Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Integr Cancer Ther ; 23: 15347354241247061, 2024.
Article in English | MEDLINE | ID: mdl-38641964

ABSTRACT

To investigate the effect of Jiedu Xiaozheng Yin (JXY) on the polarization of macrophages in colitis-associated colon cancer (CAC). An orthotopic model of CAC was established to monitor changes in the pathological state of mice. Colon length, number of colon tumors were recorded, and indices for liver, spleen, and thymus were calculated. Hematoxylin and eosin (H&E) staining was employed to observe intestinal mucosal injury and tumor formation. Immunohistochemistry (IHC) staining was utilized to investigate the effect of JXY on M1 and M2 polarization of macrophages in the colonic mucosa of CAC mice. For in vitro experiments, RT-qPCR (Reverse Transcription-quantitative PCR) and flow cytometry were used to observe the effect of JXY on various M1-related molecules such as IL-1ß, TNF-α, iNOS, CD80, CD86, and its phagocytic function as well as M2-related molecules including Arg-1, CD206, and IL-10. Subsequently, after antagonizing the TLR4 pathway with antagonists (TAK242, PDTC, KG501, SR11302, LY294002), the expression of IL-6, TNF-α, iNOS, and IL-1ß mRNA were detected by RT-qPCR. In vivo experiments, the results showed that JXY improved the pathological condition of mice in general. And JXY treatment decreased the shortening of colon length and number of tumors as compared to non-treated CAC mice. Additionally, JXY treatment improved the lesions in the colonic tissue and induced a polarization of intestinal mucosal macrophages towards the M1 phenotype, while inhibiting polarization towards the M2 phenotype. In vitro experiments further confirmed that JXY treatment promoted the activation of macrophages towards the M1 phenotype, leading to increased expression of IL-1ß, TNF-α, iNOS, CD80, CD86, as well as enhanced phagocytic function. JXY treatment concomitantly inhibited the expression of M2-phenotype related molecules Arginase-1 (Arg-1), CD206, and IL-10. Furthermore, JXY inhibited M1-related molecules such as IL-6, TNF-α, iNOS, and IL-1ß after antagonizing the TLR4 pathway. Obviously, JXY could exhibit inhibitory effects on the development of colon tumors in mice with CAC by promoting M1 polarization through TLR4-mediated signaling and impeding M2 polarization of macrophages.


Subject(s)
Colitis-Associated Neoplasms , Drugs, Chinese Herbal , Macrophages , Animals , Mice , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Interleukin-10/metabolism , Interleukin-6/metabolism , Macrophages/drug effects , Macrophages/metabolism , Phenotype , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Zhen Ci Yan Jiu ; 48(12): 1249-1257, 2023 Dec 25.
Article in English, Chinese | MEDLINE | ID: mdl-38146248

ABSTRACT

OBJECTIVES: To observe the effect of electroacupuncture (EA) at "Zusanli"(ST36) on intestinal mucosal damage, intestinal mucosal oxidative stress injury and apoptosis induced by 5-fluorouraeil (5-FU) chemotherapy in colorectal cancer-bearing mice. METHODS: Thirty male BALB/c mice were randomly divided into normal control, colorectal cancer (CT26), 5-FU, non-acupoint and ST36 groups, with 6 mice in each group. Except for those of the normal control group, mice of the remaining 4 groups received subcutaneous implantation of colorectal CT26 cell suspension (0.1 mL) in the right armpit for establishing colorectal cancer model. Rats of the 5-FU group, non-acupoint group and ST36 group were given with 5 mg/mL 5-FU solution once every 3 days for a total of 21 days. For mice of the non-acupoint group and ST36 group, EA (2 Hz, 1-2 mA) was applied to bilateral ST36 or non-acupoints (the bilateral sunken spots about 3 mm to the midpoint between the tail root and the anus) for 5 min after each intraperitoneal infusion of 5-FU, once every 3 days, for a total of 21 days. After the intervention, the diarrhea index was assessed. The length of colon (from the endpoint of cecum to the anal orifice) was measured. Histopathological changes of colonic mucosa were observed by H.E. staining, and the length of colonic villi was measured. The content of malondialdehyde (MDA), and activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) of colonic tissue were detected by thibabituric acid, xanthine oxidase and colorimetric method, respectively. The rate of cell apoptosis in the colonic tissue was measured by TUNEL assay. The positive expressions of Bax and Bcl-2 in colonic tissue were determined by immunohistochemistry. RESULTS: The CT26 model group didn't show any significant changes in the diarrhea index, colon length, colon villus length, MDA content, SOD and GSH-Px activities, colonic cell apoptosis rate, and Bax and Bcl-2 expression levels when compared with the normal group. Compared with the CT26 group, the 5-FU group had a remarkable increase in the diarrhea index, MDA content, colonic cell apoptosis rate and Bax expression level (P<0.01, P<0.05), and a marked decrease in the colon length, colon villus length, SOD and GSH-Px activities and Bcl-2 expression level (P<0.01), suggesting the side effects of administration of 5-FU. Compared with the 5-FU group, the diarrhea index, MDA content, colonic cell apoptosis rate and Bax expression level were markedly decreased (P<0.05, P<0.01) and those of the colon length, colon villus length, SOD and GSH-Px activities and Bcl-2 expression level were obviously increased (P<0.01) in the ST36 group. Compared with the 5-FU group, the non-acupoint group also had an increase in the colon villus length, SOD and GSH-Px activities (P<0.01, P<0.05) and a decrease in the cell apoptosis rate (P<0.01). CONCLUSIONS: EA at ST36 has a positive effect in reducing intestinal mucosal damage induced by 5-FU chemotherapy in cancer-bearing mice, which may be related to its function in relieving oxidative stress injury and inhibiting apoptosis of colonic tissue.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Electroacupuncture , Rats , Male , Mice , Animals , bcl-2-Associated X Protein/metabolism , Acupuncture Points , Oxidative Stress , Apoptosis , Superoxide Dismutase/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Diarrhea , Fluorouracil/adverse effects
3.
Biomed Pharmacother ; 166: 115387, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37643486

ABSTRACT

Adjuvant chemotherapy based on 5-fluorouracil (5-FU), such as FOLFOX, is suggested as a treatment for gastrointestinal cancer. Yet, intestinal damage continues to be a prevalent side effect for which there are no practical prevention measures. We investigated whether Babao Dan (BBD), a Traditional Chinese Medicine, protects against intestinal damage induced by 5-FU by controlling immune response and gut microbiota. 5-FU was injected intraperitoneally to establish the mice model, then 250 mg/kg BBD was gavaged for five days straight. 5-FU led to marked weight loss, diarrhea, fecal blood, and histopathologic intestinal damage. Administration of BBD reduced these symptoms, inhibited proinflammatory cytokine (IL-6, IL-1ß, IFN-γ, TNF-α) secretion, and upregulated the ratio of CD3(+) T cells and the CD4(+)/CD8(+) ratio. According to 16S rRNA sequencing, BBD dramatically repaired the disruption of the gut microbiota caused in a time-dependent way, and increased the Firmicutes/Bacteroidetes (F/B) ratio. Transcriptomic results showed that the mechanism is mainly concentrated on the NF-κB pathway, and we found that BBD reduced the concentration of LPS in the fecal suspension and serum, and inhibited TLR4/MyD88/NF-κB pathway activation. Furthermore, at the genus level on the fifth day, BBD upregulated the abundance of unidentified_Corynebacteriaceae, Aerococcus, Blautia, Jeotgalicoccus, Odoribacter, Roseburia, Rikenella, Intestinimonas, unidentified_Lachnospiraceae, Enterorhabdus, Ruminiclostridium, and downregulated the abundance of Bacteroides, Parabacteroides, Parasutterella, Erysipelatoclostridium, which were highly correlated with intestinal injury or the TLR4/MyD88/NF-κB pathway. In conclusion, we established a network involving 5-FU, BBD, the immune response, gut microbiota, and key pathways to explain the pharmacology of oral BBD in preventing 5-FU-induced intestinal injury.


Subject(s)
Microbiota , NF-kappa B , Animals , Mice , Myeloid Differentiation Factor 88 , Toll-Like Receptor 4 , RNA, Ribosomal, 16S , Adaptor Proteins, Signal Transducing
4.
Biomed Pharmacother ; 154: 113630, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36058147

ABSTRACT

Gastric cancer (GC) is one of the most common gastrointestinal malignancies in the world. Growing evidence emphasizes the critical role of long non-coding RNA (lncRNA) in GC tumorigenesis. The aim of the research was to elucidate the effect and mechanism of Babao Dan (BBD) on lymphangiogenesis of GC in vitro and in vivo via lncRNA-ANRIL/VEGF-C/VEGFR-3 signaling axis. The present study investigated BBD significantly decreased the expression of lncRNA-ANRIL and VEGF-C in GC cells (AGS, BGC823, and MGC80-3) by using real-time quantitative polymerasechain reaction (RT-qPCR) and the secretion and expression of VEGF-C by (enzyme linked immunosorbent assay) ELISA and western blot (WB). BBD significantly inhibited the tumor xenograft of GC growth and the expression of lncRNA-ANRIL, VEGF-C, VEGFR-3 and LYVE-1 in vivo. BBD reduced serum VEGF-C level. In vitro, BBD inhibited the tube formation and decreased the cell viability, proliferation and migration of HLECs by using tube formation, MTT, Hoechst and Transwell assays. In addition, WB assay found that BBD decreased the expression levels of VEGF-C, VEGFR-3, matrix metallopeptidase 2 (MMP-2) and matrix metallopeptidase 9 (MMP-9), and RT-qPCR assay found that the mRNA expression levels of lncRNA-ANRIL, VEGF-C, VEGFR-3, MMP-2, MMP-9, CDK4, Cyclin D1, and Bcl-2 were down-regulated, and the expression of p21 and Bax were increased. Taken together, these results demonstrated that BBD inhibited lymphangiogenesis of GC in vitro and in vivo via the lncRNA-ANRIL/VEGF-C/VEGFR-3 signaling axis.


Subject(s)
RNA, Long Noncoding , Stomach Neoplasms , Cell Line, Tumor , Drugs, Chinese Herbal , Humans , Lymphangiogenesis/genetics , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , RNA, Long Noncoding/genetics , RNA, Long Noncoding/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
5.
Theranostics ; 12(14): 6088-6105, 2022.
Article in English | MEDLINE | ID: mdl-36168633

ABSTRACT

Colorectal cancer (CRC), mostly categorized as a low immunogenic microsatellite-stable phenotype bearing complex immunosuppressive tumor microenvironment (TME), is highly resistant to immunotherapy. Seeking safe and efficient alternatives aimed at modulating tumor immunosuppressive TME to improve outcome of CRC is highly anticipated yet remains challenging. Methods: Enlightened from the drug complementary art in traditional Chinese medicine, we designed a self-assembled nanomedicine (termed LNT-UA) by the natural active ingredients of ursolic acid (UA) and lentinan (LNT) through a simple nano-precipitation method, without any extra carriers, for CRC immunotherapy. Results: UA induces immunogenic cell death (ICD), while LNT further promotes dendritic cell (DC) maturation and repolarizes tumor-associated macrophage (TAM) from a protumorigenic M2 to an antitumor M1 phenotype. Co-delivery of UA and LNT by LNT-UA effectively reshapes the immunosuppressive TME and mobilizes innate and adaptive immunity to inhibit tumor progression in the CT26 CRC tumor model. Following the principle of integrative theoretical system of traditional Chinese medicine (TCM) on overall regulation, the further combination of LNT-UA and anti-CD47 antibody (αCD47) would reinforce the antitumor immunity by promoting phagocytosis of dying tumor cells and tumor-associated antigens (TAAs), leading to effective suppression of both primary and distant tumor growth with 2.2-fold longer of median survival time in the bilateral tumor model. Most notably, this combination effect is also observed in the spontaneous CRC model induced by chemical carcinogens, with much less and smaller size of tumor nodules after sequential administration of LNT-UA and αCD47 through gavage and intraperitoneal injection, respectively. Conclusions: This study provides a promising self-assembled traditional Chinese nanomedicine to improve immunotherapy for CRC, which might be applicable for future clinical translation.


Subject(s)
Colorectal Neoplasms , Tumor Microenvironment , Carcinogens/pharmacology , China , Colorectal Neoplasms/genetics , Humans , Immunologic Factors/pharmacology , Immunotherapy/methods , Lentinan/pharmacology , Nanomedicine , Oleanolic Acid/analogs & derivatives , Ursolic Acid
6.
Article in English | MEDLINE | ID: mdl-35280511

ABSTRACT

Qingjie Fuzheng granules (QFG) exert an anticancer effect against colorectal cancers (CRC). However, the pharmacological molecular mechanisms are still unclear. This study was aimed to establish a simple method to predict targets of QFG against CRC by the network pharmacology strategy. 461 compounds and 1559 targets in QFG were enriched by BATMAN-TCM. 21 of the common targets were obtained by the groups of "Jun," "Chen," "Zuo," and "Shi" medicine in QFG. The enrichment analyses of GO functional terms, KEGG pathway, and OMIM/TTD diseases displayed the targets in the different and complementary effects of four functional medicines in QFG. Then, 613 differential targets for QFG in CRC were identified. GO functional terms and KEGG pathway analyses showed that QFG regulated the inflammatory function and lipid metabolic process. There were also targets that played a role in the binding to the receptors in membranes, in the activation of the transportation signal, and provided pain relief by regulation of the neural related pathways. Next, the protein-protein interaction network was analyzed, and the levels of the predicted targets in CRC primary tumor were explored, and 7 candidate targets of QFG against CRC were obtained. Furthermore, with real-time PCR and enzyme-linked immunosorbent assay (ELISA) analysis, downregulation of dopamine D2 receptor (DRD2) and interleukin-6 (IL-6), and upregulation of interleukin-10 (IL-10) were identified following the treatment of QFG. At last, the survival and prognosis of the potential targets of QFG in CRC patients were analyzed by GenomicScape, and IL-6 was suggested to be an index for the regulation of QFG in CRC. These results might elucidate the possible antitumor mechanism of QFG and highlight the candidate therapeutic targets and the application direction in clinical treatment for QFG.

7.
Chin J Integr Med ; 28(11): 1000-1006, 2022 Nov.
Article in English | MEDLINE | ID: mdl-33420580

ABSTRACT

OBJECTIVE: To evaluate the protective function of Babao Dan (BBD) on 5-flurouracil (5-FU)-induced intestinal mucositis (IM) and uncover the underlying mechanism. METHODS: A total of 18 male mice were randomly divided into 3 groups by a random number table, including control, 5-FU and 5-FU combined BBD groups, 6 mice in each group. A single intraperitoneal injection of 5-FU (150 mg/kg) was performed in 5-FU and 5-FU combined BBD groups on day 0. Mice in 5-FU combined BBD group were gavaged with BBD (250 mg/kg) daily from day 1 to 6. Mice in the control group were gavaged with saline solution for 6 days. The body weight and diarrhea index of mice were recorded daily. On the 7th day, the blood from the heart of mice was collected to analyze the proportional changes of immunological cells, and the mice were subsequently euthanized by mild anesthesia with 2% pentobarbital sodium. Colorectal lengths and villus heights were measured. Intestinal-cellular apoptosis and proliferation were evaluated by Tunel assay and immunohistochemical staining of proliferating cell nuclear antigen, respectively. Immunohistochemistry and Western blot were performed to investigate the expressions of components in Wnt/ß-catenin pathway (Wnt3, LRP5, ß-catenin, c-Myc, LRG5 and CD44). RESULTS: BBD obviously alleviated 5-FU-induced body weight loss and diarrhea, and reversed the decrease in the number of white blood cells, including monocyte, granulocyte and lymphocyte, and platelet (P<0.01). The shortening of colon caused by 5-FU was also reversed by BBD (P<0.01). Moreover, BBD inhibited apoptosis and promoted proliferation in jejunum tissues so as to reduce the intestinal mucosal damage and improve the integrity of villus and crypts. Mechanically, the expression levels of Wnt/ß -catenin mediators such as Wnt3, LRP5, ß-catenin were upregulated by BBD, activating the transcription of c-Myc, LRG5 and CD44 (P<0.01). CONCLUSIONS: BBD attenuates the adverse effects induced by 5-FU via Wnt/ß-catenin pathway, suggesting it may act as a potential agent against chemotherapy-induced intestinal mucositis.


Subject(s)
Antineoplastic Agents , Mucositis , Animals , Male , Mice , Antineoplastic Agents/therapeutic use , beta Catenin/metabolism , Diarrhea/drug therapy , Fluorouracil/pharmacology , Intestinal Mucosa , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/metabolism , Pentobarbital/metabolism , Pentobarbital/pharmacology , Pentobarbital/therapeutic use , Proliferating Cell Nuclear Antigen/metabolism , Saline Solution
8.
Article in English | MEDLINE | ID: mdl-34887935

ABSTRACT

Qingjie Fuzheng granule (QFG) is a traditional Chinese medicinal formula used extensively as an alternative medicine for cancer treatment, including colorectal cancer (CRC). But its pathological mechanism in CRC is unclear. To study antitumor treatment effects and mechanisms of QFG, we established a CRC HCT-116 xenograft mouse model and assessed QFG on EMT and autophagy progression in vivo. The mice were randomly divided into 2 groups (n = 10 each group) and treated with intragastric administration of 1 g/kg of QFG or saline 6 days a week for 28 days (4 weeks). Body weight was measured every other day with electronic balance. At the end of the treatment, the tumor weight was measured. Immunohistochemical (IHC) and western blot (WB) assay were used to detect the expression level of E-cadherin, N-cadherin, vimentin, and TWIST1 to evaluate the effect of QFG on tumor cell EMT progression. IHC and WB assay were also used to detect the expression level of beclin-1, LC3-II, and p62 to evaluate the effect of QFG on tumor cell autophagy progression. Furthermore, the expression level of relative proteins in mTOR pathway was detected by WB assay to investigate the mechanism of QFG effect on CRC. We discovered that QFG inhibited the rise of tumor weight while it had no effect on mice body weight, which proved that QFG could inhibit CRC growth progression without significant side effects in vivo. In addition, QFG treatment inhibited EMT and induced autophagy progression in CRC tumor cells, including that QFG upregulated the expression of E-cadherin, beclin-1, and LC3-II, but downregulated the expression of N-cadherin, vimentin, TWIST1, and p62. And, QFG decreased the ratio of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR, but increased the ratio of p-AMPK/AMPK. All findings from this research proved that QFG can induce autophagy and inhibit EMT progression in CRC via regulating the mTOR signaling pathway.

9.
J Pharm Biomed Anal ; 204: 114273, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34304010

ABSTRACT

Ba-Bao-Dan (BBD) is a well-known Traditional Chinese medicine (TCM) prescription in China. It was first formulated in approximately 1555 AD. As one of the National Protected TCM, it is widely used to treat jaundice, viral hepatitis, cholecystitis, acute urinary tract infection, cancer, and other diseases. It is a healthcare medicine that is used to prevent many diseases in China. In other Asian countries and in European and American countries, BBD is used as a drug to protect the liver. However, a systematic quality study on BBD chemical markers has not been carried out. This study aimed to establish an ultra-high performance liquid chromatography coupled with triple quadrupole mass spectrometry (UPLC-MS/MS) method for the quantitative determination of 43 compounds in BBD. Furthermore, the method was used to further find chemical markers for quality control through the combination with chemometrics. The modified chromatographic conditions were achieved on Waters Cortecs C18 column (2.1 × 100 mm, 1.6 µm) with a gradient elution consisting of 0.1 % formic acid in water and acetonitrile with methanol (1:1, V/V). All analytes were determined in the multiple reaction monitoring mode. The method was validated for linearity, detection limits, precision, repeatability, stability and accuracy. The method was used to analyze the 43 compounds in 11 batches of BBD samples. Hierarchical cluster analysis and principal component analysis were applied to evaluate intrinsic quality of BBD and to identify the potential chemical markers for quality control. In conclusion, the method rapidly and sensitively determined the 43 compounds, among which 10 compounds, namely, N-Gin R1, Gin Re, Gin Rg1, Gin Rb1, GCA, Gin Rd, CA, TCA, CDCA, and DCA, were considered as the potential chemical markers for BBD quality control.


Subject(s)
Drugs, Chinese Herbal , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Medicine, Chinese Traditional , Quality Control
10.
Article in English | MEDLINE | ID: mdl-34306145

ABSTRACT

Multidrug resistance (MDR) is a critical reason for cancer chemotherapy failure. Babaodan (BBD) is a famous traditional Chinese patent medicine reported to have antigastric cancer activity. However, the roles and molecular mechanisms of the reversal of MDR of gastric cancer by BBD have not been well described until now. Therefore, the purpose of this study was to elucidate further the role of BBD in reversing the MDR of gastric cancer cells and its specific regulatory mechanism via in vitro experiments. To verify our results, MTT, Doxorubicin (DOX) staining, Rhodamin123 (Rho123) staining, DAPI staining, Annexin V-FITC, propidium iodide (PI), Cyto-ID, and western blot assays were performed. To determine whether BBD triggers apoptosis and autophagy through the PI3K/AKT/mTOR signaling, we also applied 3-methyladenine (3-MA), chloroquine (CQ), and 740Y-P (an activator of PI3K). The results showed that BBD reversed the MDR and induced apoptosis and autophagy of SGC7901/DDP cells. Pathway analyses suggested BBD inhibits PI3K/AKT/mTOR pathway activity and subsequent apoptosis-autophagy induction. Inhibition of autophagy with 3-MA and chloroquine (CQ) was performed to confirm that BBD promoted autophagy. PI3K agonist, 740Y-P, further verified BBD inhibition of PI3K/AKT/mTOR pathway activation. In conclusion, BBD may reverse the MDR of gastric cancer cells, induce apoptosis, and promote autophagy via inactivation of the PI3K/AKT/mTOR signaling pathway.

11.
Article in English | MEDLINE | ID: mdl-32714407

ABSTRACT

Qingjie Fuzheng granule (QFG) promotes cancer cell apoptosis and ameliorates intestinal mucosal damage caused by 5-fluorouracil. However, the antitumor role of QFG in colorectal cancer (CRC) progression remains unclear. In this study, the growth of HCT-8 and HCT116 cells incubated with various concentrations of QFG for 24 and 48 h was evaluated using MTT assays; their abilities of migration and invasion were investigated through wound healing and Transwell assays. The expression of lncRNA ANRIL, let-7a, and the TGF-ß1/Smad signaling pathway components was assessed using real-time PCR and western blotting. The results elicited that QFG significantly suppressed the growth of HCT-8 and HCT116 cells; the half-maximal inhibitory concentrations (IC50) of QFG for HCT-8 and HCT116 cells for 48 h were 1.849 and 1.608 mg/mL, respectively. The abilities of wound healing, migration, and invasion of HCT-8 and HCT116 cells were dose-dependently decreased by QFG treatment for 24 h, respectively. QFG decreased the expression of lncRNA ANRIL, TGF-ß1, phosphorylated (p)-Smad2/3, Smad4, and N-cadherin and upregulated the expression of let-7a in HCT-8 and HCT116 cells. Collectively, our data demonstrated that QFG inhibited the metastasis of CRC cells by regulating the lncRNA ANRIL/let-7a/TGF-ß1/Smad axis, indicating that they might serve as an adjunctive medicine for CRC treatment.

12.
J Int Med Res ; 48(6): 300060520925598, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32529872

ABSTRACT

OBJECTIVE: To investigate the anti-metastatic effects of Babao Dan (BBD) on gastric cancer (GC) cells (AGS and MGC80-3) and explore the underlying molecular mechanisms by which it inhibits epithelial-mesenchymal transition (EMT). METHODS: AGS and MGC80-3 cells were treated with BBD. In addition, cells were treated with the EMT inducer transforming growth factor-ß1 (TGF-ß1). Cell viability was determined using the MTT assay, and the live cell ratio was calculated via cell counting. Cell invasion and migration were evaluated using the Transwell assay. Western blotting was performed to measure the protein expression of EMT biomarkers and related genes. RESULTS: BBD inhibited the viability, migration, and invasion of AGS and MGC80-3 cells, but it did not reduce the live cell ratio. Furthermore, BBD inhibited the expression of N-cadherin, vimentin, zinc finger E-box binding homeobox (ZEB)1, ZEB2, Twist1, matrix metalloproteinase (MMP)2, MMP9, TGF-ß1, and p-Smad2/3, whereas E-cadherin expression was increased in AGS and MGC80-3 cells to different degrees. Using a GC cell model of EMT induced by TGF-ß1, we proved that BBD inhibited p-Smad2/3 and N-cadherin expression, cell migration, and cell invasion. CONCLUSION: BBD suppressed cell migration and invasion by inhibiting TGF-ß-induced EMT and inactivating TGF-ß/Smad signaling in GC cells.


Subject(s)
Cell Movement/drug effects , Drugs, Chinese Herbal/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Stomach Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drugs, Chinese Herbal/therapeutic use , Humans , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta1/metabolism
13.
Biomed Res Int ; 2019: 5953619, 2019.
Article in English | MEDLINE | ID: mdl-31662984

ABSTRACT

The use of 5-fluorouracil (5-FU) has been proven benefits, but it also has adverse events in colorectal cancer (CRC) chemotherapy. In this study, we explored the mechanism of 5-FU resistance by bioinformatics analysis of the NCBI public dataset series GSE81005. Fifteen hub genes were screened out of 582 different expressed genes. Modules of the hub genes in protein-protein interaction networks gathered to TOP2α showed a decrease in HCT-8 cells but an increase in 5-FU-resistant HCT-8/5-FU cells with 5-FU exposure. Downregulation of TOP2α with siRNA or miR-494 transfection resulted in an increase of cytotoxicity and decrease of cell colonies to 5-FU for HCT-8/5-FU cells. Moreover, we found that an ethanol extract of Spica Prunellae (EESP), which is a traditional Chinese medicine with clinically beneficial effects in various cancers, was able to enhance the sensitivity of 5-FU in HCT-8/5-FU cells and partly reverse the 5-FU resistance effect. It significantly helped suppress cell growth and induced cell apoptosis in HCT-8/5-FU cells with the expression of TOP2α being significantly suppressed, which increased by 5-FU. Consistently, miR-494, which reportedly regulates TOP2α, exhibited reverse trends in EESP/5-FU combination treatment. These results suggested that Spica Prunellae may be beneficial in the treatment of 5-FU-resistant CRC patients.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , DNA Topoisomerases, Type II/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Plant Extracts/pharmacology , Poly-ADP-Ribose Binding Proteins/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colon/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Medicine, Chinese Traditional/methods , MicroRNAs , Signal Transduction/drug effects , Signal Transduction/genetics
15.
Oncol Lett ; 18(3): 3274-3282, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31452805

ABSTRACT

Ursolic acid (UA) is a biologically active compound, commonly used in traditional Chinese medicine (TCM). It has been reported to exhibit strong anticancer properties against a variety of cancers. Our previous studies showed that UA promoted apoptosis in colorectal cancer (CRC) cells and inhibited cellular proliferation and angiogenesis. However, the effect and underlying molecular mechanism of UA in CRC progression remain unclear. In the present study, the role of UA in suppressing the migration and invasion of human colon cancer HCT116 and HCT-8 cells was investigated, using Transwell assays. In addition, to evaluate whether the anticancer properties of UA were mediated by the regulation of a double-negative feedback loop consisting of the transforming growth factor-ß1 (TGF-ß1)/zinc finger E-box-binding homeobox (ZEB1) pathway and microRNA (miR)-200a/b/c, reverse transcription-quantitative PCR and western blot analysis were performed. The results indicated that UA treatment significantly suppressed cellular growth, migration and invasion in HCT116 and HCT-8 cells in a dose-dependent manner. Furthermore, following UA treatment, several crucial mediators of the TGF-ß1 signaling pathway, including TGF-ß1, phosphorylated (p)-Smad2/3, p-focal adhesion kinase and ZEB1, were significantly downregulated in the HCT116 and HCT-8 cell lines compared with the control group. Furthermore, the ratio of N-cadherin/E-cadherin, two proteins directly downstream of the TGF-ß1 signaling pathway, was found to be downregulated in UA treated CRC cells. Finally, UA significantly upregulated miR200a/b/c, with miR-200c exhibiting the highest increase in expression levels following UA treatment. Collectively, the present study suggested that inhibition of CRC cell invasion by UA occurred via regulation of the TGF-ß1/ZEB1/miR-200c signaling network, which may be one of the mechanisms by which UA appears to be an effective therapeutic agent against colon cancer.

16.
Oncol Rep ; 42(3): 1225-1236, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31322263

ABSTRACT

Colorectal cancer (CRC) is one of the most commonly diagnosed malignancies worldwide. For patients diagnosed with the presence of metastatic disease, surgery is not suitable for the majority of them. Lymphangiogenesis is a key factor during cancer metastasis and is regulated by vascular endothelial growth factor C (VEGF­C). Hedyotis diffusa Willd. (HDW) is a Chinese herb of the Rubiaceae family that reportedly inhibits tumor metastasis. However, its underlying anticancer mechanisms have not yet been elucidated. In the present study, we investigated the effects of an ethanol extract of HDW (EEHDW) on the migration capacity by wound healing and Transwell assays, and the effect on the VEGF­C expression in different CRC cell lines by western blot analysis and ELISA assays. A model of VEGF­C­stimulated human lymphatic endothelial cells (HLECs) was constructed. It was found that EEHDW suppressed lymphangiogenesis via the mediation of multiple pathways, which attenuated the migration of cells and their tube formation abilities. Multiple signaling pathways were found to be involved in the VEGF­C­mediated lymphangiogenesis. After EEHDW treatment in VEGF­C­stimulated HLECs, EEHDW was found to downregulate the expression levels of multiple signaling pathways. Taken together, these results indicate that EEHDW possesses significant anti­metastatic activities. Moreover, the suppressive effect of EEHDW on lymphangiogenesis, particularly via downregulation of VEGF­C, partly explains the potential molecular mechanism underlying the inhibitory effect of EEHDW on CRC metastasis.


Subject(s)
Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Hedyotis/chemistry , Lymphangiogenesis/drug effects , Plant Extracts/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor C/metabolism , Apoptosis , Cell Movement , Cell Proliferation , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Tumor Cells, Cultured
17.
Biomed Pharmacother ; 118: 109223, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31325706

ABSTRACT

OBJECTIVE: 5-Fluorouracil (5-FU)-based chemotherapy often causes several drawbacks including weight loss, diarrhea, myelosuppression, and the intestinal mucositis. This study aimed to evaluate the protective effect of Qingjie Fuzheng Granule (QFG) on 5-FU-induced intestinal mucositis in CT-26 tumor-bearing xenograft mice and investigated the possible molecular mechanism. METHODS: Tumor xenograft models of CT-26 cells were generated in BALB/c nude mice, the mice were randomly divided into 4 groups including control, QFG, 5-FU and 5-FU combined QFG groups. The body weight, volume of tumor and diarrhea score of each group were recorded daily. On the fifth day, the blood of mice was collected, the mice were subsequently euthanized and their thymus, spleen, intestine and tumor were removed for the following analysis. RESULTS: QFG alleviated severe diarrhea and reversed the decrease in the number of white blood cell including granulocyte and lymphocyte induced by 5-FU. QFG could also significantly improve 5-FU-induced several intestinal mucosal damages, and characterized by integrity villus and crypts, the reduction of necrotic cells. QFG decreased the serum levels of TNF-α, IL-1ß, and IL-6 and increased the levels of IL-10. Furthermore, QFG inhibited the cellular apoptosis in the jejunum tissue caused by 5-FU via the increasing Bcl-2 expression and decreasing Bax expression. In addition, QFG promoted the cell proliferation via elevating the expression of Cyclin D1 and CDK4 and reducing p21 expression. Meanwhile, QFG could not further impact on the cell apoptosis and proliferation of tumors caused by 5-FU. CONCLUSION: QFG attenuated the intestinal mucositis and diarrhea induced by 5-FU via preventive effect on inflammation and its improvement of the intestinal barrier function, inhibiting cell apoptosis and promoting cell proliferation, and without affecting the 5-FU treatment efficiency. The results suggest that QFG may act as a potential agent against chemotherapy-induced intestinal mucositis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Intestinal Mucosa/pathology , Animals , Apoptosis/drug effects , Body Weight/drug effects , Cell Proliferation/drug effects , Diarrhea/drug therapy , Drugs, Chinese Herbal/therapeutic use , Fluorouracil , Inflammation Mediators/blood , Intestinal Mucosa/drug effects , Jejunum/drug effects , Jejunum/pathology , Mice, Inbred BALB C , Mice, Nude , Spleen/drug effects , Spleen/pathology , Thymus Gland/drug effects , Thymus Gland/pathology , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/metabolism
18.
World J Gastrointest Oncol ; 11(5): 377-392, 2019 May 15.
Article in English | MEDLINE | ID: mdl-31139308

ABSTRACT

BACKGROUND: Qingjie Fuzheng granules (QFGs) are part of a traditional Chinese medicine formula, which has been widely used and found to be clinically effective with few side effects in various cancer treatments, including colorectal cancer (CRC). However, the precise mechanisms and molecular signaling pathways involved in the activity of QFGs' anticancer effect have not been reported in the literature. In this study, we hypothesized that QFGs can inhibit the growth of colorectal cancer cells, and that its mechanism is closely related to one or more intracellular signal transduction pathways. AIM: To better evaluate the mechanism underlying the anti-cancer effect of QFGs on the CRC cell lines HCT-116 and HCT-8. METHOD: First, we measured cell viability and cytotoxicity by performing MTT and lactate dehydrogenase (LDH) assays. We evaluated the role of QFGs in cell proliferation and apoptosis by assessing colony formation and analyzing Hoechst 33258 staining. Second, cell cycle and apoptosis rates were measured by fluorescence activated cell sorting, and the expression levels of survivin, cyclin D1, CDK4, p21, Bax, Bcl-2, Fas, FasL, and cleaved-caspase-3/-8/-9 were measured by performing western blots and caspase activity assays. Furthermore, inhibitors of caspase-3/-8/-9 were used to elucidate the specific apoptosis pathway induced by QFGs in cancer cells. Finally, activation of the PI3K/AKT and ERK signaling pathways was examined using the western blot assay to investigate the possible mechanism. RESULTS: MTT and LDH assays revealed that after 0.5-2.0 mg/mL of QFGs treatment, cell viability was reduced by (6.90% ± 1.03%)-(59.70% ± 1.51%) (HCT-116; P < 0.05) and (5.56% ± 4.52%)-(49.44% ± 2.47%) (HCT-8; P < 0.05), and cytotoxicity was increased from 0.52 ± 0.023 to 0.77 ± 0.002 (HCT-116; P < 0.01) and from 0.56 ± 0.054 to 0.81 ± 0.044 (HCT-8; P < 0.01) compared with the non-QFGs treatment groups. Additionally, colony formation and Hoechst 33258 staining assays showed that QFGs inhibited proliferation and induced apoptosis in CRC cells. QFGs also increased the expression levels of Bax, Fas and FasL, decreased the level of Bcl-2, and stimulated the activation of caspase-3/-8/-9, which were revealed by western blot and caspase activity assays. In contrast, when adding the three caspase inhibitors, the suppression effect of QFGs on cell viability and apoptosis were markedly inhibited. Moreover, QFGs suppressed the phosphorylation levels of PI3K, AKT and ERK. CONCLUSION: These results demonstrated that QFGs can inhibit CRC cell proliferation and induce apoptosis by suppressing the PI3K/AKT and ERK signaling pathways.

19.
Chin J Integr Med ; 25(6): 439-445, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29619747

ABSTRACT

OBJECTIVES: To investigate the protective effects of Shexiang Tongxin Dropping Pill (, STP) on Na2S2O4-induced hypoxia-reoxygenation injury in cardiomyoblast H9c2 cells. METHODS: The cell viability and levels of mRNA and protein expression in H9c2 cells were determined following Na2S2O4-induced hypoxia using Hoechst staining, annexin V/propidium iodide (PI) flow cytometry, real-time polymerase chain reaction and Western blot analysis. RESULTS: STP pretreatment significantly increased the viability and inhibited aberrant morphological changes in H9c2 cardiomyoblast cells induced by Na2S2O4 treatment (P<0.05). In addition, STP pretreatment attenuated Na2S2O4-induced hypoxic damage, down-regulated the expression of pro-apoptotic Bax, and up-regulated the expression of anti-apoptotic Bcl-2 in H9c2 cells (P<0.05). CONCLUSIONS: STP was strongly cardioprotective in hypoxia-reoxygenation injury by preventing hypoxic damage and inhibiting cellular apoptosis. These results further support the use of STP as an effective drug for the treatment of ischemic heart disease.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Oxygen/adverse effects , Protective Agents/pharmacology , Sulfates/toxicity , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , bcl-2-Associated X Protein/metabolism
20.
J Evid Based Integr Med ; 23: 2515690X18789632, 2018.
Article in English | MEDLINE | ID: mdl-30045633

ABSTRACT

In this study, hepatocellular carcinoma (HCC) mouse xenograft model, MTT assay, colony formation, nuclear staining, and Annexin-V/PI staining assays were used to evaluate the effect of Qingjie Fuzheng granules (QFG) on cell proliferation and apoptosis of HCC cell in vivo and in vitro. Furthermore, Western blotting was performed to detect the expression of Fas, FasL, Bcl-2, Bax, and the activation of caspase-3/-8/-9. The results showed that QFG reduced tumor weight ( P < .05) but had no effect on body weight gain in HCC mice in vivo. QFG significantly reduced HCC cell viability and attenuated cell proliferation in a dose-dependent manner ( P < .05). QFG increased the expression of Fas, FasL, and Bax ( P < .05). QFG downregulated the expression of Bcl-2 and promoted the activation of caspase-8, -9, and -3 ( P < .05). These results suggested that QFG possessed anticancer effects, and the mechanisms of action may involve the death receptor pathway and mitochondrion-dependent pathway-mediated apoptosis.

SELECTION OF CITATIONS
SEARCH DETAIL