Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Phytomedicine ; 116: 154877, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37267692

ABSTRACT

BACKGROUND: The flavonoid galangin (3,5,7-trihydroxyflavone) is derived from the root of Alpinia officinarum Hance, an edible and medicinal herb. Galangin has many biological activities, such as anti-inflammatory, anti-microbial, anti-viral, anti-obesogenic, and anti-oxidant effects. However, the anti-tumor mechanism of galangin remains unclear. PURPOSE: To elucidate the anti-tumor mechanisms of galangin in vitro and in vivo. METHODS: MTT, western blotting, immunoprecipitation, RT-PCR, and immunofluorescence assays were used to assess the mechanism of galangin inhibiting PD-L1 expression. The effect of galangin on T cell activity was analyzed in Hep3B/T cell co-cultures. Colony formation, EdU, migration, and invasion assays were performed to explore the effect of galangin on cancer progression and metastasis. Anti-tumor effects of galangin were investigated in a xenograft model. RESULTS: Galangin inhibited PD-L1 expression dose-dependently, which plays a major role in tumor progression. Moreover, galangin blocked STAT3 activation through the JAK1/JAK2/Src signaling pathway and Myc activation through the Ras/RAF/MEK/ERK signaling pathway. Galangin reduced PD-L1 expression by suppressing STAT3 and Myc cooperatively. Galangin increased the killing effect of T cells on tumor cells in Hep3B/T cell co-cultures. Moreover, galangin inhibited tumor cell proliferation, migration, and invasion through PD-L1. In vivo experiments showed that galangin suppressed tumor growth. CONCLUSION: Galangin enhances T-cell activity and inhibits tumor cell proliferation, migration, and invasion through PD-L1. The current study emphasizes the anti-tumor properties of galangin, offering new insights into the development of tumor therapeutics targeting PD-L1.


Subject(s)
B7-H1 Antigen , T-Lymphocytes , Humans , B7-H1 Antigen/metabolism , Ligands , Cell Line, Tumor , T-Lymphocytes/metabolism , Flavonoids/pharmacology , Apoptosis , Cell Proliferation , STAT3 Transcription Factor/metabolism
2.
Phytomedicine ; 116: 154889, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37262999

ABSTRACT

BACKGROUND: Development of clinically effective neuroprotective agents for stroke therapy is still a challenging task. Microglia play a critical role in brain injury and recovery after ischemic stroke. Traditional Chinese herbal medicines (TCHMs) are based on a unique therapeutic principle, have various formulas, and have long been widely used to treat stroke. Therefore, the active compounds in TCHMs and their underlying mechanisms of action are attracting increasing attention in the field of stroke drug development. PURPOSE: To summarize the regulatory mechanisms of TCHM-derived natural compounds on the microglial response in animal models of ischemic stroke. METHODS: We searched studies published until 10 April 2023 in the Web of Science, PubMed, and ScienceDirect using the following keywords: natural compounds, natural products or phytochemicals, traditional Chinese Medicine or Chinese herbal medicine, microglia, and ischemic stroke. This review was prepared according to PRISMA (Preferred Reporting Item for Systematic Reviews and Meta-Analysis) guidelines. RESULTS: Natural compounds derived from TCHMs can attenuate the M1 phenotype of microglia, which is involved in the detrimental inflammatory response, via inhibition of NF-κB, MAPKs, JAK/STAT, Notch, TLR4, P2X7R, CX3CR1, IL-17RA, the NLRP3 inflammasome, and pro-oxidant enzymes. Additionally, the neuroprotective response of microglia with the M2 phenotype can be enhanced by activating Nrf2/HO-1, PI3K/AKT, AMPK, PPARγ, SIRT1, CB2R, TREM2, nAChR, and IL-33/ST2. Several clinical trials showed that TCHM-derived natural compounds that regulate microglial responses have significant and safe therapeutic effects, but further well-designed clinical studies are needed. CONCLUSIONS: Further research regarding the direct targets and potential pleiotropic or synergistic effects of natural compounds would provide a more reasonable approach for regulation of the microglial response with the possibility of successful stroke drug development.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Animals , Brain Ischemia/drug therapy , Ischemic Stroke/drug therapy , Microglia , Phosphatidylinositol 3-Kinases , Plant Extracts/pharmacology , Stroke/drug therapy
3.
Phytother Res ; 37(4): 1293-1308, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36751854

ABSTRACT

Citrus peel has long been used in traditional medicine in Asia to treat common cold, dyspepsia, cough, and phlegm. Narirutin-a flavanone-7-O-glycoside-is the major flavonoid in citrus peel, and has anti-oxidative, anti-allergic, and anti-inflammatory activities. However, the anti-inflammatory mechanism of narirutin has not been fully elucidated. This study is aimed to investigate the effects of narirutin on the Nod-like receptor protein 3 (NLRP3)-mediated inflammatory response in vitro and in vivo, and determine the underlying mechanism. THP-1 differentiated macrophages and bone marrow-derived macrophages (BMDMs) were used for in vitro experiments, while dextran sulfate sodium (DSS)-induced colitis and alum-induced peritonitis mouse models were constructed to test inflammation in vivo. Narirutin suppressed secretion of interleukin (IL)-1ß and pyroptosis in lipopolysaccharide (LPS)/ATP-stimulated macrophages. Narirutin decreased the expression of NLRP3 and IL-1ß in the LPS-priming step through inhibition of NF-κB, MAPK and PI3K /AKT signaling pathways. Narirutin inhibited NLRP3-ASC interaction to suppress NLRP3 inflammasome assembly. Furthermore, oral administration of narirutin (300 mg/kg) alleviated inflammation symptoms in mice with peritonitis and colitis. These results suggest that narirutin exerts its anti-inflammatory activity by suppressing NLRP3 inflammasome activation via inhibition of the NLRP3 inflammasome priming processes and NLRP3-ASC interaction in macrophages.


Subject(s)
Colitis , Flavanones , Peritonitis , Animals , Mice , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophages , Flavanones/pharmacology , Colitis/chemically induced , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology , Peritonitis/metabolism
4.
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
5.
J Ethnopharmacol ; 283: 114715, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34648898

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The use of Panax ginseng C.A.Mey. in traditional Chinese medicine dates back to about 5000 years ago thanks to its several beneficial and healing properties. Panaxadiol is a triterpenoid sapogenin monomer found in the roots of Panax ginseng C.A.Mey. and has been proven to have various bio-activities such as anti-inflammatory, anti-tumour and neuroprotective effects. AIM OF THE STUDY: The present study focuses on investigating the inflammation inhibitory effect and mechanism of panaxadiol by regulating zinc finger protein 91-regulated activation of non-canonical caspase-8 inflammasome and MAPKs in macrophages. MATERIALS AND METHODS: In vitro, the underlying mechanisms by which panaxadiol inhibits ZFP91-regulated IL-1ß expression were investigated using molecular docking, western blotting, RT-PCR, ELISA, immunofluorescence, and immunoprecipitation assays. In vivo, colitis was induced by oral administration of DSS in drinking water, and peritonitis was induced by an intraperitoneal injection of alum. Recombinant adeno-associated virus (AAV serotype 9) vector was used to establish ZFP91 knockdown mouse. RESULTS: We confirmed that panaxadiol inhibited IL-1ß secretion by suppressing ZFP91 in macrophages. Further analysis revealed that panaxadiol inhibited IL-1ß secretion by suppressing ZFP91-regulated activation of non-canonical caspase-8 inflammasome. Meanwhile, panaxadiol inhibited IL-1ß secretion by suppressing ZFP91-regulated activation of MAPKs. In vivo, prominent anti-inflammatory effects of panaxadiol were demonstrated in a DSS induced acute colitis mouse model and in an alum-induced peritonitis model by suppressing ZFP91-regulated secretion of inflammatory mediators, consistent with the results of the AAV-ZFP91 knockdown in mice. CONCLUSIONS: We report for the first time that panaxadiol inhibited IL-1ß secretion by suppressing ZFP91-regulated activation of non-canonical caspase-8 inflammasome and MAPKs, providing evidence for anti-inflammation mechanism of panaxadiol treatment for inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ginsenosides/pharmacology , Neuroprotective Agents/pharmacology , Panax/chemistry , Animals , Anti-Inflammatory Agents/isolation & purification , Caspase 8/metabolism , Colitis/drug therapy , Gene Knockdown Techniques , Ginsenosides/isolation & purification , HEK293 Cells , Humans , Inflammasomes/metabolism , Inflammation/drug therapy , Interleukin-1beta/metabolism , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Neuroprotective Agents/isolation & purification , THP-1 Cells , Ubiquitin-Protein Ligases/genetics
6.
Br J Pharmacol ; 179(9): 1887-1907, 2022 05.
Article in English | MEDLINE | ID: mdl-34825365

ABSTRACT

BACKGROUND AND PURPOSE: ZFP91 positively regulates IL-1ß production in macrophages and may be a potential therapeutic target to treat inflammatory-related diseases. We investigated whether this process is modulated by convallatoxin, which is a cardiac glycoside isolated from the traditional Chinese medicinal plant Adonis amurensis Regel et Radde. EXPERIMENTAL APPROACH: In vitro, the mechanisms by which convallatoxin inhibits ZFP91-regulated IL-1ß expression were investigated using molecular docking, western blotting, RT-PCR, ELISA, immunofluorescence and immunoprecipitation assays.In vivo, mice liver injury was induced by an intraperitoneal injection of D-GalN and LPS, colitis was induced by oral administration of dextran sulfate sodium (DSS) in drinking water and peritonitis was induced by an intraperitoneal injection of alum. KEY RESULTS: We confirmed that convallatoxin inhibited the release of IL-1ß by down-regulating ZFP91. Importantly, we found that convallatoxin significantly reduced K63-linked polyubiquitination of pro-IL-1ß regulated by ZFP91 and decreased the efficacy of pro-IL-1ß cleavage. Moreover, convallatoxin suppressed ZFP91-mediated activation of the non-canonical cysteine-requiring aspartate protease-8 (caspase-8) inflammasome and MAPK signalling pathways in macrophages. Furthermore, we showed that ZFP91 promoted the assembly of the caspase-8 inflammasome complex, whereas convallatoxin treatment reversed this result. Mice in vivo studies further demonstrated that convallatoxin ameliorated D-GalN/LPS-induced liver injury, DSS-induced colitis and alum-induced peritonitis by down-regulating ZFP91. CONCLUSION AND IMPLICATIONS: We show for the first time that convallatoxin-mediated inhibition of ZFP91 is an important regulatory event that prevents inappropriate inflammatory responses to maintain immune homeostasis. This mechanism provides new insight for the development of convallatoxin as a novel anti-inflammatory drug targeting ZFP91. LINKED ARTICLES: This article is part of a themed issue on Inflammation, Repair and Ageing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.9/issuetoc.


Subject(s)
Caspase 8 , Inflammasomes , Interleukin-1beta , NLR Family, Pyrin Domain-Containing 3 Protein , Strophanthins , Animals , Caspase 1/metabolism , Caspase 8/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Strophanthins/pharmacology , Transcription Factors/antagonists & inhibitors , Ubiquitination , Zinc Fingers
7.
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
8.
Artif Cells Nanomed Biotechnol ; 49(1): 219-229, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33666536

ABSTRACT

Diabetes associated injury healing and other tissue irregularities are viewed as a significant concern. The purpose of the study is to design the wound regeneration activity of Ficus carica extract (FFE) loaded amphiphilic polymeric scaffold of poly(xylitol-g-adipate-co-glutamide) (PXAG)-polyhydroxybutyrate (PHB) for potential diabetic affected wound regeneration. The PXAG copolymer was prepared by the condensation method, and the polymeric scaffolds of PXAG-PHB, PXAG-PHB/FFE were developed through the ultra-sonication process and magnetic stirrer processes. The chemical structure, crystalline nature, thermal stability, size, surface charge and surface morphology of PXAG-PHB and PXAG-PHB/FFE polymeric scaffolds were investigated. The PXAG-PHB/FFE exhibits 99.0% free radical scavenging activity which was determined by the DPPH method. The inhibition zones by the PXAG-PHB/FFE indicate it had a higher antibacterial activity with the Escherichia coli (gram-negative) and Staphylococcus aureus (gram-positive) pathogens. The PXAG, PXAG-PHB and PXAG-PHB/FFE polymeric scaffolds exhibited good viability against diabetic induced wound cells (WS1) in 100 µg/mL concentrations up to 72 h incubation. Since the synthesized PXAG-PHB/FFE polymeric scaffolds possess excellent thermal stability, bioactivity, biocompatibility and antioxidant activity along with potent antimicrobial activity, they play a potential role in diabetic wound tissue regenerations.


Subject(s)
Diabetes Mellitus/physiopathology , Ficus/chemistry , Hydrophobic and Hydrophilic Interactions , Plant Extracts/chemistry , Polymers/chemistry , Polymers/pharmacology , Tissue Engineering/methods , Wound Healing/drug effects
9.
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
10.
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
11.
J Ethnopharmacol ; 257: 112835, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32278762

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Curcuma wenyujin is a Chinese traditional herbal medicine that is commonly used as an anti-oxidant, anti-proliferative, and anti-tumorigenic agent. Curcumol is a representative index component for the quality control of the essential oil of Curcuma wenyujin, which is currently used as an anti-cancer drug, and is included in the State Pharmacopoeia Commission of the People's Republic of China (2005). However, the mechanisms of action and molecular functions of curcumol are not yet fully elucidated. AIM OF THE STUDY: This study aimed to identify new effects of curcumol from the perspective of cancer immunotherapy. MATERIALS AND METHODS: The underlying mechanism of the inhibition of programmed cell death-ligand 1 (PD-L1) activation by curcumol was investigated in vitro via homology modeling, molecular docking experiments, luciferase reporter assays, MTT assays, RT-PCR, western blotting, and immunofluorescence assays. Changes in cellular proliferation, angiogenesis, and the tumor-killing activity of T-cells were analyzed via EdU labeling, colony formation, flow cytometry, wound-healing, Matrigel Transwell invasion, tube formation, and T-cell killing. The anti-tumor activity of curcumol was assessed in vivo in a murine xenograft model using Hep3B cells. RESULTS: Curcumol reduced the expression of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) via JAK1, JAK2, and Src pathways and inhibited hypoxia-inducible factor-1α (HIF-1α) protein synthesis via mTOR/p70S6K/eIF4E and MAPK pathways. Furthermore, we revealed crosstalk between STAT3 and HIF-1α pathways, which collaboratively regulated PD-L1 activation, and that curcumol played a role in this regulation. Curcumol inhibited cell proliferation, S-phase progression, tube formation, invasion, and metastasis by inhibiting PD-L1. In addition, curcumol restored the activity of cytotoxic T-cells and their capacity for tumor cell killing by inhibiting PD-L1. In vivo experiments confirmed that curcumol inhibited tumor growth in a xenograft model. CONCLUSIONS: These results illustrated that curcumol inhibits the expression of PD-L1 through crosstalk between HIF-1α and p-STAT3 (T705) signaling pathways in hepatic cancer. Thus, curcumol might represent a promising lead compound for the development of new targeted anti-cancer therapeutics.


Subject(s)
B7-H1 Antigen/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Sesquiterpenes/pharmacology , A549 Cells , Animals , Cell Line, Tumor , HeLa Cells , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Janus Kinase 2 , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Neovascularization, Pathologic/drug therapy , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , T-Lymphocytes/drug effects , TOR Serine-Threonine Kinases/metabolism
12.
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
13.
Phytomedicine ; 68: 153172, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32004989

ABSTRACT

BACKGROUND: Aberrant activation of STAT3 is frequently encountered and promotes survival, cellular proliferation, migration, invasion and angiogenesis in tumor cell. Convallatoxin, triterpenoid ingredient, exhibits anticancer pharmacological properties. PURPOSE: In this work, we investigated the anticancer potential of convallatoxin and explored whether convallatoxin mediates its effect through interference with the STAT3 activation in colorectal cancer cells. METHODS: In vitro, the underlying mechanisms of convallatoxin at inhibiting STAT3 activation were investigated by homology modeling and molecular docking, luciferase reporter assay, MTT assay, RT-PCR, Western blotting and immunofluorescence assays. Changes in cellular proliferation, apoptosis, migration, invasion and angiogenesis were analyzed by EdU labeling assay, colony formation assay, flow cytometry assay, wound-healing assay, matrigel transwell invasion assay and tube formation assays. And in vivo, antitumor activity of convallatoxin was assessed in a murine xenograft model of HCT116 cells. RESULTS: Convallatoxin decreased the viability of colorectal cancer lines. Moreover, convallatoxin reduced the P-STAT3 (T705) via the JAK1, JAK2, and Src pathways and inhibited serine-727 phosphorylation of STAT3 via the PI3K-AKT-mTOR-STAT3 pathways in colorectal cancer cells. Interestingly, we discovered the crosstalk between mTOR and JAK2 in mTOR/STAT3 and JAK/STAT3 pathways, which collaboratively regulated STAT3 activation and convallatoxin play a role in it. Convallatoxin also downregulated the expression of target genes involved cell survival (e.g., Survivin, Bcl-xl, Bcl-2), proliferation (e.g., Cyclin D1), metastasis (e.g., MMP-9), and angiogenesis (e.g., VEGF). Indeed, we found that convallatoxin inhibited tube formation, migration, and invasion of endothelial cells, and inhibited the proliferation. Finally, in vivo observations were confirmed by showing antitumor activity of convallatoxin in a murine xenograft model. CONCLUSION: The result of the current study show that convallatoxin promotes apoptosis and inhibits proliferation and angiogenesis through crosstalk between JAK2/STAT3 (T705) and mTOR/STAT3 (S727) signaling pathways in colorectal cancer cells and indicate that convallatoxin could be a valuable candidate for the development of colorectal cancer therapeutic.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , Strophanthins/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Janus Kinase 2/metabolism , Male , Mice, Nude , Molecular Docking Simulation , Neovascularization, Pathologic/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Strophanthins/chemistry , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
14.
J Transl Med ; 17(1): 410, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31805963

ABSTRACT

BACKGROUND: Currently, many clinical trials have shown that inulin-type fructans (ITF) supplementation is associated with glycemic control; nevertheless, the results are inconclusive. The aim of this meta-analysis of randomized controlled trials was to assess the effects of ITF supplementation on glycemic control. METHODS: PubMed, EMBASE and the Cochrane Library were searched for eligible articles up to March 6, 2019. A random-effects model was used to analyze the pooled results, and the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) system was applied to assess the quality of evidence. The dose-response model was used to recommend the daily dose and duration for ITF supplementation. RESULTS: Thirty-three trials involving 1346 participants were included. Overall, ITF supplementation could significantly reduce concentrations of fasting blood glucose (FBG), glycosylated hemoglobin (HbA1c), fasting insulin (FINS) and homeostasis model assessment-insulin resistance (HOMA-IR). In the prediabetes and type 2 diabetes (T2DM) population, a more significant reduction in FBG [weighted mean difference (WMD): - 0.60 mmol/l; 95% CI - 0.71, - 0.48 mmol/l; high rate], HbA1c (WMD: - 0.58%; 95% CI - 0.83, - 0.32%; high rate), FINS (WMD: - 1.75 µU/ml; 95% CI - 2.87, - 0.63 µU/ml; low rate), and HOMA-IR (WMD: - 0.69; 95% CI - 1.10, - 0.28; low rate) were observed, and ITF supplementation with a daily dose of 10 g for a duration of 6 weeks and longer was recommended. Moreover, subgroup analyses suggested that the effects of glycemic control were significantly influenced by the sex of the subjects and the type and the method of intake of ITF. CONCLUSIONS: Our analyses confirmed that these four main glycemic indicators were significantly reduced by ITF supplementation, particularly in the prediabetes and T2DM population. Evidence supports that reasonable administration of ITF supplementation may have potential clinical value as an adjuvant therapy for prediabetes and T2DM management. Trial registration The trial was registered at PROSPERO as CRD42018115875 on November 23, 2018.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Dietary Supplements , Fructans/therapeutic use , Inulin/therapeutic use , Prediabetic State/drug therapy , Randomized Controlled Trials as Topic , Adult , Aged , Diabetes Mellitus, Type 2/blood , Dose-Response Relationship, Drug , Fasting/blood , Female , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Nonlinear Dynamics , Prediabetic State/blood , Publication Bias , Treatment Outcome , Young Adult
15.
Chin J Integr Med ; 25(4): 246-251, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31236888

ABSTRACT

OBJECTIVE: To assess the efficacy and safety of the Chinese medicine Dingkun Pill (, DKP) on insulin resistance in women with polycystic ovary syndrome (PCOS). METHODS: A total of 117 women with PCOS were randomly assigned to Group A (38 women), Group B (40 women), or Group C (39 women) in a randomization sequence with SAS software and a 1:1:1 allocation ratio using random block sizes of 6, and were given 7 g of oral DKP daily (Group A), 1 tablet of Diane-35 orally daily (Group B), or 7 g of oral DKP daily plus 1 tablet of Diane-35 orally daily (Group C). Patients took all drugs cyclically for 21 consecutive days, followed by 7 drug-free days. The treatment course for the 3 groups was continued for 3 consecutive months. Oral glucose tolerance tests (OGTT) were performed before treatment and again after 2 and 3 months of therapy, respectively, and homeostasis model assessment for insulin resistance (HOMA-IR) and quantitative insulin sensitivity check index (QUICKI) were calculated. RESULTS: Of 117 women with PCOS, 110 completed the entire course of therapy: 35 in Group A, 36 in Group B, and 39 in Group C. After treatment, all three groups showed significant decreases in fasting glucose: at 1 h glucose decreased significantly in Group A (by 0.5 ± 1.4 mmol/L, P=0.028) and Group C (by 0.5 ± 1.2 mmol/L, P=0.045); while showing a tendency to increase in Group B (by 0.4 ± 1.9 mmol/L, P=0.238). HOMA-IR decreased significantly in Group C [by 0.5 (-2.2 to 0.5) mIU mmol/L2, P=0.034]. QUICKI was significantly increased in Groups A and C (by 0.009 ± 0.02, P=0.033 and by 0.009 ± 0.027, P=0.049, respectively), while no change was observed in Group B. Repeated-measure ANOVA showed that the absolute changes in all parameters (except for glucose at 1 h), including glucose and insulin levels at all time-points during OGTT and in HbA1c, HOMA-IR, and QUICKI, were not significantly different among the 3 groups after treatment (P>0.05). CONCLUSION: DKP or DKP combined with Diane-35 produce a slight improvement in insulin sensitivity compared with Diane-35 alone in PCOS patients (Trial Registration: ClinicalTrials.gov, NCT03264638).


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Insulin Resistance , Polycystic Ovary Syndrome/drug therapy , Adult , Drugs, Chinese Herbal/adverse effects , Female , Glucose Tolerance Test , Glycated Hemoglobin/metabolism , Humans
16.
Pharmacol Res ; 135: 166-180, 2018 09.
Article in English | MEDLINE | ID: mdl-30103001

ABSTRACT

Dictamnus dasycarpus is a traditional Chinese medicine thathas been commonly used in the treatment of cancer. Fraxinellone is a natural product isolated from the D. dasycarpus plant, which has been shown to exhibit neuroprotective and anti-inflammatory activities. However, whether fraxinellone exerts anticancer effects and the mechanisms by which it may inhibit tumor growth remain unknown. Here, we found that fraxinellone, in a dose-dependented manner, inhibited the expression of programmed cell death ligand-1 (PD-L1), which plays a pivotal role in tumorigenesis. It was subsequently shown that fraxinellone reduced HIF-1α protein synthesis via the mTOR/p70S6K/eIF4E and MAPK pathways. It also inhibited activation of STAT3 via the JAK1, JAK2, and Src pathways. Immunoprecipitation and western blotting assays showed that fraxinellone inhibited PD-L1 expression by reducing STAT3 and HIF-1α cooperatively. Flow cytometry, colony formation, and EdU incorporation assays demonstrated that fraxinellone inhibited cell proliferation through suppression of PD-L1. Tube formation, migration, and invasion assays showed that fraxinellone inhibits angiogenesis by suppressing PD-L1. In vivo studies further supported anticancer role for fraxinellone, demonstrating that fraxinellone treatment inhibited the growth of tumor xenografts. We concluded that fraxinellone inhibits PD-L1 expression by downregulating the STAT3 and HIF-1α signaling pathways, subsequently inhibiting proliferation and angiogenesis in cancer cells. These studies reveal previously unknown characteristics of fraxinellone and provide new perspectives into the mechanism of cancer inhibition of the compound.


Subject(s)
Antineoplastic Agents/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Benzofurans/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Benzofurans/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/metabolism , STAT3 Transcription Factor/metabolism
17.
Pathol Res Pract ; 214(9): 1324-1329, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30031586

ABSTRACT

Schisandrin, derived from the Chinese medicinal herb Schisandra chinensis, has been found to confer protective effects on circulation systems. But the underlying molecular mechanisms remain unclear. The aim of this study was to investigate the effects of a high level of glucose on RhoA and eNOS activity in human umbilical vein endothelial cells(HUVECs) and how Schisandrin plays a role in mediating these effects. To find the optimal treatment time, HUVECs were cultured at a high glucose concentration (30 mM) for different lengths of time (0, 12, 24, and 48 h). Subsequently, the cells were randomized into five groups: a normal group, a high glucose group, and three high glucose groups that were given different doses (5, 10, and 20 µM) of Schisandrin. The cells were pretreated with Schisandrin for 24 h before stimulation with high glucose. The morphology of HUVECs in the various groups was assessed under a light microscope. Immunocytochemical staining was used to detect the level of p-MYPT1 expression. The levels of RhoA activity were determined using the RhoA Activation Assay Biochem Kit. The levels of eNOS activity were examined using a nitrate reduction test. The results showed that in the high glucose group, the activity of RhoA was increased and the activity of eNOS was reduced, thus decreasing the secretion of NO. However, after pretreatment with Schisandrin (10, 20 µM), the activity of RhoA was inhibited and the activity of eNOS increased, which led to an increase in NO production compared with the high glucose group. There was no evident difference between the 5 µM Schisandrin group and the high glucose group. Taken together, these findings indicate that Schisandrin can improve the function of endothelial cells by lowering the activity of RhoA/Rho kinase and raising both the activity of eNOS and the production of NO.


Subject(s)
Cyclooctanes/pharmacology , Glucose/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Lignans/pharmacology , Nitric Oxide Synthase Type III/metabolism , Polycyclic Compounds/pharmacology , rhoA GTP-Binding Protein/metabolism , Humans
18.
Clin Rehabil ; 32(4): 429-439, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28901172

ABSTRACT

OBJECTIVE: To investigate the effectiveness of traditional manual acupuncture combined with rehabilitation therapy versus rehabilitation therapy alone for shoulder hand syndrome after stroke. DATA SOURCES: PubMed, EMBASE, the Cochrane Library, Chinese Biomedicine Database, China National Knowledge Infrastructure, VIP Information Database, Wan Fang Database and reference lists of the eligible studies were searched up to July 2017 for relevant studies. METHODS: Randomized controlled trials that compared the combined effects of traditional manual acupuncture and rehabilitation therapy to rehabilitation therapy alone for shoulder hand syndrome after stroke were included. Two reviewers independently screened the searched records, extracted the data and assessed risk of bias of the included studies. The treatment effect sizes were pooled in a meta-analysis using RevMan 5.3 software. RESULTS: A total of 20 studies involving 1918 participants were included in this study. Compared to rehabilitation therapy alone, the combined therapy significantly reduced pain on the visual analogue scale and improved limb movement on the Fugl-Meyer Assessment scale and the performance of activities of daily living (ADL) on the Barthel Index scale or Modified Barthel Index scale. Of these, the visual analogue scale score changes were significantly higher (mean difference = 1.49, 95% confidence interval = 1.15-1.82, P < 0.00001) favoring the combined therapy after treatment, with severe heterogeneity ( I2 = 71%, P = 0.0005). CONCLUSION: Current evidence suggests that traditional manual acupuncture integrated with rehabilitation therapy is more effective in alleviating pain, improving limb movement and ADL. However, considering the relatively low quality of available evidence, further rigorously designed and large-scale randomized controlled trials are needed to confirm the results.


Subject(s)
Acupuncture Therapy/methods , Exercise Therapy/methods , National Health Programs , Reflex Sympathetic Dystrophy/rehabilitation , Stroke/complications , Activities of Daily Living , Aged , China , Combined Modality Therapy , Female , Humans , Male , Medicine, Traditional , Middle Aged , Randomized Controlled Trials as Topic , Recovery of Function , Reflex Sympathetic Dystrophy/etiology , Severity of Illness Index , Stroke Rehabilitation/methods
19.
Phytother Res ; 32(1): 65-75, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29044876

ABSTRACT

Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is a complex that regulates several hundreds of genes, including those involved in immunity and inflammation, survival, proliferation, and the negative feedback of NF-κB signaling. Chelidonine, a major bioactive, isoquinoline alkaloid ingredient in Chelidonium majus, exhibits antiinflammatory pharmacological properties. However, its antiinflammatory molecular mechanisms remain unclear. In this work, we explored the effect of chelidonine on TNF-induced NF-κB activation in HCT116 cells. We found chelidonine inhibited the phosphorylation and degradation of the inhibitor of NF-κB alpha and nuclear translocation of RELA. Furthermore, by inhibiting the activation of NF-κB, chelidonine downregulated target genes involved in inflammation, proliferation, and apoptosis. Chelidonine also inhibited mitogen-activated protein kinase pathway activation by blocking c-Jun N-terminal kinase and p38 phosphorylation. These results suggest that chelidonine may be a potential therapeutic agent against inflammatory diseases in which inhibition of NF-κB activity plays an important role.


Subject(s)
Benzophenanthridines/therapeutic use , Berberine Alkaloids/therapeutic use , HCT116 Cells/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis , Benzophenanthridines/administration & dosage , Benzophenanthridines/pharmacology , Berberine Alkaloids/administration & dosage , Berberine Alkaloids/pharmacology , Humans , Signal Transduction , Transfection
20.
Molecules ; 22(9)2017 Sep 11.
Article in English | MEDLINE | ID: mdl-28891979

ABSTRACT

Ophiocordyceps xuefengensis, a recently described species of Ophiocordycepsthat is associated with the larvae of Phassusnodus (Hepialidae) in the living root or trunk of the medicinal plant Clerodendrumcyrtophyllum, isthe largest known Cordycepsspecies and is recognized as a desirable alternative for natural Ophiocordycepssinensis. This study investigated the main nucleosides and nucleobases in natural and cultured Ophiocordycepsxuefengensis. The contents of the nucleosides and nucleobases in the natural and cultured samples were determined by reverse phase HPLC. The highest concentration of adenosine was found in the natural fruit body and the cultured stroma, with almost no adenosine in the cadaver of Phassusnodus. The contents of adenine, guanosine, uridine and uracil in the cultured mycelium were significantly higher than those in the natural sample. Inosine was only detected in the natural samples. Thymidine and 2-deoxyadenosine were only found in the cadaver of Phassusnodus. Cordycepin was not detected in the five samples examined. These results suggested that the cultured mycelium and cultured stroma of Ophiocordycepsxuefengensis might be a promising substitute for natural O. xuefengensis.


Subject(s)
Clerodendrum/microbiology , Cordyceps/chemistry , Fruiting Bodies, Fungal/chemistry , Moths/microbiology , Nucleosides/isolation & purification , Adenine/isolation & purification , Adenine/metabolism , Adenosine/isolation & purification , Adenosine/metabolism , Animals , Chromatography, High Pressure Liquid/methods , Clerodendrum/parasitology , Cordyceps/metabolism , Fruiting Bodies, Fungal/metabolism , Guanosine/isolation & purification , Guanosine/metabolism , Inosine/isolation & purification , Inosine/metabolism , Larva/microbiology , Nucleosides/metabolism , Uracil/isolation & purification , Uracil/metabolism , Uridine/isolation & purification , Uridine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL