Abstract Hepatic fibrosis is a common disease with high morbidity and mortality rates. The complex and poorly understood mechanisms underlying hepatic fibrosis represent a significant challenge for the development of more effective therapeutic strategies. MKP5 is a potential regulator of multiple fibrotic diseases. However, its precise role and mechanism of action in hepatic fibrosis remains unclear. This study identified a reduction in MKP5 expression in fibrotic liver tissues of mice treated with CCl[4] and observed that MKP5 knockout mice exhibited a more pronounced development of hepatic fibrosis. In addition, RNA-seq data indicated activation of protein processing in the endoplasmic reticulum signalling pathway in fibrotic liver tissues of mice lacking MKP5. Mechanistically, MKP5 inhibits the activation of hepatic stellate cells (HSCs) and hepatocyte apoptosis through the regulation of the IRE/XBP1 pathway. Based on these findings, we developed PLGA-MKP5 nanoparticles coated with a mesenchymal stem cell membrane (MSCM). Our results demonstrated that MSCM-PLGA-MKP5 was most effective in attenuating hepatic inflammation and fibrosis in murine models by modulating the IRE/XBP1 axis. This study contributes to the current understanding of the pathogenesis of hepatic fibrosis, suggesting that the targeted delivery of MKP5 via a nano-delivery system may represent a promising therapeutic approach to treat hepatic fibrosis. Graphical abstract [44]graphic file with name 12951_2024_3029_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-03029-8. Keywords: Hepatic fibrosis; MKP5; HSCs activation; Hepatocytes apoptosis; Nano-delivery system, Mesenchymal stem cell membrane Introduction Hepatic fibrosis (HF) is a pathological process of repair and remodelling that can result from several factors, including viral infection, alcohol abuse, non-alcoholic steatohepatitis, cholestasis, and autoimmune diseases. It is a common precursor to cirrhosis and hepatocellular carcinoma [[45]1, [46]2]. HF has emerged as a significant public health concern because of its high prevalence and adverse clinical outcomes. Given the current dearth of definitive pharmaceuticals targeting HF and incomplete understanding of its pathogenesis, it is imperative to explore the molecular mechanisms underlying its prevention and treatment. It is widely accepted that inflammatory disorders are the primary initiators of HF. During this phase, macrophages and other cells secrete inflammatory cytokines and release TGF-β, which activates HSCs. These cells are characterised by cell proliferation and chemotaxis as well as a transition of cell morphology to myofibroblast-like cells. Concurrently, hepatocytes are damaged or undergo death, releasing damage-related molecular patterns (DAMPs), which destroy the normal liver structure and ultimately help to form HF [[47]2–[48]5]. Recent studies have identified endoplasmic reticulum stress (ERS) as a key factor in the pathogenesis of HF, with evidence that regulating ERS may offer a promising avenue for improving HF outcomes [[49]6, [50]7]. Moreover, the specific inhibitor of IRE1a, STF-083010, has been demonstrated to mitigate CCl[4]-induced HF by upregulating the expression of miR-122 [[51]8]. It has been demonstrated that, during HF, ERS induces hepatocyte apoptosis [[52]9] and activates HSCs [[53]10], promoting the progression of HF. Both inflammation and ERS play pivotal roles in the development of HF. Consequently, modulation of these processes through the regulation of associated gene expression represents a pivotal strategy in the treatment of HF. Dual-specific mitogen-activated protein kinase phosphatases (MKPs) inactivates the mitogen-activated protein kinase (MAPK) pathway by selectively dephosphorylating serine/threonine residues [[54]11]. Our previous studies have demonstrated that MKP5 exerts inhibitory effects on obesity-induced inflammation [[55]12] and ERS-mediated type 1/2 diabetes [[56]13, [57]14]. MKP5 has been linked to the modulation of pulmonary and cardiac fibrosis [[58]15, [59]16], indicating a potential involvement of MKP5 in the pathogenesis of metabolic disorders. However, the regulatory role of MKP5 in HF remains unclear. Therefore, it is imperative to explore the involvement of MKP5 in HF and to elucidate the molecular mechanisms involved in the prevention and treatment of HF. The complex aetiology of HF presents a significant challenge for the development of effective therapeutic options. Nucleic acid, as a universal genetic component encoding proteins, offer a promising avenue for treating a wide range of diseases, particularly compared to traditional drug targets [[60]17]. Nevertheless, clinical applications of nucleic acid drugs remain constrained by their inherent instability, intrinsic obstacles, and lack of selective targeting [[61]18]. In recent years, an increasing number of studies have demonstrated that chemical modification of nucleic acid drugs or the utilisation of carrier-delivery systems can enhance the therapeutic efficacy of nucleic acid drugs for disease treatment. Using nanoparticles (NPs) for drug delivery has emerged as a novel approach to treat diseases. It has the potential to enhance the stability of nucleic acid molecules, prevent their degradation by nucleases, and improve their effectiveness in penetrating cells [[62]19]. Nevertheless, NPs exhibit certain limitations regarding their biostability, targeting, and rapid clearance from the body, which present challenges for their use in clinical applications. Accordingly, a biomimetic approach utilising cell membranes has been identified as a potential solution [[63]20, [64]21]. Compared to other cell membranes, the mesenchymal stem cell membrane (MSCM) represents a novel carrier with active targeting, favourable biocompatibility, and immune escape characteristics of MSC. This characteristic can effectively mitigate the risk of immune rejection and enhance the treatment safety. Furthermore, MSCM protects nucleic acid drugs from degradation by nucleases in the bloodstream, prolonging their circulation [[65]22]. The coating of NPs with MSCM represents an efficient method for selective targeting, enabling drugs to enter specific cells, improving treatment accuracy. This approach has a wide range of applications in tumour therapy, inflammatory diseases, and tissue regeneration [[66]23]. This study aimed to investigate the role of MKP5 in HF. Initially, a notable decline in MKP5 expression was observed in murine fibrotic liver tissue. Subsequently, MKP5 knockout mice were generated, and the absence of MKP5 resulted in a more pronounced CCl[4]-induced HF. Moreover, our results demonstrate that MKP5 plays a regulatory role in TGF-β1-induced HSC activation and hepatocyte apoptosis through the IRE/XBP1 pathway. Based on these findings, we developed a biomimetic PLGA-MKP5 coated with MSCM. This delivery system exhibited enhanced efficacy of MKP5 in targeting damaged liver tissue and demonstrated a pronounced anti-fibrotic effect in CCl[4]-induced HF without overt toxicity. This provides a promising strategy for effective anti-fibrotic therapy. Results MKP5 knockout exacerbates CCl[4]-induced hepatic fibrosis To investigate the relationship between MKP5 and HF, a CCl[4]-induced HF mouse model was established. After 8 weeks of CCl[4] stimulation, the liver exhibited structural abnormalities characterised by fibrotic scarring and substantial collagen deposition (Fig. S1A). Furthermore, fibronectin, the primary component of the extracellular matrix (ECM), and α-smooth muscle actin (α-SMA), a fibrosis marker protein, exhibited a notable increase in expression in CCl[4]-stimulated liver tissue relative to that in control mice (Fig. S1B). The results demonstrated that mice exhibited signs of HF following CCl[4] stimulation. It is noteworthy that the protein and mRNA levels of MKP5 were significantly decreased in HF liver tissues (Fig. [67]1A–C, S1C). Moreover, the relationship between MKP5 and collagen deposition, as well as the liver function markers alanine aminotransferase (ALT) and aspartate aminotransferase (AST), was investigated in mice with HF. The results demonstrated that an increase in collagen deposition and the levels of AST and ALT in the liver tissue were accompanied by a decrease in MKP5 levels (Fig. [68]1D), indicating the potential involvement of MKP5 in the regulation of CCl[4]-induced HF. Fig. 1. [69]Fig. 1 [70]Open in a new tab MKP5 knockout exacerbates CCl[4]-induced hepatic fibrosis. The WT and KO mice were subjected to CCl[4]-induce hepatic fibrosis. A, B The protein level of MKP5 in liver tissues was determined by western blotting (A), and statistical analysis was performed using ImageJ software (B). C The mRNA level of MKP5 in liver tissues was detected by real-time PCR. (n = 6/group). D Line graphs and histograms summarising MKP5 expression samples compared to an indicator of collagen deposition, and line graphs and histogram summarising MKP5 expression compared to an indicator of ALT and AST in liver tissues (n = 3/group). E Liver weight and ratios of liver weight to body weight (M: n = 