Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.874
Filter
Add more filters

Publication year range
1.
Cell ; 154(4): 748-62, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23910378

ABSTRACT

Binding of dsDNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) triggers formation of the metazoan second messenger c[G(2',5')pA(3',5')p], which binds the signaling protein STING with subsequent activation of the interferon (IFN) pathway. We show that human hSTING(H232) adopts a "closed" conformation upon binding c[G(2',5')pA(3',5')p] and its linkage isomer c[G(2',5')pA(2',5')p], as does mouse mSting(R231) on binding c[G(2',5')pA(3',5')p], c[G(3',5')pA(3',5')p] and the antiviral agent DMXAA, leading to similar "closed" conformations. Comparing hSTING to mSting, 2',5'-linkage-containing cGAMP isomers were more specific triggers of the IFN pathway compared to the all-3',5'-linkage isomer. Guided by structural information, we identified a unique point mutation (S162A) placed within the cyclic-dinucleotide-binding site of hSTING that rendered it sensitive to the otherwise mouse-specific drug DMXAA, a conclusion validated by binding studies. Our structural and functional analysis highlights the unexpected versatility of STING in the recognition of natural and synthetic ligands within a small-molecule pocket created by the dimerization of STING.


Subject(s)
Antiviral Agents/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nucleotides, Cyclic/metabolism , Xanthones/pharmacology , Animals , Crystallography, X-Ray , Cyclic GMP/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Models, Molecular , Mutagenesis , Protein Conformation , Signal Transduction , Structure-Activity Relationship
2.
Proc Natl Acad Sci U S A ; 121(14): e2318039121, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38536750

ABSTRACT

Melanosomes are specific organelles dedicated to melanin synthesis and accumulation in melanocytes. Autophagy is suggestively involved in melanosome degradation, although the potential underlying molecular mechanisms remain elusive. In selective autophagy, autophagy receptors and E3-ligases are the key factors conferring cargo selectivity. In B16F10 cells, ß-mangostin efficiently induced melanosome degradation without affecting other organelles such as mitochondria, peroxisomes, and the endoplasmic reticulum. Among various autophagy receptors, optineurin (OPTN) contributes TANK-binding kinase 1 (TBK1)-dependently to melanosome degradation and its knockdown inhibited ß-mangostin-mediated melanosome degradation. OPTN translocation to melanosomes was dependent on its ubiquitin-binding domain. Moreover, OPTN-mediated TBK1 activation and subsequent TBK1-mediated S187 OPTN phosphorylation were essential for melanosome degradation. ß-mangostin increased K63-linked melanosome ubiquitination. Finally, the E3-ligase RCHY1 knockdown inhibited the melanosome ubiquitination required for OPTN- and TBK1-phosphorylation as well as melanosome degradation. This study suggests that melanophagy, melanosome-selective autophagy, contributes to melanosome degradation, and OPTN and RCHY1 are an essential autophagy receptor and a E3-ligase, respectively, conferring cargo selectivity in melanophagy.


Subject(s)
Autophagy , Melanosomes , Melanosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Xanthones , Melanoma, Experimental , Animals , Mice
3.
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
4.
Plant Physiol ; 192(4): 2971-2988, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37061818

ABSTRACT

Polyprenylated xanthones are natural products with a multitude of biological and pharmacological activities. However, their biosynthetic pathway is not completely understood. In this study, metabolic profiling revealed the presence of 4-prenylated 1,3,5,6-tetrahydroxyxanthone derivatives in St. John's wort (Hypericum perforatum) root extracts. Transcriptomic data mining led to the detection of 5 variants of xanthone 4-prenyltransferase (HpPT4px) comprising 4 long variants (HpPT4px-v1 to HpPT4px-v4) and 1 short variant (HpPT4px-sh). The full-length sequences of all 5 variants were cloned and heterologously expressed in yeast (Saccharomyces cerevisiae). Microsomes containing HpPT4px-v2, HpPT4px-v4, and HpPT4px-sh catalyzed the addition of a prenyl group at the C-4 position of 1,3,5,6-tetrahydroxyxanthone; 1,3,5-trihydroxyxanthone; and 1,3,7-trihydroxyxanthone, whereas microsomes harboring HpPT4px-v1 and HpPT4px-v3 additionally accepted 1,3,6,7-tetrahydroxyxanthone. HpPT4px-v1 produced in Nicotiana benthamiana displayed the same activity as in yeast, while HpPT4px-sh was inactive. The kinetic parameters of HpPT4px-v1 and HpPT4px-sh chosen as representative variants indicated 1,3,5,6-tetrahydroxyxanthone as the preferred acceptor substrate, rationalizing that HpPT4px catalyzes the first prenylation step in the biosynthesis of polyprenylated xanthones in H. perforatum. Dimethylallyl pyrophosphate was the exclusive prenyl donor. Expression of the HpPT4px transcripts was highest in roots and leaves, raising the question of product translocation. C-terminal yellow fluorescent protein fusion of HpPT4px-v1 localized to the envelope of chloroplasts in N. benthamiana leaves, whereas short, truncated, and masked signal peptides led to the disruption of plastidial localization. These findings pave the way for a better understanding of the prenylation of xanthones in plants and the identification of additional xanthone-specific prenyltransferases.


Subject(s)
Dimethylallyltranstransferase , Hypericum , Xanthones , Hypericum/genetics , Hypericum/metabolism , Dimethylallyltranstransferase/genetics , Dimethylallyltranstransferase/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Xanthones/metabolism , Xanthones/pharmacology , Plant Extracts/pharmacology
5.
J Org Chem ; 89(11): 7692-7704, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38768258

ABSTRACT

A MS/MS-based molecular networking approach compared to the Global Natural Product Social Molecular Networking library, in association with genomic annotation of natural product biosynthetic gene clusters within a marine-derived fungus, Aspergillus sydowii, identified a suite of xanthone metabolites. Chromatographic techniques applied to the cultured fungus led to the isolation of 11 xanthone-based alkaloids, dubbed sydoxanthones F-M. The structures of these alkaloids were elucidated using extensive spectroscopic data, including electronic circular dichroism and single-crystal X-ray diffraction data for configurational assignments. Among these analogues, sydoxanthones F-K exhibit structure features typical of nucleobase-coupled xanthones, with sydoxanthone H being an N-bonded xanthone dimer. Notably, (±)sydoxanthones F (1a/1b), (±)sydoxanthones H (3b/3a), and (±)sydoxanthones J (5b/5a) are enantiomeric pairs, while sydoxanthones G (2), I (4), and K (6) are stereoisomers of 1, 3, and 5, respectively. Furthermore, (+)sydoxanthone H (3a) demonstrated significant rescue of cell viability in H2O2-injuried SH-SY5Y cells by inhibiting reactive oxygen species production, suggesting its potential for neuroprotection.


Subject(s)
Aspergillus , Reactive Oxygen Species , Xanthones , Xanthones/chemistry , Xanthones/pharmacology , Xanthones/isolation & purification , Aspergillus/chemistry , Humans , Reactive Oxygen Species/metabolism , Molecular Structure , Cell Line, Tumor
6.
Bioorg Med Chem Lett ; 100: 129620, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38280655

ABSTRACT

Six amino derivatives of xanthone were obtained via chemical synthesis. Biochemical studies revealed their SIRT2 inhibitory activity ranging from 48.5 % (compound 4, 5-chloro-2-((4-(3-methoxyphenyl)piperazin-1-yl)methyl)-9H-xanthen-9-one hydrochloride) to 93.2 % (compound 3, 5-chloro-2-(((2-methoxyphenethyl)amino)methyl)-9H-xanthen-9-one hydrochloride). The structure-activity analysis showed favourable properties of secondary amines relative to tertiary piperazine derivatives. The tested compounds do not possess additional SIRT1 activating activity and no antioxidant activity (DPPH in vitro assay). Comprehensive analysis of the lipophilicity of the obtained compounds was also performed. For compound 3 potential molecular targets and similar active compounds were predicted in order to facilitate further research in this group of compounds.


Subject(s)
Sirtuin 2 , Xanthones , Piperazine , Xanthones/pharmacology , Xanthones/chemistry , Structure-Activity Relationship
7.
Org Biomol Chem ; 22(29): 5886-5890, 2024 07 24.
Article in English | MEDLINE | ID: mdl-38804835

ABSTRACT

Neutral rhodol-based red emitters are shown to efficiently localize in mitochondria, as demonstrated by confocal microscopy and co-localization studies. A simple model is proposed to explain the localization mechanism of neutral molecules. The model takes into account the strong coupling between the molecular dipole moment and the electric field of the inner mitochondrial membrane.


Subject(s)
Fluorescent Dyes , Mitochondria , Mitochondria/metabolism , Mitochondria/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Humans , Microscopy, Confocal , Xanthones/chemistry , Molecular Structure , HeLa Cells
8.
Bioorg Med Chem ; 103: 117655, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38493728

ABSTRACT

Caged xanthones represent a class of natural secondary metabolites exhibiting significant potential as antitumor agents. These compounds are characterized by their distinct cage-like structures, which offer novel and compelling frameworks for drug design. Nonetheless, there exists a dearth of research focused on the structural modification of these compounds, particularly in relation to their cage-like architectures. This study aims to address this gap by introducing an innovative synthetic method for constructing a novel caged structure that incorporates a widely employed maleimide group. Drawing upon the well-established synthetic approach for dihydroxanthones previously developed within our research group, we successfully synthesized 13 new caged xanthones using the Diels-Alder reaction. Subsequently, we evaluated their anti-proliferative activity against HepG2, A549, and MDA-MB-231 cell lines. The results revealed that compound 10i exhibited IC50 values of 15.86 µM ± 1.29, 19.27 µM ± 1.58, and 12.96 µM ± 0.09 against these cell lines, respectively. Further investigations into the mechanism of action of 10i demonstrated its ability to induce G2/M cell cycle arrest and initiate mitochondria-mediated apoptosis in breast cancer cells.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Xanthones , Humans , Female , Xanthones/pharmacology , Xanthones/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis , Drug Screening Assays, Antitumor , Structure-Activity Relationship , Molecular Structure
9.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 40-47, 2024 03 31.
Article in English | MEDLINE | ID: mdl-38650157

ABSTRACT

The penicillin binding protein 2a (PBP2a) is a key enzyme associated with bacterial cell wall synthesis and bacterial infection. Therefore, targeting PBPa2 offers a promising approach for the therapeutics of bacterial resistance and infection. This study presents a comprehensive analysis of alpha-mangostin as a potential inhibitor of PBPa2. Molecular docking simulations revealed a strong binding affinity between alpha-mangostin and PBP2a, with an affinity score of -6.01 kcal/mol. Notably, alpha-mangostin formed a preferential hydrogen bond with THR216 of PBP2a, alongside several other polar and hydrophobic interactions. ADME and Toxicity predictions indicated that alpha-mangostin possesses favourable pharmacokinetic properties, suggesting its potential as a therapeutic agent. PASS analysis further highlighted its broad range of favourable biological properties. SwissTargetPrediction analysis reinforced these findings, indicating alpha-mangostin's association with various biological processes. Cell toxicity assays demonstrated that alpha-mangostin had no significant impact on the viability of HEK-293 cells, suggesting its potential safety for further development. The IC50 value for alpha-mangostin was found to be 33.43µM. Fluorescence-based binding assays showed that alpha-mangostin effectively inhibited PBP2a activity in a concentration-dependent manner, supporting its role as an inhibitor. In conclusion, the results suggest alpha-mangostin as a promising candidate for inhibiting PBP2a. Further,  extensive studies are warranted to explore its clinical applications.


Subject(s)
Anti-Bacterial Agents , Methicillin-Resistant Staphylococcus aureus , Molecular Docking Simulation , Penicillin-Binding Proteins , Xanthones , Penicillin-Binding Proteins/antagonists & inhibitors , Penicillin-Binding Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Humans , Xanthones/chemistry , Xanthones/pharmacology , HEK293 Cells , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Protein Binding
10.
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
11.
J Nat Prod ; 87(2): 266-275, 2024 02 23.
Article in English | MEDLINE | ID: mdl-38251859

ABSTRACT

Four cytotoxic heptacyclic caged-xanthones [gambogefic acids B-E (1-4)], a cytotoxic hexacyclic caged-xanthone [garcilatelic acid (5)], and four biphenyl derivatives [garcilatelibiphenyls A-D (6-9)] were newly isolated in a phytochemical study of a 50% MeOH/CH2Cl2 extract of Garcinia lateriflora (Clusiaceae). The isolated compounds were evaluated for antiproliferative activity against five human tumor cell lines including a vincristine-resistant line. The new caged-xanthones displayed potent activity with IC50 values from 0.5 to 6.7 µM against all tested tumor cell lines.


Subject(s)
Antineoplastic Agents, Phytogenic , Garcinia , Xanthones , Humans , Biphenyl Compounds , Cell Line, Tumor , Xanthones/pharmacology , Molecular Structure , Antineoplastic Agents, Phytogenic/pharmacology
12.
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
13.
J Nat Prod ; 87(2): 238-251, 2024 02 23.
Article in English | MEDLINE | ID: mdl-38354306

ABSTRACT

Xanthone-chromanone homo- or heterodimers are regarded as a novel class of topoisomerase (Topo) inhibitors; however, limited information about these compounds is currently available. Here, 14 new (1-14) and 6 known tetrahydroxanthone chromanone homo- and heterodimers (15-20) are reported as isolated from Penicillium chrysogenum C-7-2-1. Their structures and absolute configurations were unambiguously demonstrated by a combination of spectroscopic data, single-crystal X-ray diffraction, modified Mosher's method, and electronic circular dichroism analyses. Plausible biosynthetic pathways are proposed. For the first time, it was discovered that tetrahydroxanthones can convert to chromanones in water, whereas chromone dimerization does not show this property. Among them, compounds 5, 7, 8, and 16 exhibited significant cytotoxicity against H23 cell line with IC50 values of 6.9, 6.4, 3.9, and 2.6 µM, respectively.


Subject(s)
Antineoplastic Agents , Chromones , Penicillium chrysogenum , Penicillium , Xanthones , Molecular Structure , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Topoisomerase Inhibitors , Xanthones/pharmacology , Xanthones/chemistry , Penicillium/chemistry
14.
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
15.
Acta Pharmacol Sin ; 45(5): 1002-1018, 2024 May.
Article in English | MEDLINE | ID: mdl-38225395

ABSTRACT

Diabetes mellitus results in numerous complications. Diabetic pulmonary fibrosis (DPF), a late pulmonary complication of diabetes, has not attracted as much attention as diabetic nephropathy and cardiomyopathy. Mangiferin (MF) is a natural small molecular compound that exhibits a variety of pharmacological effects including anti-inflammatory, anti-cancer, anti-diabetes, and anti-fibrosis effects. In this study, we investigated whether long-term diabetes shock induces DPF, and explored whether MF had a protective effect against DPF. We first examined the lung tissues and sections of 20 diabetic patients obtained from discarded lung surgical resection specimens and found that pulmonary fibrosis mainly accumulated around the pulmonary vessels, accompanied by significantly enhanced endothelial-mesenchymal transition (EndMT). We established a mouse model of DPF by STZ injections. Ten days after the final STZ injection, the mice were administered MF (20, 60 mg/kg, i.g.) every 3 days for 4 weeks, and kept feeding until 16 weeks and euthanized. We showed that pulmonary fibrotic lesions were developed in the diabetic mice, which began around the pulmonary vessels, while MF administration did not affect long-term blood glucose levels, but dose-dependently alleviated diabetes-induced pulmonary fibrosis. In human umbilical vein endothelial cells (HUVECs), exposure to high glucose (33.3 mM) induced EndMT, which was dose-dependently inhibited by treatment with MF (10, 50 µM). Furthermore, MF treatment promoted SIRT3 expression in high glucose-exposed HUVECs by directly binding to AMPK to enhance the activity of FoxO3, which finally reversed diabetes-induced EndMT. We conclude that MF attenuates DPF by inhibiting EndMT through the AMPK/FoxO3/SIRT3 axis. MF could be a potential candidate for the early prevention and treatment of DPF.


Subject(s)
AMP-Activated Protein Kinases , Diabetes Mellitus, Experimental , Forkhead Box Protein O3 , Mice, Inbred C57BL , Pulmonary Fibrosis , Sirtuin 3 , Xanthones , Animals , Xanthones/pharmacology , Xanthones/therapeutic use , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Sirtuin 3/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Forkhead Box Protein O3/metabolism , Male , Humans , Mice , AMP-Activated Protein Kinases/metabolism , Epithelial-Mesenchymal Transition/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Streptozocin , Signal Transduction/drug effects , Endothelial-Mesenchymal Transition
16.
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 , Rats, Inbred SHR , Xanthones , Animals , Xanthones/pharmacology , Xanthones/therapeutic use , Rats , Epithelial-Mesenchymal Transition/drug effects , Humans , Blood Pressure/drug effects , Hypertension/drug therapy , Hypertension/pathology , Fibrosis/drug therapy , Male , Cell Line , Garcinia mangostana/chemistry , Signal Transduction/drug effects , Hypertension, Renal/drug therapy , Hypertension, Renal/pathology , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Nephritis
17.
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
18.
J Nanobiotechnology ; 22(1): 216, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698399

ABSTRACT

The enhanced permeability and retention (EPR) effect has become the guiding principle for nanomedicine against cancer for a long time. However, several biological barriers severely resist therapeutic agents' penetration and retention into the deep tumor tissues, resulting in poor EPR effect and high tumor mortality. Inspired by lava, we proposed a proteolytic enzyme therapy to improve the tumor distribution and penetration of nanomedicine. A trypsin-crosslinked hydrogel (Trypsin@PSA Gel) was developed to maintain trypsin's activity. The hydrogel postponed trypsin's self-degradation and sustained the release. Trypsin promoted the cellular uptake of nanoformulations in breast cancer cells, enhanced the penetration through endothelial cells, and degraded total and membrane proteins. Proteomic analysis reveals that trypsin affected ECM components and down-regulated multiple pathways associated with cancer progression. Intratumoral injection of Trypsin@PSA Gel significantly increased the distribution of liposomes in tumors and reduced tumor vasculature. Combination treatment with intravenous injection of gambogic acid-loaded liposomes and intratumoral injection of Trypsin@PSA Gel inhibited tumor growth. The current study provides one of the first investigations into the enhanced tumor distribution of liposomes induced by a novel proteolytic enzyme therapy.


Subject(s)
Hydrogels , Liposomes , Polyethylene Glycols , Trypsin , Xanthones , Liposomes/chemistry , Animals , Polyethylene Glycols/chemistry , Hydrogels/chemistry , Humans , Trypsin/metabolism , Trypsin/chemistry , Female , Mice , Cell Line, Tumor , Mice, Inbred BALB C , Breast Neoplasms/drug therapy , Proteolysis
19.
Biotechnol Lett ; 46(1): 127-142, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38150096

ABSTRACT

Rhodomyrtus tomentosa leaf (RT)-incorporated transferosomes were developed with lecithin and cholesterol blends with edge activators at different ratios. RT-transferosomes were characterized and employed in transferosomal gel formulations for the management of skin and soft-tissue infections. The optimized formulation entrapped up to 81.90 ± 0.31% of RT with spherical vesicles (405.3 ± 2.0 nm), polydispersity index value of 0.16 ± 0.08, and zeta potential of - 61.62 ± 0.86 mV. Total phenolic and flavonoid contents of RT-transferosomes were 15.65 ± 0.04 µg GAE/g extract and 43.13 ± 0.91 µg QE/g extract, respectively. RT-transferosomes demonstrated minimum inhibitory and minimum bactericidal concentrations at 8-256 and 64-1024 µg/mL, respectively. Free radical scavenging assay showed RT-transferosomes with high scavenging activity against DPPH and ABTS radicals. Moreover, RT-transferosomes demonstrated moderate activity against mushroom tyrosinase, with IC50 values of 245.32 ± 1.32 µg/mL. The biocompatibility results against L929 fibroblast and Vero cells demonstrated IC50 at 7.05 ± 0.17 and 4.73 ± 0.13 µg/mL, respectively. In addition, nitric oxide production significantly decreased by 6.78-88.25% following the treatment with 31.2-500 ng/mL RT-transferosomes (p < 0.001). Furthermore, the freeze-thaw stability study displayed no significant change in stability in the sedimentation and pH of gel fortified with RT-transferosomes. The results suggested that RT-transferosome formulation can be effectively employed as natural biomedicines for scar prevention and the management of skin soft-tissue infections.


Subject(s)
Liposomes , Phosphatidylcholines , Xanthones , Animals , Chlorocebus aethiops , Hydrogels , Vero Cells , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Leaves/chemistry
20.
Mar Drugs ; 22(6)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38921589

ABSTRACT

Overwhelming evidence points to an aberrant Wnt/ß-catenin signaling as a critical factor in hepatocellular carcinoma (HCC) and cervical cancer (CC) pathogenesis. Dicerandrol C (DD-9), a dimeric tetrahydroxanthenone isolated from the endophytic fungus Phomopsis asparagi DHS-48 obtained from mangrove plant Rhizophora mangle via chemical epigenetic manipulation of the culture, has demonstrated effective anti-tumor properties, with an obscure action mechanism. The objective of the current study was to explore the efficacy of DD-9 on HepG2 and HeLa cancer cells and its functional mechanism amid the Wnt/ß catenin signaling cascade. Isolation of DD-9 was carried out using various column chromatographic methods, and its structure was elucidated with 1D NMR. The cytotoxicity of DD-9 on HepG2 and HeLa cells was observed with respect to the proliferation, clonality, migration, invasion, apoptosis, cell cycle, and Wnt/ß-catenin signaling cascade. We found that DD-9 treatment significantly reduced tumor cell proliferation in dose- and time-dependent manners in HepG2 and HeLa cells. The subsequent experiments in vitro implied that DD-63 could significantly suppress the tumor clonality, metastases, and induced apoptosis, and that it arrested the cell cycle at the G0/G1 phase of HepG2 and HeLa cells. Dual luciferase assay, Western blot, and immunofluorescence assay showed that DD-9 could dose-dependently attenuate the Wnt/ß-catenin signaling by inhibiting ß-catenin transcriptional activity and abrogating ß-catenin translocated to the nucleus; down-regulating the transcription level of ß-catenin-stimulated Wnt target gene and the expression of related proteins including p-GSK3-ß, ß-catenin, LEF1, Axin1, c-Myc, and CyclinD1; and up-regulating GSK3-ß expression, which indicates that DD-9 stabilized the ß-catenin degradation complex, thereby inducing ß-catenin degradation and inactivation of the Wnt/ß-catenin pathway. The possible interaction between DD-9 and ß-catenin and GSK3-ß protein was further confirmed by molecular docking studies. Collectively, DD-9 may suppress proliferation and induce apoptosis of liver and cervical cancer cells, possibly at least in part via GSK3-ß-mediated crosstalk with the Wnt/ß-catenin signaling axis, providing insights into the mechanism for the potency of DD-9 on hepatocellular and cervical cancer.


Subject(s)
Apoptosis , Cell Proliferation , Wnt Signaling Pathway , Humans , HeLa Cells , Apoptosis/drug effects , Wnt Signaling Pathway/drug effects , Cell Proliferation/drug effects , Hep G2 Cells , beta Catenin/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Liver Neoplasms/drug therapy , Xanthones/pharmacology , Xanthones/chemistry , Xanthones/isolation & purification , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL