Abstract Background The metabolic patterns of human placental-derived mesenchymal stem cell (hP-MSC) treatment for primary sclerosing cholangitis (PSC) remain unclear, and therapeutic effects significantly vary due to individual differences. Therefore, it is crucial to investigate the serological response to hP-MSC transplantation through small molecular metabolites and identify easily detectable markers for efficacy evaluation. Methods Using Mdr2^−/− mice as a PSC model and Mdr2^+/+ mice as controls, the efficacy of hP-MSC treatment was assessed based on liver pathology, liver enzymes, and inflammatory factors. Serum samples were collected for ^12C-/^13C-dansylation and DmPA labeling LC–MS analysis to investigate changes in metabolic pathways after hP-MSC treatment. Key metabolites and regulatory enzymes were validated by qRT-PCR and Western blotting. Potential biomarkers of hP-MSC efficacy were identified through correlation analysis and machine learning. Results Collectively, the results of the liver histology, serum liver enzyme levels, and inflammatory factors supported the therapeutic efficacy of hP-MSC treatment. Based on significant differences, 41 differentially expressed metabolites were initially identified; these were enriched in bile acid, lipid, and hydroxyproline metabolism. After treatment, bile acid transport was accelerated, whereas bile acid production was reduced; unsaturated fatty acid synthesis was upregulated overall, with increased FADS2 and elongase expression and enhanced fatty acid β-oxidation; hepatic proline 4-hydroxylase expression was decreased, leading to reduced hydroxyproline production. Correlation analysis of liver enzymes and metabolites, combined with time trends, identified eight potential biomarkers: 2-aminomuconate semialdehyde, l-1-pyrroline-3-hydroxy-5-carboxylic acid, l-isoglutamine, and maleamic acid were more abundant in model mice but decreased after hP-MSC treatment. Conversely, 15-methylpalmitic, eicosenoic, nonadecanoic, and octadecanoic acids were less abundant in model mice but increased after hP-MSC treatment. Conclusions This study revealed metabolic regulatory changes in PSC model mice after hP-MSC treatment and identified eight promising biomarkers, providing preclinical evidence to support therapeutic applications of hP-MSC. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-024-03967-y. Keywords: Mesenchymal stem cell, Metabolomics, Mdr2^−/− mouse, Primary sclerosing cholangitis, Chemical isotope-labeled liquid chromatography-mass spectrometry, Efficacy biomarker Introduction Primary sclerosing cholangitis (PSC) is a liver condition characterized by damage to bile ducts within or outside the liver, or both. The disease process involves an interaction of inflammation, fibrosis, and cholestasis [[40]1]. No genetic or environmental factors are associated with or known to trigger PSC [[41]2, [42]3]. Several drugs targeting biliary components, immunomodulation, fibrosis, or the gut microbiome have been tested in PSC patients [[43]4, [44]5]. Ursodeoxycholic acid (UDCA) has limited efficacy in altering the long-term course of the disease and improving survival [[45]6]. Immunosuppressive medications, including steroids, azathioprine, and cyclosporine, are effective in specific cases according to some reports; however, their overall efficacy remains uncertain and they have substantial adverse effects [[46]7]. Currently, liver transplantation remains the sole definitive treatment. The 10-year recurrence rate after liver transplantation is approximately 20% [[47]8]. Consequently, it is imperative to develop novel therapeutic interventions. Recently, cellular therapy, particularly mesenchymal stem cell (MSC) therapy [[48]9, [49]10], has received considerable attention as a promising therapeutic modality. Friedenstein et al. first identified MSCs in mouse bone marrow, and subsequent evidence has demonstrated that they originate from pericytes and adventitial progenitor cells in almost all tissues [[50]11, [51]12]. Human placenta-derived mesenchymal stem cells (hP-MSCs), found in the fetal membranes of the full-term placenta, are easily obtained using non-invasive techniques. Studies have investigated the potential therapeutic applications of MSCs in mouse models of PSC and organoid models [[52]13–[53]15]. MSCs ameliorate bile duct hyperplasia, peribiliary fibrosis, and inflammation in PSC; they are neither toxic or carcinogenic [[54]15], partially confirming the safety and efficacy of this therapeutic approach. However, the metabolic patterns after MSC therapy remain unclear, and the response to MSC therapy is variable. MSCs improve the clinical phenotype in some individuals, whereas others do not respond or even deteriorate after receiving cell therapy. Thus, it is imperative to discover new biomarkers suitable for evaluating the preclinical safety and effectiveness of MSC transplantation in PSC. Furthermore, the precise pattern of changes in serologic substances after MSC treatment remains unclear. Metabolomics is the extensive examination of biomolecules with molecular weights below 1000 Da in biological fluids, cells, and tissues [[55]16, [56]17]. This approach is invaluable for identifying potential biomarkers [[57]17]. Chemical isotope-labeled liquid chromatography-mass spectrometry (CIL LC–MS) is widely used in metabolomics. This method classifies metabolites based on their chemical groups, uses specific reagents to label each type of metabolite, and subsequently performs LC–MS analysis [[58]18]. Differential isotope labeling enables relative quantification of labeled metabolites in samples [[59]19]. This approach offers enhanced detectability, comprehensive coverage of organismal metabolites, and precise quantification [[60]20]. Specifically, ^12C-/^13C-danylation labeled LC–MS can be applied to amine/phenol metabolite analysis, whereas 2,2-dihydroxy-propanethial (DmPA) labeling is suitable for carboxylate compounds [[61]18]. This study used Mdr2^−/− mice as a PSC disease model and transplanted hP-MSCs as a therapeutic intervention. The efficacy of hP-MSC therapy was validated through comprehensive pathological and biochemical assessments. Mouse serum was collected at multiple time points, and semi-quantitative metabolomics analysis was conducted using Danyl and DmPA labeling LC–MS. Through the identification of metabolic pathways enriched in differentially abundant metabolites and validation of key metabolites and enzymes, we elucidated changes in the metabolic regulatory network after hP-MSC treatment. Furthermore, bioinformatics analysis enabled us to identify potential biomarkers for assessing hP-MSC therapeutic efficacy. Materials and methods Animal models of primary sclerosing cholangitis and hP-MSC treatment Male multidrug resistance gene 2 knockout (Mdr2^−/− or Abcb4^−/−) mice (FVB.129P2-Abcb4tm1Bor/J) and Mdr2^+/+ mice (wild-type, WT) were procured from The Jackson Laboratory (Bar Harbor, ME, USA) and allocated to three groups: Mdr2^+/+ mice as a control group, Mdr2^−/− mice treated or not with hP-MSCs as treatment groups, and the PSC model group. At week 8, the control and model groups received a caudal vein injection of 100 μL phosphate-buffered saline (PBS), whereas the treatment group received an injection of 5 × 10^5 hP-MSCs/100 μL PBS. Each group used to assess treatment effectiveness contained six mice; for serum metabolomics research, the control group comprised 12 mice, and the model and treatment groups each contained 15 mice. All mice were housed in a specific pathogen-free (SPF) facility at the Laboratory Animal Center of The First Affiliated Hospital of Zhejiang University School of Medicine, under a 12 h light/dark cycle with standard mouse chow and water. Mice were anesthetized by isoflurane (95% oxygen and 5% isoflurane, RWD, Shenzhen, China) inhalation and euthanized via cervical dislocation when necessary. All operations were performed with humane care. The experimental animal protocol was approved by the hospital’s Ethics Committee (Approval No: 2020-1088). Serum sample pretreatment Blood samples were collected from mice at weeks 0, 2, 4, and 8 after treatment (mouse ages 8, 10, 12, and 16 weeks). After incubation at room temperature for 1 h, samples were centrifuged at 4 °C and 3500 rpm for 15 min to separate the serum layer. Isolated serum was stored at − 80 °C for future use. To precipitate metabolites, serum samples were mixed with three volumes of methanol (Fisher Chemical, Waltham, MA, USA) chilled to − 20 °C and incubated at − 80 °C for 5 min. Then, samples were centrifuged at 15,000 rpm for 30 min at 4 °C and freeze-dried with a Labconco machine (Kansas City, MO, USA) to produce powdered serum metabolites. An 80-μL aliquot of serum was aspirated from each sample; all aliquots were combined to create a pooled sample that was subjected to the same procedure to obtain powdered metabolites. Dansylation and DmPA-labeling Individual samples were labeled with ^12C-dansyl chloride (Dns-Cl) (Nova Medical Testing, AB, Canada); pooled samples were labeled with ^13C-dansyl chloride (Nova Medical Testing). The metabolite extract (25 μL) was mixed with sodium carbonate/sodium bicarbonate buffer (0.5 M, pH 9.5, Sigma–Aldrich; 12.5 μL) and acetonitrile (ACN; Sigma–Aldrich; 12.5 μL). After thorough mixing, a freshly prepared solution of either ^12C-dandyl chloride (18 mg/mL in ACN, Sigma–Aldrich) or ^13C-dandyl chloride (18 mg/mL in ACN) was added (25 μL). The dansylation reaction was carried out at 40 °C for 60 min using a water bath (Thermo Fisher Scientific, Waltham, MA, USA). Subsequently, NaOH solution (250 mM, 5 μL; Sigma–Aldrich) was added and incubated at 40 °C for 10 min to remove excess dansyl chloride. Finally, formic acid prepared with ACN/H[2]O (425 mM, 25 μL) was added to neutralize and acidify the solution. DmPA bromide labeling was performed according to the method of Peng et al. [[62]21] with modifications. Single samples were labeled with ^12C-DmPA bromide (Nova Medical Testing), whereas pooled samples were labeled with ^13C-DmPA bromide (Nova Medical Testing). Pooled samples were prepared by combining equimolar amounts of individual samples. Both labeled quality control (QC) samples consisted of a mixture containing equimolar masses of ^12C-labeled and ^13C-labeled pooled samples. LC–MS analysis Labeled samples were analyzed using the UltiMate 3000 UHPLC system (Thermo Scientific) coupled with an Impact II Quadrupole Time-of-flight (QTOF) mass spectrometer (Bruker, Billerica, MA, USA). Mobile phase A (MPA) consisted of 0.1% (v/v) formic acid in water; mobile phase B (MPB) contained 0.1% (v/v) formic acid in ACN. The gradient profile for metabolite separation was as follows: at t = 0, the composition was 25% B; at t = 10 min, it reached 99% B and was held constant until t = 13 min; at t = 13.1 min, it returned to the initial condition of 25% B and was held constant until t = 16 min. The flow rate was set to 400 μL/min. To assess instrument repeatability and stability before destination sample analysis, five consecutive QC samples were tested. All samples were randomly arranged, with a blank sample and a labeled standard sample included after every 10 completed runs of destination samples. MS spectra were acquired in positive ion mode with a spectral acquisition rate of 1 Hz. Mouse serum and liver biochemical tests Serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), triglyceride (TG), and high-density lipoprotein cholesterol (HDL) were quantified using a dry biochemical analyzer (DRI-CHEM 4000ie; Fujifilm, Tokyo, Japan). Serum free fatty acid (FFA), total cholesterol (T-CHO), low-density lipoprotein cholesterol (LDL), liver malondialdehyde (MDA), and superoxide dismutase (SOD) levels were assessed with commercial kits (Jian Cheng Bio-engineering Institution, Nanjing, China). Serum interleukin-6 (IL-6), IL-10, IL-1β, and tumor necrosis factor-α (TNF-α) levels were determined using the LEGENDplex™ multifactor detection kit (BioLegend, San Diego, CA, USA), then analyzed using CytoFLEX LX (Beckman Coulter, Brea, CA, USA). Data analysis Using tools provided by OECloud ([63]https://cloud.oebiotech.com), data underwent clustering heatmap, correlation, and metabolic pathway quantitative enrichment analyses. SIMCA-P 14.1 software was used for principal component analysis (PCA) and orthogonal partial least squares discrimination analysis (OPLS-DA). Machine learning analysis was conducted using Orange software (ver. 3.35.0). All experimental data are reported as means ± standard errors of the mean. Statistical significance was assessed through one- or two-way analysis of variance (ANOVA) or a two-tailed Student’s t-test. All statistical analyses were performed using GraphPad Prism software (GraphPad Software, San Diego, CA, USA). In all analyses, P-values < 0.05 were deemed statistically significant. Additional methods Details regarding hP-MSC culture and identification, flow cytometry, immunohistochemical and immunofluorescence staining, oil red O staining, Western blotting, RNA extraction, and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) are provided in the Supplementary Materials (Supplementary Methods, Table S1). Results Liver injury was substantially reduced in the hP-MSC treatment group Osteogenic, chondrogenic, and adipogenic multipotential differentiation (Additional file [64]1: Fig. [65]S1A–D) and flow cytometry immunophenotype profiles (Additional file [66]1: Fig. [67]S1E) were used to confirm the MSC lineage. We monitored body weights of Mdr2^+/+ and Mdr2^−/− mice (Additional file [68]1: Fig. [69]S2A) at four time points and assessed the liver-weight-to-body-weight ratio at the final time point. There was no significant difference in body weight among the three groups (Fig. [70]1A, Additional file [71]1: Fig. [72]S2B), but the liver/body weight ratio decreased (hP-MSC treatment group vs. model group, Fig. [73]1B). Hematoxylin and eosin (H&E) staining of liver sections showed reduced immune cell infiltration in the confluent area and a significant decline in hepatocyte necrosis (Fig. [74]1C). Furthermore, progressive worsening of bile duct and liver lesions was observed in the model group over time. The levels of inflammatory factors IL-6, IL-10, IL-1β, and TNF-α were significantly decreased in 16-week-old mice (treatment group vs. model group, Fig. [75]1D). Serum ALT, AST, and ALP levels increased over time, whereas hP-MSC treatment partially alleviated hepatocyte injury (Fig. [76]1E–G). These findings provide evidence that hP-MSCs are effective for treating PSC. Fig. 1. [77]Fig. 1 [78]Open in a new tab Effectiveness of hP-MSC therapy for PSC. A Mdr2^+/+ mice, Mdr2^−/− mice treated with PBS, and Mdr2^−/− mice treated with hP-MSC, all via tail vein injection, served as the control, PSC model, and treatment groups, respectively. B Liver-weight-to-body-weight ratio in mice at 16 weeks. Compared to the control group, the treatment group showed an increased ratio, but it remained lower than that of the model group; one-way ANOVA. C Hematoxylin and eosin (H&E) staining of mouse livers at 8, 10, 12, and 16 weeks; scale bar = 100 μm. D Serum IL-6, TNF-α, IL-1β, and IL-10 levels in 16-week-old mice; one-way ANOVA. E Changes in ALT, F AST, and G ALP levels; two-way ANOVA; ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001. (D)–(G), n = 6 Serum metabolite profiling indicated that hP-MSC treatment affected endogenous metabolism Serum samples were isotopically labeled and subjected to CIL LC–MS analysis (Additional file [79]1: Fig. [80]S3A). Instrument performance remained stable throughout testing, and the signal responses of samples within each group were consistent (Additional file [81]1: Fig. [82]S3B–D). In dansyl-labeled samples, 1698 peak pairs were identified, compared with 1854 peak pairs in DmPA-labeled samples. This analysis produced 1338 (78.9%) accurately identified or inferred peak pairs with respect to their carbon isotope composition (^12C-/^13C). Additional file [83]1: Table S2 and S3 lists the accurately identified substances. The PCA score plot revealed that QC samples clustered together, indicating excellent LC–MS stability and repeatability, ensuring reliable data. The metabolic profile of the treatment group was more similar to the control than to the model group (Fig. [84]2A). The robustness of the OPLS-DA supervised model was confirmed by 200-replacement validation (Additional file [85]1: Fig. [86]S3E and F). The OPLS-DA score plot demonstrated discernible intergroup separation and intragroup aggregation among the three groups (Fig. [87]2B), suggesting that Mdr2^−/− mice undergoing MSC treatment experienced substantial changes in amino/phenol hydroxyl and carboxyl metabolites compared with untreated mice. Fig. 2. [88]Fig. 2 [89]Open in a new tab Metabolomics analysis of serum. A Score plots of PCA and B OPLS-DA in the control, model, and treatment groups and QC samples. C Volcano plots of the control vs. model groups and model vs. treatment groups at 10 weeks. |FC|> 1.2, P < 0.05. D Venn diagram of metabolites that differed at consecutive time points. E Heatmap of mean relative abundances of 41 metabolites that differed at consecutive time points. F Quantitative enrichment analysis of metabolic pathways based on the 41 differentially expressed metabolites To identify significant metabolites responsible for metabolomic changes, paired t-tests and fold change (FC) values were used to compare changes in peak pairs. The volcano plot revealed 1896, 1678, and 1539 differential peak pairs between the control and model groups at 10, 12, and 16 weeks, respectively. Similarly, there were 888, 491, and 762 differential peak pairs between the model and treatment groups at the same time points (Fig. [90]2C, Additional file [91]1: Fig. [92]S4A and B). Venn diagram analysis of these differentially expressed metabolites from intergroup comparisons clarified common changes across all groups. At week 10, 636 differential peak pairs were shared between the control and model groups and model and treatment groups; there were 319 peaks at week 12 and 525 peaks at week 16 (Additional file [93]1: Fig. [94]S4C). From these shared peaks, we identified corresponding substances at each time point: 149, 64, and 138 substances at weeks 10, 12, and 16, respectively. Furthermore, the intersection of differentially expressed metabolites among these three groups yielded metabolites that consistently changed across all three time points (17 species), as well as those that changed over two consecutive time points (18 + 6 species) (Fig. [95]2D). Ultimately, this approach identified 41 differentially expressed metabolites (Additional file [96]1: Table S4). Temporal changes in these metabolites were visualized using heat maps (Fig. [97]2E), revealing that MSC treatment induced significant differences in the metabolic profiles of the model and treatment groups at subsequent time points. These findings suggested a shift in the metabolic profile of the mice after hP-MSC treatment. Next, we performed quantitative enrichment analysis based on chemical structures to obtain an overview of the main classes of differential metabolites. The most enriched classes between the treatment and model groups were amino acids and peptides, followed by fatty acids and their derivatives (Additional file [98]1: Fig. [99]S4D). To investigate changes in metabolic pathways, we performed quantitative enrichment analysis of 41 different metabolites using the KEGG database ([100]https://www.kegg.jp/) and identified 27 significantly perturbed pathways between the treatment and model groups (Fig. [101]2F and Additional file [102]1: Table S5). The major metabolic pathways encompassed biosynthesis of unsaturated fatty acids, tryptophan metabolism, alanine, aspartate, and glutamate metabolism, lysine degradation, nicotinate and nicotinamide metabolism, and arginine and proline metabolism, consistent with the prominent amino acids and fatty acids identified in the differential metabolite categories. Because the 41 differential metabolites included bile acids, alcohols and derivatives, fatty acids, and fatty acids with proline that play crucial roles in pathway analysis, we hypothesized that MSC treatment influences specific metabolic processes, such as dysregulation of bile acid, lipid, and proline metabolism. Disrupted bile acid metabolism was restored after hP-MSC treatment Of the 41 metabolites identified in the initial screening, changes in the glycine derivatives glycodeoxycholic acid and N-heptanoylglycine suggested alterations in bile acid content after hP-MSC treatment. We investigated the regulatory effects of hP-MSC on bile metabolism and its efficacy in alleviating bile stasis. Immunohistochemical analysis of cytokeratin 19 (CK19) from 16-week-old mice (Fig. [103]3A and B) showed significant bile duct cell proliferation and bile duct reaction in the hepatic parenchyma of the model group. MSC treatment reduced epithelial cell proliferation. Furthermore, serum total bile acid levels of 16-week-old mice revealed a steady decrease after MSC treatment (Fig. [104]3C). To elucidate specific changes in bile acids, we comprehensively searched the identified compounds for additional bile acids. This search revealed 17 bile acids (Additional file [105]1: Table S6). The thermograms from all time points in three groups (Additional file [106]1: Fig. [107]S5A and B) showed that the model group had higher levels of bile acid sludge and toxic bile acids compared with the control group, but these levels improved after treatment. Most bile acids exhibited the greatest improvement at 16 weeks (Fig. [108]3D). Various bile acids, including α-phocaecholic acid, significantly decreased in the treatment group (Fig. [109]3E). Notably, reduced gene expression levels of cytochrome P450 family 7 subfamily A member 1 (Cyp7a1) and cytochrome P450 family 27 subfamily B member 1 (Cyp27b1) (Fig. [110]3F) in the treatment group suggested a decrease in bile acid biosynthesis. The gene expression levels of basolateral transporter multidrug resistance-associated protein 2 (Mrp2), Na ( +)/bile acid cotransporter (Ntcp), and organic anion transporting polypeptides (Oatp4) were reduced (Fig. [111]3G), indicating increased bile acid excretion. Collectively, MSC treatment improved bile acid metabolism and alleviated cholestasis in Mdr2^−/− mice. Fig. 3. [112]Fig. 3 [113]Open in a new tab hP-MSC treatment remedied bile acid metabolism disorders. A Immunohistochemical staining of CK19 in mouse livers; scale bar = 100 μm. B The quantification of CK19 positive area in mouse livers (n = 5). C Serum total bile acid levels in 16-week-old mice (n = 6). D Heat map of the 17 bile acids identified at 16 weeks. E Typical bile acids altered at 16 weeks (n = 12–15). F Hepatic mRNA levels of Cyp7a1 and Cyp27b1 and G Mrp2, Ntcp, and OATP4 in three groups of mice (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA hP-MSC treatment strongly influenced lipid metabolism Metabolic pathway enrichment analysis indicated that unsaturated fatty acid synthesis underwent significant changes after MSC treatment. A comprehensive database search identified the main unsaturated fatty acids, particularly ω-3 and ω-6 polyunsaturated fatty acids (PUFAs). The majority of PUFAs increased post-treatment, approaching levels observed in the control group (Additional file [114]1: Fig. [115]S6). To identify the mechanisms regulating differential fatty acid expression, we assessed the gene expression level of key regulatory enzymes associated with unsaturated fatty acids based on KEGG regulatory networks (Fig. [116]4A). Delta-5 desaturase (Fads1) and acyl-CoA 6-desaturase (Fads2) belong to the family of fatty acid desaturases, which introduce double bonds at specific carbon atoms in fatty acid chains to regulate their degree of unsaturation. After MSC treatment, the expression of Fads1 increased. Conversely, no significant differences relative to the control group were noted in the expression levels of stearoyl-CoA desaturase 1 (Scd1) and acyl-CoA thioesterase 2 (Acot2), which regulate saturated fatty acid production to form monounsaturated fatty acids and catalyze acyl-CoA hydrolysis into free fatty acids and CoA, respectively. Furthermore, upregulated expression levels of ELOVL2 (elongation of very long chain fatty acids-like 2) and ELOVL5, involved in regulating omega-3 and omega-6 PUFA elongation, were observed after MSC treatment. Figure [117]4B shows the integrated network depicting the unsaturated fatty acid synthesis process and its regulation by key enzymes. In summary, overall upregulation was observed in the unsaturated fatty acid synthesis pathway after treatment. Because unsaturated fatty acids had a substantial impact on overall lipid metabolism, we next evaluated a series of lipid-related parameters in mice. Fig. 4. [118]Fig. 4 [119]Open in a new tab hP-MSC treatment moderated lipid metabolism disorders. A Expression of hepatic genes involved in unsaturated fatty acid synthesis. B Network of ω-3 and ω-6 series unsaturated fatty acid biosynthesis. In the ω-3 series, α-linolenic acid (ALA, 18:3) is primarily converted into stearidonic (18:4), eicosapentaenoic (EPA, 20:5), docosapentaenoic (DPA, 22:5), and docosahexaenoic (DHA, 22:6) acids. In the ω-6 series, linoleic acid (LA, 18:2) is mainly converted into γ-linolenic (GLA, 18:3), dihomo-γ-linolenic (DGLA, 20:3), arachidonic (ARA, 20:4), adrenic (ADA, 22:4), and docosapentaenoic (DPA, 22:5) acids. Red indicates an increase after treatment, blue indicates no difference after treatment, and gray indicates undetermined substances or untested genes. Δ represents desaturase. (C) Serum FFA, T-CHO, LDL, HDL, and TG levels (n = 6). D Oil red O staining showed that the TG level declined after hP-MSC treatment; scale bar = 100 μm. E The quantification of lipid droplet area in mouse livers (n = 5). F Expression of hepatic genes involved in cholesterol synthesis, G lipogenesis, and H mitochondrial β-oxidation (n = 6). Liver MDA I and SOD J levels (n = 6). K Immunohistochemistry staining showed that γ-H2AX declined after hP-MSC treatment. L The quantification of γ-H2AX positive area in mouse livers (n = 5). M Nrf2, HO-1, and NQO1 gene expression levels in mouse livers (n = 6). ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA Initially, we assessed serum lipid-related biochemical indices. Serum FFA and T-CHO levels were reduced in Mdr2^−/− mice, whereas MSC treatment increased these levels (Fig. [120]4C). This change was attributed to reduced hepatic lipid accumulation; serum T-CHO levels were lower in Mdr2^−/− mice than in control mice [[121]22]. LDL levels were reduced in both the control and treatment groups, whereas they were elevated in the model. HDL was decreased in Mdr2^−/− mice, but its level did not increase after MSC treatment (Fig. [122]4C). MSCs may selectively internalize LDL. There were no pronounced fluctuations in TG levels among the three groups. Examination of oil red O-stained liver samples (Fig. [123]4D and E) confirmed the absence of significant changes. Analysis of key genes involved in lipid and cholesterol metabolism—sterol regulatory element binding transcription factor 2 (Srebp2) and 3-hydroxy-3-methylglutaryl-CoA reductase (Hmgcr)—did not show significant changes after MSC treatment (Fig. [124]4F). The lipogenic gene sterol regulatory element-binding protein 1c (Srebp1c) also showed no significant change compared with the model group (Fig. [125]4G). Previous studies have consistently revealed reduced mitochondrial function during cholestasis [[126]23, [127]24], characterized by impaired fatty acid oxidation [[128]23]. Therefore, we investigated pivotal genes in fatty acid oxidation. The mRNA levels of peroxisome proliferator activated receptor α (Pparα) and its target genes PPARγ coactivator 1α (Pgc1α), carnitine palmitoyltransferase 1A (Cpt1α), and acyl-coenzyme alternative oxidase (Aox) (Fig. [129]4H) were decreased in the model group; these levels increased after MSC treatment. This finding indicated decreased fatty acid breakdown in Mdr2^−/− mice during cholestasis, with potential MSC-induced improvement through the promotion of β-oxidation. Furthermore, β-oxidation of fatty acids enhanced unsaturated fatty acid synthesis. Excessive lipid deposition led to toxic injury; changes in hepatic MDA levels (F[130]ig. [131]4I) and hepatic SOD activity (Fig. [132]4J) indicated an antioxidant response. Immunohistochemical analysis of histone γ-H2AX staining (Fig. 4K, L) also indicated an attenuated oxidative damage response in MSC-treated mice compared with model mice. Increased levels of heme oxygenase 1 (Hox1), nuclear factor erythroid 2-related factor 2 (Nrf2), and NAD(P)H quinone dehydrogenase 1 (NQO1) in Fig. [133]4M indicated initiation of the antioxidant defense mechanism. hP-MSC treatment improved hydroxyproline-dependent collagen metabolic disorders Analysis of key components in the arginine and proline metabolic pathways revealed significant downregulation of proline l-1-pyrroline-3-hydroxy-5-carboxylate, trans-4-hydroxyl-proline, and 1-pyrroline-5-carboxylic acid (P5C) after MSC treatment. P5C is an intermediary product derived from glutamate and ornithine, bridging the tricarboxylic acid and urea cycles. However, no significant changes were observed in glutamate or ornithine levels after treatment (Fig. [134]5A and B). The intermediate step in these pathways involves enzymatic conversion of proline to hydroxyproline, catalyzed by prolyl 4-hydroxylase (Fig. [135]5A). Considering the close association of proline and hydroxyproline metabolism with collagen formation and degradation [[136]25], Mdr2^−/− mice might exhibit disrupted collagen fiber metabolism. To assess fibrosis, we performed Sirius red and Masson staining (Fig. [137]5C and D), which revealed a significant increase in hyperplastic collagen fibers surrounding bile ducts in model mice; this phenomenon was considerably improved in the treated mice. Alpha-smooth muscle actin (α-SMA) staining showed myofibroblast proliferation and aggregation (Fig. [138]5C and D). Hepatic mRNA levels of α-SMA, collagen alpha 1 chain type I (Col1a1), TIMP metallopeptidase inhibitor 1 (Timp1), and transforming growth factor beta 1 (Tgf-β1)—all markers of fibrosis—also indicated that MSC treatment ameliorated collagen fiber metabolic disorders in mice (Fig. [139]5E). Paha2 encodes a critical subunit of prolyl 4-hydroxylase, an essential enzyme involved in collagen synthesis, which facilitates the 4-hydroxyproline production necessary for correct folding of recently formed procollagen molecules. Studies have revealed elevated P4HA2 mRNA levels in individuals with PSC, highlighting its contributions to biliary tract reactivity and fibrosis development [[140]26]. Using qRT-PCR, we observed upregulated gene expression in the livers of model mice; however, after MSC treatment, the Mdr2^−/− mice showed decreased expression (Fig. [141]5F). These changes were validated at the protein level (Fig. [142]5G). Immunohistochemical analysis revealed predominant expression of P4HA2 in the fibrotic tissue areas, with decreased expression after treatment (Fig. [143]5 and I). According to reports, P4HA2-positive cells display the morphological characteristics of biliary reactive cells [[144]26]. To further validate this observation, we performed immunofluorescence staining for P4HA2 and CK7 (Fig. [145]5J). CK7 immunostaining delineated clusters of biliary cells in the peribiliary region that responded to biliary liver injury. Consistent with the immunohistochemical findings, there was extensive overlap in the distribution of P4HA2^+ cells and CK7^+ biliary cells. Furthermore, P4HA2 expression was rarely detected in normal mice. In control group mice, P4HA2 expression increased, while it decreased in the treatment group. Consequently, our findings suggest that hP-MSC therapy affected hydroxyproline metabolism in mice. Fig. 5. [146]Fig. 5 [147]Open in a new tab hP-MSC treatment alleviated proline‑dependent collagen metabolism disorders. A Schematic diagram of proline metabolic pathways. Blue, decreased after treatment; yellow, no change after treatment; gray, not identified; red, key enzyme. Solid lines indicate direct transformation, whereas dotted lines represent omitted intermediates. B Relative composition of key metabolites involved in proline metabolism (n = 12–15). C Sirius red, Masson, and immunohistochemical staining of α-SMA in mouse livers; scale bar = 100 μm. D Assessment of fibrosis based on Sirius red-, Masson-stained and α-SMA positive regions (n = 5). E Hepatic expression of α-SMA, COL1A1, TIMP1, and TGF-β1 (n = 6). F Hepatic expression of P4HA2 (n = 6). G Western blot analysis of the liver showed greater P4HA2 expression in the model group, which was downregulated by hP-MSC treatment. H Immunohistochemical staining of P4HA2 in mouse livers. Arrows indicate positive areas. ns, not significant. I The quantification of P4HA2 positive area in mouse livers (n = 5). J Representative confocal images of CK7, P4HA2 and DAPI in mouse livers. ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA Differential metabolites associated with clinical phenotypes The above experiments revealed the effect of MSC treatment for PSC from a metabolomic perspective and further illustrated the credibility of serum sample metabolomics in this study. We obtained 41 differentially expressed metabolites as potential biomarkers of treatment effectiveness. To filter these potential biomarkers and determine whether any are linked to disease severity, we conducted Spearman correlation analysis of their relative abundances with clinical phenotypes; specifically, we monitored the ALT, AST, and ALP levels of the three groups at each time point (Additional file [148]1: Fig. [149]S7A–C). This analysis showed that the effect of MSC treatment was more pronounced at 16 weeks. Therefore, we regarded liver enzyme data at 16 weeks as the clinical phenotype to further analysis. The calculated correlation coefficients and significance levels are recorded in Additional file [150]1: Table S7. As shown in Fig. [151]6A and Additional file [152]1: Fig. [153]S7D, most metabolite factors were associated with disease severity. Among these, l-1-pyrroline-3-hydroxy-5-carboxylic acid, a breakdown product of hydroxyproline, was positively correlated with liver enzymes (Fig. [154]6B), and monounsaturated eicosenoic acid was negatively correlated with liver enzymes (Fig. [155]6C). Next, we used the criterion |ρ|> 0.6 and p < 0.05 to screen the 41 differential metabolites, identifying eight differential metabolites: l-1-pyrroline-3-hydroxy-5-carboxylic acid, maleamic acid, 2-aminomuconate semialdehyde, l-isoglutamine, 15-methylpalmitic acid, octadecanoic acid, eicosenoic acid, and nonadecanoic acid. Generally, metabolite levels became normalized after treatment. A significant difference at two or three consecutive time points might indicate a treatment-induced change in metabolites. Additional file [156]1: Fig. [157]S8 shows that all eight metabolites followed the expected trend, confirming their suitability as potential biomarkers of MSC treatment effectiveness. Fig. 6. [158]Fig. 6 [159]Open in a new tab Associations between metabolites and clinical phenotypes. A Correlation heatmap of metabolites and liver enzymes (Spearman analysis, n = 42). Red signifies a positive correlation, whereas blue denotes a negative correlation. Gray represents correlation coefficients |ρ|< 0.6 or p < 0.05. B–C Examples of individual liver enzyme–metabolite associations. Logistic regression classifiers comprising liver enzymes, metabolites, and their combination could discriminate effective and ineffective individuals in the D training and E test sets. *P < 0.05, **P < 0.01, ***P < 0.001 Eight metabolites as potential biomarkers for evaluating hP-MSC therapy efficacy To investigate whether these eight serum metabolites could serve as biomarkers to determine MSC treatment effectiveness, we performed machine learning modeling. Serum samples from Mdr2^−/− mice administered either PBS or hP-MSCs were randomly assigned in a 2:1 ratio to the training and test sets, respectively. We studied seven fivefold cross-validated machine learning models: logistic regression (LR), support vector machine (SVM), random forest (RF), neural network (NN), naive Bayes (NB), adaBoost (AB), and k-nearest neighbor (kNN) (Additional file [160]1: Tables S8–S10). Machine learning model classification effectiveness was assessed using metrics such as the area under the receiver operating characteristic curve (AUC), accuracy, F1-score, precision, and recall. For comprehensive comparison, we selected the logistic regression model. Figure [161]6D shows that metabolites were more effective than liver enzymes in distinguishing the effectiveness of MSC treatment (AUC 0.933 and 0.843). The combination of liver enzymes and metabolites yielded better results (AUC 0.947). Our conclusions were verified by data from the testing set (Fig. [162]6E). The AUCs for liver enzymes, metabolites, and their combination were 0.757, 0.932, and 0.978, respectively. The confusion matrix also confirmed the validity of the model and the reliability of the selected biomarkers (Additional file [163]1: Fig. [164]S9). Our study indicated that these biomarkers (Table [165]1 and Fig. [166]7) can effectively evaluate MSC efficacy in PSC treatment. This finding provides a reliable scientific basis for individualized treatment decisions and will inform future clinical practice. Table 1. Eight identified serum biomarkers No RT (s) Normalized RT (s) m/z_light m/z_heavy Neutral mass (Da) Intensity nCharge nTag External Identifier Compound S144 138.6 128.7 291.1338 293.1405 129.0424 1,002,302.47 1 1 [167]C04281 L-1-Pyrroline-3-hydroxy-5-carboxylic acid S192 159.8 151.9 277.1183 279.1251 115.0269 65,500 1 1 [168]C01596 Maleamic acid S372 221.9 213.7 303.1341 305.141 141.0426 28,216 1 1 [169]C03824 2-Aminomuconate Semialdehyde S376 222.1 214 469.2444 473.2578 146.069 1,942,488.886 1 2 [170]C16673 L-Isoglutamine S1832 746.6 777.9 432.3462 434.3533 270.2559 710,872 1 1 HMDB0061709 15-Methylpalmitic Acid S1846 769 814.3 446.3621 448.3688 284.2707 2,706,476.994 1 1 [171]C01530 Octadecanoic acid S1847 769.9 815.2 472.3777 474.3844 310.2864 1,362,167.2 1 1 [172]C16526 Icosenoic acid S1854 792.9 838.2 460.3778 462.3846 298.2865 41,572.52632 1 1 [173]C16535 Nonadecanoic acid [174]Open in a new tab No.: individual peak pair number coded from the LC–MS experiment; RT (s): the retention time of the peak pair detected in sec; Normalized RT (s): the corrected retention time of the peak pair with Universal RT Calibrant data; m/z light: the mass of light-chain (^12C) labeled metabolite in the peak pair; m/z heavy: the mass of heavy-chain (^13C) labeled metabolite in the peak pair; Neutral Mass (Da): the neutral monoisotope mass of the metabolite (i.e., labeled mass—the mass of the labeling group); Intensity: the average intensity of the heavy labeled metabolite in the peak pair in all samples; nCharge: the number of charge in the peak pair detected; nTag: the number of labeling groups or tags in the labeled metabolite; External Identifier and Compound: the external identifier and name of metabolites positively identified using accurate mass and retention time matches against a labeled standard compound library, respectively Fig. 7. [175]Fig. 7 [176]Open in a new tab Relative levels of eight potential biomarkers at weeks 8, 10, 12, and 16. n = 12–15. ns, not significant; *P < 0.05, **P < 0.01, ***P < 0.001; unpaired t-test with Welch correction Discussion This study demonstrated that hP-MSC treatment affects metabolic disorders in Mdr2^−/− mice, a PSC model, with particularly strong influences on bile acid, lipid, and hydroxyproline metabolism. Although the beneficial effects of hP-MSCs on PSC have been confirmed through pathological and biochemical analysis [[177]27–[178]29], the precise underlying mechanism has been unclear. Therefore, a multi-timepoint metabolomics analysis was conducted to elucidate changes in small molecule metabolites during treatment. Through a series of bioinformatics analyses, 41 differential metabolites were initially identified; quantitative metabolic pathway enrichment analysis was then performed. Differences between MSC treatment and model groups were primarily observed in pathways such as unsaturated fatty acid synthesis, proline metabolism, and tryptophan metabolism. Differential metabolites included one bile acid and three glycine derivatives. Thus, it is reasonable to hypothesize that MSC therapy influences the metabolic profiles of bile acids, lipids, and proline. The balance of bile acid levels is disrupted in various liver disorders, especially those involving cholestasis and bile duct damage. Bile acids play a crucial role in PSC progression [[179]30]. The primary route for bile acid synthesis, the classical or neutral pathway, is initiated by CYP7A1-mediated cholesterol 7α-hydroxylation. This enzyme regulates the overall rate of bile acid production and participates in approximately 90% of all bile acid synthesis. Bile acids are amphiphilic molecules synthesized via hepatic cholesterol metabolism, comprising primary bile acids directly produced by hepatocytes and secondary bile acids formed upon intestinal secretion. In humans, most bile acids are conjugated with glycine, and the remainder are conjugated with taurine [[180]31]. In hepatocytes, bile acid transport between blood and bile is facilitated by hepatic uptake transporters, such as NTCP and OATPs. After attachment to intracellular bile acid-binding proteins, bile acids are released into tubules via efflux transporters, such as bile salt export pump (BSEP) and MRP. Subsequently, bile acids are transported via bile ducts to the intestinal lumen, where they aid lipid breakdown and cholesterol emulsification for absorption [[181]32]. Yang et al. investigated metabolic changes in three cholestatic mouse models, revealing elevated bile acid levels and reduced arginine concentrations [[182]33]. In this study, we identified 17 bile acids and their intermediates within the metabolic group; most bile acids exhibied significant elevation in the model group and subsequent reduction after hP-MSC treatment. MSCs modulate various bile acid transporters and key enzymes involved in synthesis, thereby regulating the entire bile acid metabolism network and mitigating the detrimental systemic effects of cholestasis. Additionally, comprehensive bile acid data throughout the study period demonstrated that MSCs had the greatest therapeutic efficacy at 16 weeks. Considering the crucial role of bile acids in regulating lipid and energy balance, changes in lipid metabolism are inevitable during cholestatic liver disease [[183]22, [184]34, [185]35]. Lipid levels in PSC typically exceed those recommended for treatment with lipid-lowering drugs [[186]36]. In the analysis of 41 differential metabolites and their pathway enrichment, we observed that unsaturated fatty acid synthesis plays a central role in metabolic processes. Most unsaturated fatty acids were found at lower levels in model group, whereas they showed significant increases in the control and treatment groups. PUFAs are widely acknowledged for their health-promoting properties and crucial physiological functions, particularly in terms of regulating cholesterol levels and inflammatory responses [[187]37, [188]38]. Studies have demonstrated that PPAR binds to the ω-3 fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), ultimately inhibiting cytokine production associated with the NF-κB inflammatory master transcription factor [[189]39]. To elucidate the impact of hp-MSCs on lipid metabolism, we conducted supplementary experiments. Serum biochemical data demonstrated that hP-MSCs effectively reduced LDL content in the model group, but no significant impact on HDL content was observed. We attributed this finding to MSCs’ preferential affinity for LDL loading. Furthermore, metabolic profiles indicated that cholestasis led to disrupted lipid metabolism. Detection of key genes involved in lipid metabolism showed that MSC treatment primarily enhances fatty acid β-oxidation. Mitochondrial dysfunction plays a pivotal role in cholestatic disorders and is frequently associated with impaired fatty acid β-oxidation [[190]40]. Reduced PUFA levels promote oxidative stress, thereby exacerbating hepatic injury [[191]41, [192]42]. Lipid accumulation in hepatocytes and subsequent reactive oxygen species production can cause liver inflammation and injury by activating Kupffer cells and hepatic stellate cells [[193]43]. The primary mechanism underlying this hepatotoxicity is believed to be the inhibition of fatty acid β-oxidation [[194]44]. Assessments of liver MDA and SOD levels, along with γ-H2AX pathological staining, revealed that hP-MSCs effectively mitigated oxidative stress-induced hepatocyte damage. Furthermore, we observed upregulation of the Nrf2 signaling pathway after treatment. Recent findings indicate that the transcription factor Nrf2 plays a role in maintaining lipid balance, along with its known antioxidant and anti-inflammatory properties [[195]45, [196]46]. Nrf2 depletion resulted in reduced β-oxidation gene expression [[197]47].We speculate that the alleviation of lipid accumulation-induced oxidative injury results from activation of the Nrf2/HO-1 pathway facilitated by hP-MSCs. Proline metabolism is important in the development of chronic cholestatic disorders [[198]48, [199]49]. We observed a substantial decrease in the amount of l-1-pyrroline-3-hydroxy-5-carboxylic acid, a breakdown product of hydroxyproline, as well as the levels of hydroxyproline in the control and treatment groups. Collagen synthesis regulation is intricately linked to proline and hydroxyproline metabolism [[200]50]. Based on collagen staining and the assessment of key fibrosis-related genes, we propose that hP-MSCs effectively ameliorate aberrant collagen turnover in mice. Prolyl 4-hydroxylase is a crucial regulatory node in the proline metabolism network that catalyzes the formation of 4-hydroxyproline, a pivotal enzyme in collagen synthesis [[201]51, [202]52]. Zhang et al. reported that the livers of individuals with PSC express more P4HA2, specifically in close proximity to cells involved in the ductal response, suggesting that P4HA2 targeting could be a useful therapeutic approach for biliary response and fibrosis [[203]26]. P4HA2 targeting may also reduce collagen deposition in hepatocellular carcinoma [[204]53]. Our investigation revealed significantly elevated P4HA2 expression in treatment group livers compared with those of model mice; this phenomenon was reversed after treatment intervention with hP-MSCs. Therefore, we hypothesize that hP-MSCs can improve collagen metabolic disorders by P4HA2 targeting, but the specific mechanism requires further investigation. We identified altered metabolic pathways and sought potential biomarkers from the differentially expressed metabolites. After initial screening, we detected 41 differential metabolites. Through correlation analysis and validation of machine learning models, we ultimately assessed the efficacy of eight potential biomarkers. Of these, maleamic, 15-methylpalmitic, octadecanoic, eicosenoic, and nonadecanoic acids are fatty acids or their derivatives; conversely, 2-aminomuconic acid semialdehyde, l-isoglutamine, and l-1-pyrroline-3-hydroxy-5-carboxylic acid are amino acids or their derivatives. A notable upward trend in the levels of all fatty acids or derivatives was observed post-treatment, except for maleamic acid, which may be attributed to increased unsaturated fatty acid synthesis. In contrast, a downward trend was evident in several amino acid derivatives after treatment, especially concerning l-1-pyrroline-3-hydroxy-5-carboxylic acid, a hydroxyproline metabolite for which reduction may be linked to decreased collagen hydroxyproline synthesis. Furthermore, 2-aminomuconic acid semialdehyde is a byproduct of tryptophan metabolism, suggesting that its alteration is associated with changes in gut microbiota activity [[205]54]. The abundances of serum biomarkers may be useful in establishing an effective system for MSC product monitoring and assist future PSC applications. Our AUC analysis indicated that the accuracy of these biomarkers is superior to that of liver enzymes. Nevertheless, these results should be verified in larger studies and human participants; this need for validation constitutes a limitation of our study. In addition, further studies are needed to uncover the precise mechanisms behind the metabolic changes during hP-MSC treatment. Conclusion We clarified the therapeutic benefits of hP-MSCs in PSC by improving imbalances in bile acid, lipid, and collagen metabolism in a hydroxyproline-dependent manner. Furthermore, we identified eight potential biomarkers that can evaluate hP-MSC therapy effectiveness in PSC. These findings offer novel insights into clinical applications of stem cell therapy. Supplementary Information [206]Additional file 1.^ (3.7MB, docx) Acknowledgements