1.Current Situation, Problems and Countermeasures of Experimental Research on Traditional Chinese Medicine Regulating PI3K/Akt Signaling Pathway in Rats with Polycystic Ovary Syndrome
Pengxuan YAN ; Yiqing LIU ; Nanxing XIAN ; Linjing PENG ; Kun LI ; Jingchun ZHANG ; Yukun ZHAO
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(1):259-266
Polycystic ovary syndrome(PCOS) and its resulting infertility is one of the common diseases of gynecology and reproductive endocrinology. The phosphatidylinositol 3-kinase/protein kinase B(PI3K/Akt) signaling pathway is relatively well-studied in the development of intervention in PCOS, and the experiments on PCOS in rats conducted by traditional Chinese medicine through this signaling pathway is also the main direction of mechanistic research. In this paper, 20 articles published in academic journals in the past 5 years were selected through the corresponding criteria, and the objective situation and existing problems of the selected research projects were analyzed from five aspects, namely, baseline data, modeling and treatment, grouping, evaluative indexes, and pharmacodynamic indexes. It is found that there were different degrees of problems in each research project, such as the observation indicators of modeling, criteria for judging the success of the model, the treatment period, the calculation of dosage of prescription/active ingredients and specific dosage were not clearly defined, which could easily lead the bias of the results or reduce the validity of experimental data. Based on this, the list of PCOS rat experimental research operations was formed, involving five categories of experimental rats, model construction, study implementation, outcome measures and analysis and report with a total of 21 operation lists, with a view to provide a reference for the subsequent PCOS experiments related to scientific research and helping to form high-quality results.
2.Research on Regulatory Mechanism of Verbenalin on HCoV-229E-infected Macrophage Injury Based on Mitophagy
Qiyue SUN ; Lei BAO ; Zihan GENG ; Ronghua ZHAO ; Shuran LI ; Xihe CUI ; Jingsheng ZHANG ; Xian LIU ; Rui XIE ; Xiaolan CUI ; Shanshan GUO ; Jing SUN
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(21):29-37
ObjectiveTo investigate the protective effect and mechanism of verbenalin on mouse mononuclear macrophage leukemia cells (RAW264.7) damaged by human coronavirus (HCoV)-229E infection, thereby providing experimental evidence for its development and application. MethodsRAW264.7 macrophages were infected with different concentrations of HCoV-229E to establish a coronavirus-induced macrophage injury model using the cell counting kit-8 (CCK-8) assay for assessing cell proliferation and viability. Cells were randomly divided into four groups: normal control, verbenalin group (125 μmol·L-1), model group (HCoV-229E), and HCoV-229E + verbenalin group (HCoV-229E + 125 μmol·L-1 verbenalin). Cell viability was measured using the CCK-8 assay, and the maximum non-toxic concentration (CC0), half-maximal cytotoxic concentration (CC50), half-maximal effective concentration (EC50), and selectivity index (SI) of verbenalin were calculated. Calcein/PI double staining was used to assess cell viability and cytotoxicity, and JC-1 staining was applied to evaluate changes in mitochondrial membrane potential (MMP). mito-Keima adenovirus labeling was used to assess mitophagy levels in each group. ResultsA macrophage infection model was successfully established by infecting RAW264.7 cells with the original concentration of HCoV-229E for 36 h. The CC0 of verbenalin was 125 μmol·L-1. The CC50 was 448.25 μmol·L-1. The EC50 against HCoV-229E-infected cells was 46.28 μmol·L-1, and the SI was 9.68. Compared with the normal group, the model group showed significantly reduced cell survival rate (P<0.01), increased cell death rate (P<0.01), decreased MMP (P<0.01), and suppressed mitophagy (P<0.01). In contrast, verbenalin treatment significantly improved cell survival rate (P<0.01), reduced cell death rate (P<0.01), alleviated MMP loss (P<0.01), and enhanced mitophagy levels (P<0.01) compared with the model group. ConclusionVerbenalin can enhance the survival rate of macrophages following HCoV-229E infection. The underlying mechanism may be associated with the activation of mitophagy, maintenance of MMP stability, and alleviation of mitochondrial damage.
3.Research on Regulatory Mechanism of Verbenalin on HCoV-229E-infected Macrophage Injury Based on Mitophagy
Qiyue SUN ; Lei BAO ; Zihan GENG ; Ronghua ZHAO ; Shuran LI ; Xihe CUI ; Jingsheng ZHANG ; Xian LIU ; Rui XIE ; Xiaolan CUI ; Shanshan GUO ; Jing SUN
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(21):29-37
ObjectiveTo investigate the protective effect and mechanism of verbenalin on mouse mononuclear macrophage leukemia cells (RAW264.7) damaged by human coronavirus (HCoV)-229E infection, thereby providing experimental evidence for its development and application. MethodsRAW264.7 macrophages were infected with different concentrations of HCoV-229E to establish a coronavirus-induced macrophage injury model using the cell counting kit-8 (CCK-8) assay for assessing cell proliferation and viability. Cells were randomly divided into four groups: normal control, verbenalin group (125 μmol·L-1), model group (HCoV-229E), and HCoV-229E + verbenalin group (HCoV-229E + 125 μmol·L-1 verbenalin). Cell viability was measured using the CCK-8 assay, and the maximum non-toxic concentration (CC0), half-maximal cytotoxic concentration (CC50), half-maximal effective concentration (EC50), and selectivity index (SI) of verbenalin were calculated. Calcein/PI double staining was used to assess cell viability and cytotoxicity, and JC-1 staining was applied to evaluate changes in mitochondrial membrane potential (MMP). mito-Keima adenovirus labeling was used to assess mitophagy levels in each group. ResultsA macrophage infection model was successfully established by infecting RAW264.7 cells with the original concentration of HCoV-229E for 36 h. The CC0 of verbenalin was 125 μmol·L-1. The CC50 was 448.25 μmol·L-1. The EC50 against HCoV-229E-infected cells was 46.28 μmol·L-1, and the SI was 9.68. Compared with the normal group, the model group showed significantly reduced cell survival rate (P<0.01), increased cell death rate (P<0.01), decreased MMP (P<0.01), and suppressed mitophagy (P<0.01). In contrast, verbenalin treatment significantly improved cell survival rate (P<0.01), reduced cell death rate (P<0.01), alleviated MMP loss (P<0.01), and enhanced mitophagy levels (P<0.01) compared with the model group. ConclusionVerbenalin can enhance the survival rate of macrophages following HCoV-229E infection. The underlying mechanism may be associated with the activation of mitophagy, maintenance of MMP stability, and alleviation of mitochondrial damage.
4.Proteomics-based Investigation of Therapeutic Effect and Mechanism of Verbenalin on Lung Injury in Mice Infected with Human Coronavirus-229E
Qiyue SUN ; Shanshan GUO ; Shuangrong GAO ; Lei BAO ; Zihan GENG ; Shuran LI ; Ronghua ZHAO ; Jingsheng ZHANG ; Xian LIU ; Rui XIE ; Xiaolan CUI ; Jing SUN
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(24):69-78
ObjectiveTo evaluate the pharmacological effects of verbenalin on both in vitro and in vivo infection models of human coronavirus 229E (HCoV-229E) and to preliminarily explore the antiviral mechanism of verbenalin through proteomic analysis. MethodsIn vitro, the cell counting kit-8 (CCK-8) for cell proliferation and viability assessment was used to establish a model of HCoV-229E-induced injury in human lung adenocarcinoma cells(A549). A549 cells were divided into five groups: normal group, model group, and three verbenalin treatment groups (125, 62.5, and 31.25 μmol·L-1). The cell protective activity of verbenalin was evaluated through cell viability assay and immunofluorescence staining. In vivo, 30 BALB/c mice were randomly divided into normal group, model group, chloroquine group, and high-dose, low-dose verbenalin groups (40 and 20 mg·kg-1), with six mice per group. An HCoV-229E-induced mouse lung injury model was established to evaluate the therapeutic effects of verbenalin. Lung injury was assessed by detecting the lung index and lung inhibition rate. The severity of pulmonary inflammation cytokines was measured by enzyme-linked immunosorbent assay (ELISA), while the lung morphology and structure were analyzed by micro-computed tomography (Micro-CT). Hematoxylin and eosin (HE) staining was used to assess histopathological changes in lung tissue. Additionally, four-dimensional data-independent acquisition (4D-DIA) proteomics was employed to preliminarily explore the potential mechanisms of verbenalin in treating HCoV-229E-induced lung injury in mice, through differential protein expression screening, functional annotation, enrichment analysis, and protein-protein interaction network analysis. ResultsThe A549 cells were infected with HCoV-229E at the original viral titer for 36 hours to establish an in vitro infection model. The maximum non-toxic concentration of verbenalin was 125 μmol·L-1, and the half-maximal cytotoxic concentration (CC50) was 288.8 μmol·L-1. Compared with the normal group, the model group showed a significant decrease in cell viability (P<0.01), a significant increase in the proportion of dead cells (P<0.01), mitochondrial damage, and a significant reduction in mitochondrial membrane potential (P<0.01). After treatment with different concentrations of verbenalin (125, 62.5, and 31.25 μmol·L-1), cell viability was significantly increased (P<0.01), and the proportion of dead cells was reduced (P<0.01), with mitochondrial membrane potential restored (P<0.01). In vivo experiments further confirmed the therapeutic effect of verbenalin on HCoV-229E-infected mice. Compared to the normal group, the model group showed a significant increase in the lung index (P<0.01), severe lung tissue injury, lung volume enlargement, and a significant increase in the expression of inflammatory cytokines, including interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) (P<0.01). In contrast, in the verbenalin treatment groups, these pathological changes were significantly improved, with a reduction in the lung index (P<0.01), alleviation of lung tissue injury, reduced lung volume enlargement, and a significant decrease in inflammatory cytokine expression (P<0.01). Proteomics analysis revealed that, compared to the normal group, the model group showed enrichment in several antiviral immune-related signaling pathways, including the nuclear factor-κB (NF-κB) signaling pathway (P<0.05). Compared to the model group, the verbenalin treatment group showed enrichment in several signaling pathways related to inflammatory response and autophagy (P<0.05), suggesting that verbenalin may exert its antiviral and anti-inflammatory effects by regulating these pathways. ConclusionVerbenalin demonstrates significant therapeutic effects in both in vitro and in vivo HCoV-229E infection models, with its mechanism likely related to the NOD-like receptor protein 3 (NLRP3) inflammasome pathway and mitochondrial autophagy.
5.Mechanism related to bile acids metabolism of liver injury induced by long-term administration of emodin.
Jing-Zhuo TIAN ; Lian-Mei WANG ; Yan YI ; Zhong XIAN ; Nuo DENG ; Yong ZHAO ; Chun-Ying LI ; Yu-Shi ZHANG ; Su-Yan LIU ; Jia-Yin HAN ; Chen PAN ; Chen-Yue LIU ; Jing MENG ; Ai-Hua LIANG
China Journal of Chinese Materia Medica 2025;50(11):3079-3087
Emodin is a hydroxyanthraquinone compound that is widely distributed and has multiple pharmacological activities, including anti-diarrheal, anti-inflammatory, and liver-protective effects. Research indicates that emodin may be one of the main components responsible for inducing hepatotoxicity. However, studies on the mechanisms of liver injury are relatively limited, particularly those related to bile acids(BAs) metabolism. This study aims to systematically investigate the effects of different dosages of emodin on BAs metabolism, providing a basis for the safe clinical use of traditional Chinese medicine(TCM)containing emodin. First, this study evaluated the safety of repeated administration of different dosages of emodin over a 5-week period, with a particular focus on its impact on the liver. Next, the composition and content of BAs in serum and liver were analyzed. Subsequently, qRT-PCR was used to detect the mRNA expression of nuclear receptors and transporters related to BAs metabolism. The results showed that 1 g·kg~(-1) emodin induced hepatic damage, with bile duct hyperplasia as the primary pathological manifestation. It significantly increased the levels of various BAs in the serum and primary BAs(including taurine-conjugated and free BAs) in the liver. Additionally, it downregulated the mRNA expression of farnesoid X receptor(FXR), retinoid X receptor(RXR), and sodium taurocholate cotransporting polypeptide(NTCP), and upregulated the mRNA expression of cholesterol 7α-hydroxylase(CYP7A1) in the liver. Although 0.01 g·kg~(-1) and 0.03 g·kg~(-1) emodin did not induce obvious liver injury, they significantly increased the level of taurine-conjugated BAs in the liver, suggesting a potential interference with BAs homeostasis. In conclusion, 1 g·kg~(-1) emodin may promote the production of primary BAs in the liver by affecting the FXR-RXR-CYP7A1 pathway, inhibit NTCP expression, and reduce BA reabsorption in the liver, resulting in BA accumulation in the peripheral blood. This disruption of BA homeostasis leads to liver injury. Even doses of emodin close to the clinical dose can also have a certain effect on the homeostasis of BAs. Therefore, when using traditional Chinese medicine or formulas containing emodin in clinical practice, it is necessary to regularly monitor liver function indicators and closely monitor the risk of drug-induced liver injury.
Emodin/administration & dosage*
;
Bile Acids and Salts/metabolism*
;
Animals
;
Male
;
Liver/injuries*
;
Chemical and Drug Induced Liver Injury/genetics*
;
Drugs, Chinese Herbal/adverse effects*
;
Humans
;
Rats, Sprague-Dawley
;
Mice
;
Rats
6.Three new chalcone C-glycosides from Carthami Flos.
Jia-Xu BAO ; Yong-Xiang WANG ; Xian ZHANG ; Ya-Zhu YANG ; Yue LIN ; Jiao-Jiao YIN ; Yun-Fang ZHAO ; Hui-Xia HUO ; Peng-Fei TU ; Jun LI
China Journal of Chinese Materia Medica 2025;50(13):3715-3745
The chemical components of Carthami Flos were investigated by using macroporous resin, silica gel column chromatography, reversed-phase octadecylsilane(ODS) column chromatography, Sephadex LH-20, and semi-preparative high-performance liquid chromatography(HPLC). The planar structures of the compounds were established based on their physicochemical properties and ultraviolet-visible(UV-Vis), infrared(IR), high-resolution electrospray ionization mass spectrometry(HR-ESI-MS), and nuclear magnetic resonance(NMR) spectroscopic technology. The absolute configurations were determined by comparing the calculated and experimental electronic circular dichroism(ECD). Six flavonoid C-glycosides were isolated from the 30% ethanol elution fraction of macroporous resin obtained from the 95% ethanol extract of Carthami Flos, and identified as saffloquinoside F(1), 5-hydroxysaffloneoside(2), iso-5-hydroxysaffloneoside(3), isosafflomin C(4), safflomin C(5), and vicenin 2(6). Among these, the compounds 1 to 3 were new chalcone C-glycosides. The compounds 1, 2, 4, and 5 could significantly increase the viability of H9c2 cardiomyocytes damaged by oxygen-glucose deprivation/reoxygenation(OGD/R) at a concentration of 50 μmol·L~(-1), showing their good cardioprotective activity.
Glycosides/pharmacology*
;
Flowers/chemistry*
;
Drugs, Chinese Herbal/pharmacology*
;
Carthamus tinctorius/chemistry*
;
Chalcones/pharmacology*
;
Animals
7.Stir-fried Semen Armeniacae Amarum Suppresses Aristolochic Acid I-Induced Nephrotoxicity and DNA Adducts.
Cheng-Xian LI ; Xiao-He XIAO ; Xin-Yu LI ; Da-Ke XIAO ; Yin-Kang WANG ; Xian-Ling WANG ; Ping ZHANG ; Yu-Rong LI ; Ming NIU ; Zhao-Fang BAI
Chinese journal of integrative medicine 2025;31(2):142-152
OBJECTIVE:
To investigate the protective effects of stir-fried Semen Armeniacae Amarum (SAA) against aristolochic acid I (AAI)-induced nephrotoxicity and DNA adducts and elucidate the underlying mechanism involved for ensuring the safe use of Asari Radix et Rhizoma.
METHODS:
In vitro, HEK293T cells overexpressing Flag-tagged multidrug resistance-associated protein 3 (MRP3) were constructed by Lentiviral transduction, and inhibitory effect of top 10 common pairs of medicinal herbs with Asari Radix et Rhizoma in clinic on MRP3 activity was verified using a self-constructed fluorescence screening system. The mRNA, protein expressions, and enzyme activity levels of NAD(P)H quinone dehydrogenase 1 (NQO1) and cytochrome P450 1A2 (CYP1A2) were measured in differentiated HepaRG cells. Hepatocyte toxicity after inhibition of AAI metabolite transport was detected using cell counting kit-8 assay. In vivo, C57BL/6 mice were randomly divided into 5 groups according to a random number table, including: control (1% sodium bicarbonate), AAI (10 mg/kg), stir-fried SAA (1.75 g/kg) and AAI + stir-fried SAA (1.75 and 8.75 g/kg) groups, 6 mice in each group. After 7 days of continuous gavage administration, liver and kidney damages were assessed, and the protein expressions and enzyme activity of liver metabolic enzymes NQO1 and CYP1A2 were determined simultaneously.
RESULTS:
In vivo, combination of 1.75 g/kg SAA and 10 mg/kg AAI suppressed AAI-induced nephrotoxicity and reduced dA-ALI formation by 26.7%, and these detoxification effects in a dose-dependent manner (P<0.01). Mechanistically, SAA inhibited MRP3 transport in vitro, downregulated NQO1 expression in vivo, increased CYP1A2 expression and enzymatic activity in vitro and in vivo, respectively (P<0.05 or P<0.01). Notably, SAA also reduced AAI-induced hepatotoxicity throughout the detoxification process, as indicated by a 41.3% reduction in the number of liver adducts (P<0.01).
CONCLUSIONS
Stir-fried SAA is a novel drug candidate for the suppression of AAI-induced liver and kidney damages. The protective mechanism may be closely related to the regulation of transporters and metabolic enzymes.
Aristolochic Acids/toxicity*
;
Animals
;
Humans
;
NAD(P)H Dehydrogenase (Quinone)/genetics*
;
HEK293 Cells
;
Kidney/pathology*
;
Cytochrome P-450 CYP1A2/genetics*
;
Mice, Inbred C57BL
;
DNA Adducts/drug effects*
;
Male
;
Kidney Diseases/drug therapy*
;
Drugs, Chinese Herbal/therapeutic use*
;
Mice
;
Prunus armeniaca
;
Plant Extracts
8.A thermo-sensitive hydrogel targeting macrophage reprogramming for sustained osteoarthritis pain relief.
Yue LIU ; Kai ZHOU ; Xinlong HE ; Kun SHI ; Danrong HU ; Chenli YANG ; Jinrong PENG ; Yuqi HE ; Guoyan ZHAO ; Yi KANG ; Yujun ZHANG ; Yue'e DAI ; Min ZENG ; Feier XIAN ; Wensheng ZHANG ; Zhiyong QIAN
Acta Pharmaceutica Sinica B 2025;15(11):6034-6051
Osteoarthritis (OA) causes chronic pain that significantly impairs quality of life, with current treatments often proving insufficient and accompanied by adverse effects. Recent research has identified the dorsal root ganglion (DRG) and its resident macrophages as crucial mediators of chronic OA pain through neuroinflammation driven by macrophage polarization. We present a novel injectable thermo-sensitive hydrogel system, KAF@PLEL, designed to deliver an anti-inflammatory peptide (KAF) specifically to the DRG. This biodegradable hydrogel enables sustained KAF release, promoting the reprogramming of DRG macrophages from pro-inflammatory to anti-inflammatory phenotypes. Through comprehensive in vitro and in vivo studies, we evaluated the hydrogel's biocompatibility, effects on macrophage polarization, and therapeutic efficacy in chronic OA pain management. The system demonstrated significant capabilities in preserving macrophage mitochondrial function, suppressing neuroinflammation, alleviating chronic OA pain, reducing cartilage degradation, and improving motor function in OA rat models. The sustained-release properties of KAF@PLEL enabled prolonged therapeutic effects while minimizing systemic exposure and side effects. These findings suggest that KAF@PLEL represents a promising therapeutic approach for improving outcomes in OA patients through targeted, sustained treatment.
9.Overexpression of multimerin-2 promotes cutaneous melanoma cell invasion and migration and is associated with poor prognosis.
Jinlong PANG ; Xinli ZHAO ; Zhen ZHANG ; Haojie WANG ; Xingqi ZHOU ; Yumei YANG ; Shanshan LI ; Xiaoqiang CHANG ; Feng LI ; Xian LI
Journal of Southern Medical University 2025;45(7):1479-1489
OBJECTIVES:
To investigate the inhibitory effect of multimerin-2 (MMRN2) overexpression on growth and metastasis of cutaneous melanoma cells.
METHODS:
Clinical data of patients with cutaneous melanoma were obtained from the GEO database to compare MMRN2 expressions between normal and tumor tissues. A protein-protein interaction network was constructed using the STRING database, and the intersecting genes from GEPIA2.0 were subjected to GO and KEGG enrichment analysis. The prognostic relevance of MMRN2 expression level was assessed using Cox regression and "timeROC". The correlations of MMRN2 expression level with immune infiltration and angiogenesis-related genes were analyzed using GSCA database and the ssGSEA algorithm. Colony-forming assay, Transwell assay, and wound healing assay were used to examine the changes in proliferation and migration of cultured cutaneous melanoma cells following MMRN2 knockdown. In a mouse model bearing cutaneous melanoma xenograft, the effect of MMRN2 knockdown on vital organ pathologies, survival of the mice and GM-CSF, CXCL9, and TGF‑β1 protein expressions were analyzed.
RESULTS:
MMRN2 was significantly upregulated in metastatic cutaneous melanoma (P<0.001). Protein interaction network analysis identified 15 intersecting genes, which were enriched in endothelium development and cell-cell junctions. In patients with cutaneous melanoma, a high MMRN2 expression was correlated with a poor prognosis, an advanced T stage, a greater Breslow depth, and ulceration (P<0.05). MMRN2 expression level was strongly correlated with 24 immune cell types (P<0.001), fibroblasts, endothelial cells, and expressions of the pro-angiogenic genes (KCNJ8, SLCO2A1, NRP1, and COL3A1; P<0.001). In cultured B16F10 cells, MMRN2 knockdown significantly suppressed cell proliferation, migration and invasion and caused remo-deling of the immunosuppressive microenvironment.
CONCLUSIONS
MMRN2 overexpression drives progression of cutaneous melanoma by enhancing tumor metastasis, angiogenesis and immune evasion, highlighting its potential as a therapeutic target for melanomas.
Humans
;
Melanoma/metabolism*
;
Animals
;
Cell Movement
;
Prognosis
;
Skin Neoplasms/metabolism*
;
Mice
;
Cell Proliferation
;
Neoplasm Invasiveness
;
Cell Line, Tumor
;
Protein Interaction Maps
10.Huachansu injection enhances anti-colorectal cancer efficacy of irinotecan and alleviates its induced intestinal toxicity through upregulating UGT1A1-OATP1B3 expression in vitro and in vivo.
Bo JIANG ; Zhao-Yang MENG ; Yu-Jie HU ; Jun-Jun CHEN ; Ling ZONG ; Ling-Yan XU ; Xiang-Qi ZHANG ; Jing-Xian ZHANG ; Yong-Long HAN
Journal of Integrative Medicine 2025;23(5):576-590
OBJECTIVE:
Huachansu injection (HCSI), a promising anti-cancer Chinese medicine injection, has been reported to have the potential for reducing the toxicity of chemotherapy and improving the quality of life for colorectal cancer (CRC) patients. The objective of this study is to explore the synergistic and detoxifying effects of HCSI when used in combination with irinotecan (CPT-11).
METHODS:
To investigate the effect of HCSI on anti-CRC efficacy and intestinal toxicity of CPT-11, we measured changes in the biological behavior of LoVo cells in vitro, and anti-tumor effects in LoVo cell xenograft nude mice models in vivo. Meanwhile, the effect of HCSI on intestinal toxicity and the uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1) expression was investigated in the CPT-11-induced colitis mouse model. Subsequently, we measured the effect of HCSI and its 13 constituent bufadienolides on the expression of UGT1A1 and organic anion transporting polypeptides 1B3 (OATP1B3) in HepG2 cells.
RESULTS:
The combination index (CI) results showed that the combination of HCSI and CPT-11 exhibited a synergistic effect (CI < 1), which significantly suppressing the LoVo cell migration, enhancing G2/M and S phase arrest, and inhibiting tumor growth in vivo. Additionally, the damage to intestinal tissues was attenuated by HCSI in CPT-11-induced colitis model, while the increased expression of UGT1A1 in HepG2 cells and in mouse was observed.
CONCLUSION
The co-therapy with HCSI alleviated the intestinal toxicity induced by CPT-11 and exerted an enhanced anti-CRC effect. The detoxifying mechanism may be related to the increased expression of UGT1A1 and OATP1B3 by HCSI and its bufadienolides components. The findings of this study may serve as a theoretical insights and strategies to improve CRC patient outcomes. Please cite this article as: Jiang B, Meng ZY, Hu YJ, Chen JJ, Zong L, Xu LY, Zhang XQ, Zhang JX, Han YL. Huachansu injection enhances anti-colorectal cancer efficacy of irinotecan and alleviates its induced intestinal toxicity through upregulating UGT1A1-OATP1B3 expression in vitro and in vivo. J Integr Med. 2025; 23(5):576-590.
Irinotecan/therapeutic use*
;
Animals
;
Glucuronosyltransferase/genetics*
;
Humans
;
Colorectal Neoplasms/metabolism*
;
Drugs, Chinese Herbal/therapeutic use*
;
Mice, Nude
;
Mice
;
Up-Regulation/drug effects*
;
Male
;
Xenograft Model Antitumor Assays
;
Mice, Inbred BALB C
;
Hep G2 Cells
;
Cell Line, Tumor
;
Intestines/drug effects*
;
Amphibian Venoms

Result Analysis
Print
Save
E-mail