnology, nanotechnology has been used to develop early and accurate diagnostic methods and effective treatments for various kidney diseases (75). Modified TiO2 nanotube arrays, nanosensor, nanotubes, nanorod, nanospheres and iron oxide nanoparticles have been developed, enabling researchers to sensitively, accurately and conveniently diagnose and continuously monitor the status of patients with AKI using methods such as biomarker monitoring, ultrasound detection and optical imaging (76). In addition, various delivery systems, including polymer nanoparticles, organic nanoparticles, inorganic nanoparticles, lipid nanoparticles and hydrogels, are currently available (74–76). Polymer nanoparticles can be modified with a variety of components that precisely control the characteristics of the nanoparticles, increasing the targeting ability and anti-inflammatory and antioxidant effects of the delivered exosomes (74,77). Inorganic nanoparticles, which can effectively clear ROS, provide a novel option for the treatment of various diseases caused by oxidative stress (78,79). Lipid nanoparticles use biocompatible and physiologically tolerated lipids to improve the solubility of hydrophobic drugs and enhance biocompatibility, making it easier for loaded drugs or exosomes to reach damaged sites, improving therapeutic efficacy and reducing systemic side effects (80,81).
In addition, hydrogels are also the focus of current research. Hydrogels are hydrophilic gels with 3D network structures composed of soft, biocompatible and biodegradable materials. Due to their advantages of high biocompatibility, injection flexibility and controllability, hydrogels can be easily injected (82,83). For example, hydrogels loaded with osteoinductive dental pulp stem cell-derived EVs enhance bone tissue remodeling and adhesion, and promote bone growth (82). The use of hydrogels as delivery systems has also been applied to the treatment of AKI. Studies have shown that the delivery of MSC-EVs to the injured site through hydrogels can increase the bioavailability of MSC-EVs, effectively alleviate the induced inflammatory response and reduce the remodeling of the extracellular matrix, which is expected to become an innovative treatment method for AKI (74,83–85). Finally, nanocarriers can assist in the delivery of small interfering RNAs between cells, increase their plasma stability and targeting, prolong their circulation time, and modulate their pharmacokinetics (86). Studies have prepared megalin-targeting polycationic polymyxin-polyethylenimine/DNA-nanoplexes for kidney-targeted gene delivery to improve gene transfection efficiency. This opens novel avenues for kidney gene therapy for AKI (68,87,88) (Fig. 3; Table III).
Conclusion
The present review summarizes the role and mechanism of exosomes derived from different cells and substances in AKI. Exosomes serve an important role in the occurrence and development of AKI by regulating certain pathways or targeting specific sites. For example, the exosome miR-874-3p from huMSC targets RIPK1/PGAM5 to inhibit inflammation and apoptosis. In addition, the exosome miR-216a-5p derived from hUSCs promotes the proliferation of HK-2 cells by targeting the PTEN/Akt signaling pathway. The therapeutic effects of exosomes from different sources on different pathological characteristics of AKI are also discussed by examining their reparative effects on renal tubule injury, and anti-inflammatory and anti-apoptotic effects. The roles of certain exosomes in promoting AKI are also discussed. For instance, the exosome miR-19b-3p derived from TEC can promote the activation of M1 macrophages and promote inflammation, thus promoting the development of AKI. Furthermore, future trends in the development of exosome therapy and some possible challenges, including ethical issues and unclear mechanisms of exosome action on damaged sites, are also discussed. Finally, the present review discusses the current development trends of exosome therapy and reveals that there are still several issues that remain to be solved. For instance, the field of engineering exosomes, nano-exosomes and gene-edited exosomes still needs to be explored, and the utilization of nanotechnology in these domains remains relatively rare. Unique therapeutic cargoes with biological and clinical therapeutic effects may be loaded into exosomes derived from various cells and substances, and further exploration of the pathogenesis of AKI and the mechanism of action of exosomes is needed. Subsequently, exosome therapy may provide more possibilities in regenerative medicine and the treatment of some diseases, and provides novel options with potential for the treatment of AKI.
Acknowledgements
Not applicable.
Funding
The present study was supported by the National Natural Science Foundation of China (grant no. 82070699), the Project of Medical Staff Technical Elevation of the Xijing Hospital of the Fourth Military Medical University (grant no. 2023XJSZ06) and the Discipline Promotion Project of the Xijing Hospital of the Fourth Military Medical University (grant no. XJZT24LY26).
Availability of data and materials
Not applicable.
Authors' contributions
ZZ and MB conceived the present study and revised the manuscript. ZZ and MB wrote and revised the manuscript, and constructed and revised the figures. LS revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Glossary
Abbreviations
Abbreviations:
AKI |
acute kidney injury |
S-AKI |
sepsis-induced AKI |
Cis-AKI |
cisplatin-induced AKI |
V-AKI |
vancomycin-induced AKI |
I/R-AKI |
ischemia-reperfusion-induced AKI |
ROS |
reactive oxygen species |
EV |
extracellular vesicle |
MSC |
mesenchymal stem cell |
huMSC |
human umbilical cord-derived MSC |
hAEC |
human amniotic epithelial cell |
TEC |
renal tubular epithelial cell |
rIPC |
remote ischemic preconditioning |
NEXs |
neutrophil cell membrane-engineered huMSC-secreted EVs |
References
Scholz H, Boivin FJ, Schmidt-Ott KM, Bachmann S, Eckardt KU, Scholl UI and Persson PB: Kidney physiology and susceptibility to acute kidney injury: Implications for renoprotection. Nat Rev Nephrol. 17:335–349. 2021. View Article : Google Scholar : PubMed/NCBI | |
Pan T, Jia P, Chen N, Fang Y, Liang Y, Guo M and Ding X: Delayed Remote ischemic preconditioning confersrenoprotection against septic acute kidney injury via exosomal miR-21. Theranostics. 9:405–423. 2019. View Article : Google Scholar : PubMed/NCBI | |
He Y, Li X and Huang B, Yang Y, Luo N, Song W and Huang B: Exosomal circvma21 derived from Adipose-derived stem cells alleviates Sepsis-induced acute kidney injury by targeting Mir-16-5p. Shock. 60:419–426. 2023.PubMed/NCBI | |
Jorgensen SCJ, Murray KP, Lagnf AM, Melvin S, Bhatia S, Shamim MD, Smith JR, Brade KD, Simon SP, Nagel J, et al: A multicenter evaluation of Vancomycin-associated acute kidney injury in hospitalized patients with acute bacterial skin and skin structure infections. Infect Dis Ther. 9:89–106. 2020. View Article : Google Scholar : PubMed/NCBI | |
Fathy N, Farouk S, Sayed RH and Fahim AT: Ezetimibe ameliorates cisplatin-induced nephrotoxicity: A novel therapeutic approach via modulating AMPK/Nrf2/TXNIP signaling. FASEB J. 38:e233822024. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Hu C, Zhai P, Zhang J, Jiang J, Suo J, Hu B, Wang J, Weng X, Zhou X, et al: Fibroblastic reticular cell-derived exosomes are a promising therapeutic approach for septic acute kidney injury. Kidney Int. 105:508–523. 2024. View Article : Google Scholar : PubMed/NCBI | |
Huang TY, Chien MS and Su WT: Therapeutic potential of pretreatment with exosomes derived from stem cells from the apical papilla against Cisplatin-induced acute kidney injury. Int J Mol Sci. 23:57212022. View Article : Google Scholar : PubMed/NCBI | |
Guo G, Wang Y, Kou W and Gan H: Identifying the molecular mechanisms of sepsis-associated acute kidney injury and predicting potential drugs. Front Genet. 13:10622932022. View Article : Google Scholar : PubMed/NCBI | |
Chen L, Xu JY and Tan HB: LncRNA TUG1 regulates the development of ischemia-reperfusion mediated acute kidney injury through miR-494-3p/E-cadherin axis. J Inflamm (Lond). 18:122021. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Wang J, Zhang J, Tan Y, Li Y and Peng Z: Exosomes highlight future directions in the treatment of acute kidney injury. Int J Mol Sci. 24:155682023. View Article : Google Scholar : PubMed/NCBI | |
Jiao Y, Zhang T, Zhang C, Ji H, Tong X, Xia R, Wang W, Ma Z and Shi X: Exosomal miR-30d-5p of neutrophils induces M1 macrophage polarization and primes macrophage pyroptosis in sepsis-related acute lung injury. Crit Care. 25:3562021. View Article : Google Scholar : PubMed/NCBI | |
Wang C, Zhu G, He W, Yin H, Lin F, Gou X and Li X: BMSCs protect against renal ischemia-reperfusion injury by secreting exosomes loaded with miR-199a-5p that target BIP to inhibit endoplasmic reticulum stress at the very early reperfusion stages. FASEB J. 33:5440–5456. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang W, Zhou B, Yang X, Zhao J, Hu J, Ding Y, Zhan S, Yang Y, Chen J, Zhang F, et al: Exosomal circEZH2_005, an intestinal injury biomarker, alleviates intestinal ischemia/reperfusion injury by mediating Gprc5a signaling. Nat Commun. 14:5437–5453. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhu G, Pei L, Lin F, Yin H, Li X, He W, Liu N and 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. 234:23736–23749. 2019. View Article : Google Scholar : PubMed/NCBI | |
Herman M, Randall GW, Spiegel JL, Maldonado DJ and Simoes S: Endo-lysosomal dysfunction in neurodegenerative diseases: Opinion on current progress and future direction in the use of exosomes as biomarkers. Philos Trans R Soc Lond B Biol Sci. 379:202203872024. View Article : Google Scholar : PubMed/NCBI | |
Ma Y, Brocchini S and Williams GR: Extracellular Vesicle-embedded materials. J Control Release. 361:280–296. 2023. View Article : Google Scholar : PubMed/NCBI | |
Canney M, Clark EG and Hiremath S: Biomarkers in acute kidney injury: On the cusp of a new era? J Clin Invest. 133:e1714312023. View Article : Google Scholar : PubMed/NCBI | |
Cao JY, Wang B, Tang TT, Wen Y, Li ZL, Feng ST, Wu M, Liu D, Yin D, Ma KL, et al: Exosomal miR-125b-5p deriving from mesenchymal stem cells promotes tubular repair by suppression of p53 in ischemic acute kidney injury. Theranostics. 11:5248–5266. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wu YL, Li HF, Chen HH and Lin H: MicroRNAs as biomarkers and therapeutic targets in Inflammation- and Ischemia-Reperfusion-related acute renal injury. Int J Mol Sci. 21:67382020. View Article : Google Scholar : PubMed/NCBI | |
Uccelli A, Moretta L and Pistoia V: Mesenchymal stem cells in health and disease. Nat Rev Immunol. 8:726–736. 2008. View Article : Google Scholar : PubMed/NCBI | |
Li X, Li C, Zhang L, Wu M, Cao K, Jiang F, Chen D, Li N and Li W: The significance of exosomes in the development and treatment of hepatocellular carcinoma. Mol Cancer. 19:12020. View Article : Google Scholar : PubMed/NCBI | |
Vicencio JM, Yellon DM, Sivaraman V, Das D, Boi-Doku C, Arjun S, Zheng Y, Riquelme JA, Kearney J, Sharma V, et al: Plasma exosomes protect the myocardium from ischemia-reperfusion injury. J Am Coll Cardiol. 65:1525–1236. 2015. View Article : Google Scholar : PubMed/NCBI | |
Damania A, Jaiman D, Teotia AK and Kumar A: Mesenchymal stromal Cell-derived Exosome-rich fractionated secretome confers a hepatoprotective effect in liver injury. Stem Cell Res Ther. 9:312018. View Article : Google Scholar : PubMed/NCBI | |
Huang Y and Yang L: Mesenchymal stem cells and extracellular vesicles in therapy against kidney diseases. Stem Cell Res Ther. 12:219–230. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gao F, Zuo B, Wang Y, Li S, Yang J and Sun D: Protective function of exosomes from adipose tissue-derived mesenchymal stem cells in acute kidney injury through SIRT1 pathway. Life Sci. 255:1177192020. View Article : Google Scholar : PubMed/NCBI | |
Elahi FM, Farwell DG, Nolta JA and Anderson JD: Preclinical translation of exosomes derived from mesenchymal stem/stromal cells. Stem Cells. 38:15–21. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yu Y, Chen M, Guo Q, Shen L, Liu X, Pan J, Zhang Y, Xu T, Zhang D and 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. 28:1202023. View Article : Google Scholar | |
Zhang W, Zhang J and Huang H: Exosomes from adipose-derived stem cells inhibit inflammation and oxidative stress in LPS-acute kidney injury. Exp Cell Res. 420:1133322022. View Article : Google Scholar : PubMed/NCBI | |
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. 4:342013. View Article : Google Scholar : PubMed/NCBI | |
Xing Z, Zhao C, Liu H and Fan Y: endothelial progenitor Cell-derived extracellular vesicles: A novel candidate for regenerative medicine and disease treatment. Adv Healthc Mater. 9:e20002552020. View Article : Google Scholar : PubMed/NCBI | |
Liu W, Hu C, Zhang B, Li M, Deng F and 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. 25:102023. View Article : Google Scholar : PubMed/NCBI | |
Li W, Wang W, He X, Liao Z, Aierken A, Hua J, Wang Y, Lu D and 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. 13:3792022. View Article : Google Scholar : PubMed/NCBI | |
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. 10:9561–9578. 2020. View Article : Google Scholar : PubMed/NCBI | |
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. 8:6105872020. View Article : Google Scholar : PubMed/NCBI | |
Grange C, Papadimitriou E, Dimuccio V, Pastorino C, Molina J, O'Kelly R, Niedernhofer LJ, Robbins PD, Camussi G and Bussolati B: Urinary extracellular vesicles carrying klotho improve the recovery of renal function in an acute tubular injury model. Mol Ther. 28:490–502. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sun Z, Wu J, Bi Q and 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. 13:2972022. View Article : Google Scholar : PubMed/NCBI | |
Thapa K, Singh TG and Kaur A: Targeting ferroptosis in ischemia/reperfusion renal injury. Naunyn Schmiedebergs Arch Pharmacol. 395:1331–1341. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhou Y, Que KT, Zhang Z, Yi ZJ, Zhao PX, You Y, Gong JP and Liu ZJ: Iron overloaded polarizes macrophage to proinflammation phenotype through ROS/acetyl-p53 pathway. Cancer Med. 7:4012–4022. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wallach D, Kang TB and Kovalenko A: Concepts of tissue injury and cell death in inflammation: A historical perspective. Nat Rev Immunol. 14:51–59. 2014. View Article : Google Scholar : PubMed/NCBI | |
Liu L, Ye Y, Lin R, Liu T, Wang S, Feng Z, Wang X, Cao H, Chen X, Miao J, et al: Ferroptosis: A promising candidate for exosome-mediated regulation in different diseases. Cell Commun Signal. 22:62024. View Article : Google Scholar : PubMed/NCBI | |
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI | |
Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, et al: Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 375:1254–1261. 2022. View Article : Google Scholar : PubMed/NCBI | |
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 HuMSC sEVs alleviate cisplatin-induced AKI by enhancing cellular uptake and targeting. J Nanobiotechnology. 20:3532022. View Article : Google Scholar : PubMed/NCBI | |
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. 7:1674–1688. 2017. View Article : Google Scholar : PubMed/NCBI | |
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. 13:1650–1665. 2022. View Article : Google Scholar : PubMed/NCBI | |
Awdishu L, Le A, Amato J, Jani V, Bal S, Mills RH, Carrillo-Terrazas M, Gonzalez DJ, Tolwani A, Acharya A, et al: Urinary exosomes identify inflammatory pathways in vancomycin associated acute kidney injury. Int J Mol Sci. 22:27842021. View Article : Google Scholar : PubMed/NCBI | |
Vinas JL, Burger D, Zimpelmann J, Haneef R, Knoll W, Campbell P, Gutsol A, Carter A, Allan DS and Burns KD: Transfer of microRNA-486-5p from human endothelial colony forming cell-derived exosomes reduces ischemic kidney injury. Kidney Int. 90:1238–1250. 2016. View Article : Google Scholar : PubMed/NCBI | |
Burger D, Vinas JL, Akbari S, Dehak H, Knoll W, Gutsol A, Carter A, Touyz RM, Allan DS and Burns KD: Human endothelial colony-forming cells protect against acute kidney injury: Role of exosomes. Am J Pathol. 185:2309–2323. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Huang H, Liu W, Liu S, Wang XY, Diao ZL, Zhang AH, Guo W, Han X, Dong X and Katilov O: Endothelial progenitor cells-derived exosomal microRNA-21-5p alleviates sepsis-induced acute kidney injury by inhibiting RUNX1 expression. Cell Death Dis. 12:3352021. View Article : Google Scholar : PubMed/NCBI | |
Keung C, Nguyen TC, Lim R, Gerstenmaier A, Sievert W and Moore GT: Local fistula injection of allogeneic human amnion epithelial cells is safe and well tolerated in patients with refractory complex perianal Crohn's disease: A phase I open label study with long-term follow up. EBioMedicine. 98:10487992023. View Article : Google Scholar : PubMed/NCBI | |
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 (Lausanne). 9:8296062022. View Article : Google Scholar : PubMed/NCBI | |
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. 9:7520532021. View Article : Google Scholar : PubMed/NCBI | |
Lv LL, Feng Y, Wu M, Wang B, Li ZL, Zhong X, Wu WJ, Chen J, Ni HF, Tang TT, et al: Exosomal miRNA-19b-3p of tubular epithelial cells promotes M1 macrophage activation in kidney injury. Cell Death Differ. 27:210–226. 2020. View Article : Google Scholar : PubMed/NCBI | |
Guo C, Cui Y, Jiao M, Yao J, Zhao J, Tian Y, Dong J and Liao L: Crosstalk between proximal tubular epithelial cells and other interstitial cells in tubulointerstitial fibrosis after renal injury. Front Endocrinol (Lausanne). 14:12563752023. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Zhang C, Du Y, Yang X, Liu M, Yang W, Lei G and 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 (Engl). 135:2467–2477. 2022.PubMed/NCBI | |
Ganesh A and Testai FD: Remote ischemic conditioning for acute ischemic stroke: Does stroke etiology matter? Stroke. 55:880–882. 2024. View Article : Google Scholar : PubMed/NCBI | |
Han R, Yang X, Ji X and Zhou B: Remote ischemic preconditioning prevents high-altitude cerebral edema by enhancing glucose metabolic reprogramming. CNS Neurosci Ther. 30:e700262024. View Article : Google Scholar : PubMed/NCBI | |
Torregroza C, Gnaegy L, Raupach A, Stroethoff M, Feige K, Heinen A, Hollmann MW and Huhn R: Influence of hyperglycemia and diabetes on cardioprotection by humoral factors released after remote ischemic preconditioning (RIPC). Int J Mol Sci. 22:88802021. View Article : Google Scholar : PubMed/NCBI | |
Mukai A, Suehiro K, Kimura A, Fujimoto Y, Funao T, Mori T and Nishikawa K: Protective effects of remote ischemic preconditioning against spinal cord ischemia-reperfusion injury in rats. J Thorac Cardiovasc Surg. 163:e137–e156. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Liu X, Wang B, Sun H, Ren Y and Zhang H: Compounding engineered mesenchymal stem cell-derived exosomes: A potential rescue strategy for retinal degeneration. Biomed Pharmacother. 173:1164242024. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Huo Y, Zhao C, Liu H, Shao Y, Zhu C, An L, Chen X and Chen Z: Engineered exosomes with enhanced stability and delivery efficiency for glioblastoma therapy. J Control Release. 368:170–183. 2024. View Article : Google Scholar : PubMed/NCBI | |
Donoso-Quezada J, Ayala-Mar S and Gonzalez-Valdez J: State-of-the-art exosome loading and functionalization techniques for enhanced therapeutics: A review. Crit Rev Biotechnol. 40:804–820. 2020. View Article : Google Scholar : PubMed/NCBI | |
Piffoux M, Volatron J, Cherukula K, Aubertin K, Wilhelm C, Silva AKA and Gazeau F: Engineering and loading therapeutic extracellular vesicles for clinical translation: A data reporting frame for comparability. Adv Drug Deliv Rev. 178:1139722021. View Article : Google Scholar : PubMed/NCBI | |
Mousavi SM, Hashemi SA, Gholami A, Kalashgrani MY, Vijayakameswara Rao N, Omidifar N, Hsiao WW, Lai CW and Chiang WH: Plasma-enabled smart nanoexosome platform as emerging immunopathogenesis for clinical viral infection. Pharmaceutics. 14:10542022. View Article : Google Scholar : PubMed/NCBI | |
Tran PHL, Wang T, Yin W, Tran TTD, Nguyen TNG, Lee BJ and Duan W: Aspirin-loaded nanoexosomes as cancer therapeutics. Int J Pharm. 572:1187862019. View Article : Google Scholar : PubMed/NCBI | |
Ji P, Yang Z, Li H, Wei M, Yang G, Xing H and Li Q: Smart exosomes with lymph node homing and immune-amplifying capacities for enhanced immunotherapy of metastatic breast cancer. Mol Ther Nucleic Acids. 26:987–996. 2021. View Article : Google Scholar : PubMed/NCBI | |
Latifkar A, Hur YH, Sanchez JC, Cerione RA and Antonyak MA: New insights into extracellular vesicle biogenesis and function. J Cell Sci. 132:jcs2224062019. View Article : Google Scholar : PubMed/NCBI | |
Guo M, Ge X, Wang C, Yin Z, Jia Z, Hu T, Li M, Wang D, Han Z, Wang L, et al: Intranasal delivery of Gene-edited microglial exosomes improves neurological outcomes after intracerebral hemorrhage by regulating neuroinflammation. Brain Sci. 13:6392023. View Article : Google Scholar : PubMed/NCBI | |
Hyun J, Eom J, Im J, Kim YJ, Seo I, Kim SW, Im GB, Kim YH, Lee DH, Park HS, et al: Fibroblast function recovery through rejuvenation effect of nanovesicles extracted from human adipose-derived stem cells irradiated with red light. J Control Release. 368:453–465. 2024. View Article : Google Scholar : PubMed/NCBI | |
Xiong J, Liu Z, Jia L, Sun Y, Guo R, Xi T, Li Z, Wu M, Jiang H and Li Y: Bioinspired engineering ADSC nanovesicles thermosensitive hydrogel enhance autophagy of dermal papilla cells for androgenetic alopecia treatment. Bioact Mater. 36:112–125. 2024.PubMed/NCBI | |
Tan X, Zhang J, Heng Y, Chen L, Wang Y, Wu S, Liu X, Xu B, Yu Z and Gu R: Locally delivered hydrogels with controlled release of nanoscale exosomes promote cardiac repair after myocardial infarction. J Control Release. 368:303–317. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chen Z, Hu F, Xiang J, Zhou X, Wu B, Fan B, Tang H, Liu B and Chen L: Mesoporous microneedles enabled localized controllable delivery of stimulator of interferon gene agonist nanoexosomes for FLASH radioimmunotherapy against breast cancer. ACS Appl Mater Interfaces. 16:58180–58190. 2024. View Article : Google Scholar : PubMed/NCBI | |
Tan A, Rajadas J and Seifalian AM: Exosomes as nano-theranostic delivery platforms for gene therapy. Adv Drug Deliv Rev. 65:357–367. 2013. View Article : Google Scholar : PubMed/NCBI | |
Zhao Y, Pu M, Wang Y, Yu L, Song X and He Z: Application of nanotechnology in acute kidney injury: From diagnosis to therapeutic implications. J Control Release. 336:233–251. 2021. View Article : Google Scholar : PubMed/NCBI | |
Sun T, Jiang D, Rosenkrans ZT, Ehlerding EB, Ni D, Qi C, Kutyreff CJ, Barnhart TE, Engle JW, Huang P and Cai W: A Melanin-based natural antioxidant defense nanosystem for theranostic application in acute kidney injury. Adv Funct Mater. 29:10.1002/adfm.201904833. 2019. View Article : Google Scholar : PubMed/NCBI | |
Mi L, Wang P, Yan J, Qian J, Lu J, Yu J, Wang Y, Liu H, Zhu M, Wan Y and Liu S: A novel photoelectrochemical immunosensor by integration of nanobody and TiO2 nanotubes for sensitive detection of serum cystatin C. Anal Chim Acta. 902:107–114. 2016. View Article : Google Scholar : PubMed/NCBI | |
Rubio-Navarro A, Carril M, Padro D, Guerrero-Hue M, Tarin C, Samaniego R, Cannata P, Cano A, Villalobos JM, Sevillano ÁM, et al: CD163-macrophages are involved in Rhabdomyolysis-induced kidney injury and may be detected by MRI with targeted Gold-coated iron oxide nanoparticles. Theranostics. 6:896–914. 2016. View Article : Google Scholar : PubMed/NCBI | |
Anwar M, Muhammad F, Akhtar B, Ur Rehman S and Saleemi MK: Nephroprotective effects of curcumin loaded chitosan nanoparticles in cypermethrin induced renal toxicity in rabbits. Environ Sci Pollut Res Int. 27:14771–14779. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yu H, Jin F, Liu D, Shu G, Wang X, Qi J, Sun M, Yang P, Jiang S, Ying X and Du Y: ROS-responsive nano-drug delivery system combining mitochondria-targeting ceria nanoparticles with atorvastatin for acute kidney injury. Theranostics. 10:2342–2357. 2020. View Article : Google Scholar : PubMed/NCBI | |
Qin Y, Rouatbi N, Wang JT, Baker R, Spicer J, Walters AA and Al-Jamal KT: Plasmid DNA ionisable lipid nanoparticles as non-inert carriers and potent immune activators for cancer immunotherapy. J Control Release. 369:251–265. 2024. View Article : Google Scholar : PubMed/NCBI | |
Koo J, Lim C and Oh KT: Recent advances in intranasal administration for Brain-targeting delivery: A comprehensive review of Lipid-based nanoparticles and Stimuli-responsive gel formulations. Int J Nanomedicine. 19:1767–1807. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Wei X, He X, Xiao S, Shi Q, Chen P, Lee J, Guo X, Liu H and Fan Y: Osteoinductive dental pulp stem Cell-derived extracellular Vesicle-loaded multifunctional hydrogel for bone regeneration. ACS Nano. 18:8777–8797. 2024. View Article : Google Scholar : PubMed/NCBI | |
Peng J, Yang T, Chen S, Deng N, Luo X, Liao R and Su B: Utilization of hydrogels in mesenchymal stem cell-based therapy for kidney diseases. Tissue Eng Part B Rev. 30:315–326. 2024. View Article : Google Scholar : PubMed/NCBI | |
Han DS, Erickson C, Hansen KC, Kirkbride-Romeo L, He Z, Rodell CB and Soranno DE: Mesenchymal stem cells delivered locally to Ischemia-reperfused kidneys via injectable hyaluronic acid hydrogels decrease extracellular matrix remodeling 1 month after injury in male mice. Cells. 12:17712023. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Shang Y, Chen X, Wang Z, Zhu D, Liu Y, Zhang C, Chen P, Wu J, Wu L, et al: Delivery of MSCs with a hybrid β-Sheet peptide hydrogel consisting IGF-1C domain and D-Form peptide for acute kidney injury therapy. Int J Nanomedicine. 15:4311–4324. 2020. View Article : Google Scholar : PubMed/NCBI | |
Xue HY and Wong HL: Targeting megalin to enhance delivery of anti-clusterin small-interfering RNA nanomedicine to chemo-treated breast cancer. Eur J Pharm Biopharm. 81:24–32. 2012. View Article : Google Scholar : PubMed/NCBI | |
Oroojalian F, Rezayan AH, Mehrnejad F, Nia AH, Shier WT, Abnous K and Ramezani M: Efficient megalin targeted delivery to renal proximal tubular cells mediated by modified-polymyxin B-polyethylenimine based nano-gene-carriers. Mater Sci Eng C Mater Biol Appl. 79:770–782. 2017. View Article : Google Scholar : PubMed/NCBI | |
Oroojalian F, Rezayan AH, Shier WT, Abnous K and Ramezani M: Megalin-targeted enhanced transfection efficiency in cultured human HK-2 renal tubular proximal cells using aminoglycoside-carboxyalkyl-polyethylenimine-containing nanoplexes. Int J Pharm. 523:102–120. 2017. View Article : Google Scholar : PubMed/NCBI |