Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Ethnopharmacol ; 287: 114904, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-34952191

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Fuzheng Xiaojijinzhan (FZXJJZF) decoction is an effective prescription for treating colorectal cancer liver metastasis (LMCRC). AIM OF THE STUDY: To elucidate the pharmacological mechanism of the FZXJJZF decoction therapy on LMCRC. MATERIALS AND METHODS: Firstly, a network pharmacological approach was used to characterize the underlying targets of FZXJJZF on LMCRC. Secondly, LMCRC-related genes are obtained from the public database TCGA, and those genes are further screened and clustered through Mfuzz, an R package tool. Then, targets of FZXJJZF predicted by network pharmacology were overlapped with LMCRC related genes screened by Mfuzz. Meanwhile, FZJZXJF intervened in LMCRC model,epithelial-to-mesenchymal transition (EMT), and migration and invasion of HCT-116 cells. Thirdly, the transcriptomics data of FZJZXJF inhibited HCT-116 cells of EMT cells were overlapped with EMT database data to narrow the possible range of targets. Based on this, the potential targets and signal pathways of FZJZXJF were speculated by combining the transcriptomics data with the targets from network pharmacology-TCGA. Finally, the anti-cancer mechanism of FZXJJZF on LMCRC was verified in vitro by Real-Time PCR and Western Blot in vitro. RESULTS: By network pharmacological analysis, 282 ingredients and 429 potential targets of FZXJJZF were predicted. The 9268 LMCRC-related genes in the TCGA database were classified into 10 clusters by the Mfuzz. The two clustering genes with the most similar clustering trends were overlapped with 429 potential targets, and 32 genes were found, such as CD34, TRPV3, PGR, VDR, etc. In vivo experiments, FZJZXJF inhibited the tumor size in LMCRC models, and the EMT, migration, and invasion of HCT-116 also be inhibited. Intersecting transcriptomics dates with 32 target genes, it is speculated that the VDR-TGF-ß signaling pathway may be an effective mechanism of FZXJJZF. Additionally, it is shown that FZXJJZF up-regulated the expression levels of VDR and E-cadherin and down-regulated the expression levels of TGF-ß and Snail1 in vitro. These results confirmed that FZXJJZF plays an effective role in LMCRC mainly by inhibiting EMT phenotype via the VDR-TGF-ß signaling pathway. CONCLUSIONS: Collectively, this study reveals the anti-LMCRC effect of FZXJJZF and its potential therapeutic mechanism from the perspective of potential targets and potential pathways.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , Drugs, Chinese Herbal/pharmacology , Liver Neoplasms/prevention & control , Animals , Cell Movement/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , HCT116 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Invasiveness/prevention & control , Network Pharmacology , Receptors, Calcitriol/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism
2.
Life Sci ; 290: 120266, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34968467

ABSTRACT

AIM: Gastric cancer is a prevalent malignant tumor that seriously affects human health. Berberine (BBR), an alkaloid from Chinese herbal medicines, inhibits the proliferation of various cancers. We evaluated the effects and related mechanisms of BBR on gastric cancer. MAIN METHODS: The MTT assay, flow cytometry, scratch assays, transwell experiments and xenograft nude mice models were used to investigate the antineoplastic effects of BBR. RNA-Seq, qRT-PCR, WB and ELISA were used to investigate the underlying mechanisms of BBR on gastric cancer metastasis. KEY FINDINGS: BBR treatment inhibited the proliferation of MKN-45 and HGC-27 cells, induced their apoptosis, G0/G1 cell arrest, and suppressed the migration as well as invasion of GC cells in vitro. Moreover, BBR inhibited in vivo tumor growth in MKN-45 xenograft mice. RNA-seq showed that interactions between cytokines and their receptors was one of the greatest enrichment modulated pathways and IL-6 was a key target. IL-6 knockdown significantly inhibited the activities of MKN-45 cells. Mechanistically, these findings imply that BBR inhibits GC cell proliferation by modulating the signaling pathways related to IL-6/JAK2/STAT3. SIGNIFICANCE: This study provides a theoretical basis for the use of BBR in gastric cancer prevention.


Subject(s)
Berberine/pharmacology , Stomach Neoplasms/metabolism , Animals , Apoptosis/drug effects , Berberine/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , China , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Interleukin-6/metabolism , Janus Kinase 2/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/prevention & control , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Xenograft Model Antitumor Assays
3.
J Ethnopharmacol ; 286: 114926, 2022 Mar 25.
Article in English | MEDLINE | ID: mdl-34929308

