Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Phytother Res ; 38(1): 131-146, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37821355

ABSTRACT

Neuroblastoma and glioblastoma are primary malignant tumors of the nervous system, with frequent relapse and limited clinical therapeutic drugs. The failure of their treatment is due to the tumor cells exhibiting cancer stem-like cells (CSLCs) properties. Octamer binding transcription factor 4 (Oct4) is involved in mediating CSLCs, our previous work found that Oct4-driven reprogramming of astrocytes into induced neural stem cells was potentiated with continuous sonic hedgehog (Shh) stimulation. In this study, we aimed to study the importance of Oct4 and Shh combination in the stemness properties induction of neuroblastoma and glioblastoma cells, and evaluate the anti-stemness effect of dauricine (DAU), a natural product of bis-benzylisoquinoline alkaloid. The effect of Oct4 and Shh co-activation on cancer stemness was evaluated by tumor spheres formation model and flow cytometry analysis. Then the effects of DAU on SH-SY5Y and T98-G cells were assessed by the MTT, colony formation, and tumor spheres formation model. DAU acts on Oct4 were verified using the Western blotting, MTT, and so on. Mechanistic studies were explored by siRNA transfection assay, Western blotting, and flow cytometry analysis. We identified that Shh effectively improved Oct4-mediated generation of stemness in SH-SY5Y and T98-G cells, and Oct4 and Shh co-activation promoted cell growth, the resistance of apoptosis. In addition, DAU, a natural product, was found to be able to attenuate Oct4/Shh co-activated stemness and induce cell cycle arrest and apoptosis via blocking AKT/ß-catenin signaling in neuroblastoma and glioblastoma, which contributed to the neuroblastoma and glioblastoma cells growth inhibition by DAU. In summary, our results indicated that the treatment of DAU may be served as a potential therapeutic method in neuroblastoma and glioblastoma.


Subject(s)
Benzylisoquinolines , Biological Products , Glioblastoma , Neuroblastoma , Tetrahydroisoquinolines , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Hedgehog Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Benzylisoquinolines/pharmacology , Neoplastic Stem Cells , Cell Proliferation , Apoptosis , Biological Products/pharmacology
2.
J Transl Med ; 21(1): 597, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37670360

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) is a major subtype of breast cancer, with limited therapeutic drugs in clinical. Epidermal growth factor receptor (EGFR) is reported to be overexpressed in various TNBC cells. Cantharidin is an effective ingredient in many clinical traditional Chinese medicine preparations, such as Delisheng injection, Aidi injection, Disodium cantharidinate and vitamin B6 injection. Previous studies showed that cantharidin had satisfactory pharmacological activity on a variety of tumors. In this study, we aimed to study the therapeutic potential of cantharidin for TNBC treatment by targeting EGFR, and expound its novel regulator miR-607. METHODS: The effect of cantharidin on breast cancer in vivo was evaluated by 4T1 mice model. Then the effects of cantharidin on TNBC cells was assessed by the MTT, colony formation, and AnnexinV-PE/7AAD staining. Cantharidin acts on EGFR were verified using the cell membrane chromatography, RT-PCR, Western blotting, MTT, and so on. Mechanistic studies were explored by dual-luciferase report assay, RT-PCR, western blotting, and immunofluorescence staining assay. RESULTS: Cantharidin inhibited TNBC cell growth and induce apoptosis by targeting EGFR. miR-607 was a novel EGFR regulator and exhibited suppressive functions on TNBC cell behaviors. Mechanistic study showed that cantharidin blocked the downstream PI3K/AKT/mTOR and ERK/MAPK signaling pathway. CONCLUSION: Our results revealed that cantharidin may be served as a potential candidate for TNBC treatment by miR-607-mediated downregulation of EGFR.


Subject(s)
MicroRNAs , Triple Negative Breast Neoplasms , Animals , Mice , Humans , Cantharidin , Down-Regulation , Phosphatidylinositol 3-Kinases , ErbB Receptors , Apoptosis
3.
Phytomedicine ; 103: 154249, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35716538

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a major subtype of liver cancer, with a high mortality rate, and close relation to chronic hepatitis. The components of the NLRP3 inflammasome are poorly expressed or even lost in HCC. Downregulation of the NLRP3 inflammasome expression significantly affects the clinical stages and pathological grade of HCC. According to previous research, Shuanghua decoction (SHD), a traditional folk prescription, has an inhibitory effect on nasopharyngeal cancer. PURPOSE: This study aimed to reveal the therapeutic potential of the traditional folk recipe, SHD and its demolition recipe for HCC, and to explore the underlying mechanism. METHODS: The effect of SHD and its demolition recipe on HCC cell biological behaviors was assessed using the MTT assay, colony formation, LDH release assay, KFluor-Edu staining, annexin V-FITC/PI staining assay, Hoechst staining, wound-healing assay, transwell assay, reactive oxygen species (ROS) release assay, HPLC, nude mice model, HE staining, IHC, western blot, and immunofluorescence staining in vitro and in vivo. RESULTS: SHD was found to inhibit HCC, and Oldenlandia and OP (Oldenlandia: Prunella spike = 2.5:1) were identified as the main ingredients that inhibited the proliferation and migration of HCC cells via the activation of the ROS-mediated NLRP3 inflammasome and inhibition of the NF-κB signaling pathway in vitro and in vivo. CONCLUSION: Overall, Chinese medicine theory and pharmacology research revealed that SHD, Oldenlandia and OP may be promising traditional Chinese medicine for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nasopharyngeal Neoplasms , Animals , Carcinoma, Hepatocellular/drug therapy , Drugs, Chinese Herbal , Inflammasomes , Liver Neoplasms/drug therapy , Mice , Mice, Nude , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
4.
Adv Mater ; 34(15): e2110219, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35170096

ABSTRACT

Tumorigenic environments, especially aberrantly overexpressed oncogenic microRNAs, play a critical role in various activities of tumor progression. However, developing strategies to effectively utilize and manipulate these oncogenic microRNAs for tumor therapy is still a challenge. To address this challenge, spherical nucleic acids (SNAs) consisting of gold nanoparticles in the core and antisense oligonucleotides as the shell are fabricated. Hybridized to the oligonucleotide shell is a DNA sequence to which doxorubicin is conjugated (DNA-DOX). The oligonucleotides shell is designed to capture overexpressed miR-21/miR-155 and inhibit the expression of these oncogenic miRNAs in tumor cells after tumor accumulation to manipulate genetic environment for accurate gene therapy. This process further induces the aggregation of these SNAs, which not only generates photothermal agents to achieve on-demand photothermal therapy in situ, but also enlarges the size of SNAs to enhance the retention time in the tumor for sustained therapy. The capture of the relevant miRNAs simultaneously triggers the intracellular release of the DNA-DOX from the SNAs to deliver tumor-specific chemotherapy. Both in vivo and in vitro results indicate that this combination strategy has excellent tumor inhibition properties with high survival rate of tumor-bearing mice, and can thus be a promising candidate for effective tumor treatment.


Subject(s)
Metal Nanoparticles , MicroRNAs , Nanoparticles , Neoplasms , Nucleic Acids , Animals , Carcinogenesis , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Gold , Metal Nanoparticles/therapeutic use , Mice , MicroRNAs/genetics , Neoplasms/drug therapy , Phototherapy
5.
Phytother Res ; 35(8): 4567-4578, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34058790

ABSTRACT

Regorafenib (RGF), a second-line multi-kinase inhibitor in the treatment of HCC (hepatocellular carcinoma) after sorafenib failure, exposes to the risk of drug resistance and subsequent progression of HCC patients. Toosendanin (TSN), a triterpenoid has presented excellent inhibition on several tumors. The purpose of this study is to investigate the inhibitory effect of the combination of TSN and RGF on HCC cells. We identified that TSN and RGF combination (TRC) synergistically inhibited the proliferation and migration of MHCC-97L cells. The upregulation of WWOX (WW-domain containing oxidoreductase) played a vital role in the HCC cell growth treated with TRC. TRC suppressed the phosphorylation of Stat3 and expression of DVL2, negatively regulated the activity of ß-catenin by promoting the phosphorylation of GSK3ß. In addition, the intranuclear proteins, including MMP2, MMP9, and C-MYC were significantly inhibited by TRC. The in vivo xenograft models confirmed that TRC effectually prevented the tumor growth through upregulating WWOX. Therefore, the treatment of TRC may be a potential solution of RGF resistance and promising therapeutic method in malignant HCC.


Subject(s)
Carcinoma, Hepatocellular , Drugs, Chinese Herbal/pharmacology , Liver Neoplasms , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Tumor Suppressor Proteins/metabolism , WW Domain-Containing Oxidoreductase/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Glycogen Synthase Kinase 3 , Humans , Liver Neoplasms/drug therapy , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Cancer Lett ; 513: 50-62, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34015398

ABSTRACT

Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related deaths worldwide. Loss of WW-domain containing oxidoreductase (WWOX) has been proven to be associated with malignant metastasis in patients with HCC. In this study, by using a non-biased CRISPR knockout genetic screen targeting 19,050 human genes, we found that toosendanin (TSN) is a novel druggable WWOX candidate agonist for metastatic HCC patients. We also found that TSN exhibited significant anti-proliferative and anti-metastatic effects on HCC cells in a WWOX-dependent manner. Overexpression and knockdown of WWOX in vitro and in vivo confirmed that the suppression of HCC by TSN involved WWOX. TSN regulated Stat3, DVL2, and GSK3ß by transforming their interactions with WWOX as demonstrated by a Co-IP assay. TSN accelerated the degradation of ß-catenin by promoting the function of APC, AXIN1, CK1, and GSK3ß complex. Nuclear translocation of p-Stat3 Y705 and ß-catenin was impeded by the TSN-induced blockade of JAK2/Stat3 and Wnt/ß-catenin signaling, accompanied by the inhibition of MMPs and C-MYC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drugs, Chinese Herbal/therapeutic use , Janus Kinase 2/metabolism , Liver Neoplasms/drug therapy , STAT3 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , WW Domain-Containing Oxidoreductase/metabolism , Carcinoma, Hepatocellular/pathology , Drugs, Chinese Herbal/pharmacology , Humans , Liver Neoplasms/pathology , Neoplasm Metastasis , Signal Transduction , Wnt Signaling Pathway
7.
Biomaterials ; 262: 120357, 2020 12.
Article in English | MEDLINE | ID: mdl-32911253

ABSTRACT

Chemotherapy is one of the most commonly utilized approaches to treat malignant tumor. However, the well-controlled chemotherapy able to accurately manipulate local drugs for on-demand tumor treatment is still a challenge. Herein, a magnet and light dual-responsive hydrogel combining thermosensitive poly(N-acryloyl glycinamide) (PNAGA), doxorubicin (DOX) loaded and polyester (PE) capped mesoporous silica nanocarriers (MSNs) as well as Fe3O4 nanoparticles (Fe3O4 NPs) grafted graphene oxide (GO) was fabricated to address above issue. The Fe3O4 NPs and GO respectively serve as magnetothermal agent and photothermal agent to perform hyperthermia, meanwhile to generate chain motion of PNAGA with varying degrees under different conditions of magnetic field and/or NIR irradiation. This strategy not only allowed the gel-sol transition of the hydrogel by prior heating for tumor injection, but performed controllable release routes of DOX-MSNs-PE (DMP for short) nanocarriers to meet various requirements from different patients and the changing states of tumor. Furthermore, these escaped DMP nanocarriers could be taken by surrounding tumor cells, and then deliver their drug to these cells after rapid hydrolysis of the PE cap triggered by esterase, resulting in accurate chemotherapy. Both in vitro and in vivo results indicated that the PNAGA-DMP-Fe3O4@GO hydrogel combining well-controllable chemotherapy and hyperthermia can eliminate more than 90% tumor cells and effectively inhibit the tumor growth in mice model, demonstrating the great candidate of our hydrogel for accurate tumor therapy.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Animals , Doxorubicin , Humans , Magnets , Mice , Nanogels , Silicon Dioxide
8.
J Chromatogr A ; 1586: 82-90, 2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30545684

ABSTRACT

Erythropoietin-producing hepatocyte B4 (EphB4) has recently been reported to be an oncogenic factor in many cancers and overexpressed in several types of tumors. Thus, EphB4 has the potential to be a therapeutic target in these malignancies. High-performance affinity chromatography has been a powerful tool to study the interaction between drugs and receptors. In this study, we constructed a novel EphB4 affinity chromatography model to investigate the active compounds which can target EphB4. More than 50 crude extracts of traditional Chinese medicine were screened by this affinity chromatography model. The active ingredients sanguinarine from celandine and macleaya cordata, and berberine from coptis chinensis and phellodendron amurense were found to selectively bind on the EphB4 affinity column. Next biological evaluatation data showed that EphB4 played a critical role in the inhibitory effect of sanguinarine and berberine on cancer cell growth. The results indicated that EphB4 affinity chromatography model constructed in this study can be used to screen the active compounds targeting EphB4 effectively and rapidly, especially in natural products.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Chromatography, Affinity/methods , Coptis/chemistry , Drugs, Chinese Herbal/pharmacology , Receptor, EphB4/antagonists & inhibitors , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Receptor, EphB4/metabolism , Tumor Cells, Cultured
9.
Phytother Res ; 33(3): 745-755, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30565332

ABSTRACT

Hepatocellular carcinoma (HCC) is a biologically complex disease. Combination chemotherapy is a good strategy after surgery treatment. In this study, we report that berberine combined with HMQ1611 (BCH) had a good synergistic effect on the HCC. Our findings concluded that BCH showed good inhibition on the HCC proliferation and colony formation, which attributed to cell cycle arrest by BCH at G1 phase through impairing the expression of cyclinD1, cyclinE, and cdc2 and downregulated the phosphorylation of Akt, mTOR, and ERK. Moreover, BCH negatively regulated Wnt signaling pathway by upregulating the Axin and inhibiting the nuclear translocation of ß-catenin. BCH suppressed the phosphorylation of LRP5/6, GSK3ß, the expression of Wnt5a, Frizzled8, CK1, and APC, as well as the nucleus protein included MMP2, MMP3, MMP9, and c-myc. The above data of Wnt signaling regulators contributed to inhibition by BCH on cell migration. In vivo studies, BCH significantly suppressed the growth of SMMC-7721 xenograft tumors through downregulating Ki67 and ß-catenin, as well as upregulating Axin and p-ß-catenin. In conclusion, the results revealed that BCH exhibited potential antitumor activities against human liver cancer in vitro and in vivo, and the potential mechanism underlying these activities depended on the inhibition of the Wnt/ß-catenin signaling pathway.


Subject(s)
Acetanilides/pharmacology , Benzamides/pharmacology , Berberine/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , beta Catenin/physiology
10.
Phytomedicine ; 51: 48-57, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30466627

ABSTRACT

BACKGROUND: Drug therapy plays an important role in the treatment of cervical cancer, which is one of the most common solid tumors in women. Therefore, it is important to seek more effective and less toxic therapies. PURPOSE: The aim of this study is to investigate the therapeutic potential of HMQ-T-F5 (1-(4-(2-aminoquinazolin-7-yl)phenyl)-3-(2­bromo­5-(trifluoromethoxy) phenyl)thiourea) (F5) for cervical cancer and explore the related mechanism. METHODS: By performing MTT assay, colony formation assay, flow cytometry, wound-healing assay, transwell assay, immunofluorescent staining and siRNA assay, we study the effect of F5 on human cervical HeLa cells. The mechanism of F5 was also investigated. RESULTS: We found that F5 significantly inhibited HeLa cell proliferation, led to accumulation of cells in the S phase, and induced apoptosis and inhibited migration. Mechanistically, F5 inhibited HeLa cell growth and migration through repressing the expression and nuclear translocation of ß-catenin, enhancing Axin expression, inhibiting the phosphorylation of LRP5/6 and GSK3ß, as well as downregulating the Wnt downstream targeted proteins. Knockdown of a checkpoint ß-catenin by siRNA significantly attenuated HeLa cell proliferation. Furthermore, XAV939, an inhibitor of ß-catenin, was used to treat HeLa cells and the results demonstrated that F5 inhibited proliferation and migration via the inhibition of the Wnt/ß-catenin pathway. CONCLUSION: Our findings demonstrated that F5 can target ß-catenin potentially and is useful in the treatment of cervical cancer.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Thiourea/pharmacology , Wnt Signaling Pathway/drug effects , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Down-Regulation , Female , Glycogen Synthase Kinase 3 beta/metabolism , HeLa Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , RNA, Small Interfering/pharmacology , Thiourea/analogs & derivatives , beta Catenin/metabolism
11.
Phytomedicine ; 46: 85-92, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-30097126

ABSTRACT

BACKGROUND: Colorectal cancer remains the third most common malignancies and migration is one of the main factors for its high mortality rate. Brucine, a natural plant alkaloid, has been proved to possess a variety of pharmacological functions including anti-tumor activities. PURPOSE: The aim of this study was to investigate the inhibitory effect of brucine on the colorectal cancer and the underlying mechanism. METHODS: In this study, colony formation assay and transwell assay were used to investigate the effect of brucine on LoVo cells viability and migration. Immunofluorescence assay, western blot assay and Gelatin zymography assay were used to study the mechanism of brucine. Xenograft model in nude mice was induced to investigate the in vivo effect of brucine on LoVo cells. RESULTS: Brucine could significantly decrease the viability, inhibit the colony formation and induce the apoptosis of LoVo cells. Brucine could also suppress the migration of LoVo cells in a dose-dependent manner. Western blot analysis elucidated that the inhibition of migration was associated with the decreasing expression of matrix metalloproteinases including MMP2, MMP3 and MMP9. Moreover, we found that treatment of brucine could downregulate the expression of Frizzled-8, Wnt5a, APC and GSNK1A1, and increase the expression of AXIN1. Meanwhile, brucine also decreased the phosphorylation level of LRP5/6 and GSK3ß, and increased the level of p-ß-catenin. Xenografted model in nude mice study also revealed that oral administration of brucine could inhibit the growth and migration of LoVo cells by activating the expression of AXIN1 and p-ß-catenin. CONCLUSION: Brucine could suppress the migration of the colorectal cancer in vitro and in vivo and the effect was associated with the inhibition of the Wnt/ß-catenin signaling pathway.


Subject(s)
Cell Movement/drug effects , Colorectal Neoplasms/drug therapy , Strychnine/analogs & derivatives , Wnt Signaling Pathway , Animals , Apoptosis/drug effects , Axin Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Down-Regulation , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Strychnine/pharmacology , Xenograft Model Antitumor Assays , beta Catenin/metabolism
12.
Phytother Res ; 31(9): 1392-1399, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28703366

ABSTRACT

TPD7, a novel biphenyl urea taspine derivative, and berberine have presented inhibition on VEGFR2 that can be regulated by ephrin-B2 reverse signaling through interactions with the PDZ domain. The purpose of this study is to investigate the inhibitory effect of the combination of TPD7 and berberine (TAB) on T-cell acute lymphoblastic leukemia cell growth. TPD7 and berberine together synergistically inhibited the proliferation of Jurkat cells. Also, the combination of TAB induced G1 -phase cell-cycle arrest by downregulating the level of cyclin D1, cyclin E, and CDC2. Furthermore, the combination of TAB significantly enhanced apoptosis in Jurkat cells, and the apoptosis most likely resulted from the modulation of the level of Bcl-2 family members. Most importantly, the concomitant treatment simultaneously regulated the ephrin-B2 and VEGFR2 signaling, as well as modulated the MEK/ERK and PTEN/PI3K/AKT/mTOR signaling. Therefore, the combination treatment of TAB may be a promising therapeutic method in treating T-cell acute lymphoblastic leukemia. Copyright © 2017 John Wiley & Sons, Ltd.


Subject(s)
Berberine/pharmacology , Carbanilides/pharmacology , Ephrin-B2/metabolism , Hydroxylamines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Synergism , Humans , Jurkat Cells , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
13.
Phytomedicine ; 25: 45-51, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28190470

ABSTRACT

BACKGROUND: Berberine, a plant-derived compound isolated from Coptis chinensis used in traditional Chinese medicine, has been shown to possess anti-cancer properties. However, no study has shown that berberine could target ephrin-B2, which plays a critical role in cell proliferation and migration. PURPOSE: The aim of this study is to investigate the effect of berberine on cancer cell growth and migration, through the regulation of ephrin-B2 and downstream signaling molecules. METHODS: In this study, a high ephrin-B2-expressing cell membrane chromatography method was developed to investigate 48 crude extracts from traditional Chinese medicine that could act on ephrin-B2. Cell proliferative and wound-healing assays were used to study the effect of berberine on cancer cell growth and migration. The mechanism of berberine was investigated using western blot. RESULTS: Berberine was isolated from C. chinensis extracts and showed activity on the HEK293/ephrin-B2 cell membrane chromatography column. Berberine showed a greater inhibitory effect in high-expressing ephrin-B2 cells (HEK293/ephrin-B2 cells) than in normal HEK293 cells, and decreased the expression of ephrin-B2 and its PDZ binding proteins, which indicates that ephrin-B2 is a target of berberine. Furthermore, berberine downregulates the phosphorylation of VEGFR2 and downstream signaling members (AKT and Erk1/2), which in turn downregulates the expression of MMP2 and MMP9. CONCLUSION: The above data confirm the inhibitory effects of berberine on ZR-75-30 cell proliferation and cell migration. Overall, our studies demonstrate that berberine inhibits cell growth and migration by targeting ephrin-B2.


Subject(s)
Berberine/pharmacology , Breast Neoplasms/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Coptis/chemistry , Ephrin-B2/metabolism , Plant Extracts/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Berberine/therapeutic use , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , HEK293 Cells , Humans , Phosphorylation , Phytotherapy , Plant Extracts/therapeutic use , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL