Abstract Background Exosomes derived from pre-stimulated mesenchymal stem cells (MSCs) have improved therapeutic effects in disease-associated microenvironments. In this study, we investigated the therapeutic potential of exosomes from MSCs stimulated with plasma from patients with liver failure (LF-Exos). Methods Untreated exosomes (NC-Exos) and LF-Exos were extracted and characterized by nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), western blotting, and miRNA sequencing. We then examined the protective effects of LF-Exos on hepatocytes acutely injured by D-galactosamine (D-GalN)/lipopolysaccharide (LPS) co-treatment and on a mouse model of acute liver failure (ALF). Apoptosis was assessed using the CCK-8 assay and flow cytometry. Liver tissue damage was examined by hematoxylin and eosin staining and immunohistochemistry. The levels of signaling pathway proteins were determined by western blotting. Results Stimulation with plasma from patients with liver failure significantly altered the morphology of MSCs and reduced their proliferative activity. Gene chip analysis identified 31 differentially expressed miRNAs, and further analysis showed that these differentially expressed miRNAs may affect the PI3K-AKT signaling pathway. Compared to NC-Exos, LF-Exos induced AKT phosphorylation in hepatocytes and liver tissues, inhibited D-GalN/LPS-induced apoptosis in hepatocytes, and reduced pathological liver injury in the mouse model of ALF. Conclusion The biological effects of Exos were improved after stimulation with plasma from patients with liver failure. LF-Exos may inhibit the activity of the NLRP3 inflammasome and activate the PI3K-AKT signaling pathway to exert protective effects on acutely injured hepatocytes and a mouse model of ALF. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-025-04163-2. Keywords: Exosomes, Mesenchymal stem cells, Liver failure Background Liver function impairment has a variety of etiologies. When liver failure develops, short-term mortality is high [[30]1, [31]2]. Currently, the most effective clinical treatment for liver failure is liver transplantation; however, there are problems such as a shortage of donors, high cost, and immune rejection [[32]3]. Therefore, additional treatment modalities are urgently needed. Mesenchymal stem cells (MSCs) are low immunogenic cells that possess self-renewal and multi-lineage differentiation potentials [[33]4]. Previous studies have shown that transplantation of MSCs is an effective treatment for liver failure, and they play a role in the treatment of various diseases by regulating immune function, promoting cell regeneration, and tissue repair [[34]5]. However, MSC transplantation is associated with the risks of capillary blockage and teratoma formation and has demanding transport and storage conditions [[35]6–[36]8]. Research has shown that MSC-derived exosomes (MSC-Exos) play a similar therapeutic function as parent cells [[37]9] and have advantages over parent cells, including a smaller size, a lower immunogenic response in affected tissue, no risk of tumor formation, and being easier to produce and store [[38]10–[39]13]. Therefore, cell-free therapy based on MSC-Exos has been widely researched in recent years. Previous studies have shown that exosomes are heterogeneous; exosomes derived from different parent cells or parent cells cultured under different conditions show differential expression of nucleic acids, lipids, proteins, and other contents [[40]14]. The heterogeneity of exosomes affects their functional impact on recipient cells and plays various roles in immunomodulation, cell proliferation, and apoptosis [[41]15, [42]16]. Applying the microenvironment of the disease to condition MSCs can enhance the therapeutic role of MSC-Exos [[43]17, [44]18]; therefore, based on the heterogeneity of exosomes, training MSCs and deriving “trained” MSC-Exos for targeted therapeutic effects were explored in the current study. In this study, we stimulated MSCs with plasma from patients with liver failure (LF plasma) and extracted plasma-stimulated MSC-derived exosomes (LF-Exos), observed the differences between LF-Exos and untreated exosomes (NC-Exos), and assessed the therapeutic effects of LF-Exos in acutely injured hepatocytes and a murine model of acute liver failure (ALF). Methods Cell culture Umbilical cord mesenchymal stem cells (hUCMSCs) were generously provided by National Engineering Research Center of Cell Products, AmCellGene Engineering Co., Ltd Tianjin China, and cultured in low-glucose Dulbecco’s modified Eagle’s medium (DMEM, Gibco, USA, 12320032) containing 10% fetal bovine serum (FBS, Vivacell, China, [45]C04400) and 1% penicillin–streptomycin (Solarbio, China, P1400). After 12 h, the medium was replaced with culture medium containing LF plasma; medium without added plasma was used as a control. When the cells reached 70% confluence, they were incubated in a serum-free medium for 48 h, and the supernatant was collected. A mouse AML12 hepatocyte cell line was obtained from Wuhan Pricella Biotechnology Co., Ltd. AML12 cells were cultured in DMEM/F12 (Gibco, USA, 11330032) containing 10% FBS; 1% penicillin–streptomycin; insulin, transferrin, and selenium (ITS) Liquid Media Supplement (Beyotime, China, C0341); and 40 ng/mL dexamethasone (MCE, China, HY-14648). Murine macrophage RAW264.7 cells were obtained from our central laboratory and cultured in DMEM containing 10% FBS and 1% penicillin–streptomycin. Proliferation analysis MSCs were cultured overnight in 96-well plates (3,000 cells/well) and treated with different concentrations of LF plasma. A CCK-8 (Dojindo, Japan, CK04) solution was added to the wells according to the manufacturer’s protocol to measure the cell proliferation, and the optical density at 450 nm (OD450) was read using a multifunctional microplate reader (Thermo, Synergy H1). Exosome isolation and identification The cell culture supernatant was centrifuged at 4 °C and 300×g for 10 min to remove cells, 3,000×g for 15 min to remove dead cells, and 10,000×g for 30 min to remove subcellular components. The supernatant was transferred to an ultracentrifuge tube and centrifuged at 120,000×g for 90 min to obtain the sediment containing exosomes, which were resuspended in pre-cooled PBS. The protein concentration of the exosomes was determined using a bicinchoninic acid (BCA) assay (Huaxingbio, China, HX18651), and exosomes were identified using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blotting. Exosome miRNA sequencing and sequence analysis Total RNA was extracted from the exosomes using TRIzol reagent (Life Technologies, Carlsbad, CA, USA) and purified using an RNeasy Mini Kit (Qiagen, Valencia, CA, USA, 74104). A GeneChip miRNA array (Thermo Fisher, USA, 952052) was used to detect the relative expression levels of exosomal miRNAs. Biotinylated cDNA was prepared according to the standard Affymetrix protocol, using the Ambion^® WT Expression Kit. Following labeling, the fragmented cDNA was hybridized, washed, and labelled using an Affymetrix Fluidics Station 450. All arrays were scanned using the Affymetrix GeneChip Command Console (AGCC). Treatment of cells with exosomes AML12 cells were cultured in cell culture plates (2.5 × 10^5 cells/mL) for 24 h. Next, 4 mg/mL D-Galactosamine (D-GalN; Solarbio, China, G8110), 40 µg/mL lipopolysaccharide (LPS; Sigma-Aldrich, L2880), and 20 µg/mL exosomes were added to the AML12 cells and incubated for 24 h. The CCK-8 and 7AAD Apoptosis Assay Kits (Elabscinence, China, E-CK-A218) were used to measure the effect of exosomes on AML12 cells, according to the manufacturer’s instructions. RAW264.7 macrophages were cultured in 6-well plates (5 × 10^5 cells/mL) overnight and then co-cultured with exosomes (20 µg/mL) for 6 h. Subsequently, 200 ng/mL LPS was added to the wells and incubated for 24 h, followed by total cell protein collection. Mouse model of ALF and treatment with exosomes To establish the ALF model, 8-week-old male C57BL/6 mice were injected with D-GalN (250 mg/kg) and LPS (2.5 mg/kg), followed by immediate tail vein injection of NC-Exos, LF-Exos (100 µg), or an equal volume of PBS. The mice were anesthetized 6 h after the intervention using 5% isoflurane (RWD Life Science, China, H-601) by inhalation, obtained retro-orbital blood sampls, and euthanized by cervical dislocation, liver samples were collected for further analysis. This study has been reported in accordance with the ARRIVE guidelines 2.0, and additional supporting documents are provided in the supplementary materials. Blood samples were centrifuged at 3,000 rpm for 15 min to collect serum, and the serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured according to the manufacturer’s protocols for the test kits (NJJCBIO, China, C009/C010). A portion of liver tissue was fixed in 4% paraformaldehyde, dehydrated with concentration gradients of ethanol, and embedded in paraffin. Successive sections were cut and underwent hematoxylin and eosin (HE) and TUNEL staining to observe the histopathological changes. The remainder of the tissue samples was preserved at -80℃ for western blot analysis. Western blot analysis Proteins from cell or tissue samples were extracted using RIPA lysate buffer containing 1% protein phosphatase inhibitor, quantified using BCA analysis, diluted with sample buffer, and boiled at 105 °C. Proteins were separated using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene fluoride (PVDF) membranes. After blocking with skim milk, membranes were incubated overnight with the primary antibody at 4 °C on a shaker. The blots were incubated with the horseradish peroxidase (HRP)-conjugated secondary antibody (1:5000) for 1 h at room temperature. The primary anti-AKT (1:1000, [46]AB179463) antibody was purchased from Abcam. Anti-phospho-AKT (1:2000, #4060), anti-phospho-PI3K (1:1000, #4228), and anti-ASC (1:1000, #67824) antibodies were purchased from CST. The anti-PI3K (1:1000, [47]R22768) antibody was purchased from Zenbio. Anti-NLRP3 (1:1000, AG-20B-0014) and anti-mouse caspase-1 (1:1000, AG-20B-0042) antibodies were purchased from Adipogen. Anti-mouse cleaved IL-1β (1:2000; AF-401-SP) was purchased from R&D Systems. Anti-GAPDH (1:2000, Proteintech, 60004-1) and anti-lamin B (1:1000, Proteintech, 10895-1-AP) antibodies were used as internal controls. Statistical analysis SPSS software (version 19) was used for statistical analyses, and GraphPad Prism (version 8.3) was used to create images. Data are presented as the mean ± standard deviation ( Inline graphic ± SD). The Student’s t-test was used for comparisons between two samples, and one-way analysis of variance (ANOVA) was used for comparisons between multiple samples. P-values < 0.05 were considered statistically significant. Results LF plasma affected the morphology and proliferation of MSCs We observed the morphology of MSCs under a microscope. Normal cultured MSCs showed a spindle-like shape, grew in spiral arrangements, were packed tightly with a uniform morphology and size, and had clear ovoid nuclei in the center of the cells. MSCs cultured with LF plasma were wider and had enlarged gaps between cells, a disordered cellular arrangement, and many coarse particles around the nuclei (Fig. [48]1A). The CCK-8 analysis showed that the proliferation curves of normal cultured MSCs showed typical “S” shapes. However, increasing concentrations of LF plasma inhibited the growth of MSCs (Fig. [49]1B). Fig. 1. [50]Fig. 1 [51]Open in a new tab Morphology and proliferation of MSCs. A. Representative photographs of MSCs stimulated with LF plasma after 48 h; B. Proliferation rate of MSCs stimulated with LF plasma in different concentrations (scale bar = 500 μm) Identification of MSC-Exos Two groups of MSC-Exos were isolated from the cell culture supernatant. TEM showed that the particles extracted from the two groups of supernatants had teacup or elliptical disc shapes and double-membrane structures typical of exosomes (Fig. [52]2A). NTA revealed that the NC-Exos and LF-Exos size was 30–200 nm, which was the size of exosomes (Fig. [53]2B). Western blotting confirmed that both NC-Exos and LF-Exos expressed the specific exosomal surface markers CD63, TSG101, and Alix and did not express calnexin, which is not expressed on exosomes (Fig. [54]2C). In addition, the protein content of LF-Exos was significantly higher than that of NC-Exos (Fig. [55]2D). These results confirm that the extracted particles were exosomes. Fig. 2. [56]Fig. 2 [57]Open in a new tab Characterization of NC-Exos and LF-Exos. A. TEM Images showed that both groups of exosomes had a teacup or ellipsoidal morphology (scale bar = 200 nm); B. NTA detected the range of particle size distribution of NC-Exos and LF-Exos (size: nm); C. Western Blot confirmed the expression of exosomal surface markers TSG101, Alix, CD63, and endoplasmic reticulum protein Calnexin in NC-Exos and LF-Exos (Full-length blots are presented in Supplementary Fig. [58]1); D. Protein concentration in NC-Exos and LF-Exos; ***P < 0.001 LF plasma induces changes in MSC-Exos miRNA We sequenced the miRNA extracted from NC-Exos and LF-Exos and screened for miRNAs that were differentially expressed between NC-Exos and LF-Exos. We identified 31 differentially expressed miRNAs; of these, 11 were upregulated and 20 were downregulated (|fold change|>1.2, P < 0.05) (Fig. [59]3A-B). Fig. 3. [60]Fig. 3 [61]Open in a new tab Analysis of differentially expressed miRNAs between NC-Exos and LF-Exos. A. Volcano plot of differentially expressed miRNAs between NC-Exos and LF-Exos (|Fold change|>1.2; P < 0.05); B. Heat map of differentially expressed miRNAs between NC-Exos and LF-Exos; C. GO and KEGG pathway enrichment analyses of target genes matched upregulated differentially expressed miRNAs; D. GO and KEGG pathway enrichment analyses of target genes matched to upregulated differentially expressed miRNAs The miRanda and RNAhybrid algorithms were used to predict target gene interaction binding sites for miRNAs to match the target relationship between the predicted miRNAs and mRNAs. Using the intersection of the matched mRNAs from the two databases, 5,933 predicted mRNAs were obtained. The predicted target mRNAs were subjected to gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. GO enrichment analysis showed that these genes were significantly enriched in the regulation of signaling, cell junctions, and protein binding. KEGG signaling pathway enrichment analysis showed that these genes were significantly enriched in the PI3K-AKT, Wnt, and other signaling pathways (Fig. [62]3C-D). MSC-Exos inhibited apoptosis in D-GalN/LPS-induced hepatocytes The CCK-8 assay results showed that MSC-Exos promoted the viability of AML12 cells under the inhibitory effect of D-GalN/LPS and that the viability of AML12 cells was significantly increased in the LF-Exos group (Fig. [63]4A). Fig. 4. [64]Fig. 4 [65]Open in a new tab Protective effect of NC-Exos and LF-Exos on D-GalN/LPS-induced AML12 cells. A. Percentage of cell proliferation of D-GalN/LPS-induced AML12 cells co-cultured with NC-Exos or LF-Exos for 24 h; B. Percentage of cell apoptosis of D-GalN/LPS-induced AML12 cells co-cultured with NC Exos or LF-Exos for 24 h; C. Annexin V/7AAD staining of D-GalN/LPS-induced AML12 cells; *P < 0.05; ***P < 0.001;****P < 0.0001 The apoptosis assay revealed that D-GalN/LPS significantly increased the proportion of apoptotic cells, and both NC-Exos and LF-Exos reversed D-GalN/LPS-induced apoptosis in AML12 cells. The apoptosis rate of AML12 cells in the LF-Exos group was significantly lower than that in the NC-Exos group (Fig. [66]4B-C). MSC-Exos alleviated liver injury in a d-GalN/LPS-induced ALF mouse model We observed the survival time of ALF mice. The mice started to die 3 h after D-GalN/LPS injection, and the mortality rate reached 100% within 8 h. Some mice survived in both groups following tail vein injection of NC-Exos or LF-Exos, but the survival analysis showed no significant difference (Fig. [67]5A). Fig. 5. [68]Fig. 5 [69]Open in a new tab MSC-Exos ameliorated D-GalN/LPS-induced ALF in mice. A. Survival curves after mice were injected with D-GalN/LPS (n = 10); B. Serum ALT level of mice in each group; C. Serum AST levels of mice in each group; D. Hepatic organ coefficient of mice in each group; E. Gross morphology of the liver and HE staining analyzed liver sections in each group (scale bar = 250 μm); F. TUNEL staining of liver sections in each group; *P < 0.05; **P < 0.005; ****P < 0.0001 Mice in the model group had significantly elevated serum ALT and AST levels, indicating that D-GalN/LPS caused significant liver injury. After injection with NC-Exos or LF-Exos, the serum ALT and AST levels were significantly decreased. However, the serum ALT level of the LF-Exos group was significantly lower than that of the NC-Exos group (Fig. [70]5B-C). This may be because ALT is mainly distributed in the cytoplasm of hepatocytes and is more sensitive to early liver injury, while AST is mainly distributed in the mitochondria of hepatocytes, and mitochondrial AST is only released when hepatocytes are severely damaged and necrotic [[71]19], Therefore, we only observed a significant decrease in ALT in the mouse model of ALF. The livers of mice in the model group were generally congested and enlarged, and the hepatic organ coefficient (liver weight/body weight; LW/BW) was significantly increased (Fig. [72]5D). The mouse liver pathology was examined. HE staining showed that the liver structure in normal mice was clear and obvious, and the hepatocytes were tightly arranged around the central vein. The livers of the model group were significantly damaged, blurred liver lobule structures, large areas of denatured and necrotic hepatocytes, and inflammatory cell infiltration. In the NC-Exos and LF-Exos groups, the hepatic lobular structure was restored and the necrotic area and inflammatory cell infiltration were reduced; the LF-Exos group had more appreciable improvements than the NC-Exos group (Fig. [73]5E). Mouse hepatocyte apoptosis was detected using TUNEL staining; TUNEL-positive hepatocytes were observed in the liver tissue of the model group, and the number of TUNEL-positive cells in the NC-Exos and LF-Exos groups was reduced (Fig. [74]5F). Effect of MSC-Exos on the PI3K-AKT signaling pathway and NLRP3 inflammasome in D-GalN/LPS-injured hepatocytes and ALF mice Based on the miRNA sequencing analysis, we speculated that LF-Exos might ameliorate ALF progression via the PI3K-AKT signaling pathway. In addition, the PI3K-AKT signaling pathway may be associated with the NLRP3 inflammasome [[75]20, [76]21], and activation of the NLRP3 inflammasome is also a factor in liver failure exacerbation [[77]22, [78]23]. We analyzed the expression of proteins in the PI3K-AKT signaling pathway and NLRP3 inflammasome and found that LF-Exos significantly increased the expression of PI3K and p-AKT in AML12 cells and ALF mice. In addition, the expression of NLRP3 in AML12 cells, RAW264.7 cells, and ALF mice was significantly reduced (Fig. [79]6). Fig. 6. [80]Fig. 6 [81]Open in a new tab LF-Exos ameliorates ALF by promoting the PI3K-AKT signaling pathway and inhibiting NLRP3 inflammasome activation. A. Western blot analysis of the protein expression in AML12 cells; B. Western blot analysis of the protein expression in RAW264.7 cells; C. Western blot analysis of the protein expression in the livers of mice in each group. Full-length blots are presented in Supplementary Figures [82]2–[83]4, respectively. *P < 0.05, **P < 0.005, ***P < 0.001 Discussion MSC-Exos have demonstrated therapeutic effects similar to those of MSCs and are an effective therapeutic prospect for ALF [[84]24–[85]26]. Researchers have found that “training” MSCs may improve the therapeutic effects of MSCs and MSC-Exos [[86]27–[87]29], and effective MSC stimulation may make MSC-Exos more targeted and efficient in treating diseases. Thus, determining effective stimulation has become a popular topic in exosome research. In this study, we selected LF plasma for MSC stimulation, observed the differences between NC-Exos and LF-Exos, and preliminarily verified the therapeutic effects of LF-Exos on ALF. Exosome heterogeneity leads to different compositions of MSC-Exos that have been stimulated in different ways [[88]30]. miRNA is one of the most abundant components of exosomes and the main biologically active substance [[89]31]. miRNA can inhibit gene expression and protein synthesis by competitively binding to mRNA [[90]32], which plays important roles in physiopathological processes. Shao et al. [[91]31] stimulated hUCMSCs with IL-6, obtained exosomes (Exos-IL6), and sequenced small RNAs extracted from Exos-IL6 and Exos-NC; they identified 37 differentially expressed miRNAs that were widely involved in the regulation of IL-6-related signaling pathways, including the PI3K-AKT signaling pathway. Ma et al. [[92]33] obtained exosomes from TNF-α-pretreated MSCs (T-Exos); some of the 180 miRNAs differentially expressed between T-Exos and NC-Exos were closely related to immunolog- and inflammation-related signaling pathways. Our study found that stimulation of MSCs with LF plasma affected their morphology and inhibited their proliferation. Sequencing results showed 31 differentially expressed miRNAs between NC-Exos and LF-Exos, and KEGG analysis predicted that these differentially expressed miRNAs were related to the PI3K-AKT signaling pathway. The mechanisms by which MSC-Exos ameliorate ALF are complex and varied; however, most are closely related to the regulation of immune homeostasis, inhibition of apoptosis, and promotion of the regenerative functions of the remaining cells. The PI3K-AKT signaling pathway is a classical pathway that is closely related to a wide range of physiological and pathological processes, such as cell growth, proliferation, migration, metabolism, and immunomodulation [[93]34, [94]35]. Previous studies have revealed that IGF-1R/PI3K/AKT signaling plays an important role in the development of acetaminophen (APAP)-induced ALF [[95]36]. The aberrant activation of the NLRP3 inflammasome mediates liver failure by stimulating procaspase-1 and pro-IL-1β signaling [[96]37]. Zhang et al. found TNF-α-pretreated hUCMSC-Exos affect downstream ALT, AST, and inflammatory factors by inhibiting the activation of the NLRP3-associated pathway [[97]38]. A relationship between the PI3K-AKT signaling pathway and the NLRP3 inflammasome has been reported. Zhao et al. [[98]20] revealed that AKT acts directly on NLRP3, inhibits NLRP3 oligomerization during LPS priming, and reduces cytokine secretion following NLRP3 activation. Wang et al. [[99]21] found that NLRP3 inflammasome activation is attributable to inhibition of the ROS-PI3K/AKT pathway. Tabaa et al. [[100]27] found that suppression of NLRP3 activation regulates the PI3K/AKT/mTOR cascade. Our results suggest that treatment with LF-Exos can reduce ALT and AST levels and alleviate the liver damage caused by ALF. Exosomes can activate the PI3K-AKT signaling pathway via miRNAs and may inhibit the activation of the NLRP3 inflammasome, which promotes cell proliferation, inhibits apoptosis, and causes immunomodulation. MSC-Exos have been widely noticed as an emerging therapeutic technology, and many studies have confirmed that MSC-Exos have a good therapeutic effect on liver failure; however, obtaining more therapeutically useful exosomes and clinically applying them remains a great challenge. Our team has conducted research on bioartificial liver therapy, which involves combining extracorporeal circulation with a bioreactor. Hepatocytes are usually inserted into the bioreactor, and the patient’s plasma is exchanged with the cells in the bioreactor and then returned to the blood, thus promoting the repair of liver damage. Our results showed that the modification of MSCs with LF plasma can alter the miRNA expression in LF-Exos and may play a role in inhibiting apoptosis and regulating immune responses in injured cells and ALF mice by activating the PI3K-AKT signaling pathway and inhibiting the activation of the NLRP3 inflammasome. LF-Exos may play an enhanced therapeutic role in ALF with the placement of MSCs in the bioreactor to create an MSC-based bioartificial liver. This will provide a basis for future research on MSC-based bioartificial livers. Conclusion In summary, our study revealed that LF plasma-stimulation of MSCs can regulate the biological function of MSC-Exos and that LF-Exos can play a protective role against acute liver injury or liver failure by activating the PI3K-AKT signaling pathway and inhibiting the activation of the NLRP3 inflammasome. This study lays the foundation for obtaining therapeutically useful MSC-Exos and developing novel bioartificial liver systems. Electronic supplementary material Below is the link to the electronic supplementary material. [101]Supplementary Material 1^ (731.1KB, xlsx) [102]Supplementary Material 2^ (9.3MB, pdf) [103]Supplementary Material 3^ (369.4KB, docx) Acknowledgements