Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Phytother Res ; 35(7): 3916-3935, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33970512

ABSTRACT

The programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway is abnormally expressed in cervical cancer cells. Moreover, PD-1/PD-L1 blockade reduces the apoptosis and exhaustion of T cells and inhibits the development of malignant tumors. Usnic acid is a dibenzofuran compound originating from Usnea diffracta Vain and has anti-inflammatory, antifungal, and anticancer activities. However, the molecular mechanism of its antitumor effects has not been fully elucidated. In this work, we first observed that usnic acid decreased the expression of PD-L1 in HeLa cells and enhanced the cytotoxicity of co-cultured T cells toward tumor cells. Usnic acid inhibited PD-L1 protein synthesis by reducing STAT3 and RAS pathways cooperatively. It was subsequently shown that usnic acid induced MiT/TFE nuclear translocation through the suppression of mTOR signaling pathways, and promoted the biogenesis of lysosomes and the translocation of PD-L1 to the lysosomes for proteolysis. Furthermore, usnic acid inhibited cell proliferation, angiogenesis, migration, and invasion, respectively, by downregulating PD-L1, thereby inhibiting tumor growth. Taken together, our results show that usnic acid is an effective inhibitor of PD-L1 and our study provide novel insights into the mechanism of its anticancer targeted therapy.


Subject(s)
B7-H1 Antigen , Benzofurans/pharmacology , Cell Proliferation/drug effects , T-Lymphocytes/immunology , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , HeLa Cells , Humans , Parmeliaceae/chemistry
2.
Pharmacol Res ; 155: 104727, 2020 05.
Article in English | MEDLINE | ID: mdl-32113874

ABSTRACT

Panaxadiol is a triterpenoid sapogenin monomeric compound found in the roots of Panax ginseng and has a variety of biological activities such as neuroprotective and anti-tumour functions. However, the mechanisms how panaxadiol exerts the anticancer effects remain unknown. The current study aimed to investigate the potential activity of panaxadiol on programmed cell death-ligand 1 (PD-L1) expression and tumour proliferation in human colon cancer cells and to identify the underlying mechanism. Results showed that panaxadiol showed little cytotoxicity as assessed by a cytotoxicity assay and significantly inhibited PD-L1 expression at the protein and mRNA level in a dose-dependent manner. Furthermore, panaxadiol supressed the hypoxia-induced synthesis of hypoxia-inducible factor (HIF)-1α via the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways without affecting HIF-1α degradation. Simultaneously, panaxadiol inhibited STAT3 activation through the JAK1, JAK2, and Src pathways. Moreover, pre-treatment with panaxadiol enhanced the activity of cytotoxic T lymphocytes (CTL) and regained their capacity of tumour cell killing in a T cell and tumour cell co-culture system. Immunoprecipitation showed that panaxadiol inhibited PD-L1 expression by blocking the interaction between HIF-1α and STAT3. The inhibitory effect of panaxadiol on tumour proliferation was further demonstrated by colony formation and EdU labelling assays. The anti-proliferative effect of panaxadiol was also proved by a xenograft assay in vivo. Taken together, the current work highlights the anti-tumour effect of panaxadiol, providing insights into development of cancer therapeutic through PD-L1 inhibition.


Subject(s)
Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Ginsenosides/therapeutic use , Animals , Antineoplastic Agents/pharmacology , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line , Cell Proliferation/drug effects , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Ginsenosides/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred BALB C , Mice, Nude , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Angew Chem Int Ed Engl ; 59(9): 3650-3657, 2020 Feb 24.
Article in English | MEDLINE | ID: mdl-31828892

ABSTRACT

The semihydrogenation of alkynes into alkenes rather than alkanes is of great importance in the chemical industry. Unfortunately, state-of-the-art heterogeneous catalysts hardly achieve high turnover frequencies (TOFs) simultaneously with almost full conversion, excellent selectivity, and good stability. Here, we used metal-organic frameworks (MOFs) containing Zr metal nodes ("UiO") with tunable wettability and electron-withdrawing ability as activity accelerators for the semihydrogenation of alkynes catalyzed by sandwiched palladium nanoparticles (Pd NPs). Impressively, the porous hydrophobic UiO support not only leads to an enrichment of phenylacetylene around the Pd NPs but also renders the Pd surfaces more electron-deficient, which leads to a remarkable catalysis performance, including an exceptionally high TOF of 13835 h-1 , 100 % phenylacetylene conversion 93.1 % selectivity towards styrene, and no activity decay after successive catalytic cycles. The strategy of using molecularly tailored supports is universal for boosting the selective semihydrogenation of various terminal and internal alkynes.

4.
Mol Hum Reprod ; 25(9): 538-549, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31553448

ABSTRACT

Baicalin, a traditional Chinese medicinal monomer whose chemical structure is known, can be used to treat female infertility. However, the effect of baicalin on embryonic development is unknown. This study investigated the effects of baicalin on in vitro development of parthenogenetically activated (PA) and in vitro fertilized (IVF) pig embryos and the underlying mechanisms involved. Treatment with 0.1 µg/ml baicalin significantly improved (P < 0.05) the in vitro developmental capacity of PA pig embryos by reducing the reactive oxygen species (ROS) levels and apoptosis and increasing the mitochondrial membrane potential (ΔΨm) and ATP level. mRNA and protein expression of sonic hedgehog (SHH) and GLI1, which are related to the SHH signaling pathway, in PA pig embryos at the 2-cell stage, were significantly higher in the baicalin-treated group than in the control group. To confirm that the SHH signaling pathway is involved in the mechanism by which baicalin improves embryonic development, we treated embryos with baicalin in the absence or presence of cyclopamine (Cy), an inhibitor of this pathway. Cy abolished the effects of baicalin on in vitro embryonic development. In conclusion, baicalin improves the in vitro developmental capacity of PA and IVF pig embryos by inhibiting ROS production and apoptosis, regulating mitochondrial activity and activating SHH signaling.


Subject(s)
Apoptosis/drug effects , Embryonic Development/drug effects , Flavonoids/pharmacology , Mitochondria/drug effects , Oocytes/drug effects , Animals , Cells, Cultured , Embryo Culture Techniques , Embryo, Mammalian , Female , Fertilization in Vitro , Hedgehog Proteins/metabolism , In Vitro Oocyte Maturation Techniques/veterinary , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/physiology , Oocytes/cytology , Oocytes/physiology , Oogenesis/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Swine/embryology
5.
J Nutr Biochem ; 100: 108899, 2022 02.
Article in English | MEDLINE | ID: mdl-34748924

ABSTRACT

A. membranaceus is a traditional Chinese medicine that regulates blood sugar levels, suppresses inflammation, protects the liver, and enhances immunity. In addition, A. membranaceus is also widely used in diet therapy and is a well-known health tonic. Formononetin is a natural product isolated from A. membranaceus that has multiple biological functions, including anti-cancer activity. However, the mechanism by which formononetin inhibits tumor growth is not fully understood. In this present study, we demonstrated that formononetin suppresses PD-L1 protein synthesis via reduction of MYC and STAT3 protein expression. Furthermore, formononetin markedly reduced the expression of MYC protein via the RAS/ERK signaling pathway and inhibited STAT3 activation through JAK1/STAT3 pathway. Co-immunoprecipitation experiments illustrated that formononetin suppresses protein expression of PD-L1 by interfering with the interaction between MYC and STAT3. Meanwhile, formononetin promoted PD-L1 protein degradation via TFEB and TFE3-mediated lysosome biogenesis. T cell killing assay revealed that formononetin could enhance the activity of cytotoxic T lymphocytes (CTLs) and restore ability to kill tumor cells in a co-culture system of T cells and tumor cells. In addition, formononetin inhibited cell proliferation, tube formation, cell migration, and promoted tumor cell apoptosis by suppressing PD-L1. Finally, the inhibitory effect of formononetin on tumor growth was confirmed in a murine xenograft model. The present study revealed the anti-tumor potential of formononetin, and the findings should support further research and development of anti-cancer drugs for cervical cancer.


Subject(s)
B7-H1 Antigen/metabolism , Carcinogenesis/drug effects , Isoflavones/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/metabolism , Uterine Cervical Neoplasms/physiopathology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , Down-Regulation , Female , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Lysosomes/metabolism , Organelle Biogenesis , Proto-Oncogene Proteins c-myc/genetics , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/immunology , Xenograft Model Antitumor Assays
6.
J Ethnopharmacol ; 273: 113989, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-33677006

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Glycyrrhiza glabra L., a traditional medicinal, has a history of thousands of years. It is widely used in clinic and has been listed in Chinese Pharmacopoeia. Licochalcone A is a phenolic chalcone compound and a characteristic chalcone of Glycyrrhiza glabra L. It has many pharmacological activities, such as anti-cancer, anti-inflammatory, anti-viral and anti-angiogenic activities. AIM OF THE STUDY: In this study, we explored the anti-tumor activity and potential mechanism of licochalcone A in vitro and in vivo. MATERIALS AND METHODS: In vitro, the mechanism of licochalcone A at inhibiting PD-L1 expression was investigated by molecular docking, western blotting, RT-PCR, flow cytometry, immunofluorescence and immunoprecipitation assays. The co-culture model of T cells and tumor cells was used to detect the activity of cytotoxic T lymphocytes. Colony formation, EdU labelling and apoptosis assays were used to detect changes in cellular proliferation and apoptosis. In vivo, anti-tumor activity of licochalcone A was assessed in a xenograft model of HCT116 cells. RESULTS: In the present study, we found that licochalcone A suppressed the expression of programmed cell death ligand-1 (PD-L1), which plays a key role in regulating the immune response. In addition, licochalcone A inhibited the expressions of p65 and Ras. Immunoprecipitation experiment showed that licochalcone A suppressed the expression of PD-L1 by blocking the interaction between p65 and Ras. In the co-culture model of T cells and tumor cells, licochalcone A pretreatment enhanced the activity of cytotoxic T lymphocytes and restored the ability to kill tumor cells. In addition, we showed that licochalcone A inhibited cell proliferation and promoted cell apoptosis by targeting PD-L1. In vivo xenograft assay confirmed that licochalcone A inhibited the growth of tumor xenografts. CONCLUSION: In general, these results reveal the previously unknown properties of licochalcone A and provide new insights into the anticancer mechanism of this compound.


Subject(s)
B7-H1 Antigen/metabolism , Cell Proliferation/drug effects , Chalcones/pharmacology , Colonic Neoplasms/drug therapy , NF-kappa B/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , B7-H1 Antigen/genetics , Coculture Techniques , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Nude , NF-kappa B/genetics , Neoplasms, Experimental , T-Lymphocytes/physiology , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/genetics , ras Proteins/metabolism
7.
J Ethnopharmacol ; 273: 113598, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-33220359

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Dendrobium chrysotoxum Lindl is a cultivation of Dendrobium which belongs to the family of Orchidaceae. D. chrysotoxum Lindl is a traditional Chinese medicine with a wide range of clinical applications including tonic, astringent, analgesic and anti-inflammatory properties as early as the 28th century B.C. Erianin is a representative index component for the quality control of the D. chrysotoxum Lindl, which is included in the Pharmacopoeia of the People's Republic of China (2020 version). AIM OF THE STUDY: To clarify the anti-tumour mechanisms of erianin in vitro and in vivo. MATERIALS AND METHODS: We detected the anti-tumour activity of erianin using in vitro HeLa cell models and in vivo cervical cancer xenograft models. We performed MTT, western blot, RT-PCR, homology modeling, flow cytometry, and immunoprecipitation assays to study the proteins, genes, and pathways related to erianin's anti-tumour activity. LysoTracker Red staining was performed to detect lysosome function. Transwell, wound healing, tube formation, colony formation and EdU labelling assays were performed to determine cell proliferation, migration and invasion abilities, respectively. Cytotoxic T lymphocytes ability was confirmed using HeLa/T-cell co-culture model. RESULTS: Experimental data demonstrated that erianin inhibited PD-L1 expression and induced the lysosomal degradation of PD-L1. Erianin suppressed HIF-1α synthesis through mTOR/p70S6K/4EBP1 pathway, and inhibited RAS/Raf/MEK/MAPK-ERK pathway. Immunoprecipitation experiments demonstrated that erianin reduced the interaction between RAS and HIF-1α. Experiments using a co-cultivation system of T cells and HeLa cells confirmed that erianin restored cytotoxic T lymphocytes ability to kill tumour cells. Erianin inhibited PD-L1-mediated angiogenesis, proliferation, invasion and migration. The anti-proliferative effects of erianin were supported using in vivo xenotransplantation experiments. CONCLUSIONS: Collectively, these results revealed previously unknown properties of erianin and provided a new basis for improving the efficacy of immunotherapy against cervical cancer and other malignant tumours through PD-L1.


Subject(s)
B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Bibenzyls/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Phenol/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bibenzyls/therapeutic use , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Lysosomes/metabolism , MAP Kinase Signaling System/drug effects , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Neovascularization, Pathologic/metabolism , Phenol/therapeutic use , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , T-Lymphocytes, Cytotoxic/drug effects , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , raf Kinases/metabolism , ras Proteins/metabolism
8.
Phytomedicine ; 81: 153425, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33310309

ABSTRACT

BACKGROUND: Programmed cell death-ligand 1 (PD-L1) is overexpressed in tumor cells, which causes tumor cells to escape T cell killing, and promotes tumor cell survival, cell proliferation, migration, invasion, and angiogenesis. Britannin is a natural product with anticancer pharmacological effects. PURPOSE: In this work, we studied the anticancer potential of britannin and explored whether britannin mediated its effect by inhibiting the expression of PD-L1 in tumor cells. METHODS: In vitro, the mechanisms underlying the inhibition of PD-L1 expression by britannin were investigated by MTT assay, homology modeling and molecular docking, RT-PCR, western blotting, co-immunoprecipitation, and immunofluorescence. The changes in tumor killing activity, cell proliferation, cell cycle, migration, invasion, and angiogenesis were analyzed by T cell killing assays, EdU labeling, colony formation, flow cytometry, wound healing, matrigel transwell invasion, and tube formation, respectively. In vivo, the antitumor activity of britannin was evaluated in the HCT116 cell xenograft model. RESULTS: Britannin reduced the expression of PD-L1 in tumor cells by inhibiting the synthesis of the PD-L1 protein but did not affect the degradation of the PD-L1 protein. Britannin also inhibited HIF-1α expression through the mTOR/P70S6K/4EBP1 pathway and Myc activation through the Ras/RAF/MEK/ERK pathway. Mechanistically, britannin inhibited the expression of PD-L1 by blocking the interaction between HIF-1α and Myc. In addition, britannin could enhance the activity of cytotoxic T lymphocytes and inhibit tumor cell proliferation and angiogenesis by inhibiting PD-L1. Finally, in vivo observations were confirmed by demonstrating the antitumor activity of britannin in a murine xenograft model. CONCLUSION: Britannin inhibits the expression of PD-L1 by blocking the interaction between HIF-1α and Myc. Moreover, britannin stabilizes T cell activity and inhibits proliferation and angiogenesis by inhibiting PD-L1 in cancer. The current work highlights the anti-tumor effect of britannin, providing insights into the development of cancer therapeutics via PD-L1 inhibition.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactones/pharmacology , Neovascularization, Pathologic/drug therapy , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Sesquiterpenes/pharmacology , T-Lymphocytes/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HCT116 Cells , Humans , Lactones/chemistry , MAP Kinase Signaling System/drug effects , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Neovascularization, Pathologic/metabolism , Programmed Cell Death 1 Ligand 2 Protein/chemistry , Programmed Cell Death 1 Ligand 2 Protein/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Sesquiterpenes/chemistry , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
9.
RSC Adv ; 8(73): 42109-42115, 2018 Dec 12.
Article in English | MEDLINE | ID: mdl-35558793

ABSTRACT

BCN (boron carbon nitride) nanosheets are promising photocatalyst materials for solar fuel production by visible light-driven water splitting and CO2 reduction due to their tunable band gap and unique properties. C-doping into h-BN by thermal annealing makes possible the preparation of BCN nanosheets with photocatalytic activity under visible light irradiation, but it generally requires a very high temperature (>1250 °C) from the thermodynamic viewpoint. Here, we report a new method to prepare BCN nanosheets with visible light-photocatalytic activity at lower annealing temperature (1000 °C) than equilibrium by adding alkaline earth metal compounds. BCN nanosheets formed in borate melt show a clear layered structure, tunable bandgap and photocatalytic activity for water splitting and CO2 reduction under visible light illumination. This provides a direction for doping other elements into h-BN at low annealing temperature by alkaline earth metal borates.

SELECTION OF CITATIONS
SEARCH DETAIL