Abstract Purpose Ursolic Acid (UA), a triterpenoid extracted from Hedyotis Diffusa Willd. (HDW), is known for its anti-inflammatory, antioxidant, and antitumor effects. Nevertheless, the mechanisms underlying UA’s anti-colorectal cancer (CRC) effects remain insufficiently understood. This study aimed to identify the key target proteins of UA and investigate their functions in CRC development. Methods The cytotoxicity of three active components of HDW (UA, oleanolic acid (OA), and quercetin) on CRC cells was evaluated using the CCK-8 assay. Tandem mass tag (TMT)-based proteomics was employed to detect differentially expressed proteins (DEPs) in CRC cells after UA treatment. Bioinformatics analysis and high-content screening were used to identify UA’s key protein targets. The expression and role of RPLP1 in CRC cells were investigated, including the effects of RPLP1 knockdown and its combination with UA treatment on cell proliferation, migration, invasion, and apoptosis. Results UA demonstrated superior inhibitory effects on CRC cells compared to OA and quercetin, highlighting it as a principal active ingredient of HDW. TMT-based proteomic analysis identified 438 upregulated and 366 downregulated proteins after UA intervention. Among these, RPLP1 was identified as a critical target. UA inhibited RPLP1 expression in CRC cells, resulting in decreased cell proliferation, migration, and invasion. Furthermore, the combination of UA treatment and RPLP1 knockdown exhibited synergistic effects in inhibiting CRC cell growth and migration, as well as promoting apoptosis. Conclusions UA, a bioactive triterpenoid of HDW, inhibits CRC development by targeting and suppressing RPLP1 expression. These findings provide novel insights into the therapeutic of UA for CRC and highlight RPLP1 as a promising target for intervention. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-03486-z. Keywords: Colorectal cancer, Hedyotis diffusa willd, Ursolic acid, RPLP1, Tandem mass tags Introduction As reported by recent studies, colorectal cancer (CRC) is the second leading cause of cancer-related mortality worldwide, accounting for a significant global health burden [[30]1–[31]3]. The disease often begins insidiously and may progress rapidly in advanced stages. Patients with advanced CRC frequently experience complications such as intestinal obstruction, systemic symptoms including weight loss and anemia, and metastases to critical organs such as liver, lungs, lymph nodes, and bones, which eventually lead to death [[32]4]. Notably, up to 90% of CRC-related fatalities are attributed to distant invasion and metastasis, highlighting the aggressive nature of advanced disease [[33]5]. Despite the availability of treatment such as surgical resection, chemoradiotherapy, and molecular-targeted therapy, these approaches are far from ideal [[34]6]. They are often accompanied by adverse effects, drug resistance, restricted survival benefits, and high financial costs, which together underscore the inadequacy of current clinical management strategies [[35]7]. Furthermore, once patients develop resistance to prescribed therapies, few alternative options remain, creating a critical gap in CRC treatment protocols. Given these challenges, there is an urgent need to develop novel therapeutic strategies that are not only effective but also demonstrate minimal toxicity and cost-effectiveness, providing hope for improved outcomes in CRC management. Growing evidence has demonstrated that traditional Chinese medicine (TCM), a valuable resource for anti-CRC medications, has been utilized for the prevention and treatment of colorectal cancer (CRC) for centuries [[36]8]. TCM offers a unique therapeutic approach by targeting multiple pathways and factors involved in CRC progression, while simultaneously alleviating the adverse effects associated with conventional treatments such as radiotherapy, chemotherapy, and targeted therapy, thereby prolonging patient survival [[37]9, [38]10]. Among the many TCM-based treatments, Hedyotis Diffusa Willd. (HDW), stands out as a widely used Chinese herbal medicine in anticancer formulations, effectively combating various human malignancies [[39]11]. HDW contains three primary active ingredients-ursolic acid (UA), oleanolic acid (OA), and quercetin-which exhibit diverse pharmacological properties, including antioxidant, anti-inflammatory, and immunomodulatory effects [[40]12]. Notably, mounting studies have highlighted the significant inhibitory effects of HDW on CRC development [[41]13–[42]15]. Among these active ingredients, UA is a bioactive triterpenoid with broad antitumor potential. It can be isolated from multiple plants species, including Oldenlandia diffusa, Rosmarinus officinalis, and Glechoma hederaceae [[43]16]. UA has been shown to effectively prevent various types of malignancies, including CRC [[44]17, [45]18]. Moreover, UA has the potential to enhance the antitumor efficacy of oxaliplatin in CRC treatment [[46]19]. Although these findings are promising, the precise molecular mechanisms and targets underlying UA’s anti-CRC effects remain insufficiently understood. Further research is needed to unravel the mechanistic basis of UA’s activity against CRC, paving the way for its potential clinical application. Quantitative proteomic analysis has emerged as a powerful tool for elucidating the pharmacological effects, identifying protein targets, and uncovering the mechanisms of action of traditional Chinese medicine (TCM) remedies in cancer treatment [[47]20]. With advancements in proteomics, particularly the use of stable isotope labeling and tandem mass tag (TMT)-based techniques, it is now possible to detect and analyze changes in thousands of proteins within minimal sample amounts, offering high sensitivity and accuracy [[48]21]. In this study, we applied TMT-based quantitative proteomics to investigate the differential protein expression profiles following ursolic acid (UA) intervention in colorectal cancer (CRC) cells. Bioinformatics analysis and high-content screening identified the 60 S acidic ribosomal protein P1 (RPLP1) as a key target of UA. Subsequent functional assays further evaluated the role of RPLP1 in CRC, examining both its individual functions and the combined effects of RPLP1 knockdown and UA treatment. By integrating proteomics with molecular biology techniques, this study provides a comprehensive understanding of the mechanisms underlying UA’s anti-CRC effects. These findings, in conjunction with previous research, offer valuable insights into the therapeutic potential of UA and lay a foundation for further exploration of its molecular targets. Materials and methods Cell culture and treatment Human CRC cell lines SW480 and HCT116 (Cell Bank, Chinese Academy of Sciences) were cultured in DMEM (Shanghai Yuanpei Biotechnology Co., LTD, China) and L15 media supplemented with 10% fetal bovine serum (FBS, Thermo Fisher, USA). The cells were maintained at 37 °C and in a 5% CO2 atmosphere. Cytotoxicity assay in vitro To investigate the cytotoxicity of the three active ingredients (UA, OA, and quercetin) of HDW, SW480 cells were exposed to varying concentrations (0, 0.1, 1, 2, 5, 10, 20, 40, and 70 µM) of UA (Shanghai Taosu Biochemical Technology Co., Ltd., China), quercetin (Sigma, USA), and OA (Shanghai Yuanye Biotechnology Co., Ltd., China). The suppressive effect on proliferation of SW480 cells by these three active ingredients was assessed using CCK-8 assay, and their IC50 values were determined. CCK-8 assay Logarithmically growing SW480 and HCT116 cells (5 × 10^4 cells/mL) were seeded into each well of a 96-well plate and cultured overnight in an incubator. After treatments with different concentrations of UA, quercetin, and OA, cells were allowed to incubate for 72 h. Then, 10 µL CCK-8 reagent was introduced into each well for 3 h. Absorbance was recorded at 450 nm with a microplate reader (Model 680; Bio-199 Rad, Hercules, CA, USA). TMT-based proteomics analysis When SW480 cells treated with UA and DMSO (negative control) reached a concentration of 4 × 10^3 cells/mL, they were lysed with RIPA buffer to extract total protein. After determining the protein concentration and purity, 200 µg protein extract was subjected to Filter-Aided Sample Preparation (FASP) digestion. UA-treated and DMSO-treated (negative control) SW480 cells were lysed with RIPA buffer to extract total protein. After determining the protein concentration and purity, 200 µg protein extract was subjected to Filter-Aided Sample Preparation (FASP) digestion. The solutions were collected and lyophilized. TMT labeling was then conducted utilizing a TMT labeling kit (Thermo Fisher Scientific). Subsequently, high-performance liquid chromatography fractionation (HPLC) and liquid chromatography–mass spectrometry/mass spectrometry (LC–MS/MS) analysis were performed. Each experimental sample underwent separation using an Easy nLC system at nanoflow rates. After equilibrating the column with 100% buffer A (0.1% formic acid aqueous solution), samples were introduced into an analytical column (Thermo Fisher Scientific) for separation at a flow rate of 300 nL/min. Following chromatographic separation, analysis was conducted using a Q Exactive Plus mass spectrometer for 90 min, operating in positive ion mode with a mass scan range of 350–1800 m/z. The first-level mass resolution was 70,000, with an AGC target of 3e6 and a first-level maximum IT of 50 ms. Peptide and peptide fragment mass-to-charge ratios were determined through a series of ten MS2 scans following each full scan. Using the HCD method, the MS2 activation was conducted, with the isolation window set to 2 m/z. The second-level mass resolution was 35,000 with 1 microscan, a maximum injection time of 45 ms, and a normalized collision energy of 30 eV.TMT-based proteomics was repeated four times. Identification of deps and functional enrichment analysis Protein identification and quantification were executed using Mascot2.6 and Proteome Discoverer 2.1 (Thermo Fisher Scientific), based on the UniProt human database (Uniprot_HomoSapiens_20386_20180905). DEPs were identified by Student’s t-test with a threshold value of |fold change (FC)| >1.2 and P < 0.05. Subsequently, functional enrichment analysis for DEPs was conducted, and significantly enriched GO terms and KEGG pathways were selected using Fisher’s Exact Test. High-content screening To screen for key DEPs in CRC, high-content screening was employed to evaluate the impact of 9 DEPs on HCT116 cell proliferation. Briefly, HCT116 cells were collected after being infected with shRNAs (shPC (Myc, positive control), shCtrl (negative control), shSTK26, shPKN1, shZC3H8, shRPLP1, shDDX20, shFOXK2, shFXR1, shPCGF2, and shTRIM24) for 72 h. See Supplementary Table [49]1 for detailed target information. Cells (1,000 cells/well) were plated into 96-well plates and photographed daily for 5 days utilizing a Celigo image cytometer (Nexcelom Bioscience, Lawrence, MA, USA). On the 5th day, the fold-change (FC) value of the cell count in the shCtrl group (negative control group) was compared with that in the sh-target gene groups. When FC ≥ 2, cell proliferation in the sh-target gene group exhibited significant inhibition in comparison to the shCtrl group, and the target gene was considered a proliferation-related positive regulator. Plasmids construction and transfection To evaluate the functional roles of RPLP1 in CRC, shRNAs targeting RPLP1 (shRNA-57068, shRNA-57069, and shRNA-57070) were designed and synthesized for RPLP1 knockdown. An shRNA non-target sequence was utilized as the negative control (NC). Using Lipofectamine 2000 (Invitrogen Life Technologies, USA), 293T packaging cells were transfected with lentiviruses coexpressing the shRNA sequences psPAX2 (Addgene, USA), and pMD2.G (Addgene). Subsequently, the cells were harvested 48 h post-transfection for further experiments. Quantitative real-time PCR Total RNA was isolated from SW480 and HCT116 cells using TRIzol reagent (Invitrogen Life Technologies, USA). Reverse transcription into cDNA was conducted using iScript cDNA synthesis kit (Bio-Rad, USA). qRT-PCR was employed to detect target gene expression using SYBR Green qPCR Master Mix (Servicebio). GAPDH was used as the internal control. Relative quantification was performed using the 2^−ΔΔCT method. Colony formation assay Logarithmic growth-phased SW480 and HCT116 cells (1000 cells/well) were inoculated into a 6-well plate. After incubation for 2–3 weeks, the formed cell clones were immobilized with 4% paraformaldehyde and stained with Giemsa (DingGuo Biotechnology) for 10–30 min. The number of clones was counted under a microscope. Edu assay BeyoClick^TM null EdU-488 proliferation assay (Beyotime) was conducted to detect cell proliferation. Briefly, cells (1 × 10^6 cells/mL) at logarithmic growth phase were seeded into a 6-well plate and incubated overnight. The cells were then incubated with EdU (10 µM) for 2 h. After being fixed and permeated, the cells were incubated with BeyoClick™ EdU-488. After washing thrice, the cell nuclei were labeled with Hoechst 33342 (RiboBio, Shanghai, China) in the dark for 10 min. Treated cells were observed under a fluorescence microscope (CKX41; Olympus, Tokyo, Japan). Wound healing assay for assessing cell migration SW480 cells (2 × 10^5 cells/well) at logarithmic growth phase were plated into 6-well plates. Upon reaching 80 to 90% confluence, a scratch was created in the center of the cell monolayer using a 100 µL pipette tip, which was aligned vertically to the well’s base to ensure a straight scratch. After washing with PBS to remove debris three times, complete culture medium was added, and cells were incubated in a 5% CO[2] incubator at 37 °C. Wound healing along the scratch was observed at 0 and 24 h. Transwell assay for detecting cell invasion and migration Cells (5 × 10^4 cells/mL) were starved under serum-free conditions for 12 h, harvested, and suspended in serum-free culture medium. The transwell chambers were coated with Matrigel. Then, cell suspension (200 µL) and culture medium supplemented with 10% serum (500 µL) were introduced into the upper and lower chamber, respectively. After 72 h, the chambers underwent fixation with 4% paraformaldehyde for 30 min. After washing, chambers were stained with 0.1% crystal violet for 5 min. Cell invasion to the lower chamber was observed using a fluorescence microscope (CKX41). For quantitative analysis, images were captured at 1 and 24 h post-scratch for each well, selecting five representative fields per well. The number of invaded cells was quantified using GraphPad Prism 7.0 software. The relative invasion was calculated as the number of cells that migrated through the Matrigel-coated membrane divided by the total number of cells seeded in the upper chamber. The migration rate was calculated as the number of cells that migrated to the lower surface of the membrane divided by the total number of cells seeded in the upper chamber.​. Apoptosis analysis Logarithmically growing cells (1 × 10^6 cells/well) were trypsinized, suspended in complete medium, and plated into a 6-well plate. After different treatments, cell apoptosis was detected using an Annexin V-FITC kit (Biosea Biotechnology Co., Beijing, China). Briefly, cells (5 × 10^5 cells/mL) were resuspended in 1 × binding buffer and incubated with Annexin V-FITC (5 µL) and propidium iodide (PI, 10 µL) in the dark for 15 min. A flow cytometer (FACSCalibur, Becton-Dickinson, San Jose, CA, USA) was used to detect cell apoptosis, and CellQuest software (Becton, Franklin Lakes, NJ, USA) was used for data analysis. Statistical analysis All experiments were repeated thrice. All data were depicted as mean ± standard deviation (SD). Statistical analysis was completed using SPSS 17.0 software, with statistical significance defined as P < 0.05. Results Identification of active ingredients of HDW To screen the active ingredients of HDW, the cytotoxicity of three active ingredients of HDW was evaluated. The results revealed that UA (Fig. [50]1A) and quercetin (Fig. [51]1B) showed strong inhibitory effects on SW480 cell proliferation, whereas OA (Fig. [52]1C) had no obvious inhibitory effect on SW480 cells. Among the three ingredients, UA exhibited the lowest IC50 value and was thus regarded as the active ingredient for subsequent experiments. Fig. 1. [53]Fig. 1 [54]Open in a new tab CCK8 assay evaluated the inhibitory effects of three effective ingredients of HDW on CRC cells. A: UA. B: Oleanolic acid. C: Quercetin. UA: ursolic acid DEPs screening and functional enrichment analysis TMT labeling-based proteomic analysis was conducted on SW480 cells treated with 12 µM UA and DMSO for 72 h, respectively. In this analysis, the labeling efficiency was 99.4%. The quality control results showed that the quality deviation of ≥ 90% peptide segments was small, indicating that the data were reliable. A total of 804 DEPs (438 upregulated and 366 downregulated) were screened in UA-treated SW480 and control cells. A volcano plot of the DEPs is shown in Fig. [55]2A. Moreover, the heatmap showed that UA-treated SW480 cells and control cells could be distinguished based on the expression of DEPs (Fig. [56]2B). RPLP1 and STK26 exhibited marked differential expression in UA-treated SW480 cells compared to control cells (Supplementary Table [57]1). Furthermore, GO enrichment analysis of the DEPs was performed (Fig. [58]2C). With respect to biological process (BP), DEPs were mainly enriched in the negative regulation of endopeptidase activity. Regarding molecular function (MF), DEPs were markedly involved in heparin binding. With respect to cellular components (CC), DEPs were primarily localized in the integral component of the plasma membrane. Moreover, these DEPs were implicated in KEGG pathways such as complement and coagulation cascades, cell adhesion molecules, and steroid biosynthesis (Fig. [59]2D). Fig. 2. [60]Fig. 2 [61]Open in a new tab Identification of DEPs after UA treatment based on TMT-based proteomic analysis and functional enrichment analysis. A: Volcano plot of DEPs. The horizontal axis is the fold change of protein expression after log2 conversion, and the vertical axis is the P value of significant difference test after -log10 conversion. B: Heatmap showed the expression of DEPs in all samples. A1, A2, A3, A4: negative control group (SW480 + DMSO); B1, B2, B3, B4: UA group (SW480 + UA). C: Gene Ontology annotation results. D: KEGG pathway enrichment results. DEPs: differentially expressed proteins; UA: ursolic acid; TMT: tandem mass tag RPLP1 was identified as a gene related to CRC cell proliferation High-content screening with a Celigo image cytometer was used to record HCT116 cell proliferation for 5 days following treatment with shRNAs targeting key DEPs (Supplementary Fig. 1). Based on FC on day 5, HCT116 cells treated with shSTK26 and shRPLP1 showed significant proliferation inhibition (Fig. [62]3). The high-content screening results showed that after HCT116 cells were treated with shRNA targeting RPLP1, their proliferation was significantly inhibited. RPLP1 was identified as a key gene related to CRC cell proliferation. Fig. 3. [63]Fig. 3 [64]Open in a new tab High-content screening showed HCT116 cell proliferation for 5 days after treating with shSTK26 and shRPLP1. ShPC was the positive control and shCtrl was the negative control UA inhibited RPLP1 expression in CRC cells The effect of UA on RPLP1 mRNA expression was detected by qPCR. Different concentrations (10 and 20 µM) of UA significantly inhibited RPLP1 mRNA expression in CRC cells (Fig. [65]4). Fig. 4. [66]Fig. 4 [67]Open in a new tab qPCR showed RPLP1 mRNA expression in HCT116 and SW480 cells after treatment with different concentrations (10 and 20 µM) of UA. ** P < 0.01 and *** P < 0.001 compared to control group. UA: ursolic acid RPLP1 knockdown inhibited the malignant behaviors of CRC cells To elucidate the role of RPLP1 in CRC, RPLP1 expression was suppressed in HCT116 and SW480 cells by stable transfection with shRPLP1, which included three specific sequences: shRNA-57,068, shRNA-57,069, and shRNA-57,070. In both cell lines, only shRNA-57,068 transfection significantly inhibited RPLP1 expression compared to sh-NC (P < 0.05, Fig. [68]5A). The impact of shRPLP1 transfection on CRC cell viability was examined using CCK-8 assays. The results revealed a notable reduction in cell viability of HCT116 and SW480 cells following transfection with shRNA-57068 and shRNA-57069 (P < 0.001), with the suppressive effect of shRNA-57068 being the strongest (Fig. [69]5B). Consequently, shRNA-57068 was selected for subsequent experiments to knock down RPLP1 expression in HCT116 and SW480 cells. Colony formation (Fig. [70]5C) and EdU (Fig. [71]5D) assays showed a significant reduction in the colony-forming ability and proliferative rate of both cell types upon RPLP1 knockdown (P < 0.001, Fig. [72]5C-D). Moreover, the transwell assay showed that RPLP1 knockdown dramatically suppressed the invasion of both cell lines (P < 0.001, Fig. [73]5E). The wound healing assay demonstrated a remarkable decrease in the migration of HCT116 and SW480 cells (P < 0.001, Fig. [74]5F). Collectively, RPLP1 knockdown attenuated the malignant behaviors of CRC cells. Fig. 5. [75]Fig. 5 [76]Open in a new tab RPLP1 knockdown inhibited the malignant behaviors of CRC cells. A: qPCR showed RPLP1 mRNA expression in HCT116 and SW480 cells after transfection of shRPLP1 (shRNA-57068, shRNA-57069, and shRNA-57070). B: CCK-8 assays revealed the viability of HCT116 and SW480 cells after transfection of shRPLP1 (shRNA-57068, shRNA-57069, and shRNA-57070). C: Colony formation assay showed the effects of RPLP1 knockdown on colony-forming abilities of HCT116 and SW480 cells. D: EdU assay demonstrated the effects of RPLP1 knockdown on the proliferation of HCT116 and SW480 cells. E: Transwell assay showed the effects of RPLP1 knockdown on HCT116 and SW480 cell invasion. F: Wound healing assay indicated the effects of RPLP1 knockdown on HCT116 and SW480 cell migration. * P < 0.05, ** P < 0.01, and *** P < 0.001 compared to NC group. UA: ursolic acid; NC: negative control RPLP1 knockdown and UA synergistically suppressed the malignant behaviors of CRC cells We investigated the combined effects of RPLP1 knockdown and UA treatment on HCT116 and SW480 cells. The CCK-8 assay results revealed that both RPLP1 knockdown and UA individually reduced the viability of both cell lines (P < 0.001, Fig. [77]6A). In addition, the combination of RPLP1 knockdown and UA treatment showed stronger inhibition than single treatment alone in both cell lines (P < 0.001, Fig. [78]6A). Moreover, both colony formation (Fig. [79]6B) and EdU (Fig. [80]6C) assays showed that RPLP1 knockdown and UA synergistically decreased the colony-forming ability and proliferation of both cell lines (P < 0.001), corroborating the results of the CCK-8 assay. Moreover, the wound-healing assay revealed that the combination of RPLP1 knockdown and UA dramatically inhibited cell migration (P < 0.001). Compared with the UA group, the combined treatment exhibited a more pronounced impact in suppressing the migration of both cell types (P < 0.001, Fig. [81]6D). Furthermore, RPLP1 knockdown, UA treatment, and their combination significantly promoted apoptosis in both cell lines (P < 0.001, Fig. [82]6E). The combined treatment resulted in significantly higher levels of apoptosis than UA treatment alone (P < 0.001, Fig. [83]6E). Taken together, RPLP1 knockdown and UA treatment demonstrated synergistic effects on the inhibition of CRC cell proliferation and migration and promoting apoptosis. Fig. 6. Fig. 6 [84]Open in a new tab RPLP1 knockdown and UA synergistically inhibited the malignant behaviors of CRC cells. HCT116 and SW480 cells were treated with shRPLP1, UA and their combination. A: CCK-8 assays revealed the viability of HCT116 and SW480 cells after different treatments. B: Colony formation assay showed the colony-forming abilities of HCT116 and SW480 cells after different treatments. C: EdU assay demonstrated the proliferation of HCT116 and SW480 cells after different treatments. D: Wound healing assay displayed the migratory activities of HCT116 and SW480 cells after different treatments. E: Flow cytometry analysis detected apoptosis of HCT116 and SW480 cells after different treatments. *** P < 0.001 compared to NC group. # P < 0.05 and ## P < 0.01 compared to UA groups. UA: ursolic acid; NC: negative control Discussion In the present study, we identified UA, the major active ingredient of Polygonum cuspidatum, as having significant inhibitory effects on the malignant behaviors of CRC cells. UA markedly suppressed CRC cell proliferation, migration, and invasion, while also inducing apoptosis. To explore the active components, we screened three major ingredients of Polygonum cuspidatum (UA, quercetin, and OA) and found that UA exhibited the lowest IC50 value in SW480 cells, indicating the strongest anti-proliferative effect. Consequently, UA was selected for further investigation. Subsequently, we employed TMT-based quantitative proteomic analysis to investigate global protein expression changes and uncover the potential mechanisms of UA treatment. A total of 804 differentially expressed proteins (DEPs) were identified, comprising 438 upregulated and 366 downregulated proteins. These DEPs were enriched in biological processes such as the negative regulation of endopeptidase activity and molecular functions like heparin binding, and were localized within cellular structures such as the integral component of the plasma membrane. Pathway enrichment analysis revealed significant associations with KEGG pathways, including complement and coagulation cascades, cell adhesion molecules, and steroid biosynthesis. Mechanistically, high-content screening identified RPLP1, a ribosomal protein-related gene, as a key factor closely associated with CRC cell proliferation. qPCR analysis demonstrated that UA treatment significantly downregulated the expression of RPLP1 mRNA in CRC cells. Functional assays further confirmed that shRNA-mediated knockdown of RPLP1 suppressed CRC cell proliferation, colony formation, migration, and invasion, while promoting apoptosis. Importantly, the combination of UA treatment and RPLP1 knockdown exhibited synergistic effects on CRC cells. Compared with either treatment alone, the combined approach significantly enhanced the inhibition of cell proliferation and migration and further promoted apoptosis. These findings highlight the potential of RPLP1 as a therapeutic target for UA in the treatment of CRC. HDW is a renowned heat-clearing and detoxifying herb frequently utilized in TCM formulations, with its inclusion reported in up to 5.1% of anticancer TCM prescriptions [[85]22]. Among its bioactive components, UA has demonstrated anti-inflammatory, antioxidant, and antitumor properties [[86]23]. Previous studies have shown that UA possesses a wide range of biological functions, including improving hepatic lipid deposition and inhibiting liver fibrosis [[87]24]. scavenging free radicals and exhibiting potent anti-inflammatory and antiviral effects [[88]25, [89]26]; protecting lung tissues from heat stress-induced lung injury [[90]27]; enhancing glucose tolerance and preventing hyperlipidemia [[91]28]; anti-depressant effects [[92]29], inhibiting angiogenesis [[93]30], and antitumor effects [[94]31, [95]32]. The findings of this study further strengthen the growing body of evidence supporting the therapeutic potential of UA in colorectal cancer (CRC) treatment. Additionally, UA exerts its anti-CRC effects through various mechanisms. For instance, Lu et al. indicated that UA suppresses CRC cell growth by regulating SNTB1 to modulate the PKN2/Akt/ERK pathway [[96]33]. Zhang et al. demonstrated that UA attenuates the invasive potential of CRC cells via modulating the TGF‑β1/ZEB1/miR‑200c pathway [[97]34]. Chen et al. revealed that UA alleviates CRC by engaging the non-canonical Hedgehog pathway in a smoothened-independent fashion, which is contingent upon AKT signaling activation [[98]35]. Several studies have confirmed that UA exerts the inhibitory effect on CRC via modulation of multiple signaling pathways [[99]18, [100]36]. Proteomics analysis provides a comprehensive overview of protein profile changes and is widely used in the investigation of molecular mechanisms, biomarkers, and therapeutic targets across various diseases. In this study, TMT-based proteomics was employed to explore the potential mechanisms of UA in CRC. A total of 804 DEPs were identified, among which RPLP1 was recognized as a pivotal target protein of UA. These findings suggest that DEPs regulated by UA are primarily enriched in the regulation of extracellular matrix functions and specific metabolic pathways. For instance, cell adhesion molecules, closely associated with CRC progression [[101]37], may be regulated by UA. Cell adhesion serves as a determinant of metastasis, often leading to tumor cell arrest and extravasation [[102]37]. Moreover, cell adhesion is influenced by heparin binding and integrin function inhibition [[103]38], as highlighted by the GO analysis showing common DEPs involved in heparin-binding processes. Additionally, steroid hormone biosynthesis can inhibit MAPK signaling pathways, reducing the expression and activity of endopeptidases [[104]39, [105]40]. GO analysis of common DEPs further revealed their involvement in processes impacting the integral components of the plasma membrane. An increasing body of evidence suggests that integral components of the plasma membrane, including receptor proteins, ion channels, and transport proteins, are not only structural constituents but also play crucial functional roles in CRC progression. These membrane components participate in pharmacological nutrient absorption and tumor progression by regulating extracellular signal transduction, metabolite transmembrane transport, and interactions with the tumor microenvironment. For example, receptor proteins such as EGFR and TGF-β receptors mediate growth factor signaling pathways, promoting CRC cell proliferation and invasion [[106]41, [107]42]. Ion channels, such as calcium channels, are critical for maintaining intracellular ion homeostasis and signal transduction [[108]43]. Transport proteins, including GLUT1 and amino acid transporters from the SLC family, facilitate the uptake of essential metabolites required for rapid cancer cell proliferation [[109]44, [110]45]. Therefore, proteomics analysis serves as a powerful tool for evaluating the mechanisms of UA in CRC. In various colorectal cancer (CRC) studies, apoptosis is closely linked to cancer cell death [[111]46]. Apoptosis removes damaged cells through morphological and biochemical changes, and its occurrence in CRC cells is associated with tumor progression, metastasis, and chemotherapy resistance [[112]47]. RPLP1, a ribosomal protein, is involved in protein synthesis and apoptosis. Changes in RPLP1 expression can affect apoptosis-related pathways and regulate cell survival [[113]48]. RPLP1 is upregulated in various cancers [[114]49–[115]51], and its knockdown reduces cell proliferation and migration [[116]52]. In our study, UA intervention inhibited RPLP1 expression in CRC cells, and RPLP1 knockdown suppressed malignant behaviors. These results suggest that RPLP1 may be a key regulator in CRC progression and a crucial target of UA in CRC. Conclusion The strength of this study is the identification of RPLP1 as a protein target of UA against CRC using a TMT-based proteomic approach and experimental validation. However, this study has some limitations. Firstly, RPLP1 protein expression was not verified in clinical CRC samples. Secondly, the potential mechanisms by which RPLP1 regulates CRC development were not investigated. More studies are needed to confirm these results. In conclusion, our results indicate that UA, a bioactive triterpenoid in HDW, inhibits CRC development by inhibiting RPLP1 expression. Our findings provide new perspectives for identifying the protein targets of UA for CRC treatment. Author contributions. Li-Min Zhu, Zhen-Ye Xu carried out the conception and design of the research, Li-Min Zhu, Hai-Xia Shi participated in the acquisition of data. Li-Min Zhu, Hai-Bin Deng carried out the analysis and interpretation of data. Li-Min Zhu, Hai-Xia Shi participated in the design of the study and performed the statistical analysis. Li-Min Zhu participated in obtaining funding. Li-Min Zhu conceived of the study, and participated in its design and coordination and drafted the manuscript and revision of manuscript for important intellectual content. All authors read and approved the final manuscript. Supplementary Information [117]Supplementary Material 1.^ (23KB, xls) [118]Supplementary Material 2.^ (33.6KB, xls) [119]Supplementary Material 3.^ (113.5KB, xlsx) [120]Supplementary Material 4^ (116.5KB, png) Acknowledgements