6–8/group, F: n = 9–10/group). F Levels of serum AST, ALT, MDA, and GSH, as well as Hyp in the liver (M: n = 6–8/group, F: n = 6–10/group). G Representative staining of HE, Masson, and Sirus Red in liver tissues. Scale bar: 100 μm. H Statistical analysis of Sirus Red and Masson staining of liver tissues. I Protein levels of MKP5, fibronectin, and α-SMA in liver tissue were estimated using western blotting, and statistical analysis was performed using ImageJ software. (M: n = 6–8/group, F: n = 6/group). J The mRNA levels of MKP5, ACTA2, CoL1A1, and Fn1 were analysed using real-time PCR (n = 4–6/group). *P < 0.05, **P < 0.01, ***P < 0.001 To further investigate the role of MKP5 in HF, mice deficient in MKP5 were constructed (Fig. S1D, S1E). Subsequently, CCl[4]-induced HF models were established using both female and male mice. There were no significant differences in body weight between the male and female mice (Fig. S1F). However, liver weight and the ratio of liver weight to body weight increased significantly following CCl[4] induction, and these increases were further pronounced in the CCl[4]-treated groups after MKP5 knockout (Fig. [71]1E). As illustrated in Fig. [72]1F, liver functional indicators, ALT and AST in serum and the hydroxyproline (Hyp) content in liver, were markedly elevated in CCl[4]-treated wild-type (WT) and KO mice compared with control mice. Moreover, the absence of MKP5 resulted in more pronounced exacerbation of these liver function markers in male mice, with a similar trend observed in female mice, albeit to a lesser extent. Malondialdehyde (MDA), an end-product of lipid peroxidation, serves as an indirect indicator of free radical damage to the liver. Glutathione (GSH), a crucial antioxidant, reflects the hepatic antioxidant capacity [[73]24]. In female mice, administration of CCl[4] resulted in an increase in MDA content and a decrease in GSH content in liver tissues. These changes were further exacerbated by MKP5 knockout. Similar trends were observed in male mice, although the differences were not statistically significant (Fig. [74]1F). In addition, the liver tissue of the KO-CCl[4] group exhibited a darker and harder consistency, with a notable increase in the number of diffuse fine granular lesions on the surface and within the sections, compared to that in the WT-CCl[4] group (Fig. S1G). Haematoxylin and eosin (H&E) staining revealed that the WT-CCl[4] group exhibited structural destruction of liver tissue, disorder of hepatic lobules, deviation or loss of central veins, and vacuolation of hepatocytes when compared to the WT-Control group. Masson and Sirus Red staining revealed markedly elevated collagen deposition in the liver tissue of the WT-CCl[4] group compared to that in the WT-Control group, which was further accentuated in CCl[4]-treated KO mice (Fig. [75]1G, H)). The CCl[4]-induced increase in α-SMA and fibronectin expression in KO mice was significantly higher than that in WT mice (Fig. [76]1I). Similarly, the transcription levels of the fibrosis-related genes ACTA2, CoL1A1, and Fn1 in CCl[4]-induced KO mice were significantly higher than those in WT mice (Fig. [77]1J). Similar results were observed in female mice. These findings suggest that MKP5 knockout enhances the development of CCl[4]-induced HF. MKP5 knockout promotes HSCs activation during hepatic fibrosis The activation of HSCs represents a pivotal event in the pathogenesis of HF. Upon activation, HSCs undergo transformation into myofibroblasts, leading to excessive deposition of collagen-based ECM in the liver [[78]5]. As illustrated in Fig. [79]2B, the expression of α-SMA (a marker of activated HSCs) was elevated in the liver tissues of WT-CCl[4] mice and was further augmented in KO-CCl[4] mice. This suggests that HSCs were activated following CCl[4] induction and that MKP5 knockout exacerbated the activation of HSCs. Of particular note, the co-localisation of MKP5 and α-SMA was extensive in the liver tissue, indicating that MKP5 may regulate HF through its effect on HSC activation (Fig. [80]2A). In addition, an increase in the proliferation of HSCs was observed following CCl[4] treatment, and MKP5 knockout further promoted proliferation, as evidenced by the co-localisation of α-SMA and Ki67 (a marker of cell proliferation) (Fig. [81]2B). To gain further insight into the function of MKP5 in HSC activation, primary HSCs were isolated from WT and KO mice and treated with TGF-β1. The CCK-8 results demonstrated that MKP5 knockout enhanced the TGF-β1-induced proliferation of primary HSCs compared to the WT group (Fig. [82]2C). Similarly, inhibition of MKP5 expression resulted in a more pronounced TGF-β1-induced proliferation of LX-2 cells. Conversely, MKP5 overexpression resulted in the inhibition of LX-2 cell proliferation (Fig. [83]2D). As illustrated in Fig. [84]2E, TGF-β1 elevated the expression of ECM proteins in primary HSCs from KO mice, including collagen Iα1, fibronectin and α-SMA, compared to WT mice. The results demonstrated notable discrepancies between the experimental groups (Fig. S2A). The results of real-time polymerase chain reaction (PCR) also indicated that the mRNA levels of the fibrosis-related genes ACTA2, CoL1A1, and Fn1 in TGF-β1-induced HSCs from KO mice were significantly higher than those in WT mice (Fig. [85]2G). Similarly, inhibition of MKP5 expression resulted in the up-regulation of ECM-related proteins (Fig. [86]2F, S2B) and genes (Fig. [87]2H) in LX-2 cells induced by TGF-β1, exacerbating LX-2 cell activation. Conversely, overexpression of MKP5 reversed TGF-β1-induced activation of LX-2 cells, as previously described (Fig. [88]2F, H, S2C). In HF, activated stellate cells migrate and undergo morphological transformation into fibroblasts. In this study, TGF-β1 induced the migration of LX-2 cells, and the inhibition of MKP5 expression further promoted TGF-β1-induced LX-2 cell migration. Conversely, MKP5 overexpression in LX-2 cells reversed TGF-β1-induced migration (Fig. [89]2I, J). Furthermore, the effect of MKP5 on the TGF-β1-induced LX-2 cell cycle was investigated. As illustrated in Fig. [90]2K, L and S2D, MKP5 overexpression resulted in the arrest of the TGF-β1-induced LX-2 cell cycle in the S phase due to the up-regulation of the protein expression of S phase-specific factors, including CyclinA2 and CDK2. Consequently, this regulatory mechanism inhibits cell proliferation. In conclusion, the absence of MKP5 promoted HSC activation during HF. Fig. 2. [91]Fig. 2 [92]Open in a new tab MKP5 knockout promotes HSCs activation during hepatic fibrosis. A The co-localisation of MKP5 and α-SMA in liver tissue was visualised by immunofluorescence (IF) staining. The nuclei were stained with DAPI. Scale bar: 100 μm. B IF staining was performed to visualise the co-localisation of α-SMA and Ki67 in the liver tissue. The nuclei were stained with DAPI. Scale bar: 100 μm. C Primary HSCs isolated from WT and KO mice were treated with TGF-β1 and cell proliferation was assessed using the CCK-8 assay. D LX-2 cells were infected with retrovirus to either overexpress or knock down MKP5 and then treated with TGF-β1. The proliferation of LX-2 cells upon TGF-β1 treatment was determined using a CCK-8 assay. E, G Expression levels of fibrosis-related proteins and genes in primary HSCs were analysed by western blotting (E) and real-time PCR (G). F, H–J LX-2 cells were infected with retrovirus to either overexpress or knock down MKP5, and subsequently treated with TGF-β1. Protein levels of Collagen Iα1, fibronectin, and α-SMA were detected by western blotting (F). The mRNA levels of ACTA2, Fn1, and CoL1A1 were determined by real-time PCR (H). Transwell inserts (I), and wound healing (J) were used to evaluate the migration of LX-2 cells, and quantification was performed using ImageJ software. Scale bar: 100 μm. K, L LX-2 cells were infected with a retrovirus to overexpress MKP5 and then treated with TGF-β1. The cell cycle was quantified by Annexin V-FITC/PI staining (K). Protein levels of CDK2 and CyclinA2 were measured by western blotting and the blots from three repeated experiments were quantified using ImageJ software(L). *P < 0.05, **P < 0.01, ***P < 0.001 MKP5 knockout aggravates hepatocyte apoptosis and epithelial-to-mesenchymal transition (EMT) during hepatic fibrosis Hepatocytes constitute the primary parenchymal cell type in the liver during HF, ultimately resulting in hepatocyte apoptosis. In this study, we observed a reduction in the number of albumin-positive cells in CCl[4]-induced liver tissues (Fig. [93]3A). Furthermore, MKP5 expression declined following CCl[4] treatment, which is in accordance with the observations presented in Fig. [94]1A and S1C. Of particular note, significant co-localisation between MKP5 and albumin was observed, which suggests that MKP5 may regulate HF by affecting hepatocyte injury (Fig. [95]3A). In addition, a greater number of transferase dUTP nick-end labelling (TUNEL)-positive hepatocytes was observed in CCl[4]-stimulated WT mice, indicating that CCl[4] induced hepatocyte apoptosis and that MKP5 knockout exacerbated this process (Fig. [96]3B). Moreover, Annexin V-fluorescein (FITC)/propidium iodide (PI) results demonstrated that MKP5 knockout significantly enhanced TGF-β1-induced apoptosis in primary hepatocytes compared to that in WT mice (Fig. [97]3C). Conversely, MKP5 overexpression reversed TGF-β1-induced apoptosis in AML12 cells (Fig. [98]3D). Subsequently, the expression levels of apoptosis-related proteins were evaluated. As illustrated in Fig. [99]3E, the protein levels of cleaved Caspase3, cleaved Caspase8 and cleaved Caspase9 were markedly elevated in KO mice following TGF-β1 treatment compared to WT mice. Conversely, MKP5 overexpression led to a notable reduction in the expression of apoptotic proteins in TGF-β1-induced AML12 cells (Fig. [100]3F). Upon stimulation by pro-fibrotic factors, hepatocytes undergo EMT, causing the loss of their characteristic epithelial morphology and acquisition of a fibroblast-like phenotype, accompanied by increased secretion of the ECM and collagen [[101]25]. Our findings revealed that TGF-β1 induction resulted in a notable reduction in E-cadherin protein expression in KO primary hepatocytes compared to WT mice. Concurrently, there was an observable increase in the levels of N-cadherin and Vimentin (Fig. [102]3G). Similarly, the inhibition of MKP5 expression exacerbated EMT induced by TGF-β1 in AML12 cells. Conversely, the overexpression of MKP5 resulted in the up-regulation of E-cadherin protein expression and downregulation of N-cadherin and Vimentin protein levels in TGF-β1-induced AML12 cells, attenuating TGF-β1-induced EMT transformation (Fig. [103]3H). These findings indicate that MKP5 knockout may contribute to the exacerbation of hepatocyte apoptosis and EMT during HF. Fig. 3. [104]Fig. 3 [105]Open in a new tab MKP5 knockout aggravates hepatocytes apoptosis and EMT during hepatic fibrosis. A Representative double-staining of MKP5 and albumin in liver tissues. The nuclei were stained with DAPI. Scale bar: 100 μm. B IF staining for Albumin and TUNEL staining in the liver tissue. The nuclei were stained with DAPI. Scale bar: 100 μm. C, D The ratios of apoptotic cells in primary hepatocytes (C) and MKP5 overexpression in AML12 cells (D) treated with TGF-β1 were quantified by flow cytometry using an Annexin V-FITC/PI staining kit. E, F The protein levels of cleaved Caspase3, cleaved Caspase8, and cleaved Caspase9 in primary hepatocytes (E) and AML12 cells (F) were measured via western blotting, and quantification was performed using ImageJ software. G, H Protein levels of E-cadherin, N-cadherin, and Vimentin in primary hepatocytes (G) and AML12 cells (H) were detected by western blotting. The data were quantified using the ImageJ software. *P < 0.05, **P < 0.01, ***P < 0.001 MKP5 knockout aggravates the hepatic inflammatory response during hepatic fibrogenesis To elucidate the regulatory mechanism of MKP5 in HF, RNA sequencing was employed to analyse differentially expressed genes (DEGs) in the livers of CCl[4]-induced WT and KO mice. A total of 566 DEGs were identified, with 376 upregulated and 190 downregulated genes (Fig. [106]4A). Functional classification and pathway enrichment analysis of the DEGs were performed using the Kyoto Encyclopedia of Genes and Genomes (KEGG). Ours results demonstrated that there was a notable activation of pathways linked to inflammation, including MAPK and TNF signalling pathways (Fig. [107]4B). Furthermore, Gene set enrichment analysis (GSEA) also revealed that MAPK, TGF-β, and chemokine pathways were enriched, and there was a significant positive correlation with MKP5 KO-modulated genes (MAPK: NES = 1.6477109, P = 0.0, FDR < 0.03797602; TGF-β: NES = 1.5691792, P = 0.006369427, FDR < 0.06341224; Chemokine: NES = 1.4961016, P = 0.0013513514, FDR < 0.124319231) (Fig. [108]4C). In addition, the heat map clustered a notable increase in the expression of numerous inflammatory cytokine- and chemokine-related genes (Fig. [109]4D). Immunohistochemical staining of liver tissues revealed a significant increase in CD68 expression in KO mice after CCl[4] treatment compared to WT male and female mice (Fig. [110]4E). MKP5 knockout resulted in a notable intensification of macrophage infiltration, which contributed to the aggravation of CCl[4]-induced hepatic inflammation. The MAPK pathway is intimately associated with the inflammatory process and may play a role in the regulation of liver inflammation during HF [[111]26]. To validate the activation of MAPK pathway, hepatic proteins implicated in this signalling cascade were analysed. Elevated levels of phosphorylated ERK, JNK, and P38 were observed in the livers of WT mice following CCl[4] treatment. Furthermore, MKP5 knockout resulted in further augmentation of the phosphorylation levels (Fig. [112]4F). To further investigate the impact of MKP5 on hepatic inflammation during the development of HF, the levels of inflammatory cytokines in the serum of mice were evaluated using enzyme-linked immunosorbent assay (ELISA). It was observed that the levels of IL-6 and TNF-α were significantly increased after CCl[4] induction in both male and female mice, and that the knockout of MKP5 further exacerbated the levels of inflammatory cytokines (Fig. [113]4G). Subsequently, the inflammation-related genes from RNA sequencing were further validated in mouse liver tissues using real-time PCR. It was confirmed that the expression of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, TGF-β1, and IL-18) and chemokines (CXCL1, CXCL11, CXCL2, CXCL10, CCL2, CCL3, and CCL4) was significantly increased in CCl[4]-induced KO mice compared with WT mice. Conversely, the expression of the anti-inflammatory cytokine (IL-10) was significantly decreased. Furthermore, the expression of the pro-fibrotic cytokine TGF-β1 was significantly increased in CCl[4]-induced KO mice (Fig. [114]4H), consistent with the finding in Fig. [115]4C. During HF, pro-fibrotic factors trigger an inflammatory response in HSCs and hepatocytes. As illustrated in Fig. [116]4I, TGF-β1 treatment markedly elevated the levels of pro-inflammatory cytokines in primary HSCs from WT mice, as indicated by an increase in the mRNA expression of IL-6, IL-1β, TNF-α, and MCP-1. Moreover, these genes were further increased in TGF-β1-induced primary HSCs from KO mice. In addition, inhibition of MKP5 expression aggravated the TGF-β1-induced inflammatory response in LX-2 cells, whereas MKP5 overexpression reduced the expression of inflammatory cytokines in LX-2 cells (Fig. [117]4J). Similar results were observed in hepatocytes. The transcription levels of inflammation-related genes IL-6, IL-1β, TNF-α, and MCP-1 in TGF-β1-induced primary hepatocytes from KO mice were significantly higher than those in WT mice (Fig. [118]4K). Inhibition of MKP5 expression exacerbated the inflammatory response induced by TGF-β1 in AML12 cells. Conversely, overexpression of MKP5 resulted in downregulation of the inflammatory genes induced by TGF-β1 in AML12 cells (Fig. [119]4L). These results suggest that MKP5 may exert a regulatory effect on hepatic inflammation during HF via the MAPK pathway. Fig. 4. [120]Fig. 4 [121]Open in a new tab MKP5 knockout aggravates the hepatic inflammatory response during hepatic fibrogenesis. A Volcano plot illustrating the distribution of DEGs between WT-CCl[4] and KO-CCl[4] mouse liver tissues. B KEGG analysis highlighting the differential pathways in the liver tissue between WT-CCl[4] and KO-CCl[4]. C GSEA was performed on CCl[4]-treated WT and KO mice to analyse the TGF-β, MAPK, and chemokine signalling pathways. D Heat map illustrating the DEGs between WT-CCl[4] and KO-CCl[4] mouse liver tissue. E Representative staining of CD68 in liver tissues and statistical analyses. Scale bar: 100 μm. F Protein expression of the MAPK pathway in male liver tissue was measured by western blotting, and quantification was performed using Image J software. (n = 6/group). G Serum levels of IL-6 and TNF-α were estimated using ELISA (M: n = 4–5/group, F: n = 6–9/group). H mRNA levels of inflammatory factors and chemokines in CCl[4]-treated male WT and KO mice (n = 6/group). I–L mRNA levels of IL-6, IL-1β, TNF-α, and MCP-1 were detected in primary HSCs (I), LX-2 cells (J), primary hepatocytes (K), and AML12 cells (L) following treatment with TGF-β1 using real-time PCR. * P < 0.05, ** P < 0.01, *** P < 0.001 MKP5 regulates hepatic fibrosis through the IRE/XBP1 pathway The RNA sequencing data from the liver of CCl[4]-KO/WT mice revealed that a pronounced activation of the protein processing in the endoplasmic reticulum pathway, which emerged as the most significantly affected pathway (Fig. [122]4B). A compiled list of protein processing genes in the endoplasmic reticulum signalling pathway demonstrated that top-ranking change genes, including the molecular chaperones Hsp40, Dnajb4, Dnajb11, Dnaja1, Dnajc10, and Ern1 (the coding gene of ERS kinase IRE1α). Ern1 was identified the second most significant gene among the DEGs in terms of P value (Fig. [123]5A). Fig. 5. [124]Fig. 5 [125]Open in a new tab MKP5 regulates hepatic fibrosis via the IRE/XBP1 pathway. A List of the top six DEGs between WT-CCl[4] and KO-CCl[4] mouse liver tissues. B–D The phosphorylation level of IRE1α was measured in liver tissue (B), LX-2 (C), and AML12 cells (D) using western blotting, followed by quantification using ImageJ software (n = 6/group). Up: MKP5 overexpression, down: inhibition of MKP5 expression. E–G The mRNA levels of XBP1s were detected in liver tissue (E), LX-2 (F), and AML12 cells (G) by real-time PCR. Left: MKP5 overexpression, right: inhibition of MKP5 expression. H–K LX-2 cells were pre-treated with Kira6 (0.5 µM) for 1 h and stimulated with the corresponding inhibitor and TGF-β1 for 24 h, V: Veh, K: Kira6, T: TGF-β1, K + T: Kira6 + TGF-β1. The levels of p-IRE1α and fibrosis-associated proteins were measured using western blotting (H). The mRNA levels of XBP1s (I); fibrosis-related genes ACTA2, Fn1, and CoL1A1 (J); and inflammation-associated genes IL-6, IL-1β, TNF-α, and MCP-1 (K) were determined using real-time PCR. (L-O) AML12 cells were pre-treated with Kira6 (1 µM) for 1 h and stimulated with the corresponding inhibitor and TGF-β1 for 24 h. The levels of p-IRE1α, apoptosis-associated proteins (L), and EMT-related proteins (N) were detected using western blotting. The mRNA levels of XBP1s (M), IL-6, IL-1β, TNF-α, and MCP-1 (O) were measured by real-time PCR. *P < 0.05, **P < 0.01, ***P < 0.001 The ERS plays a pivotal role in the development of HF. In the event of an ERS, IRE1α dissociates from GRP78, leading to activation of its kinase and RNase activities. Subsequently, a 26-nucleotide intron within XBP1 is cleaved, resulting in the production of activated spliced XBP1 (XBP1s) [[126]27]. Our findings revealed that the expression of p-IRE1α was elevated in the liver tissue of CCl[4]-induced WT mice and that the phosphorylation level of IRE1α was markedly increased following MKP5 knockout (Fig. [127]5B). Similarly, inhibition of MKP5 expression resulted in a further increase in p-IRE1α expression in TGF-β1-stimulated LX-2 and AML12 cells. Conversely, overexpression of MKP5 reduced the elevated expression of p-IRE1α induced by TGF-β1 (Fig. [128]5C, D). As a downstream factor of IRE1α, XBP1 was spliced in the liver tissue of WT mice after CCl[4] induction, and the level of XBP1s increased further after MKP5 knockout (Fig. [129]5E). In TGF-β1-stimulated LX-2 and AML12 cells, inhibition of MKP5 expression resulted in a further increase in the levels of XBP1 splicing, whereas overexpression of MKP5 led to a reduction in the mRNA level of XBP1s (Fig. [130]5F, G). To further elucidate the role of IRE1α in HSC activation, hepatocyte apoptosis, and EMT, we pre-treated LX-2 and AML12 cells infected with Retro-pSIREN or Retro-shMKP5 with Kira6 (an inhibitor of the IRE1α kinase) for one hour before TGF-β1 treatment. These results demonstrated that Kira6 effectively suppressed the phosphorylation of IRE1α and the mRNA level of XBP1s in LX-2 and AML12 cells (Fig. [131]5H, I,L, M, S3A, S3B). In Retro-pSIREN-infected LX-2 cells, TGF-β1 induced a significant increase in the expression of fibrosis-associated proteins, including α-SMA, collagen Iα1, and fibronectin. Furthermore, inhibition of MKP5 expression led to a further increase in the expression of the aforementioned ECM proteins, which is consistent with the results shown in Fig. [132]2F. It is noteworthy that Kira6 markedly alleviated TGF-β1-induced overexpression of α-SMA, collagen Iα1, and fibronectin in MKP5 knockdown LX-2 cells (Fig. [133]5H). Furthermore, real-time PCR results demonstrated that in MKP5 knockdown LX-2 cells, Kira6 markedly diminished the elevated expression of TGF-β1-induced fibrosis-associated genes, including ACTA2, CoL1A1, and Fn1, as well as inflammation-related genes such as IL-6, IL-1β, TNF-α, and MCP-1 (Fig. [134]5J, K). These findings indicated that MKP5 exerts regulatory control over TGF-β1-induced HSC activation and inflammatory responses via the IRE/XBP1 pathway. Similarly, in MKP5 knockdown AML12 cells, Kira6 was observed to markedly reduce the elevated expression of TGF-β1-induced apoptotic proteins, including cleaved Caspase3, cleaved Caspase8, and cleaved Caspase9 (Fig. [135]5L, S3B), as well as EMT-related proteins such as N-cadherin and Vimentin (Fig. [136]5N, S3C). Furthermore, the expression of inflammatory mediators, including IL-6, IL-1β, TNF-α, and MCP-1, were also diminished by Kira6 (Fig. [137]5O). Our data demonstrated that MKP5 exerts regulatory effects on HSC activation, hepatocyte apoptosis, and EMT through the IRE/XBP1 pathway, which ultimately attenuates HF progression. Preparation and physicochemical characterisation of nano-formulation The use of cell membrane coatings in biomimetic designs has become increasingly prevalent in recent years. In this study, MSCM was obtained from MSCs. Subsequently, a gene-carrying nanoparticles (PLGA-MKP5) coated with MSCM was prepared (Fig. [138]6A). Agarose gel electrophoresis was performed to determine the optimal ratio of PLGA to MKP5. The results demonstrated that, at a mass ratio of 2:1 of PLGA to MKP5, complete complexes were formed with no observed band tailing (Fig. [139]6B). Transmission electron microscopy revealed that the MSCM coating was effectively applied to the surface of the PLGA-MKP5 nanoparticles, exhibiting a spherical core–shell structure comparable to that observed in its non-targeted counterparts (Fig. [140]6C). Furthermore, the protein structure remained intact, resembling that of the native MSCM proteins (Fig. [141]6D, E). Dynamic light scattering (DLS) analysis demonstrated that the particle size of MSCM-PLGA-MKP5 (131 nm) was larger than that of PLGA-MKP5 (119 nm), with a surface charge of -3.08 mV (Fig. [142]6F). Subsequently, the biosafety of biomimetic nano-formulation were evaluated in LX-2 and AML12 cells. As illustrated in Fig. [143]6G, there was no significant change in cell viability for both LX-2 and AML12 cells with varying concentrations of nano-formulation compared to the untreated group. This suggests that the nano-formulation did not induce any cytotoxic effects on cells. The cellular uptake of the nanoparticles was further evaluated using laser confocal microscopy, which demonstrated a markedly elevated red fluorescence intensity of MSCM-PLGA-MKP5 compared to PLGA-MKP5 in both cell types (Fig. [144]6H). Furthermore, the protein expression (Fig. [145]6I,J) and mRNA levels (Fig. [146]6K) of MKP5 were significantly higher in the MSCM-PLGA-MKP5 and PLGA-MKP5 groups than in the phosphate buffered saline (PBS) group in the LX-2 and AML12 cells. The expression of MKP5 was significantly higher in the MSCM-PLGA-MKP5 group than in the PLGA-MKP5 group. These findings suggest that a viable method for MKP5 delivery has been established. Fig. 6. [147]Fig. 6 [148]Open in a new tab Preparation and physicochemical characterisation of nano-formulation. A Schematic showing preparation of the nano-formulation. B The binding efficiency of PLGA to MKP5 was measured. M: Marker, 1: PLGA, 2: MKP5, 3–11: MKP5: PLGA = 10:1, 5:1, 2.5:1, 2:1, 1:1, 1:2, 1:2.5, 1:5, 1:10. C The morphology of PLGA-MKP5, MSCM-PLGA-MKP5, and MSCM were observed by TEM. Scale bar: 100 nm. D The protein bands of MSCM-PLGA-MKP5 and MSCM were detected using SDS-PAGE. E Evaluation of CD45 and CD90 expression on MSCM-PLGA-MKP5 by flow cytometry. CD45, a negative marker of MSC; CD90, a positive marker of MSC. F Particle size and potential of PLGA-MKP5, and MSCM-PLGA-MKP5. G Viability of LX-2 and AML12 cells treated with MSCM-PLGA-MKP5 was assessed using the CCK-8 assay. H Cellular uptake of PLGA-MKP5 and MSCM-PLGA-MKP5 in LX-2 and AML12 cells was assessed by confocal microscopy. Scale bar: 50 μm. I–K The expression levels of MKP5 in LX-2 cells and AML 12 cells were measured using western blotting (I, J) and real-time PCR (K). *P < 0.05, **P < 0.01 In vivo toxicity and biodistribution of nano-formulation It is imperative that the biosafety of nano-formulation be evaluated before assessing their efficacy. The in vivo toxicity and biodistribution of the nano-formulation were evaluated. As illustrated in Fig. [149]7A, mice that received tail vein injections of the nanoparticles demonstrated a consistent increase in weight over time compared to the PBS group, with no statistically significant difference in body weight among all groups. Similarly, no significant differences were observed in liver weight or liver weight/body weight among the groups (Fig. [150]7B). Furthermore, histological examination using HE staining demonstrated no discernible changes in the morphology of the five primary organs (heart, liver, spleen, lung, and kidney tissues) (Fig. [151]7C). The functionality of the liver and kidneys of mice belonging to all groups was evaluated, and no discernible liver or kidney injury was observed among the groups (Fig. [152]7D). These findings suggest that the bionic nano-formulation did not induce systemic toxicity at the administered dose, confirming their favourable biosafety profile. Furthermore, to gain additional insight into the in vivo targeting effect of the nano-formulation, Cy5.5-labelled PLGA-MKP5 and MSCM-PLGA-MKP5 were prepared and intravenously injected into mice with HF. The major organs were imaged ex vivo by using an in vivo IVIS imaging system. Twelve hours after injection, accumulation of Cy5.5-labelled PLGA-MKP5 and MSCM-PLGA-MKP5 were observed in the injured liver. The fluorescence intensity was greater in both the MSCM-PLGA-MKP5 and PLGA-MKP5 groups than in the PBS control group (Fig. [153]7E). Notably, the fluorescence intensity in the MSCM-PLGA-MKP5 group was significantly higher than that in the PLGA-MKP5 group, with statistical analysis indicating approximately twice the strength of PLGA-MKP5 (Fig. [154]7E). The fluorescence intensities of isolated tissues, including the heart, liver, spleen, lungs, and kidneys, were evaluated. In accordance with the findings of the in vivo imaging, a markedly elevated fluorescence signal was observed in the liver compared to other organs (Fig. [155]7F), suggesting a greater propensity for nano-formulation accumulation in the livers of HF mice. Moreover, the MSCM-modified nano-formulation displayed favourable biocompatibility and targeting capabilities. Fig. 7. [156]Fig. 7 [157]Open in a new tab In vivo toxicity and biodistribution of the nano-formulation. A Body weight was monitored over a 15-day period following intravenous treatment with PBS, MKP5, PLGA-MKP5, and MSCM-PLGA-MKP5 (n = 6/group). B Liver weight and the ratio of liver weight to body weight were measured in each group of mice (n = 6/group). C Major organs were collected on day 15 and assessed by HE staining. Scale bar: 100 μm (n = 3/group). D Liver and kidney function, including ALT, AST, BUN, UA, and CRE levels, were determined on day 15 (n = 4–5/group). E Biodistribution of PLGA-MKP5 and MSCM-PLGA-MKP5 were detected using an In Vivo Imaging System. F Biodistribution of isolated organs after PLGA-MKP5 and MSCM-PLGA-MKP5 administration was detected using an In Vivo Optical System. *P < 0.05, **P < 0.01, ***P < 0.001 Nano-formulation regulate CCl[4]-induced hepatic fibrosis through the IRE/XBP1 pathway An HF model was constructed and treated with MSCM-PLGA-MKP5 to evaluate the anti-fibrotic effect of the nano-formulation (Fig. [158]8A). As shown in Fig. S5A, S5B, administration of PLGA-MKP5 and MSCM-PLGA-MKP5 treatment significantly upregulated the protein and mRNA expression of MKP5 compared to the CCl[4]-PBS group. Moreover, MSCM-PLGA-MKP5 exhibited a more pronounced evlevation in MKP5 expression, indicating a greater propensity for nano-formulation accumulation in the livers of HF mice. The liver weight and liver weight/body weight ratio in the CCl[4]-PBS group were significantly higher than those in the control group. Conversely, down-regulation trends were observed in the MSCM-PLGA-MKP5 and PLGA-MKP5 groups compared with the CCl[4]-PBS group. The results of body weight analysis indicated no significant differences among the groups (Fig. [159]8B). Furthermore, the liver function of the mice was examined, and it was observed that the levels of ALT and AST in the serum and Hyp in the liver tissues gradually decreased in both the CCl[4]-PLGA-MKP5 and CCl[4]-MSCM-PLGA-MKP5 groups compared to those in the CCl[4]-PBS group. The MSCM-PLGA-MKP5 group exhibited the most pronounced effects (Fig. [160]8C). Moreover, histological examination revealed structural damage to the liver tissue, including disruption of the hepatic lobule, deviation of the central vein, and vacuolation of the hepatocytes, in the CCl[4]-PBS group compared to the PBS group. Masson's trichrome and Sirus Red staining revealed markedly elevated collagen deposition in the CCl[4]-PBS group compared with the PBS group. However, these abnormalities were reversed in CCl[4]-treated PLGA-MKP5 mice and were even more pronounced in CCl[4]-MSCM-PLGA-MKP5 mice (Fig. [161]8D, S4A). Compared to the control group, the expression of fibrosis-related proteins α-SMA and fibronectin (Fig. [162]8E, S4B) and the transcriptional levels of fibrosis-related genes ACTA2, CoL1A1, and Fn1 (Fig. [163]8F) were markedly increased in the CCl[4]-PBS group. PLGA-MKP5 and MSCM-PLGA-MKP5 demonstrated a reduction in the levels of fibrosis-related proteins and mRNA induced by CCl[4], with a more pronounced effect observed in the MSCM-PLGA-MKP5 group. In addition, PLGA-MKP5 markedly diminished the transcription levels of CCl[4]-induced inflammatory cytokines, including IL-6, IL-1β, TNF-α, TGF-β1, and MCP-1. Moreover, MSCM-PLGA-MKP5 further significantly reduced the mRNA levels of these inflammatory cytokines and chemokines compared to PLGA-MKP5, thereby alleviating the hepatic inflammatory response during HF (Fig. [164]8G). These findings indicated that PLGA-MKP5 and MSCM-PLGA-MKP5 mitigated CCl[4]-induced HF, with the effect of MSCM-PLGA-MKP5 being more pronounced. Subsequently, the expression of p-IRE1α and XBP1s in the liver of mice was examined, revealing that MSCM-PLGA-MKP5 significantly reduced the phosphorylation level of IRE1α and the mRNA level of XBP1s (Fig. [165]8H, I). This observation was consistent with the results of previous studies (Fig. [166]5C–G). These results demonstrate that MSCM-PLGA-MKP5 exerts a suppressive effect on CCl[4]-induced HF via the IRE/XBP1 pathway. Fig. 8. [167]Fig. 8 [168]Open in a new tab Nano-formulation inhibits CCl[4]-induced hepatic fibrosis through the IRE/XBP1 pathway. A Schematic illustrating the procedure of the animal experiments. B Assessment of body weight, liver weight, and the liver weight/body weight ratio in mice (n = 5–7/group). C The serum levels of AST, ALT, and Hyp levels in the liver tissue of mice were evaluated (n = 4–6/group). D Morphology and collagen deposition in liver tissue were examined using HE, Masson’s, and Sirus red staining. Scale bar: 100 μm. E Protein levels of fibronectin and α-SMA in the liver tissue were analysed by western blotting (n = 6/group). F, G The mRNA levels of fibrosis-associated genes ACTA2, CoL1A1, and Fn1 (F), as well as inflammation-related genes IL-6, IL-1β, TNF-α, TGF-β1, and MCP-1 (G) were analysed by real-time PCR (n = 4/group). H The protein expression of p-IRE1α was determined by western blotting, and quantification was performed using the ImageJ software. (n = 6/group). I The mRNA level of XBP1s was estimated by real-time PCR (n = 4/group). *P < 0.05, **P < 0.01, ***P < 0.001 Discussion HF is a reversible pathological process characterised by the excessive accumulation of ECM and a persistent inflammatory response [[169]1]. Various cell types are involved in the pathogenesis of fibrosis [[170]28]. HSCs are regarded as the principal effector cells in HF and are the primary source of pro-fibrotic cytokines and ECM [[171]1, [172]29]. Under physiological conditions, HSCs are located in the Disse space and contain vincristine lipid droplets, thus exhibiting a quiescent phenotype [[173]30]. In their quiescent state, HSCs perform the functions of pericytes and serve as primary storage sites for vitamin A [[174]31]. In the event of liver damage, HSCs undergo activation and transition from a quiescent state to an activated state. HSC activation is initiated by the release of DAMPs from the hepatocytes [[175]32]. Hepatocytes constitute approximately 80% of cells in the liver and are responsible for the primary functions of this organ. They represent the primary targets of several hepatotoxicants, alcohol metabolites, hepatitis viruses, and bile acids [[176]2]. A study has indicated that in adult mice, hepatocytes undergo EMT in response to stimulation by pro-fibrotic growth factor TGF-β, which provides direct evidence for the involvement of hepatocytes in HF formation [[177]33]. Hepatocyte-mediated apoptotic vesicles stimulate Kupffer cells (KCs) to secrete fibrotic cytokines and promote HSC activation through the interaction of toll-like receptors (TLR) with DNA [[178]34]. It has recently been demonstrated that MKP5 plays a role in the regulation of pulmonary and cardiac fibrosis [[179]15, [180]16]. In this study, MKP5 knockout resulted in a worsening of CCl[4]-induced HF, accompanied by an increase in the expression of ECM proteins. Conversely, overexpression of MKP5 inhibited HSC activation and hepatocyte apoptosis, as indicated by TGF-β1. This suggests that MKP5 plays a crucial role in the pathogenesis of HF. One study has demonstrated that inflammation is a crucial factor in the development of fibrosis. Furthermore, the presence of damaged hepatocytes alone may not be enough to directly activate HSCs and cause fibrosis [[181]3]. In instances of liver damage, inflammatory cytokines such as TGF-β, TNF-α, IL-1β, and PDGF induce HSC activation, prompting a transition from a quiescent state to myofibroblast-like cells [[182]5]. Activated HSCs subsequently migrate to the site of injury and secrete ECM, ultimately resulting in the formation of fibrous scar tissue [[183]35, [184]36]. MKP5, a phosphatase of the MAPK pathway that is closely related to inflammation, has been demonstrated in previous studies to regulate the inflammation of adipocytes and islets [[185]12, [186]13]. Moreover, another study demonstrated that the expression of a range of pro-inflammatory cytokines was elevated in MKP5-deficient mice due to T-cell activation [[187]37]. In accordance with the findings, our results indicate that the absence of MKP5 exacerbates CCl[4]-induced hepatic inflammation by upregulating the expression of inflammatory cytokines and chemokines in the liver tissue, HSCs, and hepatocytes. This study demonstrated the role of MKP5 in the amelioration of hepatic inflammation and fibrosis. Nevertheless, the regulatory function of MKP5 in the interaction between hepatocytes and HSCs in HF remains unclear and requires further investigation. ERS is intimately linked to a plethora of ailments and acts as a nexus for autophagy, inflammation, and apoptosis signalling pathways, which actively facilitate the advancement of HF [[188]38]. It has been well established that ERS is activated in a CCl[4]-induced HF model [[189]6, [190]39, [191]40]. Following the onset of ERS, the number of unfolded proteins that bind to GRP78 increases. This results in the separation of IRE1α, PERK, and ATF6 transmembrane proteins, which are typically inactivated by GRP78 binding. This process initiates an UPR, which plays a pivotal role in the pathogenesis of hepatic inflammation and fibrosis [[192]41]. UPR signalling has been observed in HSCs in response to both liver injury in vivo and fibrosis stimulation in vitro [[193]42, [194]43]. Moreover, one study demonstrated that ROS generation of reactive oxygen species disrupts ER homeostasis in HSCs, triggering the UPR and ultimately leading to HSC activation and fibrosis [[195]44]. It has been demonstrated that ERS plays a significant role in the pathogenesis of metabolic liver disease and HF [[196]45]. Furthermore, ERS has been shown to promote HSC activation in a CCl[4]-induced HF model [[197]46]. Given the abundance of hepatocytes in the ER, which is essential for their physiological function, ERS-induced hepatocyte death contributes to the progression of liver injury and HF [[198]47]. When ERS persists in cells, three activated transmembrane proteins on the ER membrane trigger apoptosis. ERS-mediated hepatocyte apoptosis represents a pivotal step in the activation of HSCs [[199]48, [200]49]. This study demonstrates that ERS activation is responsible for HSCs activation and hepatocyte apoptosis and that this ERS activation is further intensified by MKP5 knockout. As one of the three major pathways of ERS, IRE/XBP1 has been reported to be involved in regulating the development of HF [[201]8, [202]50]. Some studies have demonstrated that the IRE1α/XBP1 pathway is activated during TGF-β1-induced HSC activation, which promotes fibrosis development [[203]9, [204]51]. Furthermore, 4μ8C (a noncompetitive inhibitor that blocks IRE1α kinase and ribonucleic acid endonuclease signalling) has been shown to reduce TGF-β-induced expression of sXBP1, Bip, α-SMA, Collagen Iα1, and CTGF [[205]42, [206]43, [207]52]. Our findings support the hypothesis that the IRE/XBP1 pathway is activated during TGF-β1-induced HSCs activation. The same results were observed for TGF-β1-induced hepatocyte apoptosis. Recently, a study has demonstrated that ERS promotes endothelial cell injury through the Nox4/MKP3 interaction [[208]53]. Importantly, several studies have suggested that MKP5 plays a regulatory role in diabetes and apoptosis of epithelial cells, as well as dysfunction of tight junction through the ERS pathway [[209]14, [210]54]. Our findings demonstrate that MKP5 exerts an inhibitory effect on HSCs activation, hepatocyte apoptosis, and inflammation through regulation of the IRE1α/XBP1 pathway, attenuating the progression of HF. These findings suggest that targeting MKP5 is a pivotal strategy for the development of anti-fibrotic therapies. The current treatment for HF primarily encompasses surgical and pharmacological interventions, with the latter including antiviral therapy, interferon, malotilate, lamivudine, vitamin E, silymarin, and polyene phosphatidylcholine drugs [[211]2, [212]55]. However, anti-liver fibrotic medications may elicit a range of adverse effects, such as headaches, nausea, vomiting, and other digestive discomforts. Prolonged use of the same medication may lead to the emergence of drug resistance which diminishes therapeutic efficacy and increases the risk of disease recurrence and progression [[213]56]. Nucleic acid therapy has recently become a topic of interest in the scientific community. In contrast to conventional drug therapies that typically target proteins, gene-based nucleic acid therapies can induce therapeutic effects by regulating gene expression and targeting specific cells in the target tissue [[214]57]. Gene therapy currently enables precise targeting of specific genes or signaling pathways in the HF process, thus achieving more accurate drug delivery and minimizing toxic side effects. Nevertheless, nucleic acids have been employed as therapeutic agents is challenging because of their susceptibility to degradation by nucleases, propensity to elicit immune activation, and unfavourable physicochemical properties that impede cellular uptake [[215]57]. Nano-delivery systems are novel nucleic acid drug delivery systems that employ NPs as drug carriers to facilitate precise delivery of drugs to the targeted lesion site via electrostatic interactions [[216]58]. PLGA, a synthetic polymer with good biocompatibility and biodegradability, is widely employed as a drug delivery carrier for transporting proteins, peptides, and viral DNA [[217]59]. Cellular uptake results demonstrated that PLGA-MKP5 effectively transported MKP5 to the liver, facilitating its activity. NPs function as efficacious delivery systems that protect nucleic acid drugs from degradation, enhance drug stability, and prolong their half-lives. However, the passive targeting effect of NPs is inadequate [[218]17], and non-specific cellular uptake also significantly reduces the efficiency of nano-delivery systems. The liver contains diverse cell types such as resident KCs that produce strong non-specific uptake of NPs, accelerating endocytosis and clearance of NPs, which ultimately results in lower doses reaching the target cells [[219]60]. Coating synthetic NPs with biomimetic cell membrane materials effectively preserves the proteins and polysaccharides present on the cell membrane surface, imparting NPs with surface properties similar to natural cells. This aids in evading recognition by the immune system and subsequent clearance, leading to prolonged circulation in the body and increased accumulation of drugs or therapeutic agents at the target sites. The membranes of red blood cells, platelets, stem cells, and macrophages have been identified as effective natural carriers [[220]61]. MSCM, an important component of stem cells, inherits various biological properties such as cell recognition, low immunogenicity, signal transduction and homing ability. These properties enable the NPs modified by MSCM to accurately identify target cells in drug delivery processes for versatile applications in tumour therapy, inflammatory diseases, tissue regeneration and other fields [[221]22, [222]61, [223]62]. This study describes the development of an MSCM-coated nano-formulation that does not cause systemic toxicity. The accumulation of the MSCM-PLGA-MKP5 in CCl[4]-induced liver tissues increased the local therapeutic concentration of MKP5. Moreover, the MSCM-PLGA-MKP5 demonstrated the ability inhibited the expression of IRE/XBP1 pathway indicators in fibrotic liver tissues, enhancing the anti-hepatic fibrosis effect and preventing the development of HF. The in vivo results demonstrated that MSCM-PLGA-MKP5 enhanced the therapeutic effect of MKP5 on HF and affirmed its clinical potential in the treatment of HF. However, the passive targeting effect of MSCM-coated nano-formulation remains inadequate, necessitating further research to improve the targeting specificity and optimize therapeutic outcomes. While this study addressed some challenges of nano-formulation development for translation purposes, limitations such as uncertainty regarding long-term effects remain. Although the cell membrane-coated NPs were able to escape the initial clearance by the immune system, their long-term stability and safety have not been fully validated. It is essential for future studies to rigorously assess whether the prolong administration of MSCM-coated NPs may lead to any adverse effects on organisms and conduct additional research on biosafety and long-term efficacy. In conclusion, the results of this study demonstrated that MKP5 functions as an anti-fibrotic factor in HF, inhibiting the activation of HSCs and preventing hepatocyte apoptosis. A systemic knockout of MKP5 exacerbates hepatic inflammation and HF in mice. Moreover, administration of the MSCM-coated gene-carrying bionanomimetic NPs PLGA-MKP5 to damaged liver tissues via the nano-delivery system offers protection against CCl[4]-induced HF. Our findings indicate that MKP5 may be a promising target to treat HF. The use of MSCM-PLGA-MKP5 represents a novel strategy for effective anti-fibrotic therapy. Conclusion The current study reveals that the biomimetic MSCM-PLGA-MKP5 ameliorates CCl[4]-induced hepatic inflammation and fibrosis. MKP5 suppresses HSC activation and hepatocyte apoptosis during HF through modulating the IRE/XBP1 pathway. In conclusion, our findings demonstrate that MKP5 may play a crucial role in regulating HF, and that targeting MKP5 could potentially serve as a therapeutic strategy for managing hepatic inflammation and HF. Materials and methods Animals MKP5 knockout (KO) mice were generated by Cyagen Biosciences using CRISPR/Cas9. Briefly, the targeting gDNA-MKP5 exon 2 vector was transferred into the uterus of pseudopregnant C57BL/6J mice via high-flux electroporation of fertilised eggs for KO mouse production. Genotyping was performed by PCR with the following Table [224]1 primers. Table 1. Primers for mouse genotyping Primer sequence (5′–3′) F1:5′-GACAGGAGGTCTCTTACCGTTTC-3′ F2:5′-AGGCTCTGTGTTAAGTCCATCG-3′ R1:5′-GTTAGAAGAAGGCTGCCACTTG-3′ [225]Open in a new tab Male C57BL/6J WT mice aged 6–8 weeks were purchased from Beijing Vital River Laboratory Animal Technology Company (Beijing, CN). Mouse models of CCl[4]-induced hepatic fibrosis All 8–9 week-old WT and KO mice were randomly allocated to four groups: WT: Male: n = 7–8, Female: n = 9–10; KO: Male: n = 6–7, Female: n = 9–10. CCl[4] (Macklin, CN) was dissolved in corn oil (Macklin, CN) at a ratio of 1:9 and intraperitoneally (i. p.) administered to mice at a dose of 1 mL/kg body weight twice per week for 8 weeks, control mice were injected with equal doses of corn oil. Preparation and physicochemical characterisation of MSCM-PLGA-MKP5 One millilitre of dichloromethane containing 20 mg of PLGA-PEI nanoparticles (Xi’an Qiyue Biotechnology Company, CN) was used as the oil phase and added dropwise to 10 mL of ultrapure water for emulsification for 4 h. The internal aqueous and oil phases were combined by ultrasonic treatment for 40 s to prepare the primary lotion. The external emulsion was added to 3 mL of an external aqueous phase containing 2% (w/v) sodium cholate and sonicated in an ice bath for 2 min to form a double emulsion suspension. Dichloromethane was evaporated for 4 h at room temperature, followed by mixing of MKP5 and PLGA-PEI at different mass ratio, and incubation for 30 min. MSCs were cultured in α-MEM (Gibco, USA) supplemented with 10% foetal bovine serum (FBS) (Gibco, USA), 1% (v/v) penicillin–streptomycin solution (Biosharp, CN), and glutamine (HyCyte, CN) at 37 °C in a 5% CO[2] incubator in the following experiments. To extract the MSC membrane (MSCM), 1 × 10^8 MSCs were suspended in 2 mL of PBS and 1% HALT protease inhibitor cocktail (Thermo Fisher Scientific, USA). Cells were enucleated using a dispersant, followed by ultracentrifugation (28,000 rpm, 30 min, 4 °C) with a discontinuous sucrose density gradient dissolved in HEPES buffer (Thermo Fisher Scientific, USA) at concentrations of 55%, 40%, and 30% (w/w). After collection, MSCM was quantified using the BCA method. Subsequently, MSCM vesicles were formed by passing through a single pass of 0.2 μm avanti polar lipids (MERCK, USA) to extrude polycarbonate porous membranes. Finally, MSCM-PLGA-MKP5 was prepared by mixing 1 mg of PLGA-MKP5 with 0.2 mg MSCM, and passing the mixture through a filter membrane nine times. Evaluation of CD45 and CD90 expression on MSCM-PLGA-MKP5 by flow cytometry. The size and zeta potential of PLGA-MKP5 and MSCM-PLGA-MKP5 were measured using a Zetasizer Nano-ZS instrument (Malvern, UK). The morphologies of PLGA-MKP5, MSCM and MSCM-PLGA-MKP5 were visualised using TEM (Tokyo, Japan). In vivo toxicity of nano-formulation All WT mice were randomly assigned to four groups (n = 6/group) and received intravenous injections of PBS, MKP5, PLGA-MKP5, and MSCM-PLGA-MKP5. On the 15th day, mice were euthanised. The histopathology of the major organs (heart, liver, spleen, lungs, and kidneys) was assessed by HE staining. Liver and renal functions were also measured, including ALT, AST, BUN, UA and CRE levels, which were purchased from the Nanjing Jiancheng Bioengineering Institute (CN). Biological distribution of nano-formulation WT mice were intraperitoneally (i.p.) administered 10% CCl[4] (1 mL/kg body weight) twice a week for 8 weeks (n = 4/group). In addition, mice were intravenously injected with PLGA-MKP5 or MSCM-PLGA-MKP5. After 12 h post-injection, the biodistribution of the Cy5.5-labelled PLGA-MKP5 and MSCM-PLGA-MKP5 were analysed using an IVIS spectrum system (PerkinElmer, USA) (748 nm/780 nm). In vivo efficacy of nano-formulation After intraperitoneal injection of CCl[4] for six weeks, CCl[4]-treated WT mice were randomly divided into the following groups: CCl[4]-PBS (n = 5), CCl[4] + PLGA-MKP5 (n = 6), and CCl[4] + MSCM-PLGA-MKP5 (n = 7). The mice received intravenous injections of PBS or PLGA-MKP5 or MSCM-PLGA-MKP5 every other day, and continuing for 2 weeks. Ethics statement All mice were housed in specific pathogen-free, humidity- and temperature-controlled microisolator cages with a 12-h light/dark cycle at Jilin University. All animal experiments were conducted in accordance with the Guidelines for Experimental Animals of Jilin University and approved by the Animal Experiment Ethics Committee of Jilin University (approval number: 20240038). When the study ended, the mice were euthanised by carbon dioxide (CO[2]) asphyxiation. Blood biochemical assay Mouse serum was obtained by centrifuging at 3000 rpm for 15 min. The levels of serum ALT, AST, MDA, and GSH, as well as the Hyp content in liver tissues, were measured using commercially available detection kits from the Nanjing Jiancheng Bioengineering Institute (CN), according to the manufacturer's instructions. Enzyme-linked immunosorbent assay (ELISA) The levels of IL-6 and TNF-α in the serum were quantified using an ELISA kit (Proteintech, CN) following the manufacturer’s instructions. All assays were performed according to the manufacturer’s instructions. Hematoxylin–eosin (HE), Masson and Sirus red staining We commissioned Wuhan Servicebio Technology to perform paraffin embedding and sectioning of the liver tissue. Liver paraffin sections were stained with HE, Masson’s trichrome (Solarbio, CN), and Sirus Red (Biotopped, CN) according to the manufacturer’s instructions. To analyse hepatic collagen distribution, fibrotic septa randomly selected from the right and left liver lobes of six individual mice per group were assessed. Images were captured using Image-Pro Plus software (version 6.0; Media Cybernetics, USA). The extent of collagen staining was quantified as a percentage of the stained area in each liver section. Immunohistochemistry Liver paraffin sections were de-paraffinised and rehydrated for immunohistochemical staining for CD68 (Abclonal, CN). Briefly, paraffin sections were incubated with 3% H[2]O[2] for 30 min to remove endogenous peroxidases. Subsequently, the slides were subjected to antigen retrieval in a preheated antigen repair solution (Beyotime, CN) for 30 min. After blocking with 5% BSA, the slides were incubated overnight at 4 °C with anti-CD68 antibody (Proteintech, CN), followed by incubation with HRP-conjugated secondary antibodies (CST, USA). Finally, the liver tissue sections were visualised using a DAB horseradish peroxidase chromogenic kit (ZSGB-BIO, CN) and counterstained with haematoxylin. Image analysis software Image-Pro Plus 6.0 was used to quantify positively stained areas. Immunofluorescence Frozen tissues were fixed in cold acetone for 10 min, followed by 30-min of incubation with 0.5% Triton X-100 to permeate the cell membrane. Subsequently, the tissues were blocked with 2% goat serum (Beyotime, CN) and then incubated with anti-MKP5 (Abclonal, CN), anti-α-SMA (Santa Cruz, USA), anti-albumin (Affinity, USA), and Ki67 (BIOSS, USA) antibodies overnight at 4 °C, followed by incubation with fluorescence-conjugated secondary antibodies for 1 h. DAPI (Beyotime, CN) was used to stain cell nuclei. Fluorescent images were captured using a laser scanning confocal microscope (Nikon, Japan) and analysed using Image-Pro Plus software (version 6.0) to measure the areas of the positively stained sites. Isolation of primary hepatocytes and HSCs Primary hepatocytes were isolated from male mice using a modified two-step collagenase perfusion protocol, as previously described [[226]63]. Briefly, the mice were anaesthetised, and a V-shaped incision was made to expose the internal organs. The livers were washed with D-Hank’s (Biosharp, CN) balanced salt solution containing EDTA (0.5 mM) and perfused with collagenase IV (Sigma-Aldrich, USA) in D-Hank’s balanced salt solution. Following perfusion, the livers were removed from the mice and the digested hepatocytes were dispersed in Dulbecco’s modified Eagle’s medium (DMEM)-free medium (Corning, USA) and filtered through 100 mesh sieves to remove any undigested debris. The filtrate was centrifuged at 50 ×g for 5 min at 4 °C. The hepatocytes in the precipitate were washed three times with a DMEM-free medium and harvested for subsequent analysis. Primary HSCs were isolated, as described above. Following perfusion, digestion, and centrifugation, liver parenchyma cells were removed, and the remaining non-parenchymal cells (NPC) were collected. The NPC was then re-resuspended in 25% Percoll (GE Healthcare Biosciences AB, USA) and layered between the bottom 75% Percoll buffer and top PBS. After centrifugation at 1 400 ×g for 20 min at 4 °C, the HSCs fraction was obtained at the interface between the top and intermediate layers. Cell viability was assessed using trypan blue staining. Retrovirus packaging The MKP5 gene fragment was cloned into the pLNCX2 vector by gene cloning to construct a recombinant retroviral vector containing the target gene fragment. Briefly, the MKP5 gene with the NotI/Bgl2 site was inserted into the NotI/Bgl2 cloning site of the pLNCX2 retroviral vector via NotI/Bgl2 digestion to obtain the recombinant retroviral vector pLNCX2-MKP5. Recombinant MKP5 overexpression retroviruses were generated by co-transfecting pLNCX2-MKP5, pVSV-G envelope vector, and p-Gag-pol packaging vector into HEK-293 T cells (HyCyte, CN) using Lipofectamine 3000 (Thermo Fisher Scientific, USA). Transfected HEK-293 T cells were incubated for 48 h before viral collection. Subsequently, the virus was concentrated, and its titre was determined for further experiments. The restriction site that interfered with the expression of MKP5 was BamHI/EcoRI, and the vector was pSIREN, which was in accordance with those for MKP5 overexpression, as described above. The retrovirus overexpressing of MKP5 was designated as Retro-PLNCX2/Retro-MKP5, and the retrovirus suppressing MKP5 expression was denoted as Retro-pSIREN/Retro-shMKP5. Cell lines and treatments LX-2 (HyCyte, CN), AML12 (HyCyte, CN), and HEK-293 T (HyCyte, CN) cells were cultured in DMEM supplemented with 10% FBS (Gibco, USA), 100 U/mL penicillin, and 0.1 mg/mL streptomycin (Gibco, USA) at 37 °C with 5% CO[2]. LX-2 and AML12 cells were infected with the virus (Retro-PLNCX2/Retro-MKP5 or Retro-pSIREN/Retro-shMKP5) for 24 h, followed by stimulation with 10 ng/mL TGF-β1 (Sino Biological, CN) for an additional 24 h. For experiments involving IRE inhibition, LX-2 and AML12 cells infected with the virus were preincubated with Kira6 (MCE, USA) for 1 h before stimulation with TGF-β1 plus the aforementioned inhibitors for another 24 h. For experiments involving cellular uptake, LX-2 and AML12 cells were treated with PLGA-MKP5 or MSC-PLGA-MKP5 for 24 h and photographed using a fluorescence microscope (Nikon, Japan). Cell viability and proliferation assay Cell viability and proliferation were assessed using Cell Counting Kit-8 (Sigma, USA) according to the manufacturer’s instructions. Briefly, cells were seeded in 96-well plates at a density of 5 × 10^3 cells/well. For the cell proliferation experiments, LX-2 cells were infected with the virus for 24 h and then stimulated with TGF-β1 (10 ng/mL) for an additional 24 h. For the cell viability experiments, LX-2 and AML12 cells were exposed to PLGA (25, 50, 75, and 100 μg/mL) for 24 h. Subsequently, the cells were incubated with CCK-8 solution (Promega, USA) at 37 °C for 1 h before measuring the absorbance of each well at 490 nm. Transwell invasion and scratch-wound healing assay After infecting LX-2 cells with Retro-MKP5 or Retro-shMKP5 for 24 h, the cells were stimulated with 10 ng/mL TGF-β1 for an additional 24 h. For the Transwell invasion assay, the treated cells were digested and cultured in a petri dish with a pore size of 8 μm for 24 h, fixed, stained, and photographed using a light microscope. For the scratch-wound healing assay, the cells were scraped using a sterile 20 μL pipette head, and the cell fragments were washed with PBS. Subsequently, the cells were cultured for 24 h. Finally, scratched areas were captured at both time points (0 and 24 h) and analysed using ImageJ Pro Plus 6.0. Transferase dUTP nick-end labelling (TUNEL) assay Terminal deoxynucleotidyl TUNEL staining was performed using a TUNEL apoptosis kit (Yeasen, CN) according to the manufacturer's instructions. The area of positive staining was quantified using ImageJ Pro Plus 6.0. Flow cytometry An Annexin V-FITC/PI kit (Elabscience, CN) was used to assess apoptosis. Briefly, AML12 cells were harvested after 24 h of TGF-β1 treatment, washed twice with PBS, suspended in Annexin V-FITC and PI-labelled solution, and incubated at room temperature for 15 min. Subsequently, the ratio of apoptotic cells was quantified using flow cytometry and analysed using FlowJo software (Stanford University, USA). A PI kit (Solarbio, CN) was used to assess the cell cycle. LX-2 cells treated with TGF-β1 were collected and fixed at 4 °C for 2 h by adding precooled 70% ethanol. The samples were then centrifuged at 1500 rpm for 15 min at 4 °C, and the precipitates were collected and washed thrice with PBS. Subsequently, the cells were resuspended in a PI labelling solution and incubated on ice for 15 min. Cell cycle distribution was determined using flow cytometry and was analysed using FlowJo software. RNA preparation and quantitative real-time PCR Total RNA was extracted using TRIzol (Invitrogen, USA) and reverse transcribed with oligonucleotide primers following the manufacturer’s recommended protocol. Subsequently, real-time PCR was performed using SYBR Green Master Mix and ROX on a StepOnePlus system (Thermo Fisher Scientific, USA). The primers for detecting mRNA levels were synthesised by Sangon Biotech, and their sequences are as following Table [227]2. Table 2. Primers for Quantitative Real-time PCR Genes Primer Sequence (5′–3′) GAPDH F:5′-GGAGCCATCTTTGAGCCTTCA-3′ R:5′-GAACCAAACTGAGGAATGGATCT-3′ m ACTA2 F:5′-CGGGAGAAAATGACCCAGATT-3′ R:5′-GGACAGCACAGCCTGAATAGC-3′ m CoL IA1 F:5′-GCACGAGTCACACCGGAACT-3′ R:5′-AAGGGAGCCACATCGATGAT-3′ m Fn1 F:5′-GAAACCTGCTTCAGTGTGTCTG-3′ R:5′-TTGAATTGCCACCATAAGTCTG-3′ m MKP5 F:5′-TTAGACGACAGGGTAGTAGT-3′ R:5′-GCTGGATGAGGGCATATA-3′ m IL-6 F:5′-CCACTTCACAAGTCGGAGGCTTA-3′ R:5′-GCAAGTGCATCATCGTTGTTCATAC-3′ m IL-1β F:5′-GCAACTGTTCCTGAACTCAACT-3′ R:5′-ATCTTTTGGGGTCCGTCAACT-3′ m TNF-α F:5′-AACGCCCTCCTGGCCAA-3′ R:5′-GCAAATCGGCTGACGGTG-3′ m TGF-β1 F:5′-TTGCTTCAGCTCCACAGAGA-3′ R:5′-TGGTTGTAGAGGGCAAGGAC-3′ h IL-6 F:5′-TGAGAGTAGTGAGGAACAAG-3′ R:5′-CGCAGAATGAGATGAGTTG-3′ h MCP-1 F:5′-GATCTCAGTGCAGAGGCTCG-3′ R:5′-TGCTTGTCCAGGTGGTCCAT-3′ h TNF-α F:5′-CTCTCTCTAATCAGCCCTC-3′ R:5′-AGGACCTGGGAGTAGATGA-3′ h IL-1β F:5′-CCTGAGCTCGCCAGTGAAA-3′ R:5′-ATCCAGAGGGCAGAGGTCCA-3′ [228]Open in a new tab The relative mRNA levels normalised to GAPDH were calculated according to the comparative 2^−∆∆Ct method. Western Blotting Cells and liver tissues were lysed using RIPA buffer (EpiZyme Biotechnology, CN) supplemented with a protease inhibitor (Roche, Switzerland). Protein concentrations were determined using a bicinchoninic acid (BCA) protein assay kit (EpiZyme Biotechnology, CN) and adjusted for immunoblotting. Subsequently, the protein samples were separated using 10% SDS-PAGE and transferred to polyvinylidene fluoride (PVDF) membranes. Membranes were blocked with 5% BSA in Tris-buffered saline containing Tween 20 (TBST). The membranes were then incubated with primary antibodies overnight at 4 °C, followed by incubation with HRP-coupled secondary antibodies. Finally, the protein bands were visualised using an enhanced chemiluminescence (ECL) luminescent liquid (YEASEN, CN) on a Multi FluorChem imaging system (Tanon, CN). The primary antibodies used in this study included MKP5 (Abclonal, CN), Collagen Iα1(Rockland, USA), α-SMA (Santa Cruz, USA), Fibronectin (Santa Cruz, USA), cleaved Caspase3 (CST, USA), cleaved Caspase8 (Abclonal, CN), cleaved Caspase9 (Abclonal, CN), E-cadherin (Affinity, USA), N-cadherin (Affinity, USA), Vimentin (CST, USA), p-IRE1α (Affinity, USA), IRE1α (CST, USA), p-P38 (CST, USA), P38 (CST, USA), p-JNK (CST, USA), JNK (CST, USA), p-ERK (CST, USA), ERK (CST, USA), β-Tubulin (Affinity, USA) and β-actin (Abclonal, CN). RNA sequencing analysis RNA sequencing analysis was conducted on fibrotic liver tissues from both the WT and KO mice. Total RNA was extracted using the aforementioned method, and RNA sequencing was performed by Shanghai Jiyan Biotechnology (CN). The data obtained were subsequently analysed using Shanghai NovelBio (CN). Principal component analysis was performed using the Stats and ggplot2 packages in R (version4.0). Hisat2 served as an RNA mapping algorithm to determine the sequence location in the genome by comparing the filtered sequence (clean data) to the reference genome (mm10_Ensembl100) corresponding to the sequenced species (Taxonomy ID:10090). Algorithm software was utilised for normalisation treatment, with P < 0.05 was employed to identify the DEGs. │NES│ > 1, FDR < 0.25, and P < 0.05 was subjected to GSEA analyse. The selected DEGs were subjected to GO and KEGG enrichment analyses, and the Fisher test was used to calculate the significance level (P value) for GO and KEGG analyses. Statistical analysis All data are presented as the mean ± standard deviation (SD) and were analysed using GraphPad Prism9 software. Statistical analyses were performed using one-way ANOVA variance, followed by Tukey’s test for multiple groups. Differences between two groups were analysed using a two-tailed unpaired student’s t-test. Statistical significance was set at P < 0.05. Supplementary Information [229]Additional file 1.^ (2.3MB, docx) Acknowledgements