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Amorphophalli Rhizoma (APR) is widely used as an adjuvant treatment for advanced and metastatic triple-negative breast cancer (TNBC), but its effects, potential active ingredients, and mechanism of action on estrogen receptor-positive (ER+) and human epidermal growth factor receptor-positive (HER2+) breast cancer cells were not reported. AIM OF THE STUDY: The present study investigated the effects and mechanism of APR on ER+ and HER2+ breast cancer cells. MATERIALS AND METHODS: Rotary evaporation was used to prepare different extracts of APR. Cell activity was assessed using the cell counting kit-8 (CCK-8) method. Wound healing assays were used to assess cell migration, and a cell invasion assay was performed using a Transwell chamber with Matrigel matrix. A xenograft model was used to analyze the inhibitory effects of APR on tumor growth. Bioinformatics analyses were used to explore the potential mechanism of APR in breast cancer. RT-qPCR and Western blotting were performed to reveal the molecular mechanism. RESULTS: The ethyl acetate extract of APR showed the strongest tumor inhibitory effect on ER+ and HER2+ breast cancer cells compared to petroleum ether or N-butanol extracts. APR inhibited ER+ and HER2+ breast cancer cell growth, proliferation, migration, and invasion via the phosphatidylinositol-3 kinase (PI3K)/AKT pathway. CONCLUSIONS: APR had a significant inhibitory effect on ER+ and HER2+ breast cancer cells via the PI3K/AKT signaling pathway. Therefore, APR may be useful for preventing ER+ and HER2+ breast tumor growth, proliferation, migration, and invasion.


Subject(s)
Amorphophallus/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/prevention & control , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Rhizome , Xenograft Model Antitumor Assays
4.
Acta Pharmacol Sin ; 43(6): 1544-1555, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34426644

ABSTRACT

Metastasis is the leading cause of death in breast cancer patients. Osthole, as an active compound detected in the traditional Chinese medicine Wenshen Zhuanggu Formula, has shown a promising anti-metastatic activity in human breast cancer cells, but the underlying mechanisms remain ambiguous. In this study we elucidated the anti-metastatic mechanisms of osthole in highly metastatic breast cancer cells and a zebrafish xenograft model. We showed that the expression of integrin α3 (ITGα3) and integrin ß5 (ITGß5) was upregulated in highly metastatic MDA-MB-231, MDA-MB-231BO breast cancer cell lines but was downregulated in poorly metastatic MCF-7 breast cancer cell line, which might be the key targets of osthole's anti-metastatic action. Furthermore, we showed that knockdown of ITGα3 and ITGß5 attenuated breast cancer cell migration and invasion possibly via suppression of FAK/Src/Rac1 pathway, whereas overexpression of ITGα3 and ITGß5 caused the opposite effects. Consistently, osthole significantly inhibited breast cancer metastasis by downregulating ITGα3/ITGß5 signaling in vitro and in vivo. These results provide new evidence that osthole may be developed as a candidate therapeutic drug for metastatic breast cancer.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Coumarins/pharmacology , Coumarins/therapeutic use , Female , Humans , Neoplasm Invasiveness/prevention & control , Zebrafish
5.
PLoS Comput Biol ; 17(11): e1009171, 2021 11.
Article in English | MEDLINE | ID: mdl-34843456

ABSTRACT

Predictive approaches such as virtual screening have been used in drug discovery with the objective of reducing developmental time and costs. Current machine learning and network-based approaches have issues related to generalization, usability, or model interpretability, especially due to the complexity of target proteins' structure/function, and bias in system training datasets. Here, we propose a new method "DRUIDom" (DRUg Interacting Domain prediction) to identify bio-interactions between drug candidate compounds and targets by utilizing the domain modularity of proteins, to overcome problems associated with current approaches. DRUIDom is composed of two methodological steps. First, ligands/compounds are statistically mapped to structural domains of their target proteins, with the aim of identifying their interactions. As such, other proteins containing the same mapped domain or domain pair become new candidate targets for the corresponding compounds. Next, a million-scale dataset of small molecule compounds, including those mapped to domains in the previous step, are clustered based on their molecular similarities, and their domain associations are propagated to other compounds within the same clusters. Experimentally verified bioactivity data points, obtained from public databases, are meticulously filtered to construct datasets of active/interacting and inactive/non-interacting drug/compound-target pairs (~2.9M data points), and used as training data for calculating parameters of compound-domain mappings, which led to 27,032 high-confidence associations between 250 domains and 8,165 compounds, and a finalized output of ~5 million new compound-protein interactions. DRUIDom is experimentally validated by syntheses and bioactivity analyses of compounds predicted to target LIM-kinase proteins, which play critical roles in the regulation of cell motility, cell cycle progression, and differentiation through actin filament dynamics. We showed that LIMK-inhibitor-2 and its derivatives significantly block the cancer cell migration through inhibition of LIMK phosphorylation and the downstream protein cofilin. One of the derivative compounds (LIMKi-2d) was identified as a promising candidate due to its action on resistant Mahlavu liver cancer cells. The results demonstrated that DRUIDom can be exploited to identify drug candidate compounds for intended targets and to predict new target proteins based on the defined compound-domain relationships. Datasets, results, and the source code of DRUIDom are fully-available at: https://github.com/cansyl/DRUIDom.


Subject(s)
Lim Kinases/antagonists & inhibitors , Lim Kinases/chemistry , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Computational Biology , Computer Simulation , Drug Development , Drug Discovery , Drug Evaluation, Preclinical , Drug Interactions , Humans , In Vitro Techniques , Ligands , Lim Kinases/metabolism , Machine Learning , Molecular Docking Simulation , Neoplasm Invasiveness/prevention & control , Neoplasms/drug therapy , Neoplasms/enzymology , Network Pharmacology/statistics & numerical data , Phosphorylation/drug effects , Protein Domains , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , User-Computer Interface
6.
Recent Pat Anticancer Drug Discov ; 16(2): 285-294, 2021.
Article in English | MEDLINE | ID: mdl-34355688

ABSTRACT

BACKGROUND: In recent years, there is an increasing interest in using Traditional Chinese Medicine (TCM) and their patents for the treatment of cancers. Qigefang (QGF) is a TCM formula and has been used for the treatment of metastatic esophageal cancer in China. However, its therapeutic effect on tumors and its mechanism of action is largely unknown. The aim of this study is to explore the role of QGF in the treatment of metastasis of Esophageal Squamous Cell Carcinoma( ESCC). METHODS: Human esophageal carcinoma cell line KYSE150 was used for this study. CCK-8 assay was used to determine the cytotoxicity of QGF. The KYSE150 cells were treated with QGF to determine its effect on cell migration (cell scratch assay and imaging) and invasion (Transwell system based with Matrigel assay). Western blotting was used to investigate the effect of QGF on relevant molecules of signaling pathways. A mouse model of lung metastasis of esophageal cancer was established by injecting the KYSE150-Luc cells through the tail vein. A small animal imaging system was used to observe tumor metastasis in the mice. RESULTS: QGF reduced cell migration and invasion of KYSE150 cells. QGF significantly inhibited lung metastasis in nude mice. Further study revealed that the expression of Growth arrest-specific 6 (Gas6), Anexelekto (Axl), N-Nuclear factor-kappa B (NF-κB) and matrix metalloproteinase-9 (MMP-9) proteins were decreased both in vitro and in vivo upon treatment with QGF. CONCLUSION: QGF could prevent invasion and metastasis of esophageal cancer by inhibiting the Gas6/Axl signaling pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drugs, Chinese Herbal/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Movement/drug effects , Drugs, Chinese Herbal/administration & dosage , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/prevention & control , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Patents as Topic , Signal Transduction/drug effects
7.
J Ethnopharmacol ; 277: 114184, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-33961996

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Rubia yunnanensis Diels is a traditional Chinese medicine that has diverse pharmacological activities, including antituberculosis, antirheumatism and anticancers. Rubioncolin C (RC), a natural naphthohydroquinone dimer isolated from the roots and rhizomes of R. yunnanensis Diels, has shown potent antitumor activity. However, the antitumor activity and its potential mechanism of RC in triple-negative breast cancer (TNBC) cell lines remained unclear. AIM OF THE STUDY: This study was aim to investigate the anti-proliferation and anti-metastasis activity as well as the potential mechanism of RC on triple-negative breast cancer cells in vitro and in vivo. MATERIALS AND METHODS: The sulforhodamine B assay, colony formation assay and cell cycle analysis were used to determine the anti-proliferative activity of RC on TNBC. The anti-metastatic activity in vitro of RC was detected through the scratch wound assay, cell migration and invasion assays and gelatin zymography. The flow cytometry, JC-1, GFP-LC3B plasmid transfection, MDC, Lysotracker red and Carboxy-H2DCFDA, DHE, and MitoSOX™ Red staining were performed to investigate the effect of RC on apoptosis, autophagy and ROS level. The apoptosis inhibitor, autophagy inhibitors and ROS inhibitors were used to further verify the antitumor mechanism of RC. The protein levels related with cell cycle, apoptosis, and autophagy were examined with western blotting. In addition, the anti-tumor activity of RC in vivo was assessed in an experimental metastatic model. RESULTS: In the present study, RC suppressed the proliferation of TNBC cells in a time- and dose-dependent manner via regulating cell cycle. Further experiments showed that RC inhibited the migration and invasion of TNBC cells by downregulating MMPs and inhibiting EMT. Moreover, we demonstrated that RC induced obviously apoptotic and autophagic cell death, activated MAPK signaling pathway and inhibited mTOR/Akt/p70S6K and NF-κB signaling pathways. Furthermore, the excessive ROS was produced after treatment with RC. The antioxygen NAC and GSH could rescue the cell viability and reestablish the ability of cell metastasis, and inhibit the RC-induced apoptosis and autophagy. In a mice lung metastasis model of breast cancer, RC inhibited lung metastasis, and induced autophagy and apoptosis. CONCLUSION: These findings clarified the antitumor mechanism of RC on TNBC cell lines and suggested that RC is a key active ingredient for the cancer treatment of R. yunnanensis, which would help RC develop as a new potential chemotherapeutic agent for TNBC treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Naphthoquinones/pharmacology , Rubia/chemistry , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/isolation & purification , Autophagy/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Naphthoquinones/administration & dosage , Naphthoquinones/isolation & purification , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Reactive Oxygen Species/metabolism , Time Factors
8.
J Ethnopharmacol ; 277: 114057, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-33771643

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Cimicifuga dahurica (Turcz.) Maxim (C. dahurica) has a long history of treating breast cancer. From the Qing Dynasty to the Tang Dynasty and even earlier, C. dahurica has been documented in the treatment of breast carbuncle (Breast cancer is classified as breast carbuncle in Chinese medicine). In traditional prescriptions such as "Sheng Ge Decoction", "Sheng Ma Powder" and "Breast Carbuncle Pill", as the main medicine, C. dahurica plays an important role. At present, the systematic studies on the in vitro and in vivo effects of Cimicifuga against breast cancer are rare, especially the C. dahurica. AIM OF THE STUDY: In this article, we evaluated the in vitro activity and in vivo effects of CREE (extract of the root of C. dahurica) against breast cancer, and discussed the possible mechanism of CREE in promoting breast cancer cell apoptosis. MATERIALS AND METHODS: The main component in the CREE was analyzed by HPLC. The effects of CREE on the proliferation, migration and invasion of human breast cancer cells were evaluated through SRB, colony assay, LDH release, wound healing and transwell assay. The pro-apoptotic effect of CREE was investigated in Hochest33342 and Annexin V-FITC/PI assay. To verify the results of CREE in vivo effects, we applied nude mice subcutaneous xenograft experiments. The possible mechanism of CREE treating breast cancer was investigated through mitochondrial membrane potential and western blot experiments. RESULTS: CREE contains cycloartane triterpene saponins. CREE can significantly inhibit the proliferation, migration and invasion of human breast cancer MCF-7 and MDA-MB-231 cells in vitro and it can effectively inhibit the growth of MDA-MB-231 cell subcutaneous tumors in vivo. Besides, we also found that CREE up-regulated the expression levels of Bax, caspase-9/3 and cytochrome C, and down-regulated the expression of Bcl-2. Therefore, regulation of the mitochondrial pathway may be one of the mechanisms by which CREE promotes breast cancer cell apoptosis. CONCLUSIONS: CREE exhibits sufficient anti-breast cancer activity in vivo and in vitro, this study provides persuasive evidence for the further research and development of C. dahurica.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Cimicifuga/chemistry , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/prevention & control , Xenograft Model Antitumor Assays
9.
Oncol Rep ; 45(4)2021 04.
Article in English | MEDLINE | ID: mdl-33649865

ABSTRACT

Kaempferol (KF), a flavonoid compound isolated from herbal medicines, has been reported to play a significant role in inhibiting certain types of cancer. Although recent studies reported that KF exerted inhibitive activity on liver cancer, they failed to elucidate the signaling pathways and synergistic effects in combination with chemotherapeutic drugs currently in use in the clinical setting. In the present study, the signaling pathways and synergistic effects of KF in liver cancer cells were investigated. Nine liver cancer cell lines were used to assess the inhibitive activity and synergistic effects of KF. Cellular behavioral experiments, such as viability, colony formation, cell cycle arrest, apoptotic, wound healing, and Transwell assays were used to assess the effects of KF on the proliferation, apoptosis, migration, and invasion of liver cancer cells. Western blotting was performed to validate the key signaling pathway elements underlying those cellular behaviors. KF exhibited inhibitory effects on nine liver cancer cell lines in time­ and dose­dependent manners and was mostly nontoxic to the normal hepatocyte cells. The combination of KF and doxorubicin revealed a stronger inhibitive effect on the viability of liver cancer cells. Combination therapy also revealed higher suppressive effects on colony formation, cell cycle progression, survival, DNA damage response, and mitochondrial function. By western blotting assay, mitochondrial and caspase signaling pathways were determined to be involved in proliferation inhibition. In wound healing and Transwell invasion assays, combination therapy also exhibited more robust inhibitory activity in blocking the migration and invasion of liver cancer cells. PI3K/mTOR/MMP protein pathways were also revealed to be related to cell migration inhibition. KF alone exhibited an inhibitory effect on proliferation, migration, and invasion of liver cancer cells, and its synergistic effects revealed stronger inhibitory activities. The present data indicated that KF is a promising candidate as a complementary medicine to conventional chemotherapeutic drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Doxorubicin/pharmacology , Kaempferols/pharmacology , Liver Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Doxorubicin/therapeutic use , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Kaempferols/therapeutic use , Liver Neoplasms/pathology , Neoplasm Invasiveness/prevention & control
10.
Molecules ; 25(21)2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33158043

ABSTRACT

We sought to investigate the effect of rose petal extract (RPE) on the proliferation, migration, and invasion of cancer cells. RPE significantly inhibited the growth of lung and colorectal cancer cell lines, with rapid suppression of A549 lung cancer cells at low concentrations. These effects occurred concomitantly with downregulation of the cell proliferation mediators PCNA, cyclin D1, and c-myc. In addition, RPE suppressed the migration and invasion of A549 cells by inhibiting the expression and activity of matrix metalloproteinase-2 and matrix metalloproteinase-9 (MMP-2 and -9). We hypothesize that the suppressive activity of RPE against lung cancer cell proliferation and early metastasis occurs via the EGFR-MAPK and mTOR-Akt signaling pathways. These early results highlight the significant potency of RPE, particularly for lung cancer cells, and warrant further investigation.


Subject(s)
Adenocarcinoma of Lung , Antineoplastic Agents, Phytogenic , Cell Movement/drug effects , Cell Proliferation/drug effects , Flowers/chemistry , Lung Neoplasms , Neoplasm Proteins/metabolism , Plant Extracts , Rosa/chemistry , Signal Transduction/drug effects , A549 Cells , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , ErbB Receptors/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Plant Extracts/chemistry , Plant Extracts/pharmacology
11.
Oncol Rep ; 44(6): 2537-2546, 2020 12.
Article in English | MEDLINE | ID: mdl-33125135

ABSTRACT

Poncirus fructus (PF) is a phytochemical compound extracted from the dry, immature fruits of Poncirus trifoliate. PF is traditionally used to treat gastrointestinal disorders, allergies, and inflammatory disease. In East Asia, PF is also known for its anticancer properties. There are numerous reports on the anticancer and anti­inflammatory effects of PF in a wide range of cancers and gastrointestinal diseases, respectively. However, the role of PF in inducing apoptosis and suppressing the invasiveness of hepatocellular carcinoma (HCC) remains unclear. This study investigated the ability of PF to induce apoptosis and inhibit the invasiveness and migratory ability of HCC cell lines (Hep3B and Huh7). Wound healing, Transwell migration and invasion, and colony­formation assays, as well as flow cytometry, were used to analyze cell proliferation, migration, invasion, and apoptosis. Epithelial­mesenchymal transition (EMT)­related and apoptotic proteins were assessed by western blotting. The mitochondrial membrane potential of the Hep3B and Huh7 cells was observed with tetramethylrhodamine ethyl ester. The reactive oxygen species (ROS) level was determined by dihydroethidium (DHE) staining. PF treatment significantly decreased the proliferation of Hep3B and Huh7 cells in a dose­dependent manner, reduced the mitochondrial membrane potential, increased ROS levels, decreased the protein levels of Bcl­2, and increased the protein levels of Bax and cleaved caspase­3 and 9, suggesting that PF mediated HCC apoptosis via a mitochondrial pathway. Our findings showed that PF prevented HCC cell migration and invasion by inhibiting the EMT process and downregulating MMP­2 and MMP­9 activities. The results suggest the potential anticancer effects of PF by inhibiting proliferation, inducing apoptosis, and reducing the invasion and migration of HCC cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Plant Extracts/pharmacology , Poncirus/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Drug Screening Assays, Antitumor , Fruit/chemistry , Humans , Liver Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use
12.
Front Endocrinol (Lausanne) ; 11: 568436, 2020.
Article in English | MEDLINE | ID: mdl-33101201

ABSTRACT

Background: Oral squamous cell carcinoma (OSCC) that comprises about 90% of all oral cancer cases is associated with poor prognosis due to its highly metastatic nature. The majority of OSCC treatment options are related detrimental side-effects. Hypothesis/Purpose: The present study aimed at deciphering the effects of a bioactive phytochemical, sodium danshensu, on human oral cancer cell metastasis. Methods and Results: The treatment of FaDu and Ca9-22 cells with different doses of sodium danshensu (25, 50, and 100 µM) caused a significant reduction in cellular motility, migration, and invasion, as compared to the untreated cells. This effect was associated with a reduced expression of MMP-2, vimentin and N-cadherin, together with an enhanced expression of E-cadherin and ZO-1. Further investigation on the molecular mechanism revealed that treatment with sodium danshensu caused significant reduction in p38 phosphorylation; however, phosphorylation of ERK1/2 significantly decreased only in FaDu cells, whereas p-JNK1/2 did not show any alteration. A combination of p38 and JNK1/2 inhibitors with sodium danshensu also reduced the migration in the FaDu and Ca9-22 cell lines. Conclusion: Collectively, the present study findings reveal that sodium danshensu execute anti-metastatic effect by suppressing p38 phosphorylation in human oral cancer. The study identifies sodium danshensu as a potential natural anticancer agent that can be used therapeutically to manage highly metastatic OSCC.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Cell Movement/drug effects , Drugs, Chinese Herbal/pharmacology , Lactates/pharmacology , MAP Kinase Signaling System/drug effects , Mouth Neoplasms/enzymology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/physiology , Humans , MAP Kinase Signaling System/physiology , Mouth Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control
13.
Biomed Res Int ; 2020: 9786428, 2020.
Article in English | MEDLINE | ID: mdl-33102601

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is an underlying deadly malignancy with poor prognosis, lacking effective therapies currently available to improve the prognosis. C18H17NO6 (AUCAN), a kind of dibenzofuran extracted from a special plant in Yunnan Province (China), is identified as a natural anticancer agent exerting strong inhibitory activities on various cancers. Our study was committed to investigating the potency of AUCAN against colorectal cancers and further exploring the potential mechanisms via proteomic analysis. METHODS: Cell Counting Kit-8 assay and immunofluorescence staining were used to investigate the effect of AUCAN on the viability and proliferation of HCT-116 cells and RKO cells. The apoptosis of HCT-116 and RKO cells after AUCAN administration was determined by the flow cytometry test. The effects of AUCAN on invasion and migration of tumor cells were investigated by the colony formation assay, wound healing test, and Transwell invasion test. Meanwhile, the energy metabolism and growth of tumor tissues after AUCAN administration with 10 mg/kg and 20 mg/kg were examined by PET-CT in vivo. The side effects of AUCAN treatment were also evaluated through blood routine and liver function examination. RKO cell proliferation and apoptosis in vivo were further determined by hematoxylin and eosin staining, TUNEL staining, and immunohistochemistry. Furthermore, the differentially expressed proteins (DEPs) involved in AUCAN treatment were determined by proteomic analysis followed by functional clustering analysis. RESULTS: The results showed that AUCAN suppressed the migratory abilities and enhanced apoptosis of HCT-116 and RKO cell lines. Meanwhile, AUCAN treatment dramatically depressed the growth and volume of colorectal tumors in nude mice and suppressed the survival of RKO cells in tumor tissues without any side effects on the blood routine and liver function. In addition, twenty-four upregulated and forty-two downregulated proteins were identified. Additionally, functional clustering analysis concealed enriched biological processes, cellular components, molecular functions, and related pathways of these proteins involved in cellular metabolic. Finally, the protein-protein interaction analysis revealed the regulatory connection among these DEPs. CONCLUSIONS: Taken together, AUCAN exerted its significant antitumor effect without side effects in the blood routine and liver function and the underlying mechanisms were preliminarily investigated by proteomic analysis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , Dibenzofurans/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drugs, Chinese Herbal/pharmacology , Female , HCT116 Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Plants, Medicinal , Positron Emission Tomography Computed Tomography , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics , Proteomics , Xenograft Model Antitumor Assays
14.
Life Sci ; 260: 118291, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32810510

ABSTRACT

AIMS: Despite the advanced cancer treatments, there is increased resistance to chemotherapy and subsequent mortality. In lack of reliable data in monolayer cultures and animal models, researchers are shifting to 3D cancer spheroids, which represents the in vivo robust tumour morphology. Calcium is essential in cell signalling and proliferation. It is found that T-type calcium channels (TTCCs) are overexpressed in various cancer cells, supporting their increased proliferation. Many of the TTCCs blockers available could target other channels besides TTCCs, which can cause adverse effects. Therefore, we hypothesise that TTA-A2, a highly selective blocker towards TTCCs, can inhibit the growth of cancer spheroids, and provide an anti-cancer and an adjuvant role in cancer therapy. METHODS: We studied TTA-A2 and paclitaxel (PTX-control drug) in lung adenocarcinoma cell line- A549, cancer cells and human embryonic kidney cell line- HEK 293, control cell, in their monolayer and spheroids forms for viability, proliferation, morphology change, migration, and invasion-after 48-96 h of treatment. KEY FINDINGS: Though the results varied between the monolayer and spheroids studies, we found both anti-cancer as well as adjuvant effect of TTA-A2 in both the studies. TTA-A2 was able to inhibit the growth, viability, and metastasis of the cancer cells and spheroids. Differences in the results of two modes might explain that why drugs tested successfully in monolayer culture fail in clinical trials. SIGNIFICANCE: This study establishes the role of TTA-A2, a potent TTCC blocker as an anti-cancer and adjuvant drug in reducing the viability and metastasis of the cancer cells.


Subject(s)
Adenocarcinoma of Lung/pathology , Antineoplastic Agents , Benzeneacetamides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Lung Neoplasms/pathology , Pyridines/pharmacology , A549 Cells , Adenocarcinoma of Lung/drug therapy , Benzeneacetamides/therapeutic use , Calcium Channels, T-Type/physiology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , Humans , Lung Neoplasms/drug therapy , Neoplasm Invasiveness/prevention & control , Pyridines/therapeutic use
15.
Theranostics ; 10(17): 7906-7920, 2020.
Article in English | MEDLINE | ID: mdl-32685028

ABSTRACT

Background: Capsaicin is an active compound found in plants of the Capsicum genus; it has a range of therapeutic benefits, including anti-tumor effects. Here we aimed to delineate the inhibitory effects of capsaicin on nasopharyngeal carcinoma (NPC). Methods: The anti-cancer effects of capsaicin were confirmed in NPC cell lines and xenograft mouse models, using CCK-8, clonogenic, wound-healing, transwell migration and invasion assays. Co-immunoprecipitation, western blotting and pull-down assays were used to determine the effects of capsaicin on the MKK3-p38 axis. Cell proliferation and EMT marker expression were monitored in MKK3 knockdown (KD) or over-expression NPC cell lines treated with or without capsaicin. Finally, immunohistochemistry was performed on NPC specimens from NPC patients (n = 132) and the clinical relevance was analyzed. Results: Capsaicin inhibited cell proliferation, mobility and promoted apoptosis in NPC cells. Then we found that capsaicin directly targets p38 for dephosphorylation. As such, MKK3-induced p38 activation was inhibited by capsaicin. Furthermore, we found that capsaicin-induced inhibition of cell motility was mediated by fucokinase. Xenograft models demonstrated the inhibitory effects of capsaicin treatment on NPC tumor growth in vivo, and analysis of clinical NPC samples confirmed that MKK3 phosphorylation was associated with NPC tumor growth and lymphoid node metastasis. Conclusions: The MKK3-p38 axis represents a potential therapeutic target for capsaicin. MKK3 phosphorylation might serve as a biomarker to identify NPC patients most likely to benefit from adjunctive capsaicin treatment.


Subject(s)
Capsaicin/pharmacology , MAP Kinase Kinase 3/metabolism , MAP Kinase Signaling System/drug effects , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/drug therapy , Animals , Capsaicin/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , MAP Kinase Kinase 3/genetics , MAP Kinase Signaling System/genetics , Male , Mice , Middle Aged , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Xenograft Model Antitumor Assays
16.
Oncol Rep ; 44(3): 1049-1063, 2020 09.
Article in English | MEDLINE | ID: mdl-32705271

ABSTRACT

Increasing evidence indicates that the inflammatory tumor microenvironment can lead to cancer cell metastasis. Shikonin, which is extracted from the Chinese herb Zicao (the dried root of Lithospermum erythrorhizon), possesses various pharmacological effects, but its effect on tumor metastasis in the inflammatory microenvironment remains unknown. In the present study, we aimed to investigate the potential effect of shikonin on tumor metastasis in an inflammatory microenvironment as well as the underlying molecular mechanisms. It was found that, in the inflammatory microenvironment simulated by THP­1 cell conditioned medium (THP­1­CM) in vitro, shikonin significantly inhibited the epithelial­mesenchymal transition (EMT), migration and invasion of human lung adenocarcinoma cell lines A549 and H1299. In addition, we found that interleukin­6 (IL­6), which is expressed in THP­1­CM, promoted the EMT of lung adenocarcinoma cells, and shikonin markedly inhibited IL­6­induced EMT and cell motility. Moreover, shikonin inhibited IL­6­induced phosphorylation of signal transducer and activator of transcription 3 (STAT3), prevented phosphorylated STAT3 (p­STAT3) translocation into the nucleus, and suppressed p­STAT3 transactivation activity. Additionally, it was found that shikonin inhibited lung metastasis, EMT and expression of p­STAT3 of A549 cells in vivo. Furthermore, IL­6 levels in human lung adenocarcinoma tissues were significantly associated with tumor­node­metastasis stage and lymph node metastasis, and its expression was correlated with tumor­associated macrophage (TAM) infiltration. Together, these results suggest that shikonin suppresses the migration and invasion of human lung adenocarcinoma cells in an inflammatory microenvironment involving the IL­6/STAT3 signaling pathway.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Drugs, Chinese Herbal/pharmacology , Lung Neoplasms/drug therapy , Lymphatic Metastasis/drug therapy , Naphthoquinones/pharmacology , A549 Cells , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/secondary , Cell Movement/drug effects , Cell Movement/immunology , Drugs, Chinese Herbal/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/immunology , Female , Humans , Interleukin-6/analysis , Interleukin-6/metabolism , Lung/immunology , Lung/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphatic Metastasis/immunology , Male , Middle Aged , Naphthoquinones/therapeutic use , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/prevention & control , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , THP-1 Cells , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Xenograft Model Antitumor Assays
17.
Discov Med ; 29(156): 5-16, 2020.
Article in English | MEDLINE | ID: mdl-32598859

ABSTRACT

Traditional Chinese Medicine (TCM) has been increasingly studied for antitumor activities. Icaritin, a hydrolytic product of icariin, is an effective ingredient of the traditional Chinese herb epimedium with multiple pharmacological activities. Among them, the antitumor activity of icaritin has been widely studied and reported in tumors both in vitro and in vivo. While its exact antitumor mechanisms await revelation, icaritin has been found to regulate several key molecules and pathways concerning cell fate, including CDK-dependent pathways, mitogen-activated protein kinases (MAPKs), the serine-threonine kinase AKT, signal transducer and activator of transcription 3 (STAT3), and p53. The ability to induce cellular oxidative stress also contributes to its antitumor activity. This review outlines the results of key investigations focusing on the antitumor effects and mechanisms of icaritin.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , Neoplasms/drug therapy , Phytoestrogens/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drugs, Chinese Herbal/therapeutic use , Epimedium/chemistry , Flavonoids/therapeutic use , Humans , Neoplasm Invasiveness/prevention & control , Neoplasms/pathology , Oxidative Stress/drug effects , Phytoestrogens/therapeutic use , Signal Transduction/drug effects
18.
Int J Med Mushrooms ; 22(2): 197-209, 2020.
Article in English | MEDLINE | ID: mdl-32479007

ABSTRACT

Melanoma is among the most aggressive and treatment-resistant human cancers. Phellinus baumii, a famous medicinal mushroom, has been used to treat different diseases, including cancer, in China and other east Asian countries. The purpose of this research was to explore its anticancer effects against melanoma, and the mechanisms that might be involved. CCK-8 assay exhibited that extracts of Ph. baumii (EPB) strongly inhibited cell viability of A375 melanoma cancer cell. Typical morphological changes of cell apoptosis were observed in EPB-treated A375 cells in Hoechst staining assay. Flow cytometry analysis indicated that EPB significantly induced A375 cells apoptosis and the cell cycle was disrupted in S phase. EPB increased the expression of Bax, and decreased Bcl-2 in A375 cells. EPB remarkably caused mitochondrial membrane potential collapse and induced a mitochondrial-dependent apoptosis in A375 cells evidenced by caspase-3 activation, followed by PARP cleavage. More importantly, EPB has shown a strong inhibitory effect on the migration and aggression of the A375 cells through the healing of the wound and transwell assay. In vivo, EPB was also found to strongly inhibit the growth of tumors in BALB/c nude mice. Our results indicated that Ph. baumii might be a natural therapeutic product for aggressive melanoma because it could induce apoptosis and inhibit metastasis in A375 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Basidiomycota/chemistry , Complex Mixtures/pharmacology , Melanoma/drug therapy , Agaricales , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Complex Mixtures/chemistry , Complex Mixtures/isolation & purification , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Neoplasm Invasiveness/prevention & control
19.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188380, 2020 08.
Article in English | MEDLINE | ID: mdl-32461135

ABSTRACT

Cellular communication through gap junctions and hemichannels formed by connexins and through channels made by pannexins allows for metabolic cooperation and control of cellular activity and signalling. These channel proteins have been described to be tumour suppressors that regulate features such as cell death, proliferation and differentiation. However, they display cancer type-dependent and stage-dependent functions and may facilitate tumour progression through junctional and non-junctional pathways. The accumulated knowledge and emerging strategies to target connexins and pannexins are providing novel clinical opportunities for the treatment of cancer. Here, we provide an updated overview of the role of connexins and pannexins in malignant melanoma. We discuss how targeting of these channel proteins may be used to potentiate antitumour effects in therapeutic settings, including through improved immune-mediated tumour elimination.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Connexins/metabolism , Melanoma/secondary , Skin Neoplasms/pathology , Skin/pathology , Animals , Antineoplastic Agents, Immunological/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Communication/drug effects , Cell Communication/immunology , Cell Line, Tumor , Connexins/agonists , Connexins/antagonists & inhibitors , Disease Models, Animal , Disease Progression , Gap Junctions/drug effects , Gap Junctions/pathology , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Melanoma/drug therapy , Melanoma/immunology , Melanoma/mortality , Microbiota/immunology , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Staging , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/cytology , Skin/microbiology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
20.
Integr Cancer Ther ; 19: 1534735420906463, 2020.
Article in English | MEDLINE | ID: mdl-32248718

ABSTRACT

Chemotherapy is an effective treatment for invasive breast cancer. Paradoxically, many recently published findings showed that the first-line chemotherapeutic agent paclitaxel (PTX) showed pro-metastatic effects in the progress of treating breast cancer. Xiao-Ai-Ping (XAP) injection, composed of a traditional herbal medicine, Marsdenia tenacissimae extract, is known to exert antitumor effects on various cancers. However, there are few experimental studies on breast cancer. The underlying mechanism of the antitumor effect of XAP combined with chemotherapy agents has not been fully understood. In the present study, we sought to find the antitumor effects of XAP combined with PTX in vitro and in vivo. The data demonstrated that the combination of XAP with PTX resulted in remarkable enhancement of the pro-apoptotic, migration-inhibiting, and anti-invasive effects of PTX in vitro. Significantly, further study showed the overexpression of ATF3 in PTX-treated cell, while XAP counteracted the change of ATF3 induced by PTX. Moreover, it showed that combination treatment could promote the inhibition of tumor growth in MDA-MB-231 cell xenograft mouse model. Compared with PTX treatment, the downregulation of ATF3 indicated that ATF3 played a pivotal role in the combination of XAP with PTX to exert a synergistic effect. Overall, it is expected that PTX combined with XAP may serve as an effective agent for antitumor treatment, and dampening ATF3 maybe a potential strategy to improve the efficacy of PTX.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Paclitaxel/pharmacology , Activating Transcription Factor 3/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Neoplasm Invasiveness/prevention & control , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL