Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.287
Filter
1.
ACS Appl Mater Interfaces ; 16(24): 30900-30914, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38848495

ABSTRACT

Coumarins have great pharmacotherapeutic potential, presenting several biological and pharmaceutical applications, like antibiotic, fungicidal, anti-inflammatory, anticancer, anti-HIV, and healing activities, among others. These molecules are practically insoluble in water, and for biological applications, it became necessary to complex them with cyclodextrins (CDs), which influence their bioavailability in the target organism. In this work, we studied two coumarins, and it was possible to conclude that there were structural differences between 4,7-dimethyl-2H-chromen-2-one (DMC) and 7-methoxy-4-methyl-2H-chromen-2-one (MMC)/ß-CD that were solubilized in ethanol, frozen, and lyophilized (FL) and the mechanical mixtures (MM). In addition, the inclusion complex formation improved the solubility of DMC and MMC in an aqueous medium. According to the data, the inclusion complexes were formed and are more stable at a molar ratio of 2:1 coumarin/ß-CD, and hydrogen bonds along with π-π stacking interactions are responsible for the better stability, especially for (MMC)2@ß-CD. In vivo wound healing studies in mice showed faster re-epithelialization and the best deposition of collagen with the (DMC)2@ß-CD (FL) and (MMC)2@ß-CD (FL) inclusion complexes, demonstrating clearly that they have potential in wound repair. Therefore, (DMC)2@ß-CD (FL) deserves great attention because it presented excellent results, reducing the granulation tissue and mast cell density and improving collagen remodeling. Finally, the protein binding studies suggested that the anti-inflammatory activities might exert their biological function through the inhibition of MEK, providing the possibility of development of new MEK inhibitors.


Subject(s)
Coumarins , Wound Healing , beta-Cyclodextrins , beta-Cyclodextrins/chemistry , Coumarins/chemistry , Coumarins/pharmacology , Animals , Wound Healing/drug effects , Mice , Humans , Solubility , Male
2.
J Agric Food Chem ; 72(26): 14653-14662, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38860840

ABSTRACT

The damage to the mechanical barrier of the intestinal mucosa is the initiating factor and the core link of the progression of ulcerative colitis (UC). Protecting the mechanical barrier of the intestinal mucosa is of great significance for improving the health status of UC patients. ZO-1 is a key scaffold protein of the mechanical barrier of the intestinal mucosa, and its fusion with the membrane of the intestinal epithelium is a necessary condition to maintain the integrity of the mechanical barrier of the intestinal mucosa. Enteric glial cells (EGCs) play an important role in the maintenance of intestinal homeostasis and have become a new target for regulating intestinal health in recent years. In this study, we found that glycyrol (GC), a representative coumarin compound isolated from Licorice (Glycyrrhiza uralensis Fisch, used for medicine and food), can alleviate UC by promoting the production of neurotrophic factor GDNF in mice EGCs. Specifically, we demonstrated that GC promotes the production of GDNF, then activates its receptor RET, promotes ZO-1 fusion with cell membranes, and protects the intestinal mucosal mechanical barrier. The results of this study can provide new ideas for the prevention and treatment of UC.


Subject(s)
Colitis, Ulcerative , Glial Cell Line-Derived Neurotrophic Factor , Intestinal Mucosa , Neuroglia , Zonula Occludens-1 Protein , Animals , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Mice , Humans , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-1 Protein/genetics , Male , Neuroglia/drug effects , Neuroglia/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Cell Membrane/metabolism , Cell Membrane/drug effects , Proto-Oncogene Proteins c-ret/metabolism , Proto-Oncogene Proteins c-ret/genetics , Mice, Inbred C57BL , Coumarins/pharmacology , Coumarins/chemistry , Signal Transduction/drug effects , Glycyrrhiza/chemistry
3.
Parasitol Res ; 123(6): 246, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896311

ABSTRACT

Human toxocariasis is a neglected anthropozoonosis with global distribution. Treatment is based on the administration of anthelmintics; however, their effectiveness at the tissue level is low to moderate, necessitating the discovery of new drug candidates. Several groups of synthetic compounds, including coumarin derivatives, have demonstrated bioactivity against fungi, bacteria, and even parasites, such as Dactylogyrus intermedius, Leishmania major, and Plasmodium falciparum. The aim of this study was to evaluate the effect of ten coumarin-derived compounds against Toxocara canis larvae using in vitro, cytotoxicity, and in silico tests for selecting new drug candidates for preclinical tests aimed at evaluating the treatment of visceral toxocariasis. The compounds were tested in vitro in duplicate at a concentration of 1 mg/mL, and compounds with larvicidal activity were serially diluted to obtain concentrations of 0.5 mg/mL; 0.25 mg/mL; 0.125 mg/mL; and 0.05 mg/mL. The tests were performed in a microculture plate containing 100 T. canis larvae in RPMI-1640 medium. One compound (COU 9) was selected for cytotoxicity analysis using J774.A1 murine macrophages and it was found to be non-cytotoxic at any concentration tested. The in silico analysis was performed using computational models; the compound presented adequate results of oral bioavailability. To confirm the non-viability of the larvae, the contents of the microplate wells of COU 9 were inoculated intraperitoneally (IP) into female Swiss mice at 7-8 weeks of age. This confirmed the larvicidal activity of this compound. These results show that COU 9 exhibited larvicidal activity against T. canis larvae, which, after exposure to the compound, were non-viable, and that COU 9 inhibited infection in a murine model. In addition, COU 9 did not exhibit cytotoxicity and presented adequate bioavailability in silico, similar to albendazole, an anthelmintic, which is the first choice for treatment of human toxocariasis, supporting the potential for future investigations and preclinical tests on COU 9.


Subject(s)
Coumarins , Larva , Toxocara canis , Animals , Larva/drug effects , Toxocara canis/drug effects , Coumarins/pharmacology , Coumarins/chemistry , Anthelmintics/pharmacology , Anthelmintics/chemistry , Biological Availability , Mice , Computer Simulation , Toxocariasis/drug therapy , Toxocariasis/parasitology
4.
Pestic Biochem Physiol ; 202: 105967, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38879344

ABSTRACT

Coumarin is a natural product known for its diverse biological activities. While its antifungal properties in agricultural chemistry have been extensively studied, there is limited research on its antibacterial potential. In this study, we developed several novel coumarin derivatives by combining coumarin with pyridinium salt through molecular hybridization and chemical synthesis. Our findings reveal that most of these derivatives exhibit promising antibacterial activity. Among them, derivative A25 has been identified as the most effective compound based on three-dimensional quantitative structure-activity relationships. It demonstrates significant in vitro and in vivo activity against Xanthomonas oryzae pv. oryzae (Xoo), Xanthomonas oryzae pv. oryzicola (Xoc), and Xanthomonas campestris pv. citri (Xac), outperforming the commercially available thiediazole copper. Initial investigations into its mechanism of action suggest that A25 disrupts the cell membranes of Xoc and Xoo, thereby inhibiting bacterial growth. Additionally, A25 enhances the activity of defense enzymes in rice and modulates the expression of proteins related to the pyruvate metabolism pathway. This dual action contributes to rice's resistance against bacterial infestation. We anticipate that this study will serve as a foundation for the development of coumarin-based bactericides.


Subject(s)
Anti-Bacterial Agents , Coumarins , Microbial Sensitivity Tests , Oryza , Xanthomonas , Coumarins/pharmacology , Coumarins/chemical synthesis , Coumarins/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Xanthomonas/drug effects , Oryza/microbiology , Pyridinium Compounds/pharmacology , Pyridinium Compounds/chemistry , Pyridinium Compounds/chemical synthesis , Xanthomonas campestris/drug effects , Drug Design , Salts/pharmacology , Salts/chemistry , Structure-Activity Relationship
5.
CNS Drugs ; 38(7): 507-532, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38829443

ABSTRACT

Numerous physiological processes happening in the human body, including cerebral development and function, require the participation of biometal ions such as iron, copper, and zinc. Their dyshomeostasis may, however, contribute to the onset of Alzheimer's disease (AD) and potentially other neurodegenerative diseases. Chelation of biometal ions is therefore a therapeutic strategy against AD. This review provides a survey of natural and synthetic chelating agents that are or could potentially be used to target the metal hypothesis of AD. Since metal dyshomeostasis is not the only pathological aspect of AD, and the nature of this disorder is very complex and multifactiorial, the most efficient therapeutics should target as many neurotoxic factors as possible. Various coumarin derivatives match this description and apart from being able to chelate metal ions, they exhibit the capacity to inhibit cholinesterases (ChEs) and monoamine oxidase B (MAO-B) while also possessing antioxidant, anti-inflammatory, and numerous other beneficial effects. Compounds based on the coumarin scaffold therefore represent a desirable class of anti-AD therapeutics.


Subject(s)
Alzheimer Disease , Chelating Agents , Coumarins , Alzheimer Disease/drug therapy , Humans , Coumarins/pharmacology , Coumarins/therapeutic use , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Animals , Cholinesterase Inhibitors/pharmacology , Metals/chemistry
6.
J Neuroimmune Pharmacol ; 19(1): 32, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38886254

ABSTRACT

With the increasing resistance of Acinetobacter baumannii (A. baumannii) to antibiotics, researchers have turned their attention to the development of new antimicrobial agents. Among them, coumarin-based heterocycles have attracted much attention due to their unique biological activities, especially in the field of antibacterial infection. In this study, a series of coumarin derivatives were synthesized and screened for their bactericidal activities (Ren et al. 2018; Salehian et al. 2021). The inhibitory activities of these compounds on bacterial strains were evaluated, and the related mechanism of the new compounds was explored. Firstly, the MIC values and bacterial growth curves were measured after compound treatment to evaluate the antibacterial activity in vitro. Then, the in vivo antibacterial activities of the new compounds were assessed on A. baumannii-infected mice by determining the mice survival rates, counting bacterial CFU numbers, measuring inflammatory cytokine levels, and histopathology analysis. In addition, the ROS levels in the bacterial cells were measured with DCFH-DA detection kit. Furthermore, the potential target and detailed mechanism of the new compounds during infection disease therapy were predicted and evidenced with molecular docking. After that, ADMET characteristic prediction was completed, and novel, synthesizable, drug-effective molecules were optimized with reinforcement learning study based on the probed compound as a training template. The interaction between the selected structures and target proteins was further evidenced with molecular docking. This series of innovative studies provides important theoretical and experimental data for the development of new anti-A. baumannii infection drugs.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Anti-Bacterial Agents , Coumarins , High-Throughput Screening Assays , Microbial Sensitivity Tests , Animals , Acinetobacter baumannii/drug effects , Coumarins/pharmacology , Coumarins/chemistry , Coumarins/therapeutic use , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/therapeutic use , Acinetobacter Infections/drug therapy , High-Throughput Screening Assays/methods , Molecular Docking Simulation , Male , Mice, Inbred BALB C , Female
7.
BMC Complement Med Ther ; 24(1): 226, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858650

ABSTRACT

OBJECTIVE: Previous studies have shown that fraxetin has antitumor activity in a variety of tumors, but its role in acute myeloid leukemia (AML) remains unclear. In this study, we aimed to evaluate the anti-AML effect of fraxetin through cell experiments and network pharmacology analysis. METHODS: The inhibitory and apoptotic effects of fraxetin on AML cells were determined by CCK-8 and flow cytometry experiments. Potential targets of fraxetin and AML-related targets were screened using public databases. PPI network, GO functional enrichment and KEGG pathway enrichment analyses were performed to predict the hub targets and signaling pathways by which fraxetin alleviates AML. Molecular docking was used to determine the fraxetin binding sites on hub targets. Using the GEPIA database, the expression of hub targets was analyzed in relation to the overall survival of AML patients. RESULTS: Cell experiments showed that fraxetin inhibits AML cell proliferation and induces apoptosis. To explore the potential mechanism of fraxetin, 29 shared targets of fraxetin and AML were obtained through screening online public databases. Among them, AKT1, TNF, SRC, etc., are related to AML cell apoptosis. The expression levels of SRC, NOS3, VAV1, LYN, and PTGS1 were associated with the overall survival of AML patients (p value < 0.05). The enrichment analysis results identified the main pathways, namely, focal adhesion and the PI3K-AKT signaling pathway, that affected the proliferation and apoptosis of AML cells. The analysis of hub targets of the PPI network showed that AKT1, TNF, CTNNB1, etc., were hub targets, which were related to the proliferation and apoptosis of AML cells. The results of molecular docking showed that the hub targets had good binding with fraxetin. CONCLUSION: Fraxetin may inhibit AML cell proliferation and induce AML cell apoptosis through multiple targets, such as AKT1, SRC, and EGFR, and multiple pathways, such as focal adhesion and the PI3K-AKT signaling pathway.


Subject(s)
Apoptosis , Cell Proliferation , Leukemia, Myeloid, Acute , Molecular Docking Simulation , Network Pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Coumarins/pharmacology , Signal Transduction/drug effects
8.
Planta Med ; 90(7-08): 546-553, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38843794

ABSTRACT

Urolithin A is a gut metabolite of ellagitannins and reported to confer health benefits, e.g., by increased clearance of damaged mitochondria by macroautophagy or curbed inflammation. One targeted cell type are macrophages, which are plastic and able to adopt pro- or anti-inflammatory polarization states, usually assigned as M1 and M2 macrophages, respectively. This flexibility is tightly coupled to characteristic shifts in metabolism, such as increased glycolysis in M1 macrophages, and protein expression upon appropriate stimulation. This study aimed at investigating whether the anti-inflammatory properties of U: rolithin A may be driven by metabolic alterations in cultivated murine M1(lipopolysaccharide) macrophages. Expression and extracellular flux analyses showed that urolithin A led to reduced il1ß, il6, and nos2 expression and boosted glycolytic activity in M1(lipopolysaccharide) macrophages. The pro-glycolytic feature of UROLITHIN A: occurred in order to causally contribute to its anti-inflammatory potential, based on experiments in cells with impeded glycolysis. Mdivi, an inhibitor of mitochondrial fission, blunted increased glycolytic activity and reduced M1 marker expression in M1(lipopolysaccharide/UROLITHIN A: ), indicating that segregation of mitochondria was a prerequisite for both actions of UROLITHIN A: . Overall, we uncovered a so far unappreciated metabolic facet within the anti-inflammatory activity of UROLITHIN A: and call for caution about the simplified notion of increased aerobic glycolysis as an inevitably proinflammatory feature in macrophages upon exposure to natural products.


Subject(s)
Coumarins , Glycolysis , Lipopolysaccharides , Macrophages , Animals , Coumarins/pharmacology , Glycolysis/drug effects , Macrophages/metabolism , Macrophages/drug effects , Mice , Lipopolysaccharides/pharmacology , Anti-Inflammatory Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism
9.
Gut Microbes ; 16(1): 2367342, 2024.
Article in English | MEDLINE | ID: mdl-38889450

ABSTRACT

Alcohol-related liver disease (ALD) is recognized as a global health crisis, contributing to approximately 20% of liver cancer-associated fatalities. Dysbiosis of the gut microbiome is associated with the development of ALD, with the gut microbial metabolite urolithin A (UA) exhibiting a potential for alleviating liver symptoms. However, the protective efficacy of UA against ALD and its underlying mechanism mediated by microbiota remain elusive. In this study, we provide evidence demonstrating that UA effectively ameliorates alcohol-induced metabolic disorders and hepatic endoplasmic reticulum (ER) stress through a specific gut-microbiota-liver axis mediated by major urinary protein 1 (MUP1). Moreover, UA exhibited the potential to restore alcohol-induced dysbiosis of the intestinal microbiota by enriching the abundance of Bacteroides sartorii (B. sartorii), Parabacteroides distasonis (P. distasonis), and Akkermansia muciniphila (A. muciniphila), along with their derived metabolite propionic acid. Partial attenuation of the hepatoprotective effects exerted by UA was observed upon depletion of gut microbiota using antibiotics. Subsequently, a fecal microbiota transplantation (FMT) experiment was conducted to evaluate the microbiota-dependent effects of UA in ALD. FMT derived from mice treated with UA exhibited comparable efficacy to direct UA treatment, as it effectively attenuated ER stress through modulation of MUP1. It was noteworthy that strong associations were observed among the hepatic MUP1, gut microbiome, and metabolome profiles affected by UA. Intriguingly, oral administration of UA-enriched B. sartorii, P. distasonis, and A. muciniphila can enhance propionic acid production to effectively suppress ER stress via MUP1, mimicking UA treatment. Collectively, these findings elucidate the causal mechanism that UA alleviated ALD through the gut-microbiota-liver axis. This unique mechanism sheds light on developing novel microbiome-targeted therapeutic strategies against ALD.


Subject(s)
Coumarins , Endoplasmic Reticulum Stress , Gastrointestinal Microbiome , Liver Diseases, Alcoholic , Liver , Mice, Inbred C57BL , Gastrointestinal Microbiome/drug effects , Animals , Mice , Liver/metabolism , Liver/drug effects , Liver Diseases, Alcoholic/microbiology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/prevention & control , Male , Endoplasmic Reticulum Stress/drug effects , Coumarins/pharmacology , Coumarins/metabolism , Dysbiosis/microbiology , Humans , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification
10.
Mol Neurodegener ; 19(1): 49, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890703

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of blindness in elderly people in the developed world, and the number of people affected is expected to almost double by 2040. The retina presents one of the highest metabolic demands in our bodies that is partially or fully fulfilled by mitochondria in the neuroretina and retinal pigment epithelium (RPE), respectively. Together with its post-mitotic status and constant photooxidative damage from incoming light, the retina requires a tightly-regulated housekeeping system that involves autophagy. The natural polyphenol Urolithin A (UA) has shown neuroprotective benefits in several models of aging and age-associated disorders, mostly attributed to its ability to induce mitophagy and mitochondrial biogenesis. Sodium iodate (SI) administration recapitulates the late stages of AMD, including geographic atrophy and photoreceptor cell death. METHODS: A combination of in vitro, ex vivo and in vivo models were used to test the neuroprotective potential of UA in the SI model. Functional assays (OCT, ERGs), cellular analysis (flow cytometry, qPCR) and fine confocal microscopy (immunohistochemistry, tandem selective autophagy reporters) helped address this question. RESULTS: UA alleviated neurodegeneration and preserved visual function in SI-treated mice. Simultaneously, we observed severe proteostasis defects upon SI damage induction, including autophagosome accumulation, that were resolved in animals that received UA. Treatment with UA restored autophagic flux and triggered PINK1/Parkin-dependent mitophagy, as previously reported in the literature. Autophagy blockage caused by SI was caused by severe lysosomal membrane permeabilization. While UA did not induce lysosomal biogenesis, it did restore upcycling of permeabilized lysosomes through lysophagy. Knockdown of the lysophagy adaptor SQSTM1/p62 abrogated viability rescue by UA in SI-treated cells, exacerbated lysosomal defects and inhibited lysophagy. CONCLUSIONS: Collectively, these data highlight a novel putative application of UA in the treatment of AMD whereby it bypasses lysosomal defects by promoting p62-dependent lysophagy to sustain proteostasis.


Subject(s)
Coumarins , Animals , Mice , Coumarins/pharmacology , Autophagy/drug effects , Autophagy/physiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Retina/metabolism , Retina/drug effects , Retina/pathology , Mitophagy/drug effects , Mitophagy/physiology , Sequestosome-1 Protein/metabolism , Lysosomes/metabolism , Lysosomes/drug effects , Humans , Disease Models, Animal , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Iodates/toxicity
11.
Int Immunopharmacol ; 136: 112330, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38823180

ABSTRACT

An inflammatory response is one of the pathogeneses of depression. The anti-inflammatory and neuroprotective effects of auraptene have previously been confirmed. We established an inflammatory depression model by lipopolysaccharide (LPS) injection combined with unpredictable chronic mild stress (uCMS), aiming to explore the effects of auraptene on depressive-like behaviors in adult mice. Mice were divided into a control group, vehicle group, fluoxetine group, celecoxib group, and auraptene group. Then, behavioral tests were conducted to evaluate the effectiveness of auraptene in ameliorating depressive-like behavior. Cyclooxygenase-2 (COX-2), C-reactive protein (CRP), tumor necrosis factor (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) were examined by ELISA. Interleukin-10 (IL-10), interleukin-4 (IL-4), and transforming growth factor-ß (TGF-ß) were examined by protein chip technology. The morphology of microglia was observed by the immunohistochemical method. The data showed that, compared with the control group, the vehicle group mice exhibited a depressive-like behavioral phenotype, accompanied by an imbalance in inflammatory cytokines and the activation of microglia in the hippocampus. The depressive behaviors of the auraptene group's mice were significantly alleviated, along with the decrease in pro-inflammatory factors and increase in anti-inflammatory factors, while the activation of microglia was inhibited in the hippocampus. Subsequently, we investigated the role of auraptene in vitro-cultured BV-2 cells treated with LPS. The analysis showed that auraptene downregulated the expression of IL-6, TNF-α, and NO, and diminished the ratio of CD86/CD206. The results showed that auraptene reduced the excessive phagocytosis and ROS production of LPS-induced BV2 cells. In conclusion, auraptene relieved depressive-like behaviors in mice probably via modulating hippocampal neuroinflammation mediated by microglia.


Subject(s)
Coumarins , Cytokines , Depression , Hippocampus , Lipopolysaccharides , Microglia , Stress, Psychological , Animals , Microglia/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Depression/drug therapy , Depression/immunology , Depression/chemically induced , Mice , Stress, Psychological/drug therapy , Stress, Psychological/immunology , Coumarins/pharmacology , Coumarins/therapeutic use , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Disease Models, Animal , Behavior, Animal/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/immunology , Mice, Inbred C57BL , Inflammation Mediators/metabolism
12.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928509

ABSTRACT

Inhibitors of monoamine oxidases (MAOs) are of interest for the treatment of neurodegenerative disorders and other human pathologies. In this frame, the present work describes different synthetic strategies to obtain MAO inhibitors via the coupling of the aminocoumarin core with arylsulfonyl chlorides followed by copper azide-alkyne cycloaddition, leading to coumarin-sulfonamide-nitroindazolyl-triazole hybrids. The nitration position on the coumarin moiety was confirmed through nuclear magnetic resonance spectroscopy and molecular electron density theory in order to elucidate the molecular mechanism and selectivity of the electrophilic aromatic substitution reaction. The coumarin derivatives were evaluated for their inhibitory potency against monoamine oxidases and cholinesterases. Molecular docking calculations provided a rational binding mode of the best compounds in the series with MAO A and B. The work identified hybrids 14a-c as novel MAO inhibitors, with a selective action against isoform B, of potential interest to combat neurological diseases.


Subject(s)
Coumarins , Molecular Docking Simulation , Monoamine Oxidase Inhibitors , Monoamine Oxidase , Triazoles , Coumarins/chemistry , Coumarins/pharmacology , Coumarins/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology , Monoamine Oxidase/metabolism , Monoamine Oxidase/chemistry , Humans , Sulfonamides/chemistry , Sulfonamides/pharmacology , Structure-Activity Relationship , Molecular Structure , Density Functional Theory
13.
Molecules ; 29(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38930806

ABSTRACT

Pterocaulon polystachyum is a species of pharmacological interest for providing volatile and non-volatile extracts with antifungal and amebicidal properties. The biological activities of non-volatile extracts may be related to the presence of coumarins, a promising group of secondary metabolites. In the present study, leaves and inflorescences previously used for the extraction of essential oils instead of being disposed of were subjected to extraction with supercritical CO2 after pretreatment with microwaves. An experimental design was followed to seek the best extraction condition with the objective function being the maximum total extract. Pressure and temperature were statistically significant factors, and the optimal extraction condition was 240 bar, 60 °C, and pretreatment at 30 °C. The applied mathematical models showed good adherence to the experimental data. The extracts obtained by supercritical CO2 were analyzed and the presence of coumarins was confirmed. The extract investigated for cytotoxicity against bladder tumor cells (T24) exhibited significant reduction in cell viability at concentrations between 6 and 12 µg/mL. The introduction of green technology, supercritical extraction, in the exploration of P. polystachyum as a source of coumarins represents a paradigm shift with regard to previous studies carried out with this species, which used organic solvents. Furthermore, the concept of circular bioeconomy was applied, i.e., the raw material used was the residue of a steam-distillation process. Therefore, the approach used here is in line with the sustainable exploitation of native plants to obtain extracts rich in coumarins with cytotoxic potential against cancer cells.


Subject(s)
Carbon Dioxide , Chromatography, Supercritical Fluid , Coumarins , Plant Extracts , Coumarins/chemistry , Coumarins/isolation & purification , Coumarins/pharmacology , Carbon Dioxide/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/isolation & purification , Humans , Chromatography, Supercritical Fluid/methods , Plant Components, Aerial/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification
14.
Chem Pharm Bull (Tokyo) ; 72(6): 574-583, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38866495

ABSTRACT

In Vietnam, the stems and roots of the Rutaceous plant Paramignya trimera (Oliv.) Burkill (known locally as "Xáo tam phân") are widely used to treat liver diseases such as viral hepatitis and acute and chronic cirrhosis. In an effort to search for Vietnamese natural compounds capable of inhibiting coronavirus based on molecular docking screening, two new dimeric coumarin glycosides, namely cis-paratrimerin B (1) and cis-paratrimerin A (2), and two previously identified coumarins, the trans-isomers paratrimerin B (3) and paratrimerin A (4), were isolated from the roots of P. trimera and tested for their anti-angiotensin-converting enzyme 2 (ACE-2) inhibitory properties in vitro. It was discovered that ACE-2 enzyme was inhibited by cis-paratrimerin B (1), cis-paratrimerin A (2), and trans-paratrimerin B (3), with IC50 values of 28.9, 68, and 77 µM, respectively. Docking simulations revealed that four biscoumarin glycosides had good binding energies (∆G values ranging from -10.6 to -14.7 kcal/mol) and mostly bound to the S1' subsite of the ACE-2 protein. The key interactions of these natural ligands include metal chelation with zinc ions and multiple H-bonds with Ser128, Glu145, His345, Lys363, Thr371, Glu406, and Tyr803. Our findings demonstrated that biscoumarin glycosides from P. trimera roots occur naturally in both cis- and trans-diastereomeric forms. The biscoumarin glycosides Lys363, Thr371, Glu406, and Tyr803. Our findings demonstrated that biscoumarin glycosides from P. trimera roots hold potential for further studies as natural ACE-2 inhibitors for preventing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.


Subject(s)
Angiotensin-Converting Enzyme 2 , Coumarins , Glycosides , Molecular Docking Simulation , SARS-CoV-2 , Glycosides/chemistry , Glycosides/pharmacology , Glycosides/isolation & purification , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/chemistry , Humans , Coumarins/chemistry , Coumarins/pharmacology , Coumarins/isolation & purification , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , COVID-19/virology , Rutaceae/chemistry , COVID-19 Drug Treatment , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Plant Roots/chemistry , Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/isolation & purification
15.
Eur J Pharm Biopharm ; 200: 114334, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38768764

ABSTRACT

Functional polymer-lipid hybrid nanoparticles (H-NPs) are a promising class of nanocarriers that combine the benefits of polymer and lipid nanoparticles, offering biocompatibility, structural stability, high loading capacity, and, most importantly, superior surface functionalization. Here, we report the synthesis and design of highly functional H-NPs with specificity toward the transferrin receptor (TfR), using a small molecule ligand, gambogic acid (GA). A fluorescence study revealed the molecular orientation of H-NPs, where the lipid-dense core is surrounded by a polymer exterior, functionalized with GA. Urolithin A, an immunomodulator and anti-inflammatory agent, served as a model drug-like compound to prepare H-NPs via traditional emulsion-based techniques, where H-NPs led to smaller particles (132 nm) and superior entrapment efficiencies (70 % at 10 % drug loading) compared to GA-conjugated polymeric nanoparticles (P-NPs) (157 nm and 52 % entrapment efficiency) and solid lipid nanoparticles (L-NPs) (186 nm and 29 % entrapment efficiency). H-NPs showed superior intracellular accumulation compared to individual NPs using human small intestinal epithelial (FHs 74) cells. The in vitro efficacy was demonstrated by flow cytometry analysis, in which UA-laden H-NPs showed excellent anti-inflammatory properties in cisplatin-induced injury in healthy human proximal tubular cell (HK2) model by decreasing the TLR4, NF-κß, and IL-ß expression. This preliminary work highlights the potential of H-NPs as a novel functional polymer-lipid drug delivery system, establishing the foundation for future research on its therapeutic potential in addressing chemotherapy-induced acute kidney injury in cancer patients.


Subject(s)
Cisplatin , Nanoparticles , Polymers , Humans , Cisplatin/pharmacology , Nanoparticles/chemistry , Polymers/chemistry , Lipids/chemistry , Drug Carriers/chemistry , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Xanthones/pharmacology , Xanthones/chemistry , Xanthones/administration & dosage , Cell Line , Coumarins/chemistry , Coumarins/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Liposomes
16.
Future Med Chem ; 16(11): 1053-1073, 2024.
Article in English | MEDLINE | ID: mdl-38708686

ABSTRACT

Aim: This study focused on designing and synthesizing novel derivatives of 3,5,8-trisubstituted coumarin. Results: The synthesized compounds, particularly compound 5, exhibited significant cytotoxic effects on MCF-7 cells, surpassing staurosporine, and reduced toxicity toward MCF-10A cells, highlighting potential pharmacological advantages. Further, compound 5 altered the cell cycle and significantly increased apoptosis in MCF-7 cells, involving both early (41.7-fold) and late stages (33-fold), while moderately affecting necrotic signaling. The antitumor activity was linked to a notable reduction (4.78-fold) in topoisomerase IIß expression. Molecular modeling indicated compound 5's strong affinity for EGFR, human EGF2 and topoisomerase II proteins. Conclusion: These findings highlight compound 5 as a multifaceted antitumor agent for breast cancer.


[Box: see text].


Subject(s)
Antineoplastic Agents , Apoptosis , Breast Neoplasms , Cell Proliferation , Coumarins , Drug Screening Assays, Antitumor , Humans , Coumarins/chemistry , Coumarins/pharmacology , Coumarins/chemical synthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Female , Apoptosis/drug effects , DNA Topoisomerases, Type II/metabolism , Structure-Activity Relationship , MCF-7 Cells , Molecular Structure , Cell Line, Tumor , Cell Cycle/drug effects , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors
17.
Anticancer Res ; 44(6): 2335-2341, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821578

ABSTRACT

BACKGROUND/AIM: Coumarins are a broad class of naturally occurring oxygen-heterocyclic compounds found in plants with diverse biological properties, making them attractive for evaluation as novel therapeutic agents. We herein report the in vitro cytotoxic and monoamine oxidase (MAO) inhibitory activities of 3-acetylcoumarins (6a-e). MATERIALS AND METHODS: The cytotoxic activity was evaluated using crystal violet dye binding assay, and those compounds unable to induce cytotoxicity were further tested for the monoamine oxidase (MAO) activity using the MAO-GloTM kit. RESULTS: The 3-acetylcoumarins (6a-e) were non-cytotoxic (inactive) against MDA MB-231 (estrogen receptor-negative, ER-, highly invasive) and MCF-7 (estrogen receptor-positive, ER+, weakly invasive) breast cancer cell lines, but showed interesting MAOs inhibition activities. Among the synthesized compounds, 3-acetylcoumarin bearing dichloro (-diCl) (6d; IC50=0.31±0.04 µM) at Carbon-7, 8 positions showed higher inhibition, MAO B/A non-selectivity (selectivity index, SI=3.10), reversible inhibition against the hMAO-B enzyme, and neuroprotection against H2O2-treated human neuroblastoma (N2a) cells. CONCLUSION: Compound (6d) can be considered a promising scaffold for further investigation in developing hMAO-B inhibitors (MAOIs).


Subject(s)
Coumarins , Monoamine Oxidase Inhibitors , Monoamine Oxidase , Humans , Monoamine Oxidase/metabolism , Coumarins/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Cell Line, Tumor , MCF-7 Cells , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology
18.
Bioorg Chem ; 149: 107484, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38810482

ABSTRACT

A total of 37 characteristic terpenylated coumarins (1-25), including 17 undescribed compounds (1-5, 6a/6b, 7-10, 11a/11b-13a/13b), have been isolated from the root of Ferula ferulaeoides. Meanwhile, twelve pairs of enantiomers (6a/6b, 11a/11b-15a/15b, 17a/17b, 18a/18b, 20a/20b-22a/22b, and 25a/25b) were chirally purified. The structures of these new compounds were elucidated using HRESIMS, UV, NMR, and calculated 13C NMR with a custom DP4 + analysis. The absolute configurations of all the compounds were determined for the first time using electronic circular dichroism (ECD). Then, their inhibitory effects on nitric oxide (NO) production were evaluated with LPS-induced BV-2 microglia. Compared with the positive control minocycline (IC50 = 59.3 µM), ferulaferone B (2) exhibited stronger inhibitory potency with an IC50 value of 12.4 µM. The immunofluorescence investigation indicated that ferulaferone B (2) could inhibit Iba-1 expression in LPS-stimulated BV-2 microglia.


Subject(s)
Coumarins , Dose-Response Relationship, Drug , Ferula , Lipopolysaccharides , Microglia , Nitric Oxide , Coumarins/pharmacology , Coumarins/chemistry , Coumarins/isolation & purification , Ferula/chemistry , Microglia/drug effects , Microglia/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Animals , Molecular Structure , Mice , Structure-Activity Relationship , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Plant Roots/chemistry
19.
Sci Rep ; 14(1): 11770, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38783034

ABSTRACT

Auraptene (AUR) and umbelliprenin (UMB) are naturally occurring prenylated coumarins that have demonstrated promising anticancer effects across various human cancer cell lines. This meta-analysis aimed to systematically assess, compare, and quantify the anticancer efficacy of AUR and UMB by synthesizing evidence from in vitro studies. A comprehensive literature search identified 27 eligible studies investigating AUR or UMB against cancer cells. Mixed-effects models revealed significant negative associations between coumarin dose and viability for AUR (est. = - 2.27) and UMB (est. = - 3.990), underscoring their dose-dependent cytotoxicity. Meta-regression indicated slightly higher potency for UMB over AUR, potentially due to increased lipophilicity imparted by additional isoprenyl units. Machine learning approaches identified coumarin dose and cancer type as the most influential determinants of toxicity, while treatment duration and the specific coumarin displayed weaker effects. Moderate (AUR) to substantial (UMB) between-study heterogeneity was detected, although the findings proved robust. In summary, this meta-analysis establishes AUR and UMB as promising natural anticancer candidates with clear dose-toxicity relationships across diverse malignancies. The structural insights and quantifications of anticancer efficacy can inform forthcoming efforts assessing therapeutic potential in pre-clinical models and human trials.


Subject(s)
Antineoplastic Agents , Coumarins , Umbelliferones , Humans , Coumarins/chemistry , Coumarins/pharmacology , Umbelliferones/pharmacology , Umbelliferones/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Cell Survival/drug effects
20.
Phytomedicine ; 130: 155721, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38788395

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is the most prevalent malignancy in the world with an alarming rate of mortality. Despite the advancement in treatment strategies and drug developments, the overall survival rate remains poor. Therefore, it is imperative to develop alternative or complimentary anti cancer drugs with minimum off target effects. Urolithin A, a microbial metabolite of ellagic acid and ellagitannins produced endogenously by human gut micro biome is considered to have anti-cancerous activity. However anti tumorigenic effect of urolithin A in OSCC is yet to be elucidated. In this study, we examined whether urolithin A inhibits cell growth and induces both apoptosis and autophagy dependent cell death in OSCC cell lines. PURPOSE: The present study aims to evaluate the potential of urolithin A to inhibit OSCC and its regulatory effect on OSCC proliferation and invasion in vitro and in vivo mouse models. METHODS: We evaluated whether urolithin A could induce cell death in OSCC in vitro and in vivo mouse models. RESULTS: Flow cytometric and immunoblot analysis on Urolithin A treated OSCC cell lines revealed that urolithin A markedly induced cell death of OSCC via the induction of endoplasmic reticulum stress and subsequent inhibition of AKT and mTOR signaling as evidenced by decreased levels of phosphorylated mTOR and 4EBP1. This further revealed a possible cross talk between apoptotic and autophagic signaling pathways. In vivo study demonstrated that urolithin A treatment reduced tumor size and showed a decrease in mTOR, ERK1/2 and Akt levels along with a decrease in proliferation marker, Ki67. Taken together, in vitro as well as our in vivo data indicates that urolithin A is a potential anticancer agent and the inhibition of AKT/mTOR/ERK signalling is crucial in Urolithin A induced growth suppression in oral cancer. CONCLUSION: Urolithin A exerts its anti tumorigenic activity through the induction of apoptotic and autophagy pathways in OSCC. Our findings suggest that urolithin A markedly induced cell death of oral squamous cell carcinoma via the induction of endoplasmic reticulum stress and subsequent inhibition of AKT and mTOR signaling as evidenced by decreased levels of phosphorylated mTOR and 4EBP1. Urolithin A remarkably suppressed tumor growth in both in vitro and in vivo mouse models signifying its potential as an anticancer agent in the prevention and treatment of OSCC. Henceforth, our findings provide a new insight into the therapeutic potential of urolithin A in the prevention and treatment of OSCC.


Subject(s)
Apoptosis , Autophagy , Coumarins , Mouth Neoplasms , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases , Animals , Coumarins/pharmacology , Humans , Apoptosis/drug effects , TOR Serine-Threonine Kinases/metabolism , Autophagy/drug effects , Mouth Neoplasms/drug therapy , Cell Line, Tumor , Proto-Oncogene Proteins c-akt/metabolism , MAP Kinase Signaling System/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Mice , Mice, Nude , Mice, Inbred BALB C , Cell Proliferation/drug effects , Signal Transduction/drug effects , Antineoplastic Agents, Phytogenic/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...