Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 276: 116729, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39088998

ABSTRACT

Soluble transforming growth factor beta receptor 3 (sTGFBR3) antagonist is a new focus in the research and development of Alzheimer's disease (AD) drugs. Our previous studies have identified sTGFBR3 as a promising new target for AD, with few targeted antagonists identified. In this study, we performed structural modeling of sTGFBR3 using AlphaFold2, followed by high-throughput virtual screening and surface plasmon resonance assays. which collectively identified Xanthone as potential compounds for targeting sTGFBR3. After optimizing the sTGFBR3-Xanthone complex using molecular dynamics (MD) simulations, we prepared a series of novel Xanthone derivatives and evaluated their anti-inflammatory activity, toxicity, and structure-activity relationship in BV2 cell model induced by lipopolysaccharides (LPS) or APP/PS1/tau mouse brain extract (BE). Several derivatives with the most potent anti-inflammatory activity were tested for blood-brain barrier permeability and sTGFBR3 affinity. Derivative P24, selected for its superior properties, was further evaluated in vitro. The results indicated that P24 increased the activation of TGF-ß signaling and decreased the activation of IκBα/NF-κB signaling by targeting sTGFBR3, thereby regulating the inflammation-phagocytosis balance in microglia. Moreover, the low acute toxicity, long half-life, and low plasma clearance of P24 suggest that it can be sustained in vivo. This property may render P24 a more effective treatment modality for chronic diseases, particularly AD. The study demonstrates P24 serve as potential novel candidates for the treatment of AD via antagonizing sTGFBR3.


Subject(s)
Alzheimer Disease , Xanthones , Xanthones/chemistry , Xanthones/pharmacology , Xanthones/chemical synthesis , Animals , Humans , Mice , Structure-Activity Relationship , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Molecular Structure , Drug Discovery , Dose-Response Relationship, Drug , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis , Mice, Inbred C57BL , Male
2.
Molecules ; 29(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39125108

ABSTRACT

Hypericum beanii N. Robson, a perennial upright herb, predominantly inhabits temperate regions. This species has been utilized for the treatment of various inflammation-related diseases. One new xanthone 3,7-dihydroxy-1,6-dimethoxyxanthone (1) and twenty-three known xanthones (2-24) were isolated from the aerial parts of H. beanii. The structure of the new compound was determined based on high-resolution electrospray ionization mass spectroscopy (HR-ESIMS), nuclear magnetic resonance (NMR), Infrared Spectroscopy (IR), ultraviolet spectrophotometry (UV) spectroscopic data. The anti-inflammatory effects of all the isolates were assessed by measuring the inhibitory effect on nitric oxide (NO) production in LPS-stimulated RAW 264.7 macrophages. Compounds 3,4-dihydroxy-2-methoxyxanthone (15), 1,3,5,6-tetrahydroxyxanthone (19), and 1,3,6,7-tetrahydroxyxanthone (22) exhibited significant anti-inflammatory effects at a concentration of 10 µM with higher potency compared to the positive control quercetin. Furthermore, compounds 15, 19, and 22 reduced inducible NO synthase (iNOS), tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6, and cyclooxygenase 2 (COX-2) mRNA expression in the LPS-stimulated RAW 264.7 macrophages, suggesting that these compounds may mitigate the synthesis of the aforementioned molecules at the transcriptional level, provisionally confirming their anti-inflammatory efficacy.


Subject(s)
Anti-Inflammatory Agents , Cyclooxygenase 2 , Hypericum , Interleukin-1beta , Interleukin-6 , Macrophages , Nitric Oxide , Tumor Necrosis Factor-alpha , Xanthones , Mice , Xanthones/pharmacology , Xanthones/chemistry , Xanthones/isolation & purification , Animals , RAW 264.7 Cells , Nitric Oxide/metabolism , Nitric Oxide/biosynthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Macrophages/drug effects , Macrophages/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics , Interleukin-6/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Hypericum/chemistry , Lipopolysaccharides/pharmacology , Plant Extracts/pharmacology , Plant Extracts/chemistry
3.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000485

ABSTRACT

Cytotoxic activity has been reported for the xanthone α-mangostin (AMN) against Glioblastoma multiforme (GBM), an aggressive malignant brain cancer with a poor prognosis. Recognizing that AMN's high degree of hydrophobicity is likely to limit its systemic administration, we formulated AMN using reconstituted high-density lipoprotein (rHDL) nanoparticles. The photophysical characteristics of the formulation, including fluorescence lifetime and steady-state anisotropy, indicated that AMN was successfully incorporated into the rHDL nanoparticles. To our knowledge, this is the first report on the fluorescent characteristics of AMN with an HDL-based drug carrier. Cytotoxicity studies in a 2D culture and 3D spheroid model of LN-229 GBM cells and normal human astrocytes showed an enhanced therapeutic index with the rHDL-AMN formulation compared to the unincorporated AMN and Temozolomide, a standard GBM chemotherapy agent. Furthermore, treatment with the rHDL-AMN facilitated a dose-dependent upregulation of autophagy and reactive oxygen species generation to a greater extent in LN-229 cells compared to astrocytes, indicating the reduced off-target toxicity of this novel formulation. These studies indicate the potential therapeutic benefits to GBM patients via selective targeting using the rHDL-AMN formulation.


Subject(s)
Glioblastoma , Lipoproteins, HDL , Nanoparticles , Spheroids, Cellular , Xanthones , Humans , Xanthones/chemistry , Xanthones/pharmacology , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Cell Line, Tumor , Nanoparticles/chemistry , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Spheroids, Cellular/drug effects , Drug Carriers/chemistry , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Astrocytes/metabolism , Astrocytes/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Autophagy/drug effects
4.
Future Microbiol ; 19(13): 1157-1170, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39012219

ABSTRACT

Aim: To evaluate the antifungal activity of mangiferin against Candida spp. resistant to fluconazole.Materials & methods: The antifungal activity of mangiferin was assessed using broth microdilution and its interaction with azoles and amphotericin B was evaluated by checkerboard. The activity of mangiferin against Candida spp. biofilms was assessed using the MTT colorimetric assay and its possible mechanism of action was evaluated using flow cytometry.Results: Mangiferin showed activity against Candida albicans, Candida tropicalis and Candida parapsilosis resistant to fluconazole and showed synergism with azoles and amphotericin B. Mangiferin increased the activity of antifungals against Candida biofilms and caused depolarization of the mitochondrial membrane and externalization of phosphatidylserine, suggesting apoptosis.Conclusion: mangiferin combined with antifungals has potential against Candida spp.


Candida is a type of fungus that can make people ill. Over time, many species of Candida have found ways to resist the drugs used to kill them. It is important to find new drugs. We decided to see if a substance called mangiferin works against Candida. We found that mangiferin works against Candida and may help other drugs to work better. We still need to do more studies to find out whether mangiferin can help prevent diseases caused by Candida in the future.


Subject(s)
Amphotericin B , Antifungal Agents , Biofilms , Candida , Drug Resistance, Fungal , Drug Synergism , Fluconazole , Microbial Sensitivity Tests , Xanthones , Antifungal Agents/pharmacology , Xanthones/pharmacology , Fluconazole/pharmacology , Biofilms/drug effects , Drug Resistance, Fungal/drug effects , Amphotericin B/pharmacology , Candida/drug effects , Humans , Apoptosis/drug effects , Candida albicans/drug effects , Azoles/pharmacology
5.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(4): 444-451, 2024 Aug 01.
Article in English, Chinese | MEDLINE | ID: mdl-39049631

ABSTRACT

OBJECTIVES: This study aims to investigate the primary target and potential mechanism of mangiferin (MF) in treating oral submucous fibrosis (OSF) through Gene Expression Omnibus (GEO) database chip mining, network pharmacology, and molecular docking techniques. METHODS: Potential therapeutic targets for OSF were identified using GEO chip data. The potential targets of MF were predicted, and disease-related targets for OSF were collected from databases. A Venn diagram was created using the EVenn platform to identify overlapping targets. The protein-protein interaction (PPI) network was constructed using the STRING database. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed using the DAVID platform. Cytoscape 3.10.1 software was used to visualize a drug-target-pathway-disease network, while AutoDocktools 1.5.6 software was employed for molecular docking analysis. RESULTS: A total of 356 potential targets for MF and 360 disease-related targets for OSF were obtained from multiple databases. The top 15 key target proteins in the PPI network were selected as significant candidates. GO function and KEGG pathway enrichment analyses revealed that MF treatment primarily involved advanced glycation end products-receptor (AGE-RAGE), epidermal growth factor receptor (EGFR), and other signaling pathways associated with OSF pathogenesis. Molecular docking analysis demonstrated that MF exhibited a strong binding activity toward AKT serine kinase 1 (AKT1), tumor necrosis factor (TNF), and other core targets. CONCLUSIONS: These findings suggest that MF may exert its therapeutic effects on OSF through a multitarget approach involving various signaling pathways.


Subject(s)
Molecular Docking Simulation , Network Pharmacology , Oral Submucous Fibrosis , Protein Interaction Maps , Xanthones , Xanthones/therapeutic use , Xanthones/pharmacology , Oral Submucous Fibrosis/drug therapy , Oral Submucous Fibrosis/metabolism , Humans , Gene Ontology , Data Mining , ErbB Receptors/metabolism , Software , Signal Transduction
6.
Ann Clin Lab Sci ; 54(3): 354-362, 2024 May.
Article in English | MEDLINE | ID: mdl-39048172

ABSTRACT

OBJECTIVE: Bladder cancer (BC) is primarily treated with cisplatin-based chemotherapy, but the development of cisplatin resistance often leads to BC recurrence. This study is focused on assessing the potential of gambogic acid (GA) in mitigating BC cells' cisplatin resistance, along with an analysis of the underlying mechanism involved. METHODS: Cisplatin was administered to human bladder transitional cell carcinoma cells (T24) at various concentration gradients to induce cisplatin-resistant (T24-DDP) cells. Several experimental groups were set: T24 group, T24-DDP group, T24-DDP+DDP group, T24-DDP+GA group, T24-DDP+DDP+GA group, T24-DDP+DDP+GA+miR-NC group, and T24-DDP+DDP+GA+miR-205-5p inhibitor group. The cell counting kit-8 (CCK-8) assay, Transwell migration assay, and scratch assay were respectively carried out for assessment of cell proliferation, invasion, and migration. Western blot analysis was conducted for detection of the protein expression of E-cadherin, ZEB1, Vimentin, N-cadherin, LRP, MRP, and P-gp in the cells, while the relative expression level of miR-205-5p was determined by qRT-PCR. RESULTS: In comparison with the T24-DDP group, cells in the T24-DDP+GA group showed enhanced sensitivity to cisplatin. Furthermore, as indicated by CCK-8 assay, GA improved T24-DDP cells' sensitivity to cisplatin, potentiated the effects of cisplatin, and exerted an inhibitory effect on the invasion, proliferation, as well as migration of T24-DDP cells. Through Western blot analysis, GA was revealed to significantly inhibit the expression of N-cadherin, E-cadherin, and Vimentin, as well as that of cisplatin-resistant proteins MRP, P-gp, and LRP in BC cells. In addition, shown by further experiments, GA promoted miR-205-5p expression and simultaneously inhibited ZEB1 expression within the cells. CONCLUSION: GA alleviates BC cells' cisplatin resistance through the epithelial-mesenchymal transition pathway mediated by the miR-205-5p/ZEB1 axis.


Subject(s)
Cell Proliferation , Cisplatin , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , MicroRNAs , Urinary Bladder Neoplasms , Xanthones , Zinc Finger E-box-Binding Homeobox 1 , Humans , Cisplatin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Xanthones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Cell Movement/drug effects , Antineoplastic Agents/pharmacology
7.
BMC Complement Med Ther ; 24(1): 282, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39054443

ABSTRACT

BACKGROUND: Malaria is a major global health concern, particularly in tropical and subtropical countries. With growing resistance to first-line treatment with artemisinin, there is an urgent need to discover novel antimalarial drugs. Mesua ferrea Linn., a plant used in traditional medicine for various purposes, has previously been investigated by our research group for its cytotoxic properties. The objective of this study was to explore the compounds isolated from M. ferrea with regards to their potential antiplasmodial activity, their interaction with Plasmodium falciparum lactate dehydrogenase (PfLDH), a crucial enzyme for parasite survival, and their pharmacokinetic and toxicity profiles. METHODS: The isolated compounds were assessed for in vitro antiplasmodial activity against a multidrug-resistant strain of P. falciparum K1 using a parasite lactate dehydrogenase (pLDH) assay. In vitro cytotoxicity against Vero cells was determined using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. The interactions between the isolated compounds and the target enzyme PfLDH were investigated using molecular docking. Additionally, pharmacokinetic and toxicity properties were estimated using online web tools SwissADME and ProTox-II, respectively. RESULTS: Among the seven compounds isolated from M. ferrea roots, rheediachromenoxanthone (5), which belongs to the pyranoxanthone class, demonstrated good in vitro antiplasmodial activity, with the IC50 being 19.93 µM. Additionally, there was no toxicity towards Vero cells (CC50 = 112.34 µM) and a selectivity index (SI) of 5.64. Molecular docking analysis revealed that compound (5) exhibited a strong binding affinity of - 8.6 kcal/mol towards PfLDH and was stabilized by forming hydrogen bonds with key amino acid residues, including ASP53, TYR85, and GLU122. Pharmacokinetic predictions indicated that compound (5) possessed favorable drug-like properties and desired pharmacokinetic characteristics. These include high absorption in the gastrointestinal tract, classification as a non-substrate of permeability glycoprotein (P-gp), non-inhibition of CYP2C19, ease of synthesis, a high predicted LD50 value of 4,000 mg/kg, and importantly, non-hepatotoxic, non-carcinogenic, and non-cytotoxic effects. CONCLUSIONS: This study demonstrated that compounds isolated from M. ferrea exhibit activity against P. falciparum. Rheediachromenoxanthone has significant potential as a scaffold for the development of potent antimalarial drugs.


Subject(s)
Antimalarials , Molecular Docking Simulation , Plant Extracts , Plant Roots , Plasmodium falciparum , Xanthones , Antimalarials/pharmacology , Antimalarials/pharmacokinetics , Antimalarials/chemistry , Antimalarials/toxicity , Plasmodium falciparum/drug effects , Xanthones/pharmacology , Xanthones/chemistry , Xanthones/pharmacokinetics , Animals , Plant Extracts/pharmacology , Plant Extracts/chemistry , Chlorocebus aethiops , Vero Cells
8.
Biomed Pharmacother ; 177: 117074, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972149

ABSTRACT

Glioma, a common and highly malignant central nervous system tumor, markedly influences patient prognosis via interactions with glioma-associated macrophages. Previous research has revealed the anticancer potential of ß-mangostin, a xanthone derivative obtained from the mangosteen fruit. This research investigated the role of ß-mangostin on microglia in the glioma microenvironment and evaluated the efficacy of ß-mangostin combined with anti-PD-1 antibody (αPD-1) in glioma-bearing mice. The results showed that, ß-mangostin attenuated M2 polarization in BV2 cells and promoted M1-related interleukin (IL)-1ß and IL-6 secretion, thereby inhibiting glioma invasion. In addition, ß-mangostin improved the anti-glioma effects of αPD-1 and increased CD8+T cell and M1-type microglia infiltration. Mechanistically, ß-mangostin bound to the stimulator of interferon genes (STING) protein, which is crucial for the anti-tumor innate immune response, and promoted STING phosphorylation in microglia, both in vivo and in vitro. These results provide insights into its mode of action and supporting further investigation into ß-mangostin as a therapeutic agent.


Subject(s)
Glioma , Membrane Proteins , Microglia , Xanthones , Xanthones/pharmacology , Animals , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Glioma/drug therapy , Glioma/pathology , Glioma/metabolism , Mice , Membrane Proteins/metabolism , Cell Line, Tumor , Mice, Inbred C57BL , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Tumor Microenvironment/drug effects , Male , Humans , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Phosphorylation/drug effects
9.
Bioorg Chem ; 150: 107609, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38964145

ABSTRACT

Herein, we scrutinized the inhibitory potential of five xanthones and a flavonoid, sourced from Centaurium spicatum, against ß-glucuronidase activity. The results showed that gentisin and azaleatin emerged as the most potent inhibitors, with significantly lower IC50 values of 0.96 ± 0.10 and 0.57 ± 0.04 µM, respectively. The evaluation of enzyme kinetics unveiled that the isolated xanthones manifested inhibition of ß-glucuronidase through a mixed inhibition mode, whereas azaleatin exhibited a noncompetitive inhibition mechanism. The findings from molecular docking analysis unveiled that the compounds under investigation, particularly azaleatin, displayed comparatively diminished binding affinities towards ß-glucuronidase. Furthermore, the tested drugs were shown to occupy a common binding site as the employed reference drug. Our comprehensive Molecular Dynamics (MD) simulations analysis revealed consistent trajectories for the investigated drugs, wherein azaleatin and gentisin demonstrated notable stabilization of energy levels. Analysis of various MD parameters revealed that drugs with the lowest IC50 values maintained relatively stable interactions with ß-glucuronidase. These drugs were shown to exert notable alterations in their conformation or flexibility upon complexation with the target enzyme. Conversely, the flexibility and accessibility of ß-glucuronidase was reduced upon drug binding, particularly with azaleatin and gentisin, underscoring the stability of the drug-enzyme complexes. Analysis of Coul-SR and LJ-SR interaction energies unveiled consistent and stable interactions between certain isolated drugs and ß-glucuronidase. Azaleatin notably displayed the lowest average Coul-SR interaction energy, suggesting strong electrostatic interactions with the enzyme's active site and significant conformational variability during simulation. Remarkably, LJ-SR interaction energies across different xanthones complexes were more negative than their Coul-SR counterparts, emphasizing the predominant role of van der Waals interactions, encompassing attractive dispersion and repulsive forces, in stabilizing the drug-enzyme complexes rather than electrostatic interactions.


Subject(s)
Enzyme Inhibitors , Glucuronidase , Molecular Docking Simulation , Xanthones , Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Xanthones/chemistry , Xanthones/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Dose-Response Relationship, Drug , Molecular Dynamics Simulation , Molecular Structure , Structure-Activity Relationship , Humans , Glycoproteins
10.
Int J Med Sci ; 21(9): 1681-1688, 2024.
Article in English | MEDLINE | ID: mdl-39006850

ABSTRACT

Hypertension affects a large number of individuals globally and is a common cause of nephropathy, stroke, ischaemic heart disease and other vascular diseases. While many anti-hypertensive medications are used safely and effectively in clinic practice, controlling hypertensive complications solely by reducing blood pressure (BP) can be challenging. α-Mangostin, a xanthone molecule extracted from the pericarp of Garcinia mangostana L., has shown various beneficial effects such as anti-tumor, anti-hyperuricemia, and anti-inflammatory properties. However, the effects of α-Mangostin on hypertension remain unknown. In this study, we observed that α-Mangostin significantly decreased systolic and diastolic blood pressure in spontaneously hypertensive rats (SHR), possibly through the down-regulation of angiotensin II (Ang II). We also identified early markers of hypertensive nephropathy, including urinary N-acetyl-ß-D-glucosaminidase (NAG) and ß2-microglobulin (ß2-MG), which were reduced by α-Mangostin treatment. Mechanistic studies suggested that α-Mangostin may inhibit renal tubular epithelial-to-mesenchymal transformation (EMT) by down-regulating the TGF-ß signaling pathway, thus potentially offering a new therapeutic approach for hypertension and hypertensive nephropathy.


Subject(s)
Angiotensin II , Blood Pressure , Epithelial-Mesenchymal Transition , Hypertension , Xanthones , Animals , Humans , Male , Rats , Angiotensin II/metabolism , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Cell Line , Epithelial-Mesenchymal Transition/drug effects , Fibrosis/drug therapy , Garcinia mangostana/chemistry , Hypertension/drug therapy , Hypertension/pathology , Hypertension, Renal/drug therapy , Hypertension, Renal/pathology , Nephritis , Rats, Inbred SHR , Signal Transduction/drug effects , Xanthones/pharmacology , Xanthones/therapeutic use
11.
Sci Rep ; 14(1): 17071, 2024 07 24.
Article in English | MEDLINE | ID: mdl-39048609

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a high mortality rate; therefore, the development of effective treatments is a priority. The stimulator of interferon genes (STING) pathway enhances tumor immunity by inducing the production of type 1 interferon (IFN) and proinflammatory cytokines and chemokines and promoting the infiltration of cytotoxic T cells. To assess the function of STING on pancreatic tumorigenesis, Ptf1aER-Cre/+ LSL-KrasG12D/+ p53loxP/loxP mice (KPC mice) and Ptf1aER-Cre/+ LSL-KrasG12D/+ p53loxP/loxP/STING-/- mice (KPCS mice) were generated. However, STING deletion did not affect pancreatic tumorigenesis in mice. Because STING is expressed not only in immune cells but also in cancer-associated fibroblasts (CAFs), we evaluated the STING function in PDAC CAFs. A mouse STING agonist 5,6-Dimethyl-9-oxo-9H-xanthene-4-acetic acid (DMXAA) was administered to KPC mice and CAFs from KPC mice and the resulting immune response was evaluated. DMXAA activated STING in PDAC CAFs in KPC mice, promoting cytotoxic T cell infiltration by secreting proinflammatory cytokines and enhancing tumor immunity. We next generated STING-deficient PDAC cells and subcutaneous tumors in which STING was expressed only in CAFs by performing bone marrow transplantation and assessed the antitumor effect of STING-activated CAFs. The administration of DMXAA to subcutaneous tumors expressing STING only in CAFs sustained the antitumor effect of DMXAA. About half of human PDACs lacked STING expression in the cancer stroma, suggesting that STING activation in PDAC CAFs exerts an antitumor effect, and STING agonists can be more effective in tumors with high than in those with low STING expression in the stroma.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Membrane Proteins , Pancreatic Neoplasms , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Mice , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Humans , Xanthones/pharmacology , Cell Line, Tumor , T-Lymphocytes, Cytotoxic/immunology
12.
Biomed Chromatogr ; 38(9): e5936, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38956791

ABSTRACT

Mangifera indica peels are a rich source of diverse flavonoids and xanthonoids; however, generally these are discarded. Computational studies revealed that mangiferin significantly interacts with amino acid residues of transcriptional regulators 1IK3, 3TOP, and 4f5S. The methanolic extract of Langra variety of mangoes contained the least phenol concentrations (22.6 ± 0.32 mg/gGAE [gallic acid equivalent]) compared to the chloroform (214.8 ± 0.12 mg/gGAE) and ethyl acetate fractions (195.6 ± 0.14 mg/gGAE). Similarly, the methanolic extract of Sindhri variety contained lower phenol concentrations (42.3 ± 0.13 mg/gRUE [relative utilization efficiency]) compared with the chloroform (85.6 ± 0.15 mg/gGAE) and ethyl acetate (76.1 ± 0.32 mg/gGAE) fractions. Langra extract exhibited significant α-glucosidase inhibition (IC50 0.06 mg/mL), whereas the ethyl acetate fraction was highly active (IC50 0.12 mg/mL) in Sindhri variety. Mangiferin exhibited significant inhibition (IC50 0.026 mg/mL). A moderate inhibition of 15-LOX was observed in all samples, whereas mangiferin was least active. In advanced glycation end product inhibition assay, the chloroform fraction of Langra variety exhibited significant inhibition in nonoxidative (IC50 64.4 µg/mL) and oxidative modes (IC50 54.7 µg/mL). It was concluded that both Langra and Sindhri peel extracts and fractions possess significant antidiabetic activities. The results suggest the potential use of peel waste in the management and complications of diabetes.


Subject(s)
Antioxidants , Glycation End Products, Advanced , Hypoglycemic Agents , Mangifera , Plant Extracts , Xanthones , Xanthones/chemistry , Xanthones/pharmacology , Xanthones/analysis , Mangifera/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Antioxidants/analysis , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/analysis , Glycation End Products, Advanced/chemistry , Spectroscopy, Fourier Transform Infrared/methods , Molecular Docking Simulation , Fruit/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolase Inhibitors/analysis , Computer Simulation
13.
J Biochem Mol Toxicol ; 38(7): e23765, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967724

ABSTRACT

Mangiferin is a naturally occurring glucosylxanthone that has shown promising immunomodulatory effects. It is generally isolated from the leaves, peels, bark, and kernels of Mangifera indica Linn. Mangiferin is like a miraculous natural bioactive molecule that has an immunomodulatory function that makes it a potential therapeutic candidate for the treatment of rheumatoid arthritis (RA) and cancer. The anticancer activity of mangiferin acts by blocking NF-κB, as well as regulating the ß-catenin, EMT, MMP9, MMP2, LDH, ROS, and NO, and also by the activation of macrophages. It has no cytotoxic effect on grown chondrocytes and lowers matrix metalloproteinase levels. Additionally, it has a potent proapoptotic impact on synoviocytes. The precise molecular mechanism of action of mangiferin on RA and malignancies is still unknown. This comprehensive review elaborates on the immunomodulatory effect of mangiferin and its anticancer and anti-RA activity. This also explained the total synthesis of mangiferin and its in vitro and in vivo screening models.


Subject(s)
Arthritis, Rheumatoid , Neoplasms , Xanthones , Xanthones/pharmacology , Xanthones/therapeutic use , Xanthones/chemistry , Humans , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Animals , Neoplasms/drug therapy , Neoplasms/immunology , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry
14.
ACS Biomater Sci Eng ; 10(8): 5027-5038, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39023101

ABSTRACT

α-Mangostin-rich extract (AME) shows promise as a functional ingredient for cancer chemotherapy. Here, we encapsulated AME in our originally designed antioxidant nanoparticles (NanoAOX) to increase its solubility and prevent oxidative degradation (AME@NanoAOX). In this study, two types of self-assembled polymers containing nitroxide radicals were engineered. These polymers were self-assembled into nanoscale particles in aqueous media, entrapping AME (abbreviated as AME@NanoAOX(B) and AME@NanoAOX(G)). These formulations considerably improved the stability of AME against oxidative degradation and exhibited different release profiles of α-mangostin under different pH conditions. Furthermore, AME-encapsulated nanoparticles exhibited potent cytotoxicity against various cancer cell lines, including human breast cancer (MCF-7), human lung cancer (A549), human colon cancer (Caco-2), human cervical cancer (HeLa), and human liver cancer (HepG2) cell lines, with minimal cytotoxicity in normal human mammary epithelial cells (hTERT-HME1), thus providing a high selectivity index (SI). These results indicated the promising feature of AME-encapsulated antioxidant nanoparticles (AME@NanoAOX) for cancer chemotherapy.


Subject(s)
Antioxidants , Nanoparticles , Xanthones , Humans , Xanthones/chemistry , Xanthones/pharmacology , Antioxidants/pharmacology , Antioxidants/chemistry , Nanoparticles/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Stability , MCF-7 Cells , Cell Survival/drug effects
15.
BMC Complement Med Ther ; 24(1): 276, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033112

ABSTRACT

BACKGROUND: Plant-derived compounds have chemopreventive properties to be used as alternative medicine. Pericarp of Mangosteen (Garcinia mangostana Linn.), a tropical fruit in Southeast Asia contains a phytochemical α-mangostin (α-MG) that demonstrates potent anticancer effects against various types of cancer. α-MG has been reported to be the most effective agent in human cancer cell lines. The objectives of this study were to develop oral gel formulations containing α-MG and determine their (1) anticancer activity, (2) anti-HPV-16 and antimicrobial activities, (3) nitric oxide (NO) inhibitory activity, and (4) wound healing effect. METHODS: Formulations of oral gel containing α-MG were developed. Anticancer activity on SCC-25 was assessed. Apoptotic induction was determined using flow cytometry technique. Antiviral activity against HPV-16 pseudovirus and antimicrobial activity against S. mutans, P. gingivalis and C. albicans were investigated. NO inhibition was carried out. Fibroblast cell migration was determined by in vitro scratch assay. RESULTS: The formulation of 1% α-MG in orabase gel demonstrated anticancer activity by promoting apoptosis in SCC-25. The induction of apoptotic activity was dose dependent with pronounced effect in late apoptosis. The formulation appeared to reduce cell viability of oral keratinocytes (OKC). At CC50 it showed an inhibition against HPV-16 pseudovirus infection. The formulation had no antimicrobial activity against S. mutans, P. gingivalis and C. albicans. No significant NO inhibitory activity and wound healing effects were found. CONCLUSIONS: 1% α-MG in orabase gel exhibited anticancer activity by inducing apoptosis although low level of cytotoxicity observed in OKC was present. The appropriate carrier for novel nano-particles targeting cancer cells should be further investigated.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Garcinia mangostana , Gels , Mouth Neoplasms , Xanthones , Xanthones/pharmacology , Humans , Apoptosis/drug effects , Mouth Neoplasms/drug therapy , Garcinia mangostana/chemistry , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Plant Extracts/pharmacology , Plant Extracts/chemistry , Human papillomavirus 16/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
16.
J Cell Mol Med ; 28(11): e18466, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38847482

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome characterized by pulmonary and systemic congestion resulting from left ventricular diastolic dysfunction and increased filling pressure. Currently, however, there is no evidence on effective pharmacotherapy for HFpEF. In this study, we aimed to investigate the therapeutic effect of total xanthones extracted from Gentianella acuta (TXG) on HFpEF by establishing an high-fat diet (HFD) + L-NAME-induced mouse model. Echocardiography was employed to assess the impact of TXG on the cardiac function in HFpEF mice. Haematoxylin and eosin staining, wheat germ agglutinin staining, and Masson's trichrome staining were utilized to observe the histopathological changes following TXG treatment. The results demonstrated that TXG alleviated HFpEF by reducing the expressions of genes associated with myocardial hypertrophy, fibrosis and apoptosis. Furthermore, TXG improved cardiomyocyte apoptosis by inhibiting the expression of apoptosis-related proteins. Mechanistic investigations revealed that TXG could activate the inositol-requiring enzyme 1α (IRE1α)/X-box-binding protein 1 (Xbp1s) signalling pathway, but the knockdown of IRE1α using the IRE1α inhibitor STF083010 or siRNA-IRE1α impaired the ability of TXG to ameliorate cardiac remodelling in HFpEF models. In conclusion, TXG alleviates myocardial hypertrophy, fibrosis and apoptosis through the activation of the IRE1α/Xbp1s signalling pathway, suggesting its potential beneficial effects on HFpEF patients.


Subject(s)
Apoptosis , Endoribonucleases , Heart Failure , Protein Serine-Threonine Kinases , Signal Transduction , X-Box Binding Protein 1 , Xanthones , Animals , Endoribonucleases/metabolism , Endoribonucleases/genetics , Heart Failure/drug therapy , Heart Failure/metabolism , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Mice , Male , Xanthones/pharmacology , Xanthones/isolation & purification , Apoptosis/drug effects , Disease Models, Animal , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Diet, High-Fat/adverse effects , Fibrosis , Stroke Volume/drug effects
17.
Eur J Pharmacol ; 977: 176724, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38851559

ABSTRACT

INTRODUCTION: Mangiferin is a Chinese herbal extract with multiple biological activities. Mangiferin can penetrate the blood‒brain barrier and has potential in the treatment of nervous system diseases. These findings suggest that mangiferin protects the neurological function in ischemic stroke rats by targeting multiple signaling pathways. However, little is known about the effect and mechanism of mangiferin in alleviating poststroke cognitive impairment. METHODS: Cerebral ischemia/reperfusion (I/R) rats were generated via middle cerebral artery occlusion. Laser speckle imaging was used to monitor the cerebral blood flow. The I/R rats were intraperitoneally (i.p.) injected with 40 mg/kg mangiferin for 7 consecutive days. Neurological scoring, and TTC staining were performed to evaluate neurological function. Behavioral experiments, including the open field test, elevated plus maze, sucrose preference test, and novel object recognition test, were performed to evaluate cognitive function. Metabolomic data from brain tissue with multivariate statistics were analyzed by gas chromatography‒mass spectrometry and liquid chromatography‒mass spectrometry. RESULTS: Mangiferin markedly decreased neurological scores, and reduced infarct areas. Mangiferin significantly attenuated anxiety-like and depression-like behaviors and enhanced learning and memory in I/R rats. According to the metabolomics results, 13 metabolites were identified to be potentially regulated by mangiferin, and the differentially abundant metabolites were mainly involved in lipid metabolism. CONCLUSIONS: Mangiferin protected neurological function and relieved poststroke cognitive impairment by improving lipid metabolism abnormalities in I/R rats.


Subject(s)
Cognitive Dysfunction , Lipid Metabolism , Rats, Sprague-Dawley , Reperfusion Injury , Xanthones , Animals , Xanthones/pharmacology , Xanthones/therapeutic use , Male , Lipid Metabolism/drug effects , Rats , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Behavior, Animal/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Disease Models, Animal , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/complications , Brain/metabolism , Brain/drug effects , Cognition/drug effects
18.
J Nat Prod ; 87(6): 1628-1634, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38869194

ABSTRACT

The unfolded protein response (UPR) is a key component of fungal virulence. The prenylated xanthone γ-mangostin isolated from Garcinia mangostana (Clusiaceae) fruit pericarp, has recently been described to inhibit this fungal adaptative pathway. Considering that Calophyllum caledonicum (Calophyllaceae) is known for its high prenylated xanthone content, its stem bark extract was fractionated using a bioassay-guided procedure based on the cell-based anti-UPR assay. Four previously undescribed xanthone derivatives were isolated, caledonixanthones N-Q (3, 4, 8, and 12), among which compounds 3 and 8 showed promising anti-UPR activities with IC50 values of 11.7 ± 0.9 and 7.9 ± 0.3 µM, respectively.


Subject(s)
Calophyllum , Unfolded Protein Response , Xanthones , Xanthones/pharmacology , Xanthones/chemistry , Xanthones/isolation & purification , Unfolded Protein Response/drug effects , Calophyllum/chemistry , Molecular Structure , Humans , Plant Bark/chemistry
19.
Int J Pharm ; 660: 124303, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38848801

ABSTRACT

Although the combination of anti-vascular strategy plus immunotherapy has emerged as the optimal first-line treatment of hepatocellular carcinoma, lack of tumor targeting leads to low antitumor efficacy and serious side effect. Here, we report an ultra-pH-sensitive nanoparticle of gambogenic acid (GNA) encapsulated by poly(ethylene glycol)-poly(2-azepane ethyl methacrylate) (PEG-PAEMA) for tumor-targeting combined therapy of anti-vascular strategy plus immunotherapy. PEG-PAEMA-GNA nanoparticle was quite stable at pH 7.4 for 30 d. In contrast, it exerted size shrinkage, charge reversal and the release of GNA at pH 6.7 within 24 h. Moreover, PEG-PAEMA-GNA significantly enhanced the anti-vascular activity, membrane-disruptive capability and pro-apoptosis when pH changed from 7.4 to 6.7. Western blot analysis exhibits that PEG-PAEMA and its GNA nanoparticle facilitated the phosphorylation of STING protein. In vivo assays show that PEG-PAEMA-GNA not only displayed much higher tumor inhibition of 92 % than 37 % of free GNA, but also inhibited tumor vasculature, promoted the maturation of dendritic cells and recruited more cytotoxic t-lymphocytes for sufficient anti-vascular therapy and immunotherapy. All these results demonstrate that PEG-PAEMA-GNA displayed tumor-targeting combined treatment of anti-vascular therapy and immunotherapy. This study offers a simple and novel method for the combination of anti-vascular therapy and immunotherapy with high selectivity towards tumor.


Subject(s)
Immunotherapy , Nanoparticles , Polyethylene Glycols , Xanthenes , Animals , Immunotherapy/methods , Nanoparticles/chemistry , Hydrogen-Ion Concentration , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Xanthenes/chemistry , Xanthenes/administration & dosage , Xanthenes/pharmacology , Cell Line, Tumor , Humans , Mice , Apoptosis/drug effects , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/therapy , Liver Neoplasms/drug therapy , Mice, Inbred C57BL , Dendritic Cells/drug effects , Dendritic Cells/immunology , Mice, Inbred BALB C , Xanthones/chemistry , Xanthones/administration & dosage , Xanthones/pharmacology , Drug Liberation , Human Umbilical Vein Endothelial Cells , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects
20.
J Nanobiotechnology ; 22(1): 324, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858692

ABSTRACT

Breast cancer remains a malignancy that poses a serious threat to human health worldwide. Chemotherapy is one of the most widely effective cancer treatments in clinical practice, but it has some drawbacks such as poor targeting, high toxicity, numerous side effects, and susceptibility to drug resistance. For auto-amplified tumor therapy, a nanoparticle designated GDTF is prepared by wrapping gambogic acid (GA)-loaded dendritic porous silica nanoparticles (DPSNs) with a tannic acid (TA)-Fe(III) coating layer. GDTF possesses the properties of near-infrared (NIR)-enhanced and pH/glutathione (GSH) dual-responsive drug release, photothermal conversion, GSH depletion and hydroxyl radical (·OH) production. When GDTF is exposed to NIR laser irradiation, it can effectively inhibit cell proliferation and tumor growth both in vitro and in vivo with limited toxicity. This may be due to the synergistic effect of enhanced tumor accumulation, and elevated reactive oxygen species (ROS) production, GSH depletion, and TrxR activity reduction. This study highlights the enormous potential of auto-amplified tumor therapy.


Subject(s)
Breast Neoplasms , Glutathione , Nanoparticles , Reactive Oxygen Species , Silicon Dioxide , Breast Neoplasms/drug therapy , Female , Nanoparticles/chemistry , Animals , Glutathione/metabolism , Humans , Hydrogen-Ion Concentration , Mice , Silicon Dioxide/chemistry , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Xanthones/chemistry , Xanthones/pharmacology , Tannins/chemistry , Tannins/pharmacology , Cell Proliferation/drug effects , Mice, Inbred BALB C , Drug Liberation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL