Next Article in Journal
Spontaneous Adverse Drug Reaction Reporting of Congenital Malformations: A Danish National Register Study
Previous Article in Journal
Curcumin-like Compound Inhibits Proliferation of Adenocarcinoma Cells by Inducing Cell Cycle Arrest and Senescence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Combined Network Pharmacology, Transcriptomics and Metabolomics Strategies Reveal the Mechanism of Action of Lang Chuang Wan to Ameliorate Lupus Nephritis in MRL/lpr Mice

1
College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
2
College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
3
College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(6), 916; https://doi.org/10.3390/ph18060916
Submission received: 14 April 2025 / Revised: 2 June 2025 / Accepted: 13 June 2025 / Published: 18 June 2025
(This article belongs to the Section Pharmacology)

Abstract

:
Background: Lupus nephritis (LN) is a serious complication of systemic lupus erythematosus (SLE) and is difficult to cure. Lang Chuang Wan (LCW) has been widely used in clinical practice as a treatment for SLE and LN, but its active ingredients and mechanism of action have not been elucidated. To address this, we aim to analyze LCW’s chemical components and clarify its mechanisms in treating LN. Methods: We utilized ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS) to analyze the components of LCW and assessed its effects on MRL/lpr mice through ELISA, H&E staining, Masson’s trichrome staining, and IgG immunofluorescence. Then, we further explored the mechanisms of action through network pharmacology, transcriptomics, and metabolomics, and validated with Western blot. Results: LCW contained 1303 chemical components, primarily flavonoids and terpenoids. It significantly improved kidney pathology and normalized levels of serum ANA, anti-dsDNA, anti-Sm, C3, C4, Cr, BUN, IL-6, IL-10, IL-17, TNF-α, and urinary protein (UP) in MRL/lpr mice. Network pharmacology, transcriptomics, and metabolomics indicated that LCW’s therapeutic effect on LN involved the PI3K/AKT pathway, confirmed by Western blot showing LCW’s suppression of the PI3K/AKT/mTOR pathway. Conclusions: LCW alleviates pathological symptoms in MRL/lpr mice by inhibiting the PI3K/AKT/mTOR signaling pathway, providing insights into its therapeutic mechanisms for lupus nephritis.

1. Introduction

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that can lead to damage across various organs in the body [1]. Lupus nephritis (LN) is a prevalent and serious complication of SLE, impacting around 30 to 40% of individuals diagnosed with the disease [2]. LN primarily manifests as glomerular damage, characterized by inflammation, cell proliferation, necrosis, and injury to podocytes and tubular epithelial cells [3]. The disease has a high recurrence rate, severely impacting patients’ quality of life [4]. Its pathogenesis involves a complex interplay of genetic, epigenetic, hormonal, and other factors [5]. LN treatment is typically divided into two phases: the initial phase and the maintenance phase [6], with corticosteroids, antimalarials, immunosuppressants, and biologics being the primary therapies [7]. While these treatments are effective, their side effects cannot be overlooked. More critically, despite aggressive treatment, some patients may still progress to end-stage renal disease (ESRD) [8]. This highlights the limitations of current treatment strategies and underscores the urgent need for safer, more effective alternative or adjunct therapies.
Traditional Chinese medicine (TCM) has shown potential in treating autoimmune diseases due to its unique characteristics, including multi-component, multi-target, and bidirectional regulatory properties [9]. Lang Chuang Wan (LCW), a compound formula containing 16 TCM ingredients, has been part of the Chinese pharmacopeia since 2005. LCW is known for its heat-clearing, detoxifying, blood-cooling, and circulation-promoting effects and has demonstrated promising efficacy in treating SLE. Existing studies indicate that LCW modulates inflammation in resiquimod-induced RAW264.7 mouse cells [10]. Research by Guoxin Ji and colleagues have found that LCW significantly reduced urinary protein (UP) levels, anti-double-stranded DNA antibodies (anti-dsDNA), anti-nuclear antibodies (ANA), and inflammatory cytokines (TNF-α, IL-6) in a mouse model of SLE-like disease [11]. These results suggest that LCW may alleviate inflammation, regulate immune responses, and protect kidney function, although its underlying mechanisms warrant further investigation.
In recent years, network pharmacology has emerged as a crucial tool for uncovering the mechanisms underlying the actions of TCM [12]. Additionally, the rapid advancement of high-throughput technologies, such as metabolomics, has enabled the identification of biological processes that have already occurred [13], playing a crucial role in autoimmune disease research [14]. Transcriptomics, with its capacity for quantitative analysis and efficient screening, can identify and quantify thousands of differentially expressed genes [15], providing a powerful approach for exploring the complex pathological mechanisms of LN [16]. These advanced technologies offer new perspectives and strategies for modern research on LCW. In this study, we utilize ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS) to identify the major active components of LCW. Enzyme-linked immunosorbent assay (ELISA) is employed to assess the therapeutic effects of LCW in treating LN. Histopathological analysis using H&E staining, Masson’s trichrome staining, and immunofluorescence (IF) is conducted to observe renal improvements in MRL/lpr mice treated with LCW. By integrating visual analysis from network pharmacology, transcriptomics, and metabolomics, we predict the potential mechanisms of LCW in treating LN. Finally, Western blot is used to validate these mechanisms, clarifying LCW’s specific role in regulating the expression of key proteins.

2. Results

2.1. Active Components of LCW

Using UPLC-MS/MS technology, 1303 metabolites were identified in LCW, and their base peak chromatogram (BPC) is shown in Figure 1. The top 25 active components are listed in Table 1. The active components of LCW are predominantly natural compounds, including flavonoids, terpenoids, and phenolic acids.

2.2. LCW Regulates the Levels of Serum Autoantibodies and Inflammatory Cytokines in MRL/lpr Mice, Enhancing Renal Function and Reducing Kidney Damage

The ELISA test results (Figure 2A–L) indicate that, compared to the control group, the levels of ANA, anti-dsDNA, and anti-Sm were significantly elevated in the model group (p < 0.01). Following treatment, antibody titers in the PA group, LCW-L group, and LCW-H group showed significant reductions, with p < 0.01, p < 0.05, and p < 0.01, respectively. Regarding renal function markers, the levels of Cr, BUN, and UP in the model group were significantly higher than those in the control group (p < 0.01). After treatment with PA and LCW, these levels were significantly restored. Specifically, in the PA and LCW-H groups, Cr, BUN, and UP levels were significantly reduced (p < 0.01). The LCW-L group also showed significant reductions in Cr, BUN, and UP levels (p < 0.05, p < 0.05, p < 0.01, respectively). For inflammation markers, compared to the control group, the levels of IL-6, IL-17, and TNF-α in the model group were significantly elevated (p < 0.01), while IL-10 was significantly decreased (p < 0.01). After treatment, the levels of inflammatory cytokines in the PA group were significantly improved (p < 0.01). In the LCW-L group, although IL-17 levels did not show a significant change, IL-6 and TNF-α levels were significantly reduced (p < 0.05), and IL-10 showed a trend towards increased levels (p < 0.05). In the LCW-H group, all tested cytokines IL-17 (p < 0.05), IL-6 (p < 0.01), and TNF-α (p < 0.01) were significantly reduced, while IL-10 was significantly elevated (p < 0.01). Additionally, the levels of complement components C3 and C4 in the model group were significantly lower than those in the control group (p < 0.05), with a more significant reduction in C4 (p < 0.01). Following treatment, C3 and C4 levels in the PA group were significantly increased (p < 0.01). In the LCW-H group, both C3 and C4 levels were significantly elevated (p < 0.05). However, the LCW-L group did not significantly affect the levels of C3 and C4.
H&E staining results (Figure 2M) show that the glomeruli in the control group are intact, with no abnormal pathological structures observed. In contrast, the model group mice exhibited glomerular capillary hyperplasia, increased glomerular volume, reduced Bowman’s space, focal tubular epithelial cell edema with deformation and necrosis, and tubular lumen dilation. Compared to the model group, the severity of renal lesions in the PA, LCW-L, and LCW-H groups was significantly reduced. Masson’s staining results (Figure 2N,O) revealed that, compared to the control group, the area of renal interstitial fibrosis in the model group was significantly increased (p < 0.01). After treatment, the area of renal interstitial fibrosis in the PA, LCW-L, and LCW-H groups was significantly reduced (p < 0.01, p < 0.05, p < 0.01, respectively). IF results (Figure 2P,Q) showed that, compared to the control group, the deposition of IgG immune complexes in the model group was significantly increased (p < 0.01). After treatment, IgG immune complex deposition in the PA and LCW-H groups was significantly reduced (p < 0.01 and p < 0.05, respectively). Conversely, while the LCW-L group showed a decrease in IgG deposition, this change was not statistically significant.
Combining the serum biomarker data with the pathological analysis results, LCW-H demonstrated more significant effects in regulating immunity, exerting anti-inflammatory effects, improving renal function, and inhibiting fibrosis, when compared to LCW-L. Based on these findings, subsequent analyses will focus on exploring the mechanisms and pathways through which LCW-H exerts its effects.

2.3. Network Pharmacology Prediction for LCW Treatment of LN

A total of 965 potential targets for the active components of LCW and 1634 targets related to LN were identified. Among these two sets, 249 common targets were found (Figure 3A). These overlapping targets were imported into the STRING database and analyzed using Cytoscape 3.7.2 software to construct a PPI network (Figure 3B). The network contained 248 nodes and 2453 edges. Based on an analysis of network parameters including degree, betweenness centrality, and closeness centrality seven key targets were identified (Figure 3C). GO analysis (Figure 3D) revealed that the biological processes (BP) most commonly associated with these targets included responses to xenobiotic stimuli, lipopolysaccharide, and inflammation. Cellular components (CC) were predominantly located in extracellular exosomes, cytoplasm, and the plasma membrane. Molecular functions (MF) primarily involved enzyme binding, protein binding, and signaling receptor binding. The KEGG pathway analysis (Figure 3E) indicated that the LCW treatment for LN potentially influences several critical signaling pathways, such as the PI3K-AKT pathway, Mitogen-Activated Protein Kinase (MAPK) pathway, and the Nuclear Factor Kappa B (NF-κB) pathway.

2.4. Transcriptomics Analysis Results

RNA sequencing (RNA-seq) analysis was performed on the transcripts from the control, model, and LCW-H groups for comparison, generating a total of 114.74 Gb of clean data. Each sample’s clean data reached 5.68 Gb, with a Q30 base percentage of 93.15% or higher. The clean reads from each sample were then mapped to the designated reference genome, with alignment efficiency varying between 95.88% and 97.31%, demonstrating the high quality of the transcriptomic sequencing. PCA score plots (Figure 4A,B) revealed clear separation between the control and model groups, as well as between the model and LCW-H groups. Figure 4C–F display the DEGs between the control and model groups, and between the model and LCW-H groups, respectively. In comparison to the control group, the model group exhibited 1166 genes that were upregulated and 401 genes that were downregulated (Figure 4C). In contrast, the LCW-H group exhibited 236 upregulated genes and 987 downregulated genes relative to the model group (Figure 4D). A comparison of the two transcriptomic datasets identified 745 overlapping DEGs (Figure 4E). Figure 4F presents the hierarchical clustering analysis of DEGs, illustrating expression changes in the model group that were subsequently reversed by LCW treatment. Further Gene Ontology (GO) analysis of the DEGs, shown in Figure 4G, revealed that BP were predominantly associated with key pathways such as apoptosis, protein phosphorylation, and the positive regulation of gene expression. In terms of CC, these processes primarily occurred in essential structures like the cytoplasm, cell membrane, and mitochondria. MF analysis highlighted important functions, including protein binding, nucleotide binding, and ATP binding. KEGG pathway analysis (Figure 4H) revealed notable enrichment of the DEGs in several key pathways, including the FOXO signaling pathway, PI3K-AKT signaling pathway, and MAPK signaling pathway.

2.5. Results of Metabolomics Analysis

Metabolomics data from the control, model, and LCW-H groups were analyzed using multivariate statistical methods with an LC/MS system. PCA was conducted to offer a comprehensive overview of the data, identify potential outliers, and evaluate the variations in serum metabolites across the three groups. Figure 5A clearly illustrates the significant differences in metabolic profiles between the control, model, and LCW-H groups. The OPLS-DA score plots (Figure 5B,C) highlight the intergroup differences between control vs. model and model vs. LCW-H, with VIP values reported. These results suggest that LCW intervention effectively ameliorated the metabolic disturbances in MRL/lpr mice. Through screening, 42 biomarkers closely associated with lipid metabolism were identified, which exhibited significant changes following LCW-H treatment (Table 2). Heatmap analysis (Figure 5D) further underscores the significant impact of LCW-H on the metabolic characteristics of MRL/lpr mice, revealing its prominent regulatory effect. Using the BMKCloud platform for analysis of these candidate biomarkers (Figure 5E) showed significant enrichment in metabolic pathways, including steroid biosynthesis, steroid hormone biosynthesis, and arachidonic acid metabolism (p < 0.05).

2.6. Integrated Multi-Omics Analysis of LCW Treatment in MRL/lpr Mice

By integrating network pharmacology with transcriptomic data, 27 overlapping genes were successfully identified (Figure 6A). Figure 6B shows the correlation heatmap between 27 overlapping genes and 42 potential biomarkers. These genes were then combined with 42 candidate biomarkers and analyzed together using the MetaboAnalyst 6.0 platform (https://www.metaboanalyst.ca/) (accessed on 24 November 2024). The results (Figure 6C) revealed significant enrichment in various metabolic processes, including the generation of primary bile acids, steroid hormone biosynthesis, steroid production, formation of unsaturated fatty acids, as well as the FOXO and PI3K-AKT signaling pathways (p < 0.05).

2.7. LCW Inhibits Key Proteins in the PI3K/AKT/mTOR Pathway to Improve LN

The Western blot analysis results indicated that the levels of phosphorylated PI3K, phosphorylated AKT, and phosphorylated mTOR proteins were significantly higher in the experimental group compared to the control group (p < 0.01). Following LCW intervention, in the LCW-L group, there were no statistically significant changes in the expression of p-PI3K and p-mTOR when compared to the model group. However, the expression of p-AKT protein was significantly reduced (p < 0.05). In the LCW-H group, a significant down-regulation of p-PI3K, p-AKT, and p-mTOR protein expression was observed compared to the model group. (p < 0.05, p < 0.01, p < 0.01, respectively) (Figure 7A–D). Notably, the total PI3K, AKT, and mTOR expression remained unaltered in both groups (Figure 7E–G).

3. Discussion

LN is a kidney inflammation associated with SLE, and LCW is a traditional Chinese medicine preparation containing various bioactive components used in the treatment of LN. Although LCW has been used clinically, its mechanism of action remains unclear. This study utilized the MRL/lpr mouse model, along with UPLC-MS/MS, ELISA, histopathology, network pharmacology, transcriptomics, metabolomics, and Western blot techniques, to investigate the effective components of LCW in treating LN and its underlying mechanisms. This study is the first to elucidate the mechanisms by which LCW treats LN, providing further evidence for its clinical application, promotion, and the development of new drugs for treating LN.

3.1. Key Bioactive Components of LCW in LN Therapy

Initially, we identified 1303 active components in LCW, including quercetin, luteolin, and berberine, using UPLC-MS/MS technology. Studies have shown that quercetin and its derivatives can significantly suppress the production of pro-inflammatory cytokines, such as IL-6, IL-8, and TNF-α [17]. Additionally, quercetin can lower plasma concentrations of BUN and Cr, improving renal histopathological conditions in MRL/lpr mice [18]. Luteolin has been found to ameliorate pathological abnormalities and enhance renal function by reducing oxidative stress and UP levels in MRL/lpr mice [19]. Berberine exhibits anti-inflammatory and anti-renal fibrosis effects [20,21]. These properties suggest that the diverse bioactive constituents in LCW may play a substantial role in slowing the progression of LN.

3.2. LCW Ameliorates LN via Immune Regulation, Anti-Inflammatory Effects, and Renal Protection

To explore the potential mechanisms of action of LCW in the treatment of LN, a systematic pharmacodynamic evaluation was conducted using MRL/lpr mice [22]. ANA, as an initial indicator for screening LN, yield positive results, prompting further testing for anti-dsDNA to clarify the autoimmune response [23]. The titer of anti-dsDNA is closely associated with LN activity and serves as a key indicator for assessing disease status and monitoring therapeutic response [24]. Additionally, anti-Sm antibodies provide significant auxiliary diagnostic value [25]. Together, these three antibodies are essential markers of autoimmune activity in the diagnosis of LN. LCW treatment significantly reduced the levels of ANA, anti-dsDNA, and anti-Sm, suggesting that LCW can improve the pathological state of LN by inhibiting excessive autoimmune responses. Cr and BUN are primary indicators for assessing glomerular filtration function [26], while UP results from the destruction of the glomerular filtration barrier (GFB), which is a hallmark clinical manifestation of LN [27]. Therefore, Cr, BUN, and UP are critical markers of renal function impairment in LN. LCW treatment significantly improved the levels of Cr, BUN, and UP, indicating its substantial therapeutic effect in protecting renal function. The progression of LN is also closely linked to the abnormal levels of certain inflammatory cytokines. Studies have shown that TNF-α and IL-6 are involved in the pathogenesis of LN [28], promoting inflammatory responses and exacerbating kidney damage [29]. Moreover, IL-17 plays a dual role: it is both a disease-promoting cytokine in inflammatory conditions and an essential component of the immune system’s defense against infections [30]. IL-10, with its anti-inflammatory properties, may help restore immune balance [31]. LCW treatment reduced the levels of IL-6, IL-17, and TNF-α while increasing IL-10 levels, demonstrating its significant anti-inflammatory and immune-regulatory effects. Complement system components C3 and C4 are critical indicators of immune complex deposition and inflammatory responses [32]. LCW therapy notably increased C3 and C4 levels, suggesting its ability to inhibit excessive complement activation, which may contribute to improved management of LN.
Renal biopsy is considered the gold standard for diagnosing and predicting the prognosis of LN [33]. Inflammation is a hallmark of LN, making the control of glomerulonephritis a key therapeutic target. Fibrosis, a pathological process characterized by the continuous accumulation of connective tissue within the organ, leads to a reduction in parenchymal cells and damage to the organ’s structure and function. Organs that have sustained long-term damage and inflammation are particularly susceptible to fibrosis [34]. After LCW intervention, there was a significant reduction in inflammatory cell infiltration around the glomeruli in MRL/lpr mice. Additionally, interstitial fibrosis was alleviated, and IgG immune complex deposition decreased. These results suggest that LCW effectively improves renal lesions in MRL/lpr mice.

3.3. Network Pharmacology Reveals LCW’s Anti-LN Mechanisms

Network pharmacology analysis has identified key targets, such as the overexpression of TNF-α and IL-6, which exacerbate the symptoms of LN [35,36]. STAT3 has been proposed as a potential novel therapeutic target for LN [37]. Subsequent GO and KEGG analyses suggest that LCW may regulate gene expression and modulate the PI3K/Akt signaling pathway in the treatment of LN. Studies have shown that the activation of the PI3K/Akt/mTOR signaling pathway plays a crucial role in the pathogenesis of LN in mice [38]. These findings suggest that LCW may modulate the expression of factors such as TNF-α, IL-6, and STAT3, interfere with the PI3K/Akt signaling pathway, and exert anti-inflammatory and immune-modulatory effects, thereby slowing the progression of LN.

3.4. Transcriptomic Analysis

Transcriptomic analysis revealed that the GO and KEGG enrichment of DEGs was significantly associated with gene expression regulation, the FOXO signaling pathway, and the PI3K-AKT signaling pathway, consistent with predictions from network pharmacology. These findings suggest that LCW exerts its therapeutic effects on LN through multiple pathways and targets [10]. Previous studies have shown that FOXO3a plays a crucial role in inflammatory and autoimmune processes, and its upregulation alleviates symptoms by inhibiting the PI3K-AKT signaling pathway, a mechanism that helps reduce the severity of LN [39]. Additionally, FOXO1 overexpression significantly reduces glomerular cell proliferation and 24 h UP levels in MRL/lpr mice [40]. The PI3K/AKT/mTOR pathway is considered a key pathological feature of LN [41]. This pathway induces ribosome biogenesis, protein synthesis, cell growth, and the secretion of pro-inflammatory and mitotic factors, while inhibiting autophagy through the regulation of transcription factors such as FOXO [28]. These findings underscore the importance of targeting the PI3K/AKT/mTOR axis in the treatment of LN.

3.5. Metabolic Profiling Analysis

Metabolic profiling analysis identified 42 potential biomarkers closely linked to lipid metabolism. These biomarkers are primarily involved in lipid metabolic pathways such as steroid biosynthesis, steroid hormone biosynthesis, and arachidonic acid metabolism. This suggests that LCW-H may regulate key enzymes or metabolites in these pathways, influencing inflammatory responses and immune modulation, and subsequently correcting lipid metabolism disorders induced by LN. Existing studies have established that abnormal lipid metabolism is closely associated with the pathogenesis of LN [11], potentially disrupting lipid homeostasis in renal tissue and exacerbating glomerular and tubulointerstitial diseases [42]. Furthermore, disrupted lipid metabolism and its metabolites may stimulate the generation of IgG autoantibodies and cytokines, worsening the progression of LN. Importantly, the PI3K/AKT pathway plays a pivotal role in lipid metabolism regulation [43]. Based on these findings, we hypothesize that LCW-H may regulate lipid metabolism through the PI3K/AKT pathway to exert therapeutic effects on LN.

3.6. Integrated Multi-Omics Analysis

The integrated analysis using network pharmacology, transcriptomics, and metabolomics has revealed significant enrichment in pathways such as primary bile acid biosynthesis, the FOXO signaling pathway, and the PI3K-AKT signaling pathway in the treatment of LN with LCW. Notably, the PI3K/AKT signaling pathway plays a pivotal role in regulating metabolism, inflammation, and immune responses in immune cells [44]. In LN, the abnormal activation of the PI3K-AKT pathway and its downstream mTOR signaling pathway is directly linked to inflammation, immune dysregulation, and renal fibrosis. This pathway promotes the proliferation and activation of renal fibroblasts, which are key contributors to kidney injury [39,45]. Therefore, future studies will focus on elucidating the specific mechanisms by which LCW modulates the PI3K/AKT/mTOR signaling pathway in the treatment of LN.

3.7. LCW Ameliorates LN via Suppression of PI3K/AKT/mTOR Signaling

The PI3K pathway functions through its regulatory p85 and catalytic p110 subunits, which activate the AKT/PKB effector downstream. AKT exists in three isoforms AKT1, AKT2, and AKT3 each playing crucial roles in cell signaling. mTOR, a serine/threonine kinase, governs cellular metabolism and is vital for autophagy [46]. As a downstream target of the PI3K/AKT pathway, mTOR acts as a negative regulator of autophagy by inhibiting the UNC-51-like autophagy-activating kinase (ULK) complex and preventing the formation of autophagosomes [47]. Disruption of the autophagy exacerbates inflammatory cytokine production and impairs apoptotic cell clearance, both of which contribute to the progression of LN [48]. We employed Western blot to assess key proteins within the PI3K/AKT/mTOR signaling pathway. The results revealed significant upregulation of p-PI3K, p-AKT, and p-mTOR in kidney tissue from MRL/lpr mice, consistent with prior studies [46,49]. Increased levels of p-p85 (PI3K) suggest that its activation could be a pivotal driver of aberrant renal cell proliferation and survival. Activation of PI3K leads to the conversion of PIP2 to PIP3 on the cell membrane, recruiting AKT to the membrane and promoting its phosphorylation to p-AKT [50]. As a central molecule in this signaling cascade, p-AKT regulates cell survival and growth by phosphorylating downstream target proteins. mTOR is subsequently activated through p-AKT, and its aberrant activation disrupts immune balance, promotes autoimmune responses, induces renal fibrosis, inhibits autophagy, and contributes to kidney damage [51]. The PI3K/AKT/mTOR pathway is highly activated in MRL/lpr mice, underscoring its critical role in LN pathogenesis [45]. Following LCW treatment, we observed a significant reduction in p-AKT expression in the LCW-L group, although changes in p-PI3K and p-mTOR levels were not statistically significant. In contrast, in the LCW-H group, p-PI3K, p-AKT, and p-mTOR expression were all notably downregulated, suggesting that LCW-H effectively inhibits the abnormal activation of the PI3K/AKT/mTOR pathway. In addition, previous studies have shown that berberine can downregulate the PI3K/AKT pathway, thereby reducing apoptosis of renal tubular epithelial cells [52]. It can also inhibit mTOR, influencing energy metabolism and promoting apoptosis [53]. Luteolin and quercetin have been found to inhibit the activation of the PI3K/AKT signaling pathway and the formation of renal fibrosis [54,55,56]. This inhibitory effect may play a crucial protective role by promoting renal cell autophagy and reducing apoptosis, thereby slowing the progression of kidney damage. These findings imply that LCW may treat LN through the inhibition of the PI3K/AKT/mTOR signaling pathway, offering a potential strategy for managing fibrosis associated with LN and regulating cellular autophagy processes.

3.8. Limitations and Prospects

The present study employs the MRL/lpr mouse model to simulate LN, which is widely used in LN research. However, compared to human LN, there are still differences in its pathophysiological processes and immune responses. Therefore, future research needs to be conducted in models that are closer to human LN to further validate the therapeutic effects and mechanisms of LCW. Meanwhile, omics data primarily reflect changes in biological processes and cannot directly prove causality. Therefore, more in-depth experimental validations, such as gene knockout and signal pathway inhibitors, are required to clarify the targets and mechanisms of LCW’s action. Despite these limitations, this study provides new insights and evidence for LCW’s treatment of LN. Future research can further explore the active components and targets of LCW and conduct clinical trials to potentially offer new effective treatments for LN.
In summary, this study has revealed that the active components in LCW, including quercetin, luteolin, and berberine, play a significant role in exerting anti-inflammatory and immunomodulatory effects, while also improving renal function in MRL/lpr mice. Furthermore, our findings demonstrate that LCW-H exerts its protective effects by inhibiting the abnormal activation of the PI3K/AKT/mTOR signaling pathway. However, to fully elucidate the precise mechanisms underlying LCW’s therapeutic actions, further research is required.

4. Materials and Methods

4.1. Drugs and Reagents

Lang Chuang Wan (LCW, batch number National Medicine Approval Z22021973) was provided by Changchun Haiwai Pharmaceutical Group Co., Ltd. (Changchun, China), with a dosage of 0.5 g per tablet. Prednisone Acetate Tablets (PA, National Medicine Approval H12020689) were supplied by Tianjin Tianyao Pharmaceutical Co., Ltd. (Tianjin, China), with a dosage of 5 mg per tablet.

4.2. UPLC–MS/MS Analysis of LCW Components

Accurately weigh 15.6 mg of LCW powder and transfer it into a 2 mL centrifuge tube. Add 600 µL of methanol containing 2-amino-3-(2-chloro-phenyl)-propionic acid (at 4 ppm). Vortex for 30 s, then add steel balls and place the tube in a tissue grinder. Grind for 60 s at 55 Hz. Perform ultrasound treatment at room temperature for 15 min, followed by centrifugation at 12,000 rpm and 4 °C for 10 min. Filter the supernatant through a 0.22 µm membrane and transfer it to a detection vial for UPLC-MS/MS analysis [57].
The analysis was conducted using a Vanquish UHPLC System (Thermo Fisher Scientific, Waltham, MA, USA) for liquid chromatography. Chromatographic separation was achieved with an ACQUITY UPLC® HSS T3 column (2.1 × 100 mm, 1.8 µm) (Waters, Milford, MA, USA), maintained at a consistent temperature of 40 °C. The flow rate and injection volume were set at 0.3 mL/min and 2 μL, respectively. For LC-ESI (+)-MS analysis, the mobile phases consisted of 0.1% formic acid in acetonitrile (v/v) (B2) and 0.1% formic acid in water (v/v) (A2). The chromatographic gradient was as follows: 0–1 min, 8% B2; 1–8 min, 8% to 98% B2; 8–10 min, 98% B2; 10–10.1 min, 98% to 8% B2; 10.1–12 min, and 8% B2. For LC-ESI (−)-MS analysis, acetonitrile (B3) and 5 mM ammonium formate (A3) were used as the mobile phases. The gradient for this analysis was as follows: 0–1 min, 8% B3; 1–8 min, 8% to 98% B3; 8–10 min, 98% B3; 10–10.1 min, 98% to 8% B3; 10.1–12 min, and 8% B3 [58]. Metabolite detection was carried out using a Q Exactive mass spectrometer (Thermo Fisher Scientific, USA), equipped with an ESI ion source. Both MS1 and MS/MS data were obtained in Full MS-ddMS2 mode (data-dependent MS/MS). The instrument parameters were as follows: sheath gas pressure of 40 arb, auxiliary gas flow of 10 arb, spray voltages of 3.50 kV (ESI+) and −2.50 kV (ESI−), capillary temperature of 325 °C, MS1 scan range of m/z 100–1000, MS1 resolving power of 70,000 FWHM, 10 data-dependent scans per cycle, MS/MS resolving power of 17,500 FWHM, normalized collision energy of 30 eV, and automatic dynamic exclusion [59].

4.3. Animal and Experimental Design

Eight-week-old female BALB/c and MRL/lpr mice, each with a body weight of 20 ± 2 g, were obtained from Spero Biotech Co., Ltd. (Beijing, China), under the animal license number SCXK (Jing) 2019-0010. The mice were housed in a standard specific pathogen-free (SPF) environment, maintained on a 12 h light/dark cycle, with free access to both food and water. The study was conducted in accordance with ethical guidelines, and the protocol was approved by the Ethics Committee of Changchun University of Chinese Medicine (approval number: 2023513).
After a one-week acclimatization period, six BALB/c mice were assigned to the control group (control), and 24 MRL/lpr mice were randomly divided into four groups (n = 6) based on a random number table: the model group (model), the positive control group (PA, 5.0 mg/kg), the low-dose Lang Chuang Wan group (LCW-L, 1.6 g/kg), and the high-dose Lang Chuang Wan group (LCW-H, 3.2 g/kg). Both the control and model groups were administered the same volume of normal saline via gavage. All mice were treated daily via gavage for 8 weeks. Urine samples were collected at weeks 0, 2, 4, 6, and 8. At the conclusion of the study, all mice were euthanized, and blood samples along with kidney tissue were collected. Blood samples were centrifuged at 3000 rpm for 10 min at 4 °C to obtain serum, which was used for ELISA and serum metabolomics analyses. Kidney tissue was rapidly excised and stored as follows: portions of the tissue were immediately frozen in liquid nitrogen and stored at −80 °C for subsequent transcriptomics and Western blot analyses, while the remaining tissue was fixed in 4% paraformaldehyde for histopathological examination.

4.4. ELISA Analysis

The serum levels of ANA, anti-dsDNA, anti-Sm antibodies (anti-Sm), complement components C3 and C4, serum creatinine (Cr), blood urea nitrogen (BUN), UP, IL-6, IL-10, IL-17, and tumor necrosis factor-α (TNF-α) in mice were measured using an ELISA kit (Jiangsu Meimian Industrial Co., Ltd., Yancheng, China). All procedures were carried out in accordance with the manufacturer’s guidelines.

4.5. Histopathological Analysis of Kidney Tissue

Kidney tissues were fixed in 4% paraformaldehyde, then subjected to a dehydration process through an alcohol gradient before being immersed in wax. After being impregnated with wax, the tissues were embedded, frozen, and trimmed. The cooled wax blocks were then placed in a paraffin sectioning machine, where 4 μm thick sections were sliced and mounted onto slides. These sections were subsequently stained with hematoxylin and eosin (H&E) and Masson’s trichrome stains. After staining, the sections were scanned and analyzed for renal histopathological features.
For dewaxing and rehydration, xylene and a series of alcohol gradients were used. Antigen retrieval was carried out using an EDTA-based retrieval buffer. Following a 30 min blocking step with BSA, the sections were incubated overnight at 4 °C with the primary antibody. Following this, the samples were washed three times with PBS for 5 min each. The corresponding secondary antibody was applied and incubated at room temperature for 1 h. Cell nuclei were counterstained with DAPI, and the slides were mounted with an anti-fluorescence quencher before being scanned or photographed for detailed observation.

4.6. Network Pharmacology

The active components and targets of LCW were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP, https://www.tcmsp-e.com/load_intro.php?id=43 (accessed on 15 July 2024), using oral bioavailability (OB) >30% and drug-likeness (DL) ≥0.18 as selection criteria. Additional target information was collected from SwissTargetPrediction (http://www.swisstargetprediction.ch/ (accessed on 15 July 2024)), PubChem (https://pubchem.ncbi.nlm.nih.gov/ (accessed on 15 July 2024)), and relevant literature [10], with a screening threshold of probability > 0. Targets related to LN were obtained from the OMIM database (https://www.omim.org/), GeneCards (http://www.genecards.org/), and DrugBank (https://go.drugbank.com/) using “lupus nephritis” as the search term. The LCW targets were then mapped to LN-related targets using the Bioinformatics website (https://www.bioinformatics.com.cn/), and intersection targets were identified, representing the genes involved in the treatment of LN by LCW. The intersection targets were subsequently uploaded to the STRING 12.0 database (https://cn.string-db.org/) and analyzed using Cytoscape 3.7.2 software to construct a protein–protein interaction (PPI) network, with a high-confidence threshold of ≥0.7. Core targets were identified based on degree centrality, betweenness centrality, and closeness centrality. Finally, the intersection targets were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis using the DAVID tool (https://david.ncifcrf.gov/).

4.7. Kidney Tissue Transcriptomic Analysis

RNA was isolated from animal tissues using the TRIzol Reagent, following the protocol provided in the instruction manual. The reagent was supplied by Life Technologies, Carlsbad, California, USA. The RNA yield and purity were quantified using the NanoDrop 2000, an instrument from Thermo Fisher Scientific (Wilmington, DE, USA). RNA quality was assessed by evaluating its integrity with the RNA Nano 6000 Assay Kit, which was analyzed on the Agilent Bioanalyzer 2100 system, provided by Agilent Technologies (Santa Clara, CA, USA). This meticulous process ensured the RNA’s integrity for subsequent experimental procedures. The libraries were sequenced on an Illumina NovaSeq platform to generate 150 bp paired-end reads, in accordance with the manufacturer’s guidelines. The raw reads were then processed using the BMK Cloud bioinformatics pipeline (www.biocloud.net (accessed on 12 February 2025)). Initially, the raw data in fastq format were filtered using in-house Perl scripts to obtain clean reads by removing adapter sequences, reads containing poly-N, and low-quality sequences. During this step, key quality metrics such as Q20, Q30, GC content, and sequence duplication levels were calculated. All subsequent analyses were based on high-quality clean data. After processing, adapter sequences and low-quality reads were removed, resulting in clean reads. The clean reads were subsequently aligned to the reference genome using the Hisat2 tool. Only those reads with a perfect match, or a single mismatch, were considered for further analysis and annotation. Gene expression levels were quantified by calculating fragments per kilobase of transcript per million fragments mapped (FPKM), with the formula: FPKM = cDNA Fragments/(Mapped Fragments (Millions) × Transcript Length (kb). Differential expression analysis between two conditions was performed using DESeq2. DESeq2 provides statistical routines for determining differential expression in digital gene expression data using a model based on the negative binomial distribution. Differentially Expressed Genes (DEGs) were filtered using the criteria of FDR < 0.05 and |log2Fold Change| > 1. Subsequently, visual enrichment analysis of these DEGs was performed using the DAVID database, and weishengxin.

4.8. Serum Metabolomic Analysis

The metabolomics analysis was performed using a state-of-the-art LC/MS system, featuring a Waters Acquity I-Class PLUS UPLC, coupled with the high-definition Waters Xevo G2-XS QTof mass spectrometer. Chromatographic separation was performed using a Waters Acquity UPLC HSS T3 column (1.8 µm, 2.1 × 100 mm). The mobile phases for both positive and negative ion modes included 0.1% formic acid in water (phase A) and 0.1% formic acid in acetonitrile (phase B), with an injection volume of 2 μL. The Xevo G2-XS QTof mass spectrometer was operated in MSe mode, enabling simultaneous collection of full and fragmented mass spectra, as controlled by MassLynx V4.2 software. The system supported dual-channel acquisition with a collision energy range of 10 to 40 volts and a scan rate of 0.2 s per spectrum. The ESI ion source settings included a capillary voltage of 2500 V for positive ions and −2000 V for negative ions, a cone voltage of 30 V, and an ion source temperature of 100 °C. Desolvation gas was set to 500 °C with flow rates of 50 L/h for backflush and 800 L/h for desolvation. Raw data were collected using MassLynx V4.2 and processed with Progenesis QI software (version 4.0) for peak detection, alignment, and subsequent refinement. Compound identification was supported by the METLIN database and Biomark’s library. After normalization against the total peak area, principal component analysis (PCA) and Spearman correlation analysis were performed to assess the consistency between the samples and QC. Metabolite characterization was further conducted using the KEGG, HMDB, and Lipid Maps databases. Differential metabolites were identified through a t-test for significance (p < 0.05), and orthogonal partial least squares discriminant analysis (OPLS-DA) was performed using the “ropls” R package 4.5.1, supported by 200 permutation tests for validation. Selection of key metabolites was based on variable importance in projection (VIP) values and p-values. KEGG pathway enrichment analysis for these metabolites was conducted using a hypergeometric test (p < 0.05).

4.9. Western Blotting

The expression levels of p-PI3K, PI3K (phosphatidylinositol 3-kinase), p-AKT, AKT (protein kinase B), p-mTOR, and mTOR (mammalian target of rapamycin) in mouse kidney tissue were assessed by Western blot. The primary antibody reagents used were as follows: p-AKT (80455-1-RR), mTOR (28273-1-AP), and GAPDH (60004-1-Ig) from Wuhan Sanying Biotechnology Co., Ltd. (Wuhan, China); PI3K (AF6241) and p-PI3K (AF3241) from Affinity Biosciences Ltd. (Liyang, Jiangsu, China); AKT (A18675) from ABclonal Biotechnology Co., Ltd. (Wuhan, China); and p-mTOR (5536S) from Cell Signaling Technology, Inc. (Danvers, MA, USA).
Kidney tissue samples were carefully minced and homogenized in RIPA lysis buffer to denature the proteins. The homogenate was vortexed and centrifuged at 12,000 rpm for 10 min at 4 °C to collect the supernatant. Protein concentration was quantified using the BCA assay, and the protein supernatant was stored at −20 °C until further use. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was conducted, followed by membrane transfer and blocking. The membrane was then incubated overnight at 4 °C with primary antibodies against PI3K, p-PI3K, AKT, p-mTOR (1:1000 dilution), mTOR (1:5000 dilution), p-AKT (1:30,000 dilution), and GAPDH (1:50,000 dilution). Afterward, the membrane was incubated with HRP-conjugated secondary antibodies (1:10,000 dilution) at room temperature for 2 h. The membrane was subsequently washed, and protein bands were visualized using an ECL reagent and a chemiluminescence imaging system (Hangzhou Shenhua Technology Co., Ltd., SH-523, Hangzhou, China). Images were then captured and analyzed with ImageJ software, version 1.53a.

4.10. Statistical Analysis

All data are expressed as mean ± standard error of the mean (SEM). Statistical analyses were performed using GraphPad Prism 9.5 software (GraphPad Software, Inc., Boston, MA, USA). Group differences were assessed using one-way analysis of variance (ANOVA), with subsequent data visualization in the form of charts. Statistical significance was considered at a p-value of less than 0.05.

5. Conclusions

This study demonstrates that LCW exhibits significant pharmacological effects in MRL/lpr mice, particularly by inhibiting the PI3K/AKT/mTOR signaling pathway, thereby effectively alleviating the symptoms of LN. These findings provide strong theoretical support for the clinical application of LCW, and the development of new therapeutic agents based on its active components.

Author Contributions

Conceptualization, C.L. and G.J.; methodology, C.L. and G.J.; software, X.Z.; validation, C.L., Z.L. and X.Z.; formal analysis, Z.L.; investigation, H.Y.; resources, B.Y.; data curation, H.Y.; writing—original draft preparation, C.L.; writing—review and editing, Z.Y. and S.W. (Shilei Wang); visualization, T.J.; supervision, S.W. (Shumin Wang); project administration, S.W. (Shumin Wang); funding acquisition, T.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The animal research protocol was approved by the Laboratory Animal Ethics Committee of Changchun University of Traditional Chinese Medicine (protocol code 2023513; approval date: 25 September 2023).

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding authors.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
SLESystemic lupus erythematosus
LNLupus nephritis
LCWLang Chuang Wan
ANAAnti-nuclear antibodies
ANOVAOne-way analysis of variance
anti-dsDNAAnti-double-stranded DNA antibodies
anti-SmAnti-Sm antibody
BPBiological processes
CCCellular components
MFMolecular functions
GOGene Ontology
KEGGKyoto Encyclopedia of Genes and Genomes
BPCBase peak chromatograms
BUNBlood urea nitrogen
DAVIDDatabase for annotation, visualization and integrated discovery
DEGsDifferentially expressed genes
DEMDifferentially expressed metabolites
ELISAEnzyme-linked immunosorbent assay
HMDBHuman metabolome database
OPLS-DAOrthogonal partial least squares discriminant analysis
PCAPrincipal component analysis
SEMStandard error of the mean
UPLC-MS/MSUltra-performance liquid chromatography–tandem mass spectrometry
UPUrinary protein
VIPVariable importance for the projection

References

  1. Voelker, R. What Is Lupus? JAMA 2024, 332, 1312. [Google Scholar] [CrossRef] [PubMed]
  2. Halfon, M.; Tankeu, A.T.; Ribi, C. Mitochondrial Dysfunction in Systemic Lupus Erythematosus with a Focus on Lupus Nephritis. Int. J. Mol. Sci. 2024, 25, 6162. [Google Scholar] [CrossRef]
  3. Guo, Z.; Guo, Q.; Li, X.; Gao, X.; Zhang, L.; Xu, K. Urinary Biomarkers Associated with Podocyte Injury in Lupus Nephritis. Front. Pharmacol. 2024, 15, 1324540. [Google Scholar] [CrossRef] [PubMed]
  4. Li, Y.; Qi, W. Experimental Study of Nanostructured Lipid Carrier in the Treatment of Immune Lupus Nephritis. J. Nanosci. Nanotechnol. 2021, 21, 1099–1106. [Google Scholar] [CrossRef]
  5. Sharafaldin, E.N.K.; Sim, M.S.; Lim, S.K.; Alhussieni, K.; Huri, H.Z. Precision Medicine in Lupus Nephritis. Clin. Chim. Acta 2024, 562, 119894. [Google Scholar] [CrossRef]
  6. Dall’Era, M. Treatment of Lupus Nephritis: Current Paradigms and Emerging Strategies. Curr. Opin. Rheumatol. 2017, 29, 241–247. [Google Scholar] [CrossRef] [PubMed]
  7. Rovin, B.H.; Ayoub, I.M.; Chan, T.M.; Liu, Z.-H.; Mejía-Vilet, J.M.; Floege, J. KDIGO 2024 Clinical Practice Guideline for the Management of LUPUS NEPHRITIS. Kidney Int. 2024, 105, S1–S69. [Google Scholar] [CrossRef]
  8. Anders, H.-J.; Saxena, R.; Zhao, M.; Parodis, I.; Salmon, J.E.; Mohan, C. Lupus Nephritis. Nat. Rev. Dis. Prim. 2020, 6, 7. [Google Scholar] [CrossRef]
  9. Wang, K.; Gao, Y.; Lu, C.; Li, Y.; Zhou, B.; Qin, X.; Du, G.; Gao, L.; Guan, D.; Lu, A. Uncovering the Complexity Mechanism of Different Formulas Treatment for Rheumatoid Arthritis Based on a Novel Network Pharmacology Model. Front. Pharmacol. 2020, 11, 1035. [Google Scholar] [CrossRef]
  10. Gao, Y.; Wang, K.; Wang, P.; Li, X.; Chen, J.; Zhou, B.; Tian, J.; Guan, D.; Qin, X.; Lu, A. A Novel Network Pharmacology Strategy to Decode Mechanism of Lang Chuang Wan in Treating Systemic Lupus Erythematosus. Front. Pharmacol. 2020, 11, 512877. [Google Scholar] [CrossRef]
  11. Ji, G.; Liu, Y.; Yang, X.; Wang, R.; Bu, H.; Wang, S. Therapeutic Effect of Lupus Pills on Pristane-induced Systemic Lupus Erythematosus in Mice. Spec. Wild Econ. Anim. Plant Res. 2022, 44, 16–23. [Google Scholar] [CrossRef]
  12. Zheng, S.; Xue, T.; Wang, B.; Guo, H.; Liu, Q. Application of Network Pharmacology in the Study of the Mechanism of Action of Traditional Chinese Medicine in the Treatment of COVID-19. Front. Pharmacol. 2022, 13, 926901. [Google Scholar] [CrossRef] [PubMed]
  13. Hu, L.; Sui, X.; Dong, X.; Li, Z.; Lun, S.; Wang, S. Low Beauvericin Concentrations Promote PC-12 Cell Survival under Oxidative Stress by Regulating Lipid Metabolism and PI3K/AKT/mTOR Signaling. Ecotoxicol. Environ. Saf. 2024, 269, 115786. [Google Scholar] [CrossRef]
  14. Mishra, S.; Kell, P.; Scherrer, D.; Dietzen, D.J.; Vite, C.H.; Berry-Kravis, E.; Davidson, C.; Cologna, S.M.; Porter, F.D.; Ory, D.S.; et al. Accumulation of Alkyl-Lysophosphatidylcholines in Niemann-Pick Disease Type C1. J. Lipid Res. 2024, 65, 100600. [Google Scholar] [CrossRef]
  15. Lowe, R.; Shirley, N.; Bleackley, M.; Dolan, S.; Shafee, T. Transcriptomics Technologies. PLoS Comput. Biol. 2017, 13, e1005457. [Google Scholar] [CrossRef]
  16. Alarcón-Riquelme, M.E. Transcriptome Studies in Lupus Nephritis. Arch. Immunol. Ther. Exp. 2022, 70, 15. [Google Scholar] [CrossRef]
  17. Septembre-Malaterre, A.; Boumendjel, A.; Seteyen, A.-L.S.; Boina, C.; Gasque, P.; Guiraud, P.; Sélambarom, J. Focus on the High Therapeutic Potentials of Quercetin and Its Derivatives. Phytomedicine Plus 2022, 2, 100220. [Google Scholar] [CrossRef] [PubMed]
  18. Chen, H.-Y.; Chiang, Y.-F.; Hong, Y.-H.; Shieh, T.-M.; Huang, T.-C.; Ali, M.; Chang, H.-Y.; Wang, K.-L.; Hsia, S.-M. Quercetin Ameliorates Renal Injury and Pyroptosis in Lupus Nephritis through Inhibiting IL-33/ST2 Pathway In Vitro and In Vivo. Antioxidants 2022, 11, 2238. [Google Scholar] [CrossRef]
  19. Ding, T.; Yi, T.; Li, Y.; Zhang, W.; Wang, X.; Liu, J.; Fan, Y.; Ji, J.; Xu, L. Luteolin Attenuates Lupus Nephritis by Regulating Macrophage Oxidative Stress via HIF-1α Pathway. Eur. J. Pharmacol. 2023, 953, 175823. [Google Scholar] [CrossRef] [PubMed]
  20. Wang, S.; Xu, Z.; Cai, B.; Chen, Q. Berberine as a Potential Multi-Target Agent for Metabolic Diseases: A Review of Investigations for Berberine. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 971–979. [Google Scholar] [CrossRef]
  21. Gao, R.; Lu, Y.; Zhang, W.; Zhang, Z. The Application of Berberine in Fibrosis and the Related Diseases. Am. J. Chin. Med. 2024, 52, 753–773. [Google Scholar] [CrossRef] [PubMed]
  22. Fu, H.X.; Fan, X.P.; Li, M.; Liu, M.J.; Sun, Q.L. MiR-146a Relieves Kidney Injury in Mice with Systemic Lupus Erythematosus through Regulating NF-κB Pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7024–7032. [Google Scholar] [PubMed]
  23. Nashi, R.A.; Shmerling, R.H. Antinuclear Antibody Testing for the Diagnosis of Systemic Lupus Erythematosus. Med. Clin. N. Am. 2021, 105, 387–396. [Google Scholar] [CrossRef]
  24. Solé, C.; Royo, M.; Sandoval, S.; Moliné, T.; Gabaldón, A.; Cortés-Hernández, J. Precise Targeting of Autoantigen-Specific B Cells in Lupus Nephritis with Chimeric Autoantibody Receptor T Cells. Int. J. Mol. Sci. 2024, 25, 4226. [Google Scholar] [CrossRef] [PubMed]
  25. Saleem, A.; Zeeshan, B.; Dissanayake, G.; Zergaw, M.; Elgendy, M.; Billey, A. Anti-Smith Antibodies as a Predictive Factor for Developing Lupus Nephritis in Systemic Lupus Erythematosus Patients: A Systematic Review. Cureus 2024, 16, e66270. [Google Scholar] [CrossRef]
  26. Hokamp, J.A.; Nabity, M.B. Renal Biomarkers in Domestic Species. Vet. Clin. Pathol. 2016, 45, 28–56. [Google Scholar] [CrossRef]
  27. Liu, J.; Xu, J.; Huang, J.; Gu, C.; Liu, Q.; Zhang, W.; Gao, F.; Tian, Y.; Miao, X.; Zhu, Z.; et al. TRIM27 Contributes to Glomerular Endothelial Cell Injury in Lupus Nephritis by Mediating the FoxO1 Signaling Pathway. Lab. Investig. 2021, 101, 983–997. [Google Scholar] [CrossRef]
  28. Schmeisser, K.; Parker, J.A. Pleiotropic Effects of mTOR and Autophagy During Development and Aging. Front. Cell Dev. Biol. 2019, 7, 192. [Google Scholar] [CrossRef]
  29. Zhang, L.; Li, H.; Wu, Y.; Cheng, L.; Yan, Y.; Yang, X.; Zhu, F.; He, S.; Tang, W.; Zuo, J. (5R)-5-Hydroxytriptolide Ameliorates Lupus Nephritis in MRL/Lpr Mice by Preventing Infiltration of Immune Cells. Am. J. Physiol.-Ren. Physiol. 2017, 312, F769–F777. [Google Scholar] [CrossRef]
  30. Mills, K.H.G. IL-17 and IL-17-Producing Cells in Protection versus Pathology. Nat. Rev. Immunol. 2023, 23, 38–54. [Google Scholar] [CrossRef]
  31. Hendriks, R.W. Interleukin-10 Multitasking in Allergic Airway Inflammation. Cell. Mol. Immunol. 2023, 20, 1530–1532. [Google Scholar] [CrossRef] [PubMed]
  32. Vasilev, V.; Artero, M.R.; Petkova, M.; Mihaylova, G.; Dragon-Durey, M.-A.; Radanova, M.; Roumenina, L.T. Clinical Relevance of Anti-C3 and Anti-C4 Autoantibodies in Lupus Nephritis. Kidney Int. Rep. 2024, 9, 1429–1440. [Google Scholar] [CrossRef] [PubMed]
  33. Tang, Y.; Zhang, W.; Zhu, M.; Zheng, L.; Xie, L.; Yao, Z.; Zhang, H.; Cao, D.; Lu, B. Lupus Nephritis Pathology Prediction with Clinical Indices. Sci. Rep. 2018, 8, 10231. [Google Scholar] [CrossRef] [PubMed]
  34. Sciascia, S.; Cozzi, M.; Barinotti, A.; Radin, M.; Cecchi, I.; Fenoglio, R.; Mancardi, D.; Wilson Jones, G.; Rossi, D.; Roccatello, D. Renal Fibrosis in Lupus Nephritis. Int. J. Mol. Sci. 2022, 23, 14317. [Google Scholar] [CrossRef]
  35. Adhya, Z.; Borozdenkova, S.; Karim, M.Y. The Role of Cytokines as Biomarkers in Systemic Lupus Erythematosus and Lupus Nephritis. Nephrol. Dial. Transplant. 2011, 26, 3273–3280. [Google Scholar] [CrossRef]
  36. Qing, X.; Chinenov, Y.; Redecha, P.; Madaio, M.; Roelofs, J.J.; Farber, G.; Issuree, P.D.; Donlin, L.; Mcllwain, D.R.; Mak, T.W.; et al. iRhom2 Promotes Lupus Nephritis through TNF-α and EGFR Signaling. J. Clin. Investig. 2018, 128, 1397. [Google Scholar] [CrossRef]
  37. Yu, J.; Fan, S.; Li, X.; Hou, R.; Hu, X.; Wang, J.; Shan, R.; Dong, Z.; Xie, M.; Dong, Y.; et al. Novel Insights into STAT3 in Renal Diseases. Biomed. Pharmacother. 2023, 165, 115166. [Google Scholar] [CrossRef]
  38. Stylianou, K.; Petrakis, I.; Mavroeidi, V.; Stratakis, S.; Vardaki, E.; Perakis, K.; Stratigis, S.; Passam, A.; Papadogiorgaki, E.; Giannakakis, K.; et al. The PI3K/Akt/mTOR Pathway Is Activated in Murine Lupus Nephritis and Downregulated by Rapamycin. Nephrol. Dial. Transplant. 2010, 26, 498–508. [Google Scholar] [CrossRef]
  39. Zhao, C.; Gu, Y.; Chen, L.; Su, X. Upregulation of FoxO3a Expression through PI3K/Akt Pathway Attenuates the Progression of Lupus Nephritis in MRL/Lpr Mice. Int. Immunopharmacol. 2020, 89, 107027. [Google Scholar] [CrossRef]
  40. Feng, X.; Wu, C.; Yan, G.; Liu, Q.; Liu, J.; Hao, J.; Xing, L.; Yang, M.; Liu, S. TLR2 Plays a Critical Role in HMGB1-Induced Glomeruli Cell Proliferation Through the FoxO1 Signaling Pathway in Lupus Nephritis. J. Interferon Cytokine Res. 2016, 36, 258–266. [Google Scholar] [CrossRef]
  41. Ji, G.; Liu, Y.; Yang, X.; Wang, R.; Bu, H.; Wang, S. Network Pharmacology to Unveil the Mechanism of Astragali Radix in the Treatment of Lupus Nephritis via PI3K/AKT/mTOR Pathway. Sci. Rep. 2024, 14, 25983. [Google Scholar] [CrossRef]
  42. Hu, C.; Du, Y.; Xu, X.; Li, H.; Duan, Q.; Xie, Z.; Wen, C.; Han, X. Lipidomics Revealed Aberrant Metabolism of Lipids Including FAHFAs in Renal Tissue in the Progression of Lupus Nephritis in a Murine Model. Metabolites 2021, 11, 142. [Google Scholar] [CrossRef] [PubMed]
  43. Ruan, X.Z.; Varghese, Z.; Moorhead, J.F. An Update on the Lipid Nephrotoxicity Hypothesis. Nat. Rev. Nephrol. 2009, 5, 713–721. [Google Scholar] [CrossRef] [PubMed]
  44. Xu, J.; Yao, X.; Li, X.; Xie, S.; Chi, S.; Zhang, S.; Cao, J.; Tan, B. Farnesoid X Receptor Regulates PI3K/AKT/mTOR Signaling Pathway, Lipid Metabolism, and Immune Response in Hybrid Grouper. Fish Physiol. Biochem. 2022, 48, 1521–1538. [Google Scholar] [CrossRef] [PubMed]
  45. Gao, L.; Yuan, P.; Zhang, Q.; Fu, Y.; Hou, Y.; Wei, Y.; Zheng, X.; Feng, W. Taxifolin Improves Disorders of Glucose Metabolism and Water-Salt Metabolism in Kidney via PI3K/AKT Signaling Pathway in Metabolic Syndrome Rats. Life Sci. 2020, 263, 118713. [Google Scholar] [CrossRef]
  46. Dong, Y.; Gao, L.; Sun, Q.; Jia, L.; Liu, D. Increased Levels of IL-17 and Autoantibodies Following Bisphenol A Exposure Were Associated with Activation of PI3K/AKT/mTOR Pathway and Abnormal Autophagy in MRL/Lpr Mice. Ecotoxicol. Environ. Saf. 2023, 255, 114788. [Google Scholar] [CrossRef]
  47. Kim, Y.C.; Guan, K.-L. mTOR: A Pharmacologic Target for Autophagy Regulation. J. Clin. Investig. 2015, 125, 25–32. [Google Scholar] [CrossRef]
  48. Yang, F.; Zhang, Y.; Dong, L.; Song, Z. Cordyceps Cicadae Ameliorates Inflammatory Responses, Oxidative Stress, and Fibrosis by Targeting the PI3K/mTOR-mediated Autophagy Pathway in the Renal of MRL/Lpr Mice. Immun. Inflamm. Dis. 2024, 12, e1168. [Google Scholar] [CrossRef]
  49. Zhang, X.-M.; Qing, M.-J.; Liu, X.-K.; Peng, L. Complement Factor B Inhibitor LNP023 Mediates the Effect and Mechanism of AMPK/mTOR on Autophagy and Oxidative Stress in Lupus Nephritis. Kaohsiung J. Med. Sci. 2024, 40, 996–1005. [Google Scholar] [CrossRef]
  50. Tsay, A.; Wang, J.-C. The Role of PIK3R1 in Metabolic Function and Insulin Sensitivity. Int. J. Mol. Sci. 2023, 24, 12665. [Google Scholar] [CrossRef]
  51. Ruby, M.; Gifford, C.C.; Pandey, R.; Raj, V.S.; Sabbisetti, V.S.; Ajay, A.K. Autophagy as a Therapeutic Target for Chronic Kidney Disease and the Roles of TGF-Β1 in Autophagy and Kidney Fibrosis. Cells 2023, 12, 412. [Google Scholar] [CrossRef] [PubMed]
  52. Jin, R.; Chen, A.; Ye, Y.; Ren, Y.; Lu, J.; Xuan, F.; Zhou, W. Effect of Berberine Combined with Metformin on Autophagy in Polycystic Ovary Syndrome by Regulating AMPK/AKT/mTOR Pathway. Mol. Reprod. Dev. 2024, 91, e23768. [Google Scholar] [CrossRef] [PubMed]
  53. Ma, Z.J.; Hu, S.L.; Wang, S.S.; Guo, X.; Zhang, X.N.; Sun, B.; Chen, L.M. Effects and underlying mechanism of berberine on renal tubulointerstitial injury in diabetic rats. Zhonghua Yi Xue Za Zhi 2016, 96, 3072–3077. [Google Scholar] [CrossRef] [PubMed]
  54. Li, R.; Shi, C.; Wei, C.; Wang, C.; Du, H.; Liu, R.; Wang, X.; Hong, Q.; Chen, X. Fufang Shenhua Tablet Inhibits Renal Fibrosis by Inhibiting PI3K/AKT. Phytomedicine 2023, 116, 154873. [Google Scholar] [CrossRef]
  55. Xu, X.; Yu, Z.; Han, B.; Li, S.; Sun, Y.; Du, Y.; Wang, Z.; Gao, D.; Zhang, Z. Luteolin Alleviates Inorganic Mercury-Induced Kidney Injury via Activation of the AMPK/mTOR Autophagy Pathway. J. Inorg. Biochem. 2021, 224, 111583. [Google Scholar] [CrossRef]
  56. Prasher, P.; Sharma, M.; Singh, S.K.; Gulati, M.; Chellappan, D.K.; Zacconi, F.; De Rubis, G.; Gupta, G.; Sharifi-Rad, J.; Cho, W.C.; et al. Luteolin: A Flavonoid with a Multifaceted Anticancer Potential. Cancer Cell Int. 2022, 22, 386. [Google Scholar] [CrossRef] [PubMed]
  57. Vasilev, N.; Boccard, J.; Lang, G.; Grömping, U.; Fischer, R.; Goepfert, S.; Rudaz, S.; Schillberg, S. Structured Plant Metabolomics for the Simultaneous Exploration of Multiple Factors. Sci. Rep. 2016, 6, 37390. [Google Scholar] [CrossRef]
  58. Zelena, E.; Dunn, W.B.; Broadhurst, D.; Francis-McIntyre, S.; Carroll, K.M.; Begley, P.; O’Hagan, S.; Knowles, J.D.; Halsall, A.; Wilson, I.D.; et al. Development of a Robust and Repeatable UPLC−MS Method for the Long-Term Metabolomic Study of Human Serum. Anal. Chem. 2009, 81, 1357–1364. [Google Scholar] [CrossRef]
  59. Want, E.J.; Masson, P.; Michopoulos, F.; Wilson, I.D.; Theodoridis, G.; Plumb, R.S.; Shockcor, J.; Loftus, N.; Holmes, E.; Nicholson, J.K. Global Metabolic Profiling of Animal and Human Tissues via UPLC-MS. Nat. Protoc. 2013, 8, 17–32. [Google Scholar] [CrossRef]
Figure 1. UPLC-MS/MS analysis of LCW. (A) Positive ion base peak chromatogram of LCW based on UPLC-MS/MS. (B) Negative ion base peak chromatogram of LCW based on UPLC-MS/MS.
Figure 1. UPLC-MS/MS analysis of LCW. (A) Positive ion base peak chromatogram of LCW based on UPLC-MS/MS. (B) Negative ion base peak chromatogram of LCW based on UPLC-MS/MS.
Pharmaceuticals 18 00916 g001
Figure 2. Comprehensive analysis of serum autoantibody levels, renal function, inflammatory cytokine levels, and renal histopathology. The evaluated biomarkers include the following: (A) ANA, (B) anti-dsDNA, (C) anti-Sm, (D) Cr, (E) BUN, (F) UP, (G) IL-6, (H) IL-10, (I) IL-17, (J) TNF-α, (K) C3, and (L) C4. Histopathological assessment includes the following: (M) H&E staining results, with black arrows indicating tubular epithelial cell edema with deformation and necrosis, and white arrows indicating interstitial inflammatory cell infiltration. (N) Masson’s trichrome staining results. (O) Quantification of renal fibrosis scoring. (P) Results of IgG immune complex deposition. (Q) Quantification of IgG immune complex deposition. (Magnification 40×, Scale bar = 100 μm.) Data are presented as mean ± standard error of the mean (SEM), ## p < 0.01 indicate statistically significant differences compared to the control group; * p < 0.05 and ** p < 0.01 indicate statistically significant differences compared to the model group.
Figure 2. Comprehensive analysis of serum autoantibody levels, renal function, inflammatory cytokine levels, and renal histopathology. The evaluated biomarkers include the following: (A) ANA, (B) anti-dsDNA, (C) anti-Sm, (D) Cr, (E) BUN, (F) UP, (G) IL-6, (H) IL-10, (I) IL-17, (J) TNF-α, (K) C3, and (L) C4. Histopathological assessment includes the following: (M) H&E staining results, with black arrows indicating tubular epithelial cell edema with deformation and necrosis, and white arrows indicating interstitial inflammatory cell infiltration. (N) Masson’s trichrome staining results. (O) Quantification of renal fibrosis scoring. (P) Results of IgG immune complex deposition. (Q) Quantification of IgG immune complex deposition. (Magnification 40×, Scale bar = 100 μm.) Data are presented as mean ± standard error of the mean (SEM), ## p < 0.01 indicate statistically significant differences compared to the control group; * p < 0.05 and ** p < 0.01 indicate statistically significant differences compared to the model group.
Pharmaceuticals 18 00916 g002
Figure 3. Network pharmacology prediction of the LCW treatment for LN. (A) Venn diagram of the overlapping targets between LCW and LN. (B) PPI network of LCW acting on 249 common targets. (C) Potential core targets of LCW action. (D) GO functional enrichment analysis of LCW treatment targets for LN. (E) KEGG pathway enrichment analysis of LCW treatment targets for LN.
Figure 3. Network pharmacology prediction of the LCW treatment for LN. (A) Venn diagram of the overlapping targets between LCW and LN. (B) PPI network of LCW acting on 249 common targets. (C) Potential core targets of LCW action. (D) GO functional enrichment analysis of LCW treatment targets for LN. (E) KEGG pathway enrichment analysis of LCW treatment targets for LN.
Pharmaceuticals 18 00916 g003
Figure 4. Transcriptomics analysis results (n = 6). (A) PCA plot of control and model group samples. (B) PCA plot of model and LCW-H group samples. (C) Volcano plot of DEGs between control and model groups. (D) Volcano plot of DEGs between model and LCW-H groups. (E) Venn diagram of overlapping DEGs between control-vs.-model and model-vs.-LCW-H comparison groups. (F) Heatmap of hierarchical clustering analysis of DEGs. (G) GO analysis results of DEGs. (H) KEGG analysis results of DEGs.
Figure 4. Transcriptomics analysis results (n = 6). (A) PCA plot of control and model group samples. (B) PCA plot of model and LCW-H group samples. (C) Volcano plot of DEGs between control and model groups. (D) Volcano plot of DEGs between model and LCW-H groups. (E) Venn diagram of overlapping DEGs between control-vs.-model and model-vs.-LCW-H comparison groups. (F) Heatmap of hierarchical clustering analysis of DEGs. (G) GO analysis results of DEGs. (H) KEGG analysis results of DEGs.
Pharmaceuticals 18 00916 g004
Figure 5. Metabolomics analysis results (n = 6). (A) PCA of control, model, and LCW-H groups. (B) OPLS-DA analysis between control and model groups. (C) OPLS-DA analysis between model and LCW-H groups. (D) Heatmap visualization of metabolic biomarkers in control, model, and LCW-H groups. (E) Enrichment analysis of candidate biomarkers.
Figure 5. Metabolomics analysis results (n = 6). (A) PCA of control, model, and LCW-H groups. (B) OPLS-DA analysis between control and model groups. (C) OPLS-DA analysis between model and LCW-H groups. (D) Heatmap visualization of metabolic biomarkers in control, model, and LCW-H groups. (E) Enrichment analysis of candidate biomarkers.
Pharmaceuticals 18 00916 g005
Figure 6. Multi-omics integrated analysis of LCW treatment for LN. (A) Common genes between network pharmacology and transcriptomics. (B) Spearman correlation heatmap of 27 overlapping genes and 42 candidate biomarkers. Color gradient indicates correlation strength (red: r > 0, positive association; blue: r < 0, negative association; *, **, and *** denote statistically significant differences (p < 0.05, p < 0.01, p < 0.001) between common genes and candidate biomarkers). (C) Joint pathway analysis of network pharmacology, transcriptomics, and metabolomics.
Figure 6. Multi-omics integrated analysis of LCW treatment for LN. (A) Common genes between network pharmacology and transcriptomics. (B) Spearman correlation heatmap of 27 overlapping genes and 42 candidate biomarkers. Color gradient indicates correlation strength (red: r > 0, positive association; blue: r < 0, negative association; *, **, and *** denote statistically significant differences (p < 0.05, p < 0.01, p < 0.001) between common genes and candidate biomarkers). (C) Joint pathway analysis of network pharmacology, transcriptomics, and metabolomics.
Pharmaceuticals 18 00916 g006
Figure 7. Western blot analysis of the expression of PI3K/AKT/mTOR signaling pathway-related proteins. (A) Individual protein bands and groupings. (B) p-PI3K protein expression. (C) p-AKT protein expression. (D) p-mTOR protein expression. (E) PI3K protein expression. (F) AKT protein expression. (G) mTOR protein expression. GAPDH was used as a loading control for grayscale analysis. Data are presented as mean ± standard error (n = 3). Compared to the control group, ## p < 0.01; compared to the model group, * p < 0.05, ** p < 0.01.
Figure 7. Western blot analysis of the expression of PI3K/AKT/mTOR signaling pathway-related proteins. (A) Individual protein bands and groupings. (B) p-PI3K protein expression. (C) p-AKT protein expression. (D) p-mTOR protein expression. (E) PI3K protein expression. (F) AKT protein expression. (G) mTOR protein expression. GAPDH was used as a loading control for grayscale analysis. Data are presented as mean ± standard error (n = 3). Compared to the control group, ## p < 0.01; compared to the model group, * p < 0.05, ** p < 0.01.
Pharmaceuticals 18 00916 g007
Table 1. The 25 Active Constituents in LCW.
Table 1. The 25 Active Constituents in LCW.
No.CompoundsFormulaPrecursor TypeExact MassClassPeak Area
1Berberine[C20H18NO4]+[M + H]+336.12358Alkaloids765,955,338
2ChrysinC15H10O4[M − H]−254.05791Flavonoids959,729,637.3
3Licoisoflavone aC20H18O6[M + H]+354.11033Flavonoids343,842,417.9
4DesmethylxanthohumolC20H20O5[M + H]+340.13107Flavonoids234,716,721.9
5RutinC27H30O16[M − H]−610.15338Flavonoids192,697,182
62,4,6-trihydroxydihydrochalconeC15H14O4[M − H]−258.0892Flavonoids73,232,107.88
7BaicaleinC15H10O5[M + H]+270.05282Flavonoids28,482,364.63
82′,7-dihydroxy-4′,5′-dimethoxyisoflavoneC17H14O6[M − H]−314.07903Flavonoids18,475,453.99
9DaidzinC21H20O9[M − H2O − H]−416.11073Flavonoids7,967,367.338
10AcacetinC16H12O5[M + H]+284.06847Flavonoids6,688,437.089
11LuteolinC15H10O6[M − H2O + H]+286.04774Flavonoids4,144,178.487
12TiboloneC21H28O2[M − H]−312.20892Hormone class24,691,286.41
13Syringic acidC9H10O5[M − H]−198.05282Phenolic acids1,025,933,162
143-o-feruloylquinic acidC17H20O9[M − H]−368.11073Phenolic acids353,099,163.3
155-caffeoylquinic acidC16H18O9[M − H]−354.09508Phenolic acids272,904,467.6
165-feruloylquinic acidC17H20O9[M − H]−368.11073Phenolic acids177,445,299.3
17Quinic acidC7H12O6[M − H]−192.06339Phenolic acids112,171,658.5
18Caffeic acidC9H8O4[M − H2O + H]+180.04226Phenolic acids21,905,035.5
19Beta-sitosterolC29H50O[M − H2O + H]+414.38614phytosterols41,671,858.23
20Oleanoic acidC30H48O3[M − H]−456.36033Terpenoids2,895,688,889
21HederageninC30H48O4[M − H]−472.35524Terpenoids1,355,770,073
22Glycyrrhetinic acidC30H46O4[M + H]+470.33959Terpenoids379,591,568.6
23AlbiflorinC23H28O11[M + H]+480.16315Terpenoids178,862,591.3
24Licoricesaponin g2C42H62O17[M + H]+838.39868Terpenoids27,520,043.37
25Dihydrotanshinone iC18H14O3[M − H]−278.09429Terpenoids15,131,118.85
Table 2. Differential metabolites in MRL/lpr mouse serum between groups.
Table 2. Differential metabolites in MRL/lpr mouse serum between groups.
No.Metabolite NameMolecular FormulaHMDB IDModel/ControlLCW-H/Model
1CerebrosterolC27H46O2HMDB0001419
2Dodecanoic acidC12H24O2HMDB0000638
33alpha,7alpha-Dihydroxy-5beta-cholestanateC27H46O4HMDB0000359
44alpha-MethylzymosterolC28H46OHMDB0001217
5CalcitriolC27H44O3HMDB0001903
64,4-Dimethyl-5alpha-cholesta-8-en-3beta-olC29H50OHMDB0006840
7Nervonic acidC24H46O2HMDB0002368
82-Methoxyestrone 3-glucuronideC25H32O9HMDB0004482
92-HydroxyestroneC18H22O3HMDB0000343
10Leukotriene C4C30H47N3O9SHMDB0001198
1117alpha,21-DihydroxypregnenoloneC21H32O4HMDB0006762
12Caprylic acidC8H16O2HMDB0000482
1313(S)-HPOTC18H30O4HMDB0301803
14Prostaglandin F2alphaC20H34O5HMDB0001139
1517-Hydroxylinolenic acidC18H30O3HMDB0011108
16PregnenoloneC21H32O2HMDB0000253
17Testosterone glucuronideC25H36O8HMDB0003193
1812(S)-HPETEC20H32O4HMDB0004243
195,7,24(28)-ErgostatrienolC28H44OHMDB0060404
20(6Z,9Z,12Z)-Octadecatrienoic acidC18H30O2HMDB0003073
21CholesterolC27H46OHMDB0000067
22alpha-Linolenic acidC18H30O2HMDB0001388
2312-OPDAC18H28O3HMDB0301804
24AldosteroneC21H28O5HMDB0000037
25(9Z,11E)-(13S)-13-Hydroperoxyoctadeca-9,11-dienoic acidC18H32O4HMDB0003871
26Stearic acidC18H36O2HMDB0000827
27PregnanediolC21H36O2HMDB0004025
283alpha,7alpha,12alpha-Trihydroxy-5beta-cholestaneC27H48O3HMDB0001457
29(9Z)-Octadecenoic acidC18H34O2HMDB0000207
303alpha,7alpha-Dihydroxy-5beta-cholestaneC27H48O2HMDB0006893
31Prostaglandin G2C20H32O6HMDB0003235
3221-DeoxycortisolC21H30O4HMDB0004030
33TestololactoneC19H26O3HMDB0258855
34Cholic acidC24H40O5HMDB0000619
357-DehydrocholesterolC27H44OHMDB0000032
36AvenastenoneC29H46OHMDB0304267
3711,12-DHETC20H34O4HMDB0002314
387alpha-Hydroxy-3-oxo-4-cholestenoateC27H42O4HMDB0012458
399-OxoODEC18H30O3HMDB0004669
409(S)-HODEC18H32O3HMDB0004670
41CampesterolC28H48OHMDB0002869
42CortolC21H36O5HMDB0003180
↑ indicates upregulation of differential metabolites when comparing Model vs. Control or LCW-H vs. Model. ↓ indicates downregulation of differential metabolites when comparing Model vs. Control or LCW-H vs. Model.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, C.; Ji, G.; Zhang, X.; Yu, H.; Li, Z.; Yang, B.; Yao, Z.; Wang, S.; Jiang, T.; Wang, S. Combined Network Pharmacology, Transcriptomics and Metabolomics Strategies Reveal the Mechanism of Action of Lang Chuang Wan to Ameliorate Lupus Nephritis in MRL/lpr Mice. Pharmaceuticals 2025, 18, 916. https://doi.org/10.3390/ph18060916

AMA Style

Li C, Ji G, Zhang X, Yu H, Li Z, Yang B, Yao Z, Wang S, Jiang T, Wang S. Combined Network Pharmacology, Transcriptomics and Metabolomics Strategies Reveal the Mechanism of Action of Lang Chuang Wan to Ameliorate Lupus Nephritis in MRL/lpr Mice. Pharmaceuticals. 2025; 18(6):916. https://doi.org/10.3390/ph18060916

Chicago/Turabian Style

Li, Cuicui, Guoxin Ji, Xinru Zhang, Hang Yu, Zhimeng Li, Bo Yang, Zhuangzhuang Yao, Shilei Wang, Tongwei Jiang, and Shumin Wang. 2025. "Combined Network Pharmacology, Transcriptomics and Metabolomics Strategies Reveal the Mechanism of Action of Lang Chuang Wan to Ameliorate Lupus Nephritis in MRL/lpr Mice" Pharmaceuticals 18, no. 6: 916. https://doi.org/10.3390/ph18060916

APA Style

Li, C., Ji, G., Zhang, X., Yu, H., Li, Z., Yang, B., Yao, Z., Wang, S., Jiang, T., & Wang, S. (2025). Combined Network Pharmacology, Transcriptomics and Metabolomics Strategies Reveal the Mechanism of Action of Lang Chuang Wan to Ameliorate Lupus Nephritis in MRL/lpr Mice. Pharmaceuticals, 18(6), 916. https://doi.org/10.3390/ph18060916

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop