- Review
- Open access
- Published:
Mesenchymal stem cell-derived exosomes: an emerging therapeutic strategy for hepatic ischemia-reperfusion injury
Stem Cell Research & Therapy volume 16, Article number: 178 (2025)
Abstract
Hepatic ischemia-reperfusion injury (HIRI) severely threatens the success rates of liver surgery and transplantation. Its complex pathological process involves multiple factors such as oxidative stress, inflammatory responses, and ferroptosis, creating an urgent need for new therapeutic strategies. Exosomes derived from mesenchymal stem cells (MSCs) are emerging as a next-generation acellular therapeutic approach. With their outstanding immune-regulatory capabilities, significant reparative functions, and good biocompatibility, they are leading innovations in the field of HIRI treatment. This article provides a systematic comparison of the therapeutic characteristics of MSC-derived exosomes(MSC-EXOs) from four different sources: adipose tissue, bone marrow, umbilical cord, and induced pluripotent stem cells. Although the clinical translation of MSC-EXOs still faces challenges such as variations in isolation methods, large-scale production, and safety assessments, their remarkable therapeutic effects and vast application potential signal the arrival of a new era of precision treatment for HIRI. This review not only provides a comprehensive theoretical foundation to promote the clinical application of MSC-EXOs but also opens up innovative research directions in the field of regenerative medicine.
Introduction
In recent years, the high incidence of liver injury, liver tumors, and related chronic diseases (such as viral hepatitis and fatty liver disease) has driven the demand for liver resection surgeries [1,2,3]. However, the use of portal vein occlusion techniques to reduce intraoperative bleeding inevitably leads to HIRI [4, 5]. Due to the liver’s unique dual blood supply and complex anatomical structure, ischemia-reperfusion injury(IRI) has become a common challenge in liver surgery [6, 7]. This issue is particularly severe in patients with insufficient liver function reserve, as they are more susceptible to damage, significantly lowering postoperative prognosis and quality of life [8, 9]. The pathological process of HIRI is mainly divided into two stages: the ischemic phase and the reperfusion phase [10,11,12,13,14]. The ischemic phase typically involves a lack of liver blood supply, leading to tissue hypoxia and cellular damage [15]. The reperfusion phase occurs when blood flow is restored, triggering the complement cascade reaction, which subsequently leads to secondary injury [16, 17]. Currently, although the main clinical treatments for HIRI include pharmacological and surgical therapies [18,19,20], patients may still experience adverse reactions triggered by chemotherapy. Additionally, the high risks and costs associated with surgical treatment may result in unsatisfactory therapeutic outcomes for HIRI [11]. Therefore, the development of safe and effective treatment strategies has become an urgent clinical issue. MSC-EXOs, as a novel acellular therapeutic strategy, have become a research hotspot due to their unique biological properties. This article systematically compares, for the first time, the mechanisms and protective effects of MSC-EXOs from four different sources—adipose tissue, bone marrow, umbilical cord, and induced pluripotent stem cells (iPSCs)—in the treatment of HIRI. Additionally, we focus on the key challenges faced by the clinical translation of MSC-EXOs, such as the standardization of isolation methods, large-scale production, and safety assessment, and propose corresponding optimization strategies to provide new insights for promoting the clinical application of MSC-EXOs in the treatment of HIRI.
Mechanisms of HIRI and current therapeutic strategies
HIRI is a severe complication that occurs during surgeries for end-stage liver diseases, such as liver transplantation and liver resection. It can easily lead to acute liver injury, liver failure, and even patient death [11, 21, 22]. Severe HIRI may result in irreversible damage, trigger multiple organ dysfunction, and negatively affect patient prognosis and survival rates. The pathophysiological mechanisms of HIRI are highly complex, and current research has proposed potential mechanisms, including mitochondrial damage, oxidative stress imbalance, abnormal cell death, excessive activation of immune cells, intracellular inflammation dysregulation, and microcirculation dysfunction [11, 22,23,24]. With the progress of research, developing effective clinical prevention and treatment strategies has become possible. At present, the main treatment methods for HIRI include pharmacological interventions, surgical treatment, and machine perfusion [24,25,26,27] (Fig. 1).
The diagram shows the main factors and interventions related to HIRI. The core factors of HIRI include inflammatory dysregulation, microcirculatory dysfunction, mitochondrial damage, immune cell overactivation, oxidative stress, and abnormal cell death. Meanwhile, interventions for HIRI include surgical treatment, machine perfusion, and drug intervention
In terms of pharmacological interventions, antioxidants (such as N-acetylcysteine), anti-inflammatory drugs (such as steroids), and immunomodulators (such as cyclosporine A) have shown certain therapeutic effects in basic research by scavenging free radicals, inhibiting inflammation, and regulating the immune system [22, 28,29,30,31,32]. However, these drugs lack sufficient validation in clinical trials and have potential side effects, which limits their practical application. Surgical treatment mainly involves traditional surgical procedures, while ischemic preconditioning (IPC) and ischemic postconditioning (IPostC) are commonly used auxiliary interventions during the perioperative period to help alleviate IRI during surgery [20, 33, 34]. Traditional surgery is primarily used to repair liver damage, such as removing the damaged part or improving blood flow. However, these methods have limited long-term protective effects against IRI after surgery. IPC involves a brief period of ischemia and reperfusion of the liver before surgery, which activates the body’s self-protection mechanisms to reduce postoperative damage [35]. In contrast, IPostC is performed during the reperfusion phase after ischemia, aiming to reduce reperfusion injury by briefly interrupting blood flow. Although IPostC has shown some benefits in studies, its effectiveness depends on precise timing and implementation conditions.
Despite the benefits shown by IPC and IPostC in studies, they face significant challenges. Their effectiveness is limited by the intervention time window and individual patient factors, such as underlying diseases (e.g., hypertension, diabetes), especially in cases of acute ischemia [36, 37]. Additionally, these strategies are typically more suitable for planned surgeries rather than emergency surgeries or severe injury cases. Improving the effectiveness of these methods remains a key area of research [38,39,40]. Machine perfusion, as an emerging organ preservation method, helps reduce ischemic damage by simulating physiological conditions. Research has shown that machine perfusion can provide oxygen, nutrients, and remove metabolic byproducts, thereby maintaining organ vitality and prolonging preservation time [41, 42]. However, machine perfusion requires advanced equipment and technology, with high operational costs [43, 44]. Moreover, its long-term stability and standardized processes have not been fully established, limiting its widespread application in clinical settings.
With the development of regenerative medicine, cell therapy has gradually become a hotspot in HIRI research. Among these studies, MSCs have gained widespread attention due to their anti-inflammatory, anti-apoptotic, and tissue-repair-promoting effects. However, cell therapy still faces many challenges in clinical translation, including low graft cell survival, poor functional stability, and potential immune rejection issues [45,46,47,48]. In this context, MSC-EXOs have emerged as a novel acellular therapeutic strategy, offering new hope for the treatment of HIRI.
Functions and application potential of MSCs and their derived exosomes
MSCs are a type of stem cell with multi-lineage differentiation potential and strong immune-regulatory functions [49,50,51]. In recent years, they have become an emerging therapeutic option to mitigate IRI due to their secretion of paracrine factors and promotion of tissue repair [52,53,54]. In particular, exosomes secreted by MSCs, as an important subtype of extracellular vesicles, exhibit unique therapeutic advantages. MSC-EXOs are nanovesicles with a diameter ranging from 30 to 200 nanometers [55], rich in specific surface markers (such as CD9, CD63, and CD81) [56], and carrying various bioactive molecules, including proteins, nucleic acids (e.g., miRNA, lncRNA), lipids, and metabolites [57, 58]. These exosomes can precisely deliver bioactive substances through endocytosis, ligand-receptor binding, or membrane fusion [59,60,61,62].
In the treatment of HIRI, MSC-EXOs significantly improve liver damage through anti-inflammatory, antioxidative, cell death regulation, and tissue repair-promoting effects(Fig. 2). Compared with traditional MSC therapy, MSC-EXOs have notable advantages: as an acellular therapeutic option, they avoid the risks associated with cell transplantation (e.g., tumorigenesis) [63, 64], exhibit lower immunogenicity [65, 66], can be produced on a large scale [67, 68], and offer flexible routes of administration [69, 70], making them suitable for clinical use. MSC-EXOs have broad application prospects in HIRI treatment, applicable to both acute and chronic liver injuries [71,72,73], and can be combined with other therapeutic strategies [65, 74,75,76]. Through engineering modifications, the targeting ability of MSC-EXOs can be further enhanced, paving the way for the development of novel drug delivery systems and smart drug delivery strategies.
Recent studies have shown that Huc-MSC-exosomes(hucMSC-EXOs) modified with HSTP1 can specifically bind to activated hepatic stellate cells (aHSC), thereby improving liver fibrosis [77]. Additionally, exosomes derived from adipose-derived mesenchymal stem cells(AMSCs), modified with miR-122, also play a significant role in controlling liver injury [78]. Another study pointed out that by engineering MSC-Exos and adding quercetin and vitamin A, the therapeutic effects of exosomes were enhanced, achieving selective targeting of the liver and effectively reducing the senescence response induced by acute liver injury [79].
In personalized treatment, appropriate exosome sources can be selected based on the patient’s specific condition, and administration schemes can be optimized to achieve precise therapy. However, the clinical application of MSC-EXOs still faces challenges, such as the need to establish standardized preparation processes and quality control systems, and to assess their long-term safety and efficacy. Future research should focus on addressing these key issues to promote the clinical translation of MSC-EXOs in HIRI treatment. By analyzing the characteristics, functions, and application potential of MSC-EXOs, their unique value as a novel therapeutic strategy is gradually emerging. With further research and technological advancements, MSC-EXOs are expected to provide safer and more effective treatment options for HIRI patients.
The protective effects of exosomes derived from different sources of MSCs on HIRI
In recent years, exosomes derived from different sources of MSCs have shown significant therapeutic effects in the treatment of HIRI. This section will focus on the mechanisms and protective effects of exosomes from four sources of MSCs—adipose tissue, bone marrow, umbilical cord, and iPSCs—in the treatment of HIRI.
Protective effects of exosomes derived from AMSCs on HIRI
Research has shown that AMSCs significantly alleviate HIRI by secreting exosomes and various bioactive factors. In a rat model, Zhang et al. [80] induced liver injury by occluding the hepatic portal triad for 30 min and intervened with tail vein injection of AMSC-derived exosomes (AMSC-EXOs). The results indicated that AMSC-EXOs significantly improved liver function, reduced hepatocyte damage, primarily by inhibiting oxidative stress and apoptosis, while promoting mitochondrial dynamics and biosynthesis. In another rat study, Piao et al. [81] induced liver injury by blocking hepatic blood flow for 30 min and found that AMSC-EXOs reduced the expression of pyroptosis-related proteins (such as caspase-1 and GSDMD) by inhibiting the NLRP3 inflammasome and NF-κB pathway, activated the Wnt/β-catenin pathway, and promoted the expression of liver regeneration factors (such as Cyclin D1 and VEGF), thereby significantly reducing liver injury and promoting liver tissue repair. In a further study using a miniature pig model, Wang et al. [82,83,84] investigated the role and mechanisms of AMSC-EXOs in liver injury treatment. The study found that AMSC-EXOs significantly improved liver function and reduced tissue damage by inhibiting pro-inflammatory factors (such as TNF-α, IL-6, and CRP) and upregulating anti-inflammatory factors (such as IL-10). Additionally, AMSC-EXOs effectively protected hepatocytes from damage by inhibiting the endoplasmic reticulum stress pathways (such as GRP78, IRE1α, and PERK/eIF2α) and reducing the expression of anti-regenerative factors (SOCS3 and TGF-β), thus promoting liver regeneration (Fig. 3).
Protective effects of exosomes derived from BMSCs on HIRI
In recent years, exosomes derived from bone marrow mesenchymal stem cells (BMSCs) have gained increasing attention for their protective effects in HIRI. Wu et al. [18] used a rat liver transplant model to induce HIRI by transplanting severe fatty liver and intervened with heme oxygenase-1-modified BMSC-derived exosomes (HM-EXOs). The study found that HM-EXOs significantly reduced the necrotic area and neutrophil infiltration in liver tissue, inhibited the increase of ferroptosis-related markers (Fe²⁺ and MDA), and enhanced the expression of the antioxidant protein GPX4, protecting mitochondrial structure. In vitro experiments showed that HM-exos delivered miR-124-3p to inhibit STEAP3 expression, reduced lipid ROS and iron ion levels, suppressed lipid peroxidation, and decreased ferroptosis. This research offers a novel strategy for exosome-based organ transplantation protection.
In another mouse I/R model, Li et al. [85] induced liver injury by occluding hepatic vessels and simulated in vitro ischemia-reperfusion injury (H/R) in cells. The experiment showed that tail vein injection of BMSC-Exos enriched with miR-25-3p reduced hepatocyte apoptosis and tissue necrosis and alleviated injury by inhibiting the PTEN and p53 signaling pathways. In vitro experiments further confirmed that miR-25-3p, by targeting PTEN, alleviated hypoxia-induced apoptosis and enhanced cell viability. This study revealed that BMSC-Exos modulate the p53 signaling pathway by delivering miR-25-3p, providing new mechanistic support for the treatment of HIRI(Fig. 4).
Exosomes secreted by BMSCs, modified with HO-1, deliver miR-124-3p. miR-124-3p targets STEAP3, reducing the conversion of Fe³⁺ to Fe²⁺ and promoting the transport of Fe²⁺ via DMT1, thereby regulating the process of ferroptosis. In the Fenton reaction, Fe²⁺ induces lipid peroxidation, increases lipid ROS, and activates ferroptosis, a process that is suppressed by the antioxidant action of GPX4. Additionally, miR-25-3p in the exosomes can inhibit PTEN, thereby regulating the p53 signaling pathway and reducing cell apoptosis
Additionally, Zhang et al. [86] used Baicalin-pretreated BMSC-derived exosomes (Ba-EXO) for intervention in a mouse liver injury model. The results showed that Ba-Exo reduced liver tissue necrosis and inflammatory cell infiltration, and improved Th17/Treg cell imbalance. In vitro experiments indicated that Ba-EXO upregulated the expression of FGF21, activated FOXO1, and inhibited the JAK2/STAT3 signaling pathway, effectively alleviating Th17/Treg imbalance and significantly reducing reperfusion injury. This study provides a new strategy for the treatment of reperfusion injury, demonstrating that Ba-EXO regulates immune balance and signaling pathways to exert its protective effects.
Protective effects of exosomes derived from umbilical cord MSCs(UCMSCs) on HIRI
In recent years, the role of hUCMSCs in HIRI has also been studied. In a mouse model, Xie et al. [87] induced HIRI by occluding hepatic vessels for 90 min and intervened with tail vein injection of hUCMSC-EXOs. The study found that hUCMSC-EXOs significantly improved liver injury markers, alleviated histological damage, regulated the Th17/Treg cell balance, and exerted their effects through the miR-1246-mediated IL-6-gp130-STAT3 signaling pathway. In further studies, Xie et al. [88] explored the protective effects of hUCMSC-EXOs in both in vivo and in vitro models. In vitro, using LO2 cells to simulate I/R, hUCMSC-EXOs delivered miR-1246, which significantly increased cell viability, reduced apoptosis, and alleviated H/R injury by regulating the GSK3β/Wnt/β-catenin signaling pathway. In in vivo experiments, a mouse I/R model was established by blocking liver blood flow for 90 min and then restoring blood flow. Treatment with hUCMSC-EXOs via portal vein injection significantly improved liver function, reduced hepatocyte damage and necrosis, and decreased apoptosis and pro-inflammatory mediator levels.
Protective effects of exosomes derived from iPSC-MSCs on HIRI
iPSCs can differentiate into various somatic cells, demonstrating wide application potential. In recent years, the role of hiPSC-MSC-derived exosomes (hiPSC-MSC-EXOs) in HIRI has also been studied. In a rat model, Nong et al. [89] induced HIRI by occluding the portal vein branches for 60 min, followed by injection of hiPSC-MSC-EXOs. The study found that hiPSC-MSC-EXOs significantly reduced liver injury marker levels, alleviated inflammation and oxidative stress, and inhibited apoptosis. To further investigate the underlying mechanism, Du et al. [90] studied a mouse I/R model and found that after fusion with hepatocytes, hiPSC-MSC-EXOs activated the sphingosine kinase/sphingosine-1-phosphate (SK/S1P) signaling pathway, promoting hepatocyte proliferation and significantly protecting the liver from IRI(Fig. 5).
In conclusion, exosomes derived from various types of mesenchymal stem cells exert therapeutic effects in liver ischemia-reperfusion injury through different molecular mechanisms. AMSC-EXOs primarily regulate inflammation and oxidative stress pathways, BMSC-EXOs mainly influence ferroptosis and apoptosis, hUCMSC-EXOs focus on immune regulation and cell survival, while hiPSC-MSC-EXOs promote cell proliferation and tissue repair.
Comparative summary of exosomes from different sources
With the development of regenerative medicine, MSC-EXOs from different sources have shown unique advantages and promising application prospects in disease treatment. AMSCs have characteristics such as minimally invasive collection, high purity yield, and low immunogenicity, and are abundant in source, making them suitable for autologous transplantation with good stability [91,92,93]. However, their extraction efficiency needs improvement, and the activity and functionality of MSCs are influenced by the donor’s age [94, 95]. These features make them a potential choice for the treatment of acute liver injury and inflammatory liver diseases. BMSCs have strong resistance to inactivation and a low pathogen infection rate, demonstrating significant advantages in immune regulation and having a broad range of applications [96, 97]. However, the collection process is invasive, and large-scale production is challenging, potentially making them useful for liver transplantation and immune-related disease treatment. UCMSCs have notable advantages such as non-invasive collection, ethical acceptability, and strong proliferative capacity [98, 99]. They are relatively easy to obtain and show excellent potential in tissue damage repair, potentially applicable in the treatment of acute liver failure and neonatal liver diseases. iPSC-MSCs can be controlled for production, manufactured on a large scale, and customized for individual needs, offering unlimited sourcing [100,101,102,103,104]. However, they face challenges such as high costs and complex quality control, making them suitable for developing personalized treatment strategies.
To provide a comprehensive analysis of the characteristics of MSC-Exos from different sources in the treatment of HIRI, we conducted a systematic comparison of exosomes derived from the four sources mentioned above. The table summarizes the exosome preparation features, clinical advantages, technical limitations, and application directions, highlighting their differentiated potential in liver injury treatment (Table 1).
Discussion
Summary of key findings
This article summarizes the protective effects and potential mechanisms of MSC-EXOs from four different sources in HIRI. AMSC-EXOs exert anti-inflammatory effects by regulating multiple signaling pathways, promoting hepatocyte regeneration. However, their key active components and specific mechanisms still require further clarification. BMSC-EXOs improve liver function by reducing ferroptosis, but the exact mechanisms within the liver microenvironment remain unclear. hUCMSC-EXOs regulate signaling pathways by delivering specific molecules, restoring immune balance. However, research on the synergistic effects of other active components is still limited. hiPSC-MSC-EXOs promote hepatocyte proliferation and repair, but their storage stability and long-term safety need further investigation.
Challenges in clinical translation and potential solutions
Variability in isolation methods and standardization
Exosome isolation technology is of great significance in biomedical research and clinical applications. However, existing isolation methods still present certain challenges. Although ultracentrifugation is widely regarded as the “gold standard” for exosome isolation [105, 106], it has drawbacks such as sample loss [107], long processing times, high equipment costs [105], and the risk of structural damage to exosomes due to shear forces [105], which limit its application in high-throughput and large-scale production. Density gradient centrifugation offers high purity and no contamination, but the yield of exosomes is low, and the process is complex, making it unsuitable for handling large-volume samples [108,109,110]. Microfluidic technology, with its high resolution and high-throughput potential, has attracted significant attention, but it is currently mainly used in laboratory research, and its high cost and complexity hinder industrial advancement [111, 112]. To overcome these challenges, integrated isolation solutions can be developed in the future. For example, combining ultracentrifugation with immunoaffinity separation technology, using specific antibodies to recognize exosome markers (such as CD9, CD63, and CD81), can significantly improve the purity and recovery rate of exosomes. Additionally, the precision screening and dynamic monitoring capabilities of microfluidic technology can be utilized to optimize the isolation process. Moreover, optimizing centrifugation conditions (such as flow rate and temperature) using machine learning and artificial intelligence technologies can enhance separation efficiency and the reproducibility of results, thus driving the development of exosome isolation technology toward standardization, scalability, and industrialization.
Large-scale production and quality control
Currently, large-scale exosome production relies on bioreactor culture systems. However, traditional culture media, such as those containing fetal bovine serum (FBS), have complex components and high costs. These not only limit the yield of exosomes but also may introduce batch-to-batch variability and contamination risks (e.g., endotoxins) [113]. In the future, serum-free media with clearly defined chemical compositions and lower costs could be introduced, and microcarrier technology could be integrated to further optimize the cell culture environment, improving exosome production efficiency and quality.
In terms of quality control, standardized international guidelines should be established, including systematic characterization of particle size distribution (30–150 nm), typical marker expression levels (e.g., CD9, CD63, CD81), purity, and biological functions. Multi-omics analyses, such as proteomics and metabolomics, should be used to analyze the active components of exosomes, while also assessing their storage stability to provide a scientific basis for long-term preservation and transportation.
Long-term safety and immunogenicity
Exosomes are considered an ideal strategy for acellular therapy due to their low immunogenicity, but their long-term safety still requires in-depth study. Current research mainly focuses on acute pathological models, with a lack of long-term monitoring under chronic pathological conditions. It is recommended to introduce large animal models to conduct multi-cycle, multi-dose trials to evaluate the accumulation effects and potential toxicity of exosomes under chronic pathological conditions. Additionally, real-time imaging tracking technologies, such as MRI and fluorescence imaging, should be utilized to monitor the distribution and metabolic pathways of exosomes in vivo, providing a comprehensive assessment of their long-term safety.
Clinical application challenges
MSC-EXOs show potential in clinical applications but also face numerous challenges. Firstly, their composition and function exhibit heterogeneity due to differences in source, culture conditions, and isolation methods [114, 115], leading to inconsistent therapeutic effects. Secondly, MSC-EXOs have poor in vivo stability, are susceptible to environmental factors, and are rapidly cleared from the bloodstream, requiring multiple administrations to maintain efficacy [116,117,118,119]. Furthermore, production and storage methods have not been standardized, and there is a lack of unified quality control, which impacts batch-to-batch consistency and the reliability of clinical evaluations. In terms of administration, local injection is difficult to target precisely to the affected area, while intravenous injection faces the issues of rapid clearance and insufficient targeting. Potential solutions to these challenges include the development of efficient delivery systems, such as targeted nanoparticles or liposomes, to precisely deliver MSC-EXOs to the affected areas; optimizing storage and transportation conditions, such as using lyophilization technology to improve exosome stability; and standardizing quality control in production processes to ensure functional consistency and therapeutic efficacy.
Conclusion and future perspectives
This review demonstrates that MSC-EXOs, as a cell-free therapeutic strategy, shows great potential for the treatment of HIRI. By systematically analyzing current research findings, we have observed that MSC exosomes derived from adipose tissue, bone marrow, umbilical cord, and iPSCs, bone, umbilical cord, and iPSCs each have unique advantages in clinical applications. MSC-EXOs are particularly attractive in therapeutic development due to their cell-free nature, low immunogenicity, and high stability. However, several key challenges must still be overcome to achieve the clinical translation of MSC-EXOs, including the standardization of exosome isolation methods, the establishment of scalable production systems, and comprehensive safety assessments. To address these challenges, we recommend the adoption of advanced isolation technologies combined with standardized quality control systems, as well as systematic safety evaluations. Furthermore, future research should focus on developing engineered MSC-EXOs with enhanced targeting capabilities and therapeutic efficiency. For example, CRISPR-Cas9 gene editing technology can be used to optimize the expression of surface markers on exosomes, enhancing their targeting ability at liver injury sites. Additionally, nanotechnology can be utilized to improve the stability and targeted delivery of exosomes, such as using nanoparticles or liposomes to encapsulate exosomes, thus enhancing their accumulation and therapeutic effects in the liver. Researchers may also use high-throughput screening techniques to quickly evaluate the efficacy of MSC-EXOs from different sources, thereby identifying the most suitable exosome variants for clinical applications. To enable large-scale production of MSC-EXOs, GMP-compliant production platforms should be established, and low-cost, serum-free culture media systems should be explored, alongside the development of precise quality control measures to ensure the stability of exosome quality. Moreover, multi-center clinical trials should be conducted to comprehensively assess the long-term safety and efficacy of MSC-EXOs. These advancements are crucial for translating MSC-EXOs from laboratory research to clinical applications. The success of MSC-EXOs therapy is not only expected to revolutionize the treatment of HIRI but also open up new research directions in regenerative medicine and promote the widespread application of cell-free therapeutic strategies in various diseases.
Data availability
The datasets used and analyzed during the current study are available from the corresponding author upon reasonable request.
Abbreviations
- AMSCs:
-
Adipose-derived mesenchymal stem cells
- BMSCs:
-
Bone mesenchymal stem cells
- Cas9:
-
CRISPR-associated protein 9
- CRISPR:
-
Clustered regularly interspaced short palindromic repeats
- CRP:
-
C-reactive protein
- eIF2α:
-
Eukaryotic translation initiation factor 2α
- EV:
-
Extracellular vesicle
- FGF21:
-
Ibroblast growth factor 21
- FOXO1:
-
Forkhead box protein O1
- GPX4:
-
Glutathione peroxidase 4
- GRP78:
-
Glucose regulated protein 78
- GSDMD:
-
Gasdermin D
- GSK3β:
-
Glycogen synthase kinase-3β
- hiPSCs:
-
Human induced pluripotent stem cells
- H/R:
-
Hypoxia/re-oxygenation
- HIRI:
-
Hepatic ischemia-reperfusion injury
- hucMSCs:
-
Human umbilical cord mesenchymal stem cells
- IL-6:
-
Interleukin-6
- IL-10:
-
Interleukin-10
- IncRNA:
-
Long non-coding RNA
- IPC:
-
Ischemic preconditioning
- IPostC:
-
Ischemic postconditioning
- IRE1α:
-
Inositol-requiring enzyme 1α
- IRI:
-
Ischemic reperfusion injury
- IPSC:
-
Induced pluripotent stem cell
- JAK2:
-
Janus kinase-2
- miRNA:
-
microRNA
- MSCs:
-
Mesenchymal stem cells
- MVB:
-
Multivesicular body
- NF-kB:
-
Nuclear factor kappa-B
- NLRP3:
-
Nucleotide-binding oligomerization domain-like receptor protein 3
- PCNA:
-
Proliferating cell nuclear antigen
- PERK:
-
Protein kinase RNA–like endoplasmic reticulum kinase
- PTEN:
-
Phosphatase and tensin homolog deleted on chromosome ten
- ROS:
-
Reactive oxygen species
- SK/S1P:
-
Sphingosine kinase/sphingosine-1-phosphate
- SOCS3:
-
Suppressor of cytokine signaling 3
- STAT3:
-
Signal transducer and activator of transcription 3
- STEAP3:
-
Six-Transmembrane epithelial antigen of prostate 3
- TBIL:
-
Total bilirubin
- TGF-β:
-
Transforming growth factor-β
- Th17/Treg:
-
T helper cell 17/regulatory T cell
- TNF-α:
-
Tumor necrosis factor-α
- VEGF:
-
Vascular endothelial growth factor
References
Viganò L, Conci S, Cescon M, Fava C, Capelli P, D’Errico A, Torzilli G, Di Tommaso L, Giuliante F, Vecchio FM, et al. Liver resection for hepatocellular carcinoma in patients with metabolic syndrome: A multicenter matched analysis with HCV-related HCC. J Hepatol. 2015;63(1):93–101.
Lou TW, Yang RX, Fan JG. The global burden of fatty liver disease: the major impact of China. Hepatobiliary Surg Nutr. 2024;13(1):119–23.
Esparza-Baquer A, Labiano I, Sharif O, Agirre-Lizaso A, Oakley F, Rodrigues PM, Zhuravleva E, O’Rourke CJ, Hijona E, Jimenez-Agüero R, et al. TREM-2 defends the liver against hepatocellular carcinoma through multifactorial protective mechanisms. Gut. 2021;70(7):1345–61.
Liu Y, Li Z, Sun T, Li Z, Manyande A, Xiang H, He Z. Gut microbiota regulates hepatic ischemia-reperfusion injury-induced cognitive dysfunction via the HDAC2-ACSS2 axis in mice. CNS Neurosci Ther. 2024;30(2):e14610.
Kong E, Yuan C, Li Y, Tian T, He Y, Feng X. Protective Efficiency Comparison of Direct and Remote Ischemic Preconditioning on Ischemia Reperfusion Injury of the Liver in Patients Undergoing Partial Hepatectomy. Biomed Res Int 2023, 2023:2763320.
Rajendran L, Lenet T, Shorr R, Abou Khalil J, Bertens KA, Balaa FK, Martel G. Should cell salvage be used in liver resection and transplantation?? A systematic review and Meta-analysis. Ann Surg. 2023;277(3):456–68.
Qin Y, Li C, Ge X, Zhang Q, Wei X, Liu R. MMP2/9 downregulation is responsible for hepatic function recovery in cirrhotic rats following associating liver partition and portal vein ligation for staged hepatectomy. Ann Transl Med. 2022;10(8):468.
Xu L, Wang W, Xu Y, Yang B. Efficacy of a modified FRAIL scale in predicting the peri-operative outcome of hepatectomy in older adults (aged ≥ 75 years): a model development study. BMC Geriatr. 2023;23(1):770.
Wang Q, Li C, Chen G, Feng K, Chen Z, Xia F, Cai P, Zhang L, Sparrelid E, Brismar TB et al. Unsupervised machine learning of MRI radiomics features identifies two distinct subgroups with different liver function reserve and risks of Post-Hepatectomy liver failure in patients with hepatocellular carcinoma. Cancers (Basel) 2023, 15(12).
Tan S, Lu X, Chen W, Pan B, Kong G, Wei L. Analysis and experimental validation of IL-17 pathway and key genes as central roles associated with inflammation in hepatic ischemia-reperfusion injury. Sci Rep. 2024;14(1):6423.
Liu J, Luo R, Zhang Y, Li X. Current status and perspective on molecular targets and therapeutic intervention strategy in hepatic ischemia-reperfusion injury. Clin Mol Hepatol. 2024;30(4):585–619.
Lin CS, He MQ, An MY, Zhao QH, Zhang ZH, Deng KY, Ai Y, Xin HB. Ultra-Small Copper-Based Multienzyme-Like nanoparticles protect against hepatic Ischemia-Reperfusion injury through scavenging reactive oxygen species in mice. Small 2024:e2403313.
Guo J, Han S, Chen Q, Wang T, Yu B, Zhou J, Qiu T. Analysis of potential immune-related genes involved in the pathogenesis of ischemia-reperfusion injury following liver transplantation. Front Immunol. 2023;14:1126497.
George J, Lu Y, Tsuchishima M, Tsutsumi M. Cellular and molecular mechanisms of hepatic ischemia-reperfusion injury: the role of oxidative stress and therapeutic approaches. Redox Biol. 2024;75:103258.
Yu B, Zhang Y, Wang T, Guo J, Kong C, Chen Z, Ma X, Qiu T. MAPK signaling pathways in hepatic ischemia/reperfusion injury. J Inflamm Res. 2023;16:1405–18.
Yuan D, Collage RD, Huang H, Zhang X, Kautza BC, Lewis AJ, Zuckerbraun BS, Tsung A, Angus DC, Rosengart MR. Blue light reduces organ injury from ischemia and reperfusion. Proc Natl Acad Sci U S A. 2016;113(19):5239–44.
Dar WA, Sullivan E, Bynon JS, Eltzschig H, Ju C. Ischaemia reperfusion injury in liver transplantation: cellular and molecular mechanisms. Liver Int. 2019;39(5):788–801.
Wu L, Tian X, Zuo H, Zheng W, Li X, Yuan M, Tian X, Song H. miR-124-3p delivered by exosomes from Heme oxygenase-1 modified bone marrow mesenchymal stem cells inhibits ferroptosis to attenuate ischemia-reperfusion injury in steatotic grafts. J Nanobiotechnol. 2022;20(1):196.
Mu J, Li C, Shi Y, Liu G, Zou J, Zhang DY, Jiang C, Wang X, He L, Huang P, et al. Protective effect of platinum nano-antioxidant and nitric oxide against hepatic ischemia-reperfusion injury. Nat Commun. 2022;13(1):2513.
Mao XL, Cai Y, Chen YH, Wang Y, Jiang XX, Ye LP, Li SW. Novel targets and therapeutic strategies to protect against hepatic ischemia reperfusion injury. Front Med (Lausanne). 2021;8:757336.
Zhang W, Liu J, Li P, Wang X, Tang B. Reversible fluorescent probes for dynamic imaging of liver Ischemia-Reperfusion injury. Acc Chem Res. 2024;57(17):2594–605.
Chen H, Yin W, Yao K, Liang J, Cai J, Sui X, Zhao X, Zhang J, Xiao J, Li R, et al. Mesenchymal stem cell Membrane-Camouflaged liposomes for biomimetic delivery of cyclosporine A for hepatic Ischemia-Reperfusion injury prevention. Adv Sci (Weinh). 2024;11(32):e2404171.
Wang Y, Sun X, Han X, Sun J, Li L, Zhang D, Sun G. Resveratrol improves hepatic ischemia-reperfusion injury by inhibiting neutrophils via the ERK signaling pathway. Biomed Pharmacother. 2023;160:114358.
Liu J, Man K. Mechanistic insight and clinical implications of ischemia/reperfusion injury post liver transplantation. Cell Mol Gastroenterol Hepatol. 2023;15(6):1463–74.
Yang L, Wang W, Wang X, Zhao J, Xiao L, Gui W, Fan H, Xia J, Li Z, Yan J, et al. Creg in hepatocytes ameliorates liver ischemia/reperfusion injury in a TAK1-Dependent manner in mice. Hepatology. 2019;69(1):294–313.
Clarke G, Mergental H, Hann A, Perera M, Afford SC, Mirza DF. How machine perfusion ameliorates hepatic ischaemia reperfusion injury. Int J Mol Sci 2021, 22(14).
Brüggenwirth IMA, Mueller M, Lantinga VA, Camagni S, De Carlis R, De Carlis L, Colledan M, Dondossola D, Drefs M, Eden J, et al. Prolonged preservation by hypothermic machine perfusion facilitates logistics in liver transplantation: A European observational cohort study. Am J Transpl. 2022;22(7):1842–51.
Rüdiger HA, Graf R, Clavien PA. Sub-lethal oxidative stress triggers the protective effects of ischemic preconditioning in the mouse liver. J Hepatol. 2003;39(6):972–7.
Kageyama S, Nakamura K, Fujii T, Ke B, Sosa RA, Reed EF, Datta N, Zarrinpar A, Busuttil RW, Kupiec-Weglinski JW. Recombinant relaxin protects liver transplants from ischemia damage by hepatocyte glucocorticoid receptor: from bench-to-bedside. Hepatology. 2018;68(1):258–73.
Huang Y, Xu L, Wang N, Wei Y, Wang W, Xu M, Jiang L. Preoperative dexamethasone administration in hepatectomy of 25-min intermittent Pringle’s maneuver for hepatocellular carcinoma: a randomized controlled trial. Int J Surg. 2023;109(11):3354–64.
Glantzounis GK, Yang W, Koti RS, Mikhailidis DP, Seifalian AM, Davidson BR. Continuous infusion of N-acetylcysteine reduces liver warm ischaemia-reperfusion injury. Br J Surg. 2004;91(10):1330–9.
Chaves Cayuela N, Kiyomi Koike M, Jacysyn JF, Rasslan R, Azevedo Cerqueira AR, Pereira Costa SK. Picanço Diniz-Júnior JA, Massazo Utiyama E, Frasson de Souza Montero E: N-Acetylcysteine reduced ischemia and reperfusion damage associated with steatohepatitis in mice. Int J Mol Sci 2020, 21(11).
Wang ZY, Liu Y, Li SP, Li JJ, Zhang Z, Xiao XC, Ou Y, Wang H, Cai JZ, Yang S. Hypoxia inducible factor 1α promotes interleukin-1 receptor antagonist expression during hepatic ischemia-reperfusion injury. World J Gastroenterol. 2022;28(38):5573–88.
Chen Y, Yan J, Wang K, Zhu Z. Comparing the protective effects of local and remote ischemic preconditioning against ischemia-reperfusion injury in hepatectomy: a systematic review and network meta-analysis. Transl Gastroenterol Hepatol. 2024;9:13.
Koti RS, Seifalian AM, Davidson BR. Protection of the liver by ischemic preconditioning: a review of mechanisms and clinical applications. Dig Surg. 2003;20(5):383–96.
Robertson FP, Fuller BJ, Davidson BR. An evaluation of ischaemic preconditioning as a method of reducing ischaemia reperfusion injury in liver surgery and transplantation. J Clin Med 2017, 6(7).
Costa FL, Yamaki VN, Gonçalves TB, Coelho JV, Percário S, Brito MV. Combined remote ischemic perconditioning and local postconditioning on liver ischemia-reperfusion injury. J Surg Res. 2014;192(1):98–102.
Moskowitz MA, Waeber C. Remote ischemic preconditioning: making the brain more tolerant, safely and inexpensively. Circulation. 2011;123(7):709–11.
Kloner RA, Rezkalla SH. Preconditioning, postconditioning and their application to clinical cardiology. Cardiovasc Res. 2006;70(2):297–307.
Hausenloy DJ, Yellon DM. The therapeutic potential of ischemic conditioning: an update. Nat Rev Cardiol. 2011;8(11):619–29.
De Carlis R, Paolo M, Taner B. Donation after circulatory death: novel strategies to improve the liver transplant outcome. J Hepatol. 2023;78(6):1169–80.
Ceresa CDL, Nasralla D, Pollok JM, Friend PJ. Machine perfusion of the liver: applications in transplantation and beyond. Nat Rev Gastroenterol Hepatol. 2022;19(3):199–209.
Schlegel A, Mergental H, Fondevila C, Porte RJ, Friend PJ, Dutkowski P. Machine perfusion of the liver and bioengineering. J Hepatol. 2023;78(6):1181–98.
Endo C, van Rijn R, Huurman V, Schurink I, van den Berg A, Murad SD, van Hoek B, de Meijer VE, de Jonge J, van der Hilst CS, et al. Cost-effectiveness of dual hypothermic oxygenated machine perfusion versus static cold storage in DCD liver transplantation. Transplantation. 2024;109(2):e101–108.
Zhou T, Yuan Z, Weng J, Pei D, Du X, He C, Lai P. Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol. 2021;14(1):24.
Otsuka R, Wada H, Murata T, Seino KI. Immune reaction and regulation in transplantation based on pluripotent stem cell technology. Inflamm Regen. 2020;40:12.
Meissner TB, Schulze HS, Dale SM. Immune editing: overcoming immune barriers in stem cell transplantation. Curr Stem Cell Rep. 2022;8(4):206–18.
Chen Y, Pal S, Hu Q. Recent advances in biomaterial-assisted cell therapy. J Mater Chem B. 2022;10(37):7222–38.
Lv Y, Shi H, Liu H, Zhou L. Current therapeutic strategies and perspectives in refractory ITP: what have we learned recently? Front Immunol. 2022;13:953716.
Huang R, Chen J, Guo B, Jiang C, Sun W. Diabetes-induced male infertility: potential mechanisms and treatment options. Mol Med. 2024;30(1):11.
Hao T, Ji G, Qian M, Li QX, Huang H, Deng S, Liu P, Deng W, Wei Y, He J, et al. Intracellular delivery of nitric oxide enhances the therapeutic efficacy of mesenchymal stem cells for myocardial infarction. Sci Adv. 2023;9(48):eadi9967.
Sierra-Parraga JM, Merino A, Eijken M, Leuvenink H, Ploeg R, Møller BK, Jespersen B, Baan CC, Hoogduijn MJ. Reparative effect of mesenchymal stromal cells on endothelial cells after hypoxic and inflammatory injury. Stem Cell Res Ther. 2020;11(1):352.
Sahu A, Jeon J, Lee MS, Yang HS, Tae G. Nanozyme impregnated mesenchymal stem cells for hepatic Ischemia-Reperfusion injury alleviation. ACS Appl Mater Interfaces. 2021;13(22):25649–62.
Hong P, Yang H, Wu Y, Li K, Tang Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: a comprehensive review. Stem Cell Res Ther. 2019;10(1):242.
Shen Z, Huang W, Liu J, Tian J, Wang S, Rui K. Effects of mesenchymal stem Cell-Derived exosomes on autoimmune diseases. Front Immunol. 2021;12:749192.
Las Heras K, Royo F, Garcia-Vallicrosa C, Igartua M, Santos-Vizcaino E, Falcon-Perez JM, Hernandez RM. Extracellular vesicles from hair follicle-derived mesenchymal stromal cells: isolation, characterization and therapeutic potential for chronic wound healing. Stem Cell Res Ther. 2022;13(1):147.
Zou J, Yang W, Cui W, Li C, Ma C, Ji X, Hong J, Qu Z, Chen J, Liu A, et al. Therapeutic potential and mechanisms of mesenchymal stem cell-derived exosomes as bioactive materials in tendon-bone healing. J Nanobiotechnol. 2023;21(1):14.
Zhang Z, Zhao S, Sun Z, Zhai C, Xia J, Wen C, Zhang Y, Zhang Y. Enhancement of the therapeutic efficacy of mesenchymal stem cell-derived exosomes in osteoarthritis. Cell Mol Biol Lett. 2023;28(1):75.
Zhuo Y, Luo Z, Zhu Z, Wang J, Li X, Zhang Z, Guo C, Wang B, Nie D, Gan Y, et al. Direct cytosolic delivery of SiRNA via cell membrane fusion using cholesterol-enriched exosomes. Nat Nanotechnol. 2024;19(12):1858–68.
Xie M, Wu Y, Zhang Y, Lu R, Zhai Z, Huang Y, Wang F, Xin C, Rong G, Zhao C, et al. Membrane Fusion-Mediated loading of therapeutic SiRNA into exosome for Tissue-Specific application. Adv Mater. 2024;36(33):e2403935.
Qian X, An N, Ren Y, Yang C, Zhang X, Li L. Immunosuppressive effects of mesenchymal stem Cells-derived exosomes. Stem Cell Rev Rep. 2021;17(2):411–27.
Pan W, Chen H, Wang A, Wang F, Zhang X. Challenges and strategies: scalable and efficient production of mesenchymal stem cells-derived exosomes for cell-free therapy. Life Sci. 2023;319:121524.
Zomer HD, de Souza Lima VJ, Bion MC, Brito KNL, Rode M, Stimamiglio MA, Jeremias TDS, Trentin AG. Evaluation of secretomes derived from human dermal and adipose tissue mesenchymal stem/stromal cells for skin wound healing: not as effective as cells. Stem Cell Res Ther. 2024;15(1):15.
Zhao W, Zhang R, Zang C, Zhang L, Zhao R, Li Q, Yang Z, Feng Z, Zhang W, Cui R. Exosome derived from mesenchymal stem cells alleviates pathological scars by inhibiting the proliferation, migration and protein expression of fibroblasts via delivering miR-138-5p to target SIRT1. Int J Nanomed. 2022;17:4023–38.
Zhou X, Xu Y, Wang X, Lu W, Tang X, Jin Y, Ye J. Single and combined strategies for mesenchymal stem cell exosomes alleviate liver fibrosis: a systematic review and meta-analysis of preclinical animal models. Front Pharmacol. 2024;15:1432683.
Guo R, Zhuang H, Chen X, Ben Y, Fan M, Wang Y, Zheng P. Tissue engineering in growth plate cartilage regeneration: mechanisms to therapeutic strategies. J Tissue Eng. 2023;14:20417314231187956.
Han M, Zhang Z, Liu Z, Liu Y, Zhao H, Wang B, Zhang C, Shang H, Li Y, Wang S, et al. Three-dimensional-cultured MSC-derived exosome with hydrogel for cerebral ischemia repair. Biomater Adv. 2023;149:213396.
Cao Q, Huang C, Chen XM, Pollock CA. Mesenchymal stem Cell-Derived exosomes: toward Cell-Free therapeutic strategies in chronic kidney disease. Front Med (Lausanne). 2022;9:816656.
Hade MD, Suire CN, Suo Z. Mesenchymal stem Cell-Derived exosomes: applications in regenerative medicine. Cells 2021, 10(8).
Abid AI, Conzatti G, Toti F, Anton N, Vandamme T. Mesenchymal stem cell-derived exosomes as cell free nanotherapeutics and nanocarriers. Nanomedicine. 2024;61:102769.
Puspita R, Jusuf AA, Antarianto RD, Sianipar IR. A systematic review of the anti-inflammatory and anti-fibrotic potential of human umbilical cord mesenchymal stem cells-derived exosomes in experimental models of liver regeneration. Mol Biol Rep. 2024;51(1):999.
Lin F, Chen W, Zhou J, Zhu J, Yao Q, Feng B, Feng X, Shi X, Pan Q, Yu J, et al. Mesenchymal stem cells protect against ferroptosis via exosome-mediated stabilization of SLC7A11 in acute liver injury. Cell Death Dis. 2022;13(3):271.
Lin D, Chen H, Xiong J, Zhang J, Hu Z, Gao J, Gao B, Zhang S, Chen J, Cao H, et al. Mesenchymal stem cells Exosomal let-7a-5p improve autophagic flux and alleviate liver injury in acute-on-chronic liver failure by promoting nuclear expression of TFEB. Cell Death Dis. 2022;13(10):865.
Zhang YW, Hou LS, Xing JH, Zhang TR, Zhou SY, Zhang BL. Two-Membrane hybrid nanobiomimetic delivery system for targeted autophagy Inhibition of activated hepatic stellate cells to synergistically treat liver fibrosis. ACS Appl Mater Interfaces 2023.
You DG, Oh BH, Nguyen VQ, Lim GT, Um W, Jung JM, Jeon J, Choi JS, Choi YC, Jung YJ, et al. Vitamin A-coupled stem cell-derived extracellular vesicles regulate the fibrotic cascade by targeting activated hepatic stellate cells in vivo. J Control Release. 2021;336:285–95.
Azizsoltani A, Hatami B, Zali MR, Mahdavi V, Baghaei K, Alizadeh E. Obeticholic acid-loaded exosomes attenuate liver fibrosis through dual targeting of the FXR signaling pathway and ECM remodeling. Biomed Pharmacother. 2023;168:115777.
Lin Y, Yan M, Bai Z, Xie Y, Ren L, Wei J, Zhu D, Wang H, Liu Y, Luo J, et al. Huc-MSC-derived exosomes modified with the targeting peptide of aHSCs for liver fibrosis therapy. J Nanobiotechnol. 2022;20(1):432.
Lou G, Yang Y, Liu F, Ye B, Chen Z, Zheng M, Liu Y. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. J Cell Mol Med. 2017;21(11):2963–73.
Fang J, Liang W. ASCs -derived exosomes loaded with vitamin A and Quercetin inhibit rapid senescence-like response after acute liver injury. Biochem Biophys Res Commun. 2021;572:125–30.
Zhang Q, Piao C, Ma H, Xu J, Wang Y, Liu T, Liu G, Wang H. Exosomes from adipose-derived mesenchymal stem cells alleviate liver ischaemia reperfusion injury subsequent to hepatectomy in rats by regulating mitochondrial dynamics and biogenesis. J Cell Mol Med. 2021;25(21):10152–63.
Piao C, Sang J, Kou Z, Wang Y, Liu T, Lu X, Jiao Z, Wang H. Effects of exosomes derived from Adipose-Derived mesenchymal stem cells on pyroptosis and regeneration of injured liver. Int J Mol Sci 2022, 23(20).
Wang Y, Piao C, Liu T, Lu X, Ma Y, Zhang J, Ma H, Wang H. Exosomes derived from adipose mesenchymal stem cells promote regeneration of injured liver in minipigs. Int J Mol Sci 2024, 25(12).
Wang Y, Piao C, Liu T, Lu X, Ma Y, Zhang J, Liu G, Wang H. Effects of the exosomes of adipose-derived mesenchymal stem cells on apoptosis and pyroptosis of injured liver in miniature pigs. Biomed Pharmacother. 2023;169:115873.
Wang Y, Liu T, Jiao G, Lv Y, Piao C, Lu X, Ma H, Wang H. Exosomes from adipose-derived mesenchymal stem cells can attenuate liver injury caused by minimally invasive hemihepatectomy combined with ischemia-reperfusion in minipigs by modulating the Endoplasmic reticulum stress response. Life Sci. 2023;321:121618.
Li H, Lin W, Zhang G, Liu R, Qu M, Zhang J, Xing X. BMSC-exosomes miR-25-3p regulates the p53 signaling pathway through PTEN to inhibit cell apoptosis and ameliorate liver Ischemia–reperfusion injury. Stem Cell Rev Rep. 2023;19(8):2820–36.
Zhang B, Su L, Chen Z, Wu M, Wei J, Lin Y. Exosomes derived from Baicalin-pretreated bone mesenchymal stem cells improve Th17/Treg imbalance after hepatic ischemia-reperfusion via FGF21 and the JAK2/STAT3 pathway. IUBMB Life. 2024;76(8):534–47.
Xie K, Liu L, Chen J, Liu F. Exosomal miR-1246 derived from human umbilical cord blood mesenchymal stem cells attenuates hepatic ischemia reperfusion injury by modulating T helper 17/regulatory T balance. IUBMB Life. 2019;71(12):2020–30.
Xie K, Liu L, Chen J, Liu F. Exosomes derived from human umbilical cord blood mesenchymal stem cells improve hepatic ischemia reperfusion injury via delivering miR-1246. Cell Cycle. 2019;18(24):3491–501.
Nong K, Wang W, Niu X, Hu B, Ma C, Bai Y, Wu B, Wang Y, Ai K. Hepatoprotective effect of exosomes from human-induced pluripotent stem cell-derived mesenchymal stromal cells against hepatic ischemia-reperfusion injury in rats. Cytotherapy. 2016;18(12):1548–59.
Du Y, Li D, Han C, Wu H, Xu L, Zhang M, Zhang J, Chen X. Exosomes from Human-Induced pluripotent stem Cell-Derived mesenchymal stromal cells (hiPSC-MSCs) protect liver against hepatic ischemia/ reperfusion injury via activating sphingosine kinase and sphingosine-1-Phosphate signaling pathway. Cell Physiol Biochem. 2017;43(2):611–25.
Chen X, Zheng J, Yin L, Li Y, Liu H. Transplantation of three mesenchymal stem cells for knee osteoarthritis, which cell and type are more beneficial? A systematic review and network meta-analysis. J Orthop Surg Res. 2024;19(1):366.
Barreca MM, Cancemi P, Geraci F. Mesenchymal and induced pluripotent stem Cells-Derived extracellular vesicles: the new frontier for regenerative medicine?? Cells 2020, 9(5).
Abraham M, Kori I, Vishwakarma U, Goel S. Comprehensive assessment of goat adipose tissue-derived mesenchymal stem cells cultured in different media. Sci Rep. 2024;14(1):8380.
Trotzier C, Bellanger C, Abdessadeq H, Delannoy P, Mojallal A, Auxenfans C. Deciphering influence of donor age on adipose-derived stem cells: in vitro paracrine function and angiogenic potential. Sci Rep. 2024;14(1):27589.
Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J Transl Med. 2014;12:8.
Wang WZ, Wang YH, Bao SS, He F, Li G, Yang G, Chen J, Yang XY, Xiao Y, Tong YS, et al. iPSCs-derived iMSCs prevent osteoporotic bone loss and affect bone metabolites in ovariectomized mice. J Cell Mol Med. 2024;28(22):e70200.
Li C, Du Y, Zhang T, Wang H, Hou Z, Zhang Y, Cui W, Chen W. Genetic scissors CRISPR/Cas9 genome editing cutting-edge biocarrier technology for bone and cartilage repair. Bioact Mater. 2023;22:254–73.
Liu W, Liu A, Li X, Sun Z, Sun Z, Liu Y, Wang G, Huang D, Xiong H, Yu S, et al. Dual-engineered cartilage-targeting extracellular vesicles derived from mesenchymal stem cells enhance osteoarthritis treatment via miR-223/NLRP3/pyroptosis axis: toward a precision therapy. Bioact Mater. 2023;30:169–83.
Feng H, Liu Q, Deng Z, Li H, Zhang H, Song J, Liu X, Liu J, Wen B, Wang T. Human umbilical cord mesenchymal stem cells ameliorate erectile dysfunction in rats with diabetes mellitus through the Attenuation of ferroptosis. Stem Cell Res Ther. 2022;13(1):450.
Sun Y, Zhang W, Li X. Induced pluripotent stem cell-derived mesenchymal stem cells deliver exogenous miR-105-5p via small extracellular vesicles to rejuvenate senescent nucleus pulposus cells and attenuate intervertebral disc degeneration. Stem Cell Res Ther. 2021;12(1):286.
Mello DB, Mesquita FCP, Silva Dos Santos D, Asensi KD, Dias ML, Campos de Carvalho AC, Goldenberg R, Kasai-Brunswick TH. Mesenchymal stromal Cell-Based products: challenges and clinical therapeutic options. Int J Mol Sci 2024, 25(11).
Liu J, Chen W, Zhao Z, Xu HH. Reprogramming of mesenchymal stem cells derived from iPSCs seeded on biofunctionalized calcium phosphate scaffold for bone engineering. Biomaterials. 2013;34(32):7862–72.
Hu GW, Li Q, Niu X, Hu B, Liu J, Zhou SM, Guo SC, Lang HL, Zhang CQ, Wang Y, et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Res Ther. 2015;6(1):10.
Hirschi KK, Li S, Roy K. Induced pluripotent stem cells for regenerative medicine. Annu Rev Biomed Eng. 2014;16:277–94.
Ru Q, Chen L, Xu G, Wu Y. Exosomes in the pathogenesis and treatment of cancer-related cachexia. J Transl Med. 2024;22(1):408.
Roszkowski S. Therapeutic potential of mesenchymal stem cell-derived exosomes for regenerative medicine applications. Clin Exp Med. 2024;24(1):46.
Lobb RJ, Becker M, Wen SW, Wong CS, Wiegmans AP, Leimgruber A, Möller A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 2015;4:27031.
Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, Hu BY, Qin W, Zou TT, Fu Y, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):152.
Rashidi M, Bijari S, Khazaei AH, Shojaei-Ghahrizjani F, Rezakhani L. The role of milk-derived exosomes in the treatment of diseases. Front Genet. 2022;13:1009338.
Jia Y, Yu L, Ma T, Xu W, Qian H, Sun Y, Shi H. Small extracellular vesicles isolation and separation: current techniques, pending questions and clinical applications. Theranostics. 2022;12(15):6548–75.
Arya R, Jit BP, Kumar V, Kim JJ. Exploring the potential of exosomes as biomarkers in tuberculosis and other diseases. Int J Mol Sci 2024, 25(5).
Al-Madhagi H. The landscape of exosomes biogenesis to clinical applications. Int J Nanomed. 2024;19:3657–75.
Lee DY, Lee SY, Yun SH, Jeong JW, Kim JH, Kim HW, Choi JS, Kim GD, Joo ST, Choi I, et al. Review of the current research on fetal bovine serum and the development of cultured meat. Food Sci Anim Resour. 2022;42(5):775–99.
Lotfy A, AboQuella NM, Wang H. Mesenchymal stromal/stem cell (MSC)-derived exosomes in clinical trials. Stem Cell Res Ther. 2023;14(1):66.
Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020, 367(6478).
Zhao W, Zhang H, Liu R, Cui R. Advances in Immunomodulatory mechanisms of mesenchymal stem cells-Derived exosome on immune cells in Scar formation. Int J Nanomed. 2023;18:3643–62.
Zhai Z, Cui T, Chen J, Mao X, Zhang T. Advancements in engineered mesenchymal stem cell exosomes for chronic lung disease treatment. J Transl Med. 2023;21(1):895.
Kraińska MM, Pietrzkowska N, Turlej E, Zongjin L, Marycz K. Extracellular vesicles derived from mesenchymal stem cells as a potential therapeutic agent in acute kidney injury (AKI) in felines: review and perspectives. Stem Cell Res Ther. 2021;12(1):504.
Feng Y, Bao X, Zhao J, Kang L, Sun X, Xu B. MSC-Derived exosomes mitigate myocardial ischemia/reperfusion injury by reducing neutrophil infiltration and the formation of neutrophil extracellular traps. Int J Nanomed. 2024;19:2071–90.
Acknowledgements
Not applicable.
Funding
This work was supported by the Foundation of Hubei Provincial Health Commission (Approval No. WJ2021M091), Project Fund of Xianning Municipal Science and Technology Bureau (Approval No. 2022ZRKX074), and Project of Outstanding Young and Middle-aged Scientific and Technological Innovation Teams in Universities of Hubei Province (Approval No. T201921).
Author information
Authors and Affiliations
Contributions
Bo Zhao, Jiping Wei, and Zijian Jiang wrote, edited, and revised the manuscript and confirmed the authenticity of the raw data. Yiming Long drew the schematic illustrations. Yan Xu participated in collating article tables. Botao Jiang provided direction and guidance throughout the preparation of this manuscript. All authors read and approved the final version of the manuscript. All authors contributed to the article and approved the submitted version.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Generative AI and AI-assisted technologies in the writing process
During the preparation of this work, the authors used ChatGPT in order to improve the readability and quality of the text. After using this tool, the authors reviewed and edited the content as needed and take full responsibility for the content of the publication.
Conflict of interests
The authors declare that there is no conflict of interest.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Zhao, B., Wei, J., Jiang, Z. et al. Mesenchymal stem cell-derived exosomes: an emerging therapeutic strategy for hepatic ischemia-reperfusion injury. Stem Cell Res Ther 16, 178 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04302-9
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04302-9