Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.656
Filter
1.
Zhongguo Zhong Yao Za Zhi ; 49(17): 4650-4657, 2024 Sep.
Article in Chinese | MEDLINE | ID: mdl-39307803

ABSTRACT

Central nervous system(CNS) disorders can significantly impact patients' daily lives, impairing their ability to work and imposing a substantial financial burden on their families. In recent years, the incidence of CNS diseases has shown a significant increase with the continuous improvement of the quality of life and the aging problem. Therefore, the search for new preventive and curative drugs has been a research hotspot for this group of diseases. Osthole(OST), isolated from Umbelliferae such as Cnidium monnieri, Angelica sinensis, and Heracleum hemsleyanum, possesses a variety of pharmacological effects such as neuroprotective, antioxidant, anti-inflammatory, and antithrombotic effects. There is increasing evidence that OST has demonstrated significant preventive and curative effects in various CNS disease models. This paper systematically reviewed the research progress of OST in preventing and treating CNS diseases by reviewing domestic and international literature to provide more in-depth theoretical support for the future clinical application of OST in the prevention and treatment of CNS diseases.


Subject(s)
Central Nervous System Diseases , Coumarins , Coumarins/therapeutic use , Humans , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/prevention & control , Animals , Drugs, Chinese Herbal/therapeutic use , Neuroprotective Agents/therapeutic use
2.
Front Cell Infect Microbiol ; 14: 1403219, 2024.
Article in English | MEDLINE | ID: mdl-39253327

ABSTRACT

Introduction: Despite years of efforts to develop new antibiotics for eradicating multidrug-resistant (MDR) and multi-virulent Methicillin-Resistant Staphylococcus aureus (MRSA) and Vancomycin-Resistant Staphylococcus aureus (VRSA) infections, treatment failures and poor prognoses in most cases have been common. Therefore, there is an urgent need for new therapeutic approaches targeting virulence arrays. Our aim is to discover new anti-virulence therapies targeting MRSA and VRSA virulence arrays. Methodology: We employed phenotypic, molecular docking, and genetic studies to screen for anti-virulence activities among selected promising compounds: Coumarin, Simvastatin, and Ibuprofen. Results: We found that nearly all detected MRSA and VRSA strains exhibited MDR and multi-virulent profiles. The molecular docking results aligned with the phenotypic and genetic assessments of virulence production. Biofilm and hemolysin productions were inhibited, and all virulence genes were downregulated upon treatment with sub-minimum inhibitory concentration (sub-MIC) of these promising compounds. Ibuprofen was the most active compound, exhibiting the highest inhibition and downregulation of virulence gene products. Moreover, in vivo and histopathological studies confirmed these results. Interestingly, we observed a significant decrease in wound area and improvements in re-epithelialization and tissue organization in the Ibuprofen and antimicrobial treated group compared with the group treated with antimicrobial alone. These findings support the idea that a combination of Ibuprofen and antimicrobial drugs may offer a promising new therapy for MRSA and VRSA infections. Conclusion: We hope that our findings can be implemented in clinical practice to assist physicians in making the most suitable treatment decisions.


Subject(s)
Anti-Bacterial Agents , Biofilms , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Molecular Docking Simulation , Staphylococcal Infections , Vancomycin-Resistant Staphylococcus aureus , Virulence Factors , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Methicillin-Resistant Staphylococcus aureus/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Biofilms/drug effects , Virulence Factors/genetics , Vancomycin-Resistant Staphylococcus aureus/drug effects , Animals , Virulence/drug effects , Ibuprofen/pharmacology , Ibuprofen/therapeutic use , Humans , Coumarins/pharmacology , Coumarins/therapeutic use , Mice , Disease Models, Animal , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics , Drug Resistance, Multiple, Bacterial
3.
Int Immunopharmacol ; 142(Pt B): 113151, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39303538

ABSTRACT

The current study aimed to evaluate the preventive effects of urolithin C (Uro C), a gut microbial metabolite of ellagitannins on D-galactose (D-gal)-induced brain damage during the aging process and to elucidate the underlying mechanisms. In our study, the protective effect of Uro C on D-gal-induced BV2 microglia cell-mediated neuroinflammation damage in primary cortical neurons in vitro was confirmed. The results in an aging model in vivo induced by D-gal demonstrated that Uro C prevented D-gal-induced memory impairment, long-term potentiation (LTP) damage, and synaptic dysfunction through behavioral, electrophysiological, and histological examinations. Additionally, amyloidogenesis was observed in the central nervous system. The findings indicated that Uro C exhibited a preventive effect on the D-gal-induced elevation of ß-amyloid (1-42 specific) (Aß1-42) accumulation, APP levels, ABCE1 levels, and the equilibrium of the cholinergic system in the aging mouse brain. Moreover, Uro C demonstrated downregulation of D-gal-induced glial overactivation through inhibition of the MAPK/NF-kB pathway. This resulted in the regulation of inflammatory mediators and cytokines, including iNOS, IL-6, IL-1ß, and TNF-ɑ, in the mouse brain and BV2 microglial cells. Taken together, our results suggested that Uro C treatment could effectively mitigate the D-gal-induced memory impairment and amyloidogenesis, and the underlying mechanism might be tightly related to the improvement of neuroinflammation by suppressing the MAPK/NF-kB pathway, indicating Uro C might be an alternative and promising agent for the treatment of aging and age-associated brain diseases.


Subject(s)
Aging , Amyloid beta-Peptides , Cognitive Dysfunction , Coumarins , Galactose , Hydrolyzable Tannins , Microglia , NF-kappa B , Signal Transduction , Animals , Coumarins/pharmacology , Coumarins/therapeutic use , NF-kappa B/metabolism , Hydrolyzable Tannins/pharmacology , Hydrolyzable Tannins/therapeutic use , Aging/drug effects , Aging/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Mice , Amyloid beta-Peptides/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Signal Transduction/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Down-Regulation/drug effects , Cell Line , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured
4.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125817

ABSTRACT

Recent studies confirmed that pyroptosis is involved in the progression of pulmonary hypertension (PH), which could promote pulmonary artery remodeling. Urolithin A (UA), an intestinal flora metabolite of ellagitannins (ETs) and ellagic acid (EA), has been proven to possess inhibitory effects on pyroptosis under various pathological conditions. However, its role on PH remained undetermined. To investigate the potential of UA in mitigating PH, mice were exposed to hypoxia (10% oxygen, 4 weeks) to induce PH, with or without UA treatment. Moreover, in vitro experiments were carried out to further uncover the underlying mechanisms. The in vivo treatment of UA suppressed the progression of PH via alleviating pulmonary remodeling. Pyroptosis-related genes were markedly upregulated in mice models of PH and reversed after the administration of UA. In accordance with that, UA treatment significantly inhibited hypoxia-induced pulmonary arterial smooth muscle cell (PASMC) pyroptosis via the AMPK/NF-κB/NLRP3 pathway. Our results revealed that UA treatment effectively mitigated PH progression through inhibiting PASMC pyroptosis, which represents an innovative therapeutic approach for PH.


Subject(s)
AMP-Activated Protein Kinases , Coumarins , Hypertension, Pulmonary , Hypoxia , Myocytes, Smooth Muscle , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Pulmonary Artery , Pyroptosis , Signal Transduction , Animals , Coumarins/pharmacology , Coumarins/therapeutic use , Pyroptosis/drug effects , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Hypoxia/metabolism , Hypoxia/complications , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/etiology , Male , AMP-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Disease Models, Animal
5.
Int Immunopharmacol ; 140: 112854, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39116494

ABSTRACT

The inflammation causes the destroyed osseointegration at the implant-bone interface, significantly increasing the probability of implant loosening in osteoporotic patients. Currently, inhibiting the differentiation of M1 macrophages and the inflammatory response could be a solution to stabilize the microenvironment of implants. Interestingly, some natural products have anti-inflammatory and anti-polarization effects, which could be a promising candidate for stabilizing the implants' microenvironment in osteoporotic patients. This research aims to explore the inhibitory effect of Urolithin B(UB) on macrophage M1 polarization, which ameliorates inflammation, thus alleviating implant instability. We established an osteoporosis mouse model of implant loosening. The mouse tissues were taken out for morphological analysis, staining analysis, and bone metabolic index analysis. In in vitro experiments, RAW264.7 cells were polarized to M1 macrophages using lipopolysaccharide (LPS) and analyzed by immunofluorescence (IF) staining, Western blot (WB), and flow cytometry. The CSP100 plus chip experiments were used to explore the potential mechanisms behind the inhibiting effects of UB. Through observation of these experiments, UB can improve the osseointegration between the implants and femurs in osteoporotic mice and enhance the stability of implants. The UB can inhibit the differentiation of M1 macrophages and local inflammation via inhibiting the phosphorylation of VEGFR2, which can be further proved by the weakened inhibited effects of UB in macrophages with lentivirus-induced overexpression of VEGFR2. Overall, UB can specifically inhibit the activation of VEGFR2, alleviate local inflammation, and improve the stability of implants in osteoporotic mice.


Subject(s)
Cell Differentiation , Coumarins , Macrophages , Osteoporosis , Vascular Endothelial Growth Factor Receptor-2 , Animals , Female , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Differentiation/drug effects , Coumarins/pharmacology , Coumarins/therapeutic use , Disease Models, Animal , Down-Regulation/drug effects , Inflammation/drug therapy , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Osseointegration/drug effects , Osteoporosis/drug therapy , Osteoporosis/immunology , Phosphorylation/drug effects , Prostheses and Implants , RAW 264.7 Cells , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Int Immunopharmacol ; 140: 112839, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39126737

ABSTRACT

OBJECTIVES: Treatment of hemorrhagic shock (HS) induced multi-organ injury remains a challenge. Bergapten (BeG) is a bioactive coumarin-derived compound, and previous articles have suggested that BeG may serve as a prospective therapeutic modality for HS. This study was designed to investigate the efficacy of BeG in the treatment of HS and its underlying mechanisms. METHODS: In this research, we established a rat model of HS, following which we assessed the protective effects of BeG on HS induced multi-organ injury. Subsequently, we scrutinized the activation of NLRP3 inflammasomes and pyroptosis in damaged organs. Additionally, we conducted examinations of AMPK and the downstream mitophagy pathway in damaged organs. Finally, we established a hypoxia/reoxygenation (H/R) model in HK-2 cells to simulate the in vitro HS process. Following AMPK inhibition with compound C, we evaluated the levels of mitophagy and cellular pyroptosis in BeG-treated HK-2 cells subjected to H/R. RESULTS: BeG treatment alleviated HS induced multi-organ injury. Subsequent analyses indicated that the therapeutic effects of BeG were related to the attenuation of NLRP3 inflammasome activation and pyroptosis. Additionally, we found BeG treatment stimulated the phosphorylation of AMPK, thereby enhancing mitophagy. Lastly, we found that the inhibition of AMPK in vitro attenuates BeG's enhancement of mitophagy and its suppression of pyroptosis. CONCLUSION: Our research indicates that BeG has the potential to alleviate multi-organ injury induced by HS. The protective effect of BeG is likely associated with its promotion of mitophagy through AMPK activation, thereby inhibiting NLRP3 inflammasome-mediated pyroptosis.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Shock, Hemorrhagic , Animals , Male , Rats , AMP-Activated Protein Kinases/metabolism , Cell Line , Coumarins/pharmacology , Coumarins/therapeutic use , Disease Models, Animal , Inflammasomes/metabolism , Mitophagy/drug effects , Multiple Organ Failure/drug therapy , Multiple Organ Failure/prevention & control , Multiple Organ Failure/etiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Rats, Sprague-Dawley , Shock, Hemorrhagic/drug therapy , Shock, Hemorrhagic/complications
7.
Molecules ; 29(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39124919

ABSTRACT

The main feature of neurodegenerative diseases, including Alzheimer's disease, is the network of complex and not fully recognized neuronal pathways and targets involved in their onset and progression. The therapeutic treatment, at present mainly symptomatic, could benefit from a polypharmacological approach based on the development of a single molecular entity designed to simultaneously modulate different validated biological targets. This strategy is principally based on molecular hybridization, obtained by linking or merging different chemical moieties acting with synergistic and/or complementary mechanisms. The coumarin core, widely found in nature, endowed with a recognized broad spectrum of pharmacological activities, large synthetic accessibility and favourable pharmacokinetic properties, appears as a valuable, privileged scaffold to be properly modified in order to obtain compounds able to engage different selected targets. The scientific literature has long been interested in the multifaceted profiles of coumarin derivatives, and in this review, a survey of the most important results of the last four years, on both natural and synthetic coumarin-based compounds, regarding the development of anti-Alzheimer's compounds is reported.


Subject(s)
Alzheimer Disease , Coumarins , Drug Discovery , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Coumarins/chemistry , Coumarins/therapeutic use , Coumarins/pharmacology , Humans , Animals , Biological Products/chemistry , Biological Products/therapeutic use , Biological Products/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(7): 1370-1381, 2024 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-39051083

ABSTRACT

OBJECTIVE: To observe the therapeutic effects of urolithin A (UA) on respiratory syncytial virus (RSV)-induced lung infection in neonatal mice and explore the underlying mechanisms. METHODS: Babl/c mice (5-7 days old) were subjected to nasal instillation of RSV and received intraperitoneal injection of saline or 2.5, 5 and 10 mg/kg UA 2 h after the infection and then once daily for 2 weeks. Bronchoalveolar lavage fluid (BALF) was then collected for detection of inflammatory cells and mediators, and lung pathology was evaluated with HE staining. RSV-infected BEAS-2B cells were treated with 2.5, 5 or 10 µmol/ L UA. Inflammatory factors, cell viability, apoptosis and autophagy were analyzed using ELISA, CCK-8 assay, TUNEL staining, flow cytometry, Western blotting and immunofluorescence staining. The cellular expressions of miR-136 and Sirt1 mRNAs were detected using qRT-PCR. A dual-luciferase reporter system was used to verify the binding between miR-136 and Sirt1. RESULTS: In neonatal Babl/c mice, RSV infection caused obvious lung pathologies, promoted pulmonary cell apoptosis and LC3-Ⅱ/Ⅰ, Beclin-1 and miR-136 expressions, and increased the total cell number, inflammatory cells and factors in the BALF and decreased p62 and Sirt1 expressions. All these changes were alleviated dose-dependently by UA. In BEAS-2B cells, RSV infection significantly increased cell apoptosis, LC3B-positive cells and miR-136 expression and reduced Sirt1 expression (P<0.01), which were dose-dependently attenuated by UA. Dual-luciferase reporter assay confirmed the binding between miR-136 and Sirt1. In RSV-infected BEAS-2B cells with UA treatment, overexpression of miR-136 and Ex527 treatment both significantly increased the inflammatory factors and cell apoptosis but decreased LC3B expression, and these changes were further enhanced by their combined treatment. CONCLUSION: UA ameliorates RSV-induced lung infection in neonatal mice by activating miR-136-mediated Sirt1 signaling pathway.


Subject(s)
Animals, Newborn , Apoptosis , Mice, Inbred BALB C , MicroRNAs , Respiratory Syncytial Virus Infections , Respiratory Syncytial Viruses , Signal Transduction , Sirtuin 1 , Animals , Mice , Sirtuin 1/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Signal Transduction/drug effects , Apoptosis/drug effects , Lung/pathology , Lung/metabolism , Coumarins/pharmacology , Coumarins/therapeutic use , Bronchoalveolar Lavage Fluid , Autophagy/drug effects , Humans
9.
Inflamm Res ; 73(10): 1657-1670, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39052062

ABSTRACT

OBJECTIVE: Nordalbergin is a coumarin extracted from Dalbergia sissoo DC. To date, the biological effects of nordalbergin have not been well investigated. To investigate the anti-inflammatory responses and the anti-oxidant abilities of nordalbergin using lipopolysaccharide (LPS)-activated macrophages and LPS-induced sepsis mouse model. MATERIALS AND METHODS: Production of nitrite oxide (NO), prostaglandin E2 (PGE2), pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1ß), reactive oxygen species (ROS), tissue damage and serum inflammatory markers, and the activation of the NLRP3 inflammasome were examined. RESULTS: Our results indicated that nordalbergin reduced the production of NO and pro-inflammatory cytokines in vitro and ex vivo. Nordalbergin also suppressed iNOS and cyclooxygenase-2 expressions, decreased NF-κB activity, and attenuated MAPKs signaling pathway activation by decreasing JNK and p38 phosphorylation by LPS-activated J774A.1 macrophages. Notably, nordalbergin diminished NLRP3 inflammasome activation via repressing the maturation of IL-1ß and caspase-1 and suppressing ROS production by LPS/ATP- and LPS/nigericin-activated J774A.1 macrophages. Furthermore, nordalbergin exhibited protective effects against the infiltration of inflammatory cells and also inhibited the levels of organ damage markers (AST, ALT, BUN) by LPS-challenged mice. CONCLUSION: Nordalbergin possesses anti-inflammatory effects in macrophage-mediated innate immune responses, alleviates ROS production, decreases NLRP3 activation, and exhibits protective effects against LPS-induced tissue damage in mice.


Subject(s)
Endotoxemia , Inflammasomes , Lipopolysaccharides , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species , Animals , Reactive Oxygen Species/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , NF-kappa B/metabolism , Male , Endotoxemia/chemically induced , Endotoxemia/drug therapy , Inflammasomes/drug effects , Inflammasomes/metabolism , Mice, Inbred C57BL , Cytokines/metabolism , Signal Transduction/drug effects , Coumarins/pharmacology , Coumarins/therapeutic use , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Line , Macrophages/drug effects , Macrophages/metabolism , Nitric Oxide/metabolism , MAP Kinase Signaling System/drug effects
10.
Cell Mol Gastroenterol Hepatol ; 18(4): 101379, 2024.
Article in English | MEDLINE | ID: mdl-39038605

ABSTRACT

BACKGROUND & AIMS: Cirrhotic portal hypertension (CPH) is the leading cause of mortality in patients with cirrhosis. Over 50% of patients with CPH treated with current clinical pharmacotherapy still present variceal bleeding or sometimes death owing to insufficient reduction in portal pressure. Elevated intrahepatic vascular resistance (IHVR) plays a fundamental role in increasing portal pressure. Because of its potent effect in reducing portal pressure and maintaining normal portal inflow to preserve liver function, lowering the IHVR is acknowledged as an optimal anti-CPH strategy but without clinical drugs. We aimed to investigate the protective effect of microbial-derived Urolithin A (UroA) in IHVR and CPH. METHODS: Carbon tetrachloride or bile duct ligation surgery was administered to mice to induce liver fibrosis and CPH. 16S rRNA gene sequencing was used for microbial analysis. Transcriptomics and metabolomics analyses were employed to study the host and cell responses. RESULTS: UroA was remarkably deficient in patients with CPH and was negatively correlated with disease severity. UroA deficiency was also confirmed in CPH mice and was associated with a reduced abundance of UroA-producing bacterial strain (Lactobacillus murinus, L. murinus). Glutaminolysis of hepatic stellate cells (HSCs) was identified as a previously unrecognized target of UroA. UroA inhibited the activity of glutaminase1 to suppress glutaminolysis, which counteracted fibrogenesis and contraction of HSCs and ameliorated CPH by relieving IHVR. Supplementation with UroA or L. murinus effectively ameliorated CPH in mice. CONCLUSIONS: We for the first time identify the deficiency of gut microbial metabolite UroA as an important cause of CPH. We demonstrate that UroA exerts an excellent anti-CPH effect by suppressing HSC glutaminolysis to lower the IHVR, which highlighted its great potential as a novel therapeutic agent for CPH.


Subject(s)
Coumarins , Glutaminase , Hypertension, Portal , Liver Cirrhosis , Animals , Mice , Hypertension, Portal/drug therapy , Hypertension, Portal/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Humans , Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Male , Coumarins/pharmacology , Coumarins/therapeutic use , Glutamine/metabolism , Glutamine/pharmacology , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Vascular Resistance/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology
11.
Eur J Pharmacol ; 979: 176829, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39053867

ABSTRACT

Alzheimer's disease (AD) and osteoporosis (OP) are both serious degenerative diseases, with the potential for concurrent occurrence in clinical settings, and they share certain pathological correlations. Osthole (OST) and notopterol (NOT) are the main active ingredients in traditional Chinese medicine, Angelica pubescens and Notopterygium incisum, respectively, and they exhibit neuroprotective and osteoprotective effects. However, whether the combination of OST and NOT produces a synergistic effect against AD and/or OP remains unclear. The aim of this study was to investigate whether the combination of OST and NOT could produce synergistic anti-AD and/or OP effects using the previously constructed zebrafish AD/OP comorbidity model. Active compounds with anti-AD and OP effects were screened from Angelica pubescens and Notopterygium incisum through network pharmacology, identifying OST and NOT, respectively. Then, the AlCl3-induced (Aluminum chloride, AlCl3) AD combined with OP zebrafish model, in conjunction with the Chou-Talalay synergy evaluation model, was employed to assess whether the OST and NOT combination produced synergistic effects against AD and/or OP. Furthermore, a CuSO4-induced (Copper sulfate, CuSO4) inflammation zebrafish model was used to investigate whether the combination of OST and NOT produced synergistic anti-inflammatory effects, thereby resulting in synergistic anti-AD and/or OP effects. The results demonstrated that the OST-NOT combined treatment produced a synergistic anti-AD and OP effect. Moreover, the combined treatment of OST and NOT significantly inhibited nitric oxide (NO) and reactive oxygen species (ROS) release more effectively than OST or NOT alone, indicating a synergistic anti-inflammatory effect of the OST and NOT combined treatment.


Subject(s)
Alzheimer Disease , Coumarins , Disease Models, Animal , Drug Synergism , Osteoporosis , Zebrafish , Animals , Coumarins/pharmacology , Coumarins/therapeutic use , Alzheimer Disease/drug therapy , Osteoporosis/drug therapy , Drug Therapy, Combination , Comorbidity , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide/metabolism
12.
J Med Chem ; 67(15): 13231-13251, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39049433

ABSTRACT

Increasing evidence has demonstrated that oxidative phosphorylation (OXPHOS) is closely associated with the progression of pancreatic cancer (PC). Given its central role in mitochondrial transcription, the human mitochondrial RNA polymerase (POLRMT) is a promising target for developing PC treatments. Herein, structure-activity relationship exploration led to the identification of compound S7, which was the first reported POLRMT inhibitor possessing single-digit nanomolar potency of inhibiting PC cells proliferation. Mechanistic studies showed that compound S7 exerted antiproliferative effects without affecting the cell cycle, apoptosis, mitochondrial membrane potential (MMP), or intracellular reactive oxygen species (ROS) levels specifically in MIA PaCa-2 cells. Notably, compound S7 inhibited tumor growth in MIA PaCa-2 xenograft tumor model with a tumor growth inhibition (TGI) rate of 64.52% demonstrating significant improvement compared to the positive control (44.80%). In conclusion, this work enriched SARs of POLRMT inhibitors, and compound S7 deserved further investigations of drug-likeness as a candidate for PC treatment.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Coumarins , DNA-Directed RNA Polymerases , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Animals , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Coumarins/pharmacology , Coumarins/chemistry , Coumarins/chemical synthesis , Coumarins/therapeutic use , Cell Proliferation/drug effects , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , Cell Line, Tumor , Mice , Mice, Nude , Fluorine/chemistry , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Drug Screening Assays, Antitumor
13.
J Pharm Pharmacol ; 76(8): 1038-1050, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38848454

ABSTRACT

OBJECTIVE: Breast cancer is a malignant tumor with high invasion and metastasis. TGF-ß1-induced epithelial-mesenchymal transition (EMT) is crucially involved in the growth and metastasis of breast cancer. Wedelolactone (Wed) is extracted from herbal medicine Ecliptae Herba, which is reported to have antineoplastic activity. Here, we aimed to elucidate the efficacy and mechanism of Wed against breast cancer. METHODS: The effects of Wed on migration and invasion of 4T1 were detected. The expression of EMT-related markers was detected by Western blot and qPCR. The 4T1 orthotopic murine breast cancer model was established to evaluate the therapeutic effect of Wed on the growth and metastasis of breast cancer through TGF-ß1/Smad pathway. RESULTS: Wed inhibited the proliferation, migration and invasion of 4T1. It exhibited concentration-dependent inhibition of p-Smad2/3. Wed also reversed the expression of EMT-markers induced by TGF-ß1. In addition, Wed suppressed the growth and metastasis of breast cancer in mice. It also affected p-Smad3 expression as well as EMT-related genes, suggesting that its anti-breast cancer effect may be related to the TGF-ß1/Smad pathway. CONCLUSION: Wed reverses EMT by regulating TGF-ß1/Smad pathway, potentially serving as a therapeutic agent for breast cancer. Wed is expected to be a potential drug to inhibit TGF-ß1/Smad pathway-related diseases.


Subject(s)
Breast Neoplasms , Cell Movement , Cell Proliferation , Coumarins , Epithelial-Mesenchymal Transition , Mice, Inbred BALB C , Signal Transduction , Transforming Growth Factor beta1 , Animals , Transforming Growth Factor beta1/metabolism , Female , Signal Transduction/drug effects , Epithelial-Mesenchymal Transition/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Movement/drug effects , Coumarins/pharmacology , Coumarins/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Smad Proteins/metabolism , Neoplasm Metastasis , Smad3 Protein/metabolism , Neoplasm Invasiveness , Humans , Smad2 Protein/metabolism
14.
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
15.
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
16.
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
17.
Cell Biol Int ; 48(8): 1111-1123, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38741282

ABSTRACT

Polycystic ovary syndrome (PCOS) is the primary cause of female infertility with a lack of universal therapeutic regimen. Although osthole exhibits numerous pharmacological activities in treating various diseases, its therapeutic effect on PCOS is undiscovered. The present study found that application of osthole improved the symptoms of PCOS mice through preventing ovarian granulosa cells (GCs) production of more estrogen and alleviating the liberation of pro-inflammatory cytokine interleukin (IL)-1ß, IL-6, and tumor necrosis factor alpha. Meanwhile, osthole enhanced ovarian antioxidant capacity and alleviated intracellular reactive oxygen species (ROS) accumulation with a concurrent attenuation for oxidative stress, while intervention of antioxidant enzymic activity and glutathione (GSH) synthesis neutralized the salvation of osthole on GCs secretory disorder and chronic inflammation. Further analysis revealed that osthole restored the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and forkhead box O 1 (Foxo1) whose repression antagonized the amelioration of osthole on the insufficiency of antioxidant capacity and accumulation of ROS. Moreover, Nrf2 served as an intermedium to mediate the regulation of osthole on Foxo1. Additionally, osthole restricted the phosphorylation of IκBα and nuclear factor kappa B (NF-κB) subunit p65 by DHEA and weakened the transcriptional activity of NF-κB, but this effectiveness was abrogated by the obstruction of Nrf2 and Foxo1, whereas adjunction of GSH renewed the redemptive effect of osthole on NF-κB whose activation caused an invalidation of osthole in rescuing the aberration of GCs secretory function and inflammation response. Collectively, osthole might relieve the symptoms of PCOS mice via Nrf2-Foxo1-GSH-NF-κB pathway.


Subject(s)
Coumarins , Forkhead Box Protein O1 , Glutathione , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Polycystic Ovary Syndrome , Reactive Oxygen Species , Signal Transduction , Animals , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/drug therapy , Female , NF-E2-Related Factor 2/metabolism , Mice , Coumarins/pharmacology , Coumarins/therapeutic use , NF-kappa B/metabolism , Forkhead Box Protein O1/metabolism , Signal Transduction/drug effects , Glutathione/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Disease Models, Animal
18.
Alzheimers Dement ; 20(6): 4212-4233, 2024 06.
Article in English | MEDLINE | ID: mdl-38753870

ABSTRACT

BACKGROUND: Compromised autophagy, including impaired mitophagy and lysosomal function, plays pivotal roles in Alzheimer's disease (AD). Urolithin A (UA) is a gut microbial metabolite of ellagic acid that stimulates mitophagy. The effects of UA's long-term treatment of AD and mechanisms of action are unknown. METHODS: We addressed these questions in three mouse models of AD with behavioral, electrophysiological, biochemical, and bioinformatic approaches. RESULTS: Long-term UA treatment significantly improved learning, memory, and olfactory function in different AD transgenic mice. UA also reduced amyloid beta (Aß) and tau pathologies and enhanced long-term potentiation. UA induced mitophagy via increasing lysosomal functions. UA improved cellular lysosomal function and normalized lysosomal cathepsins, primarily cathepsin Z, to restore lysosomal function in AD, indicating the critical role of cathepsins in UA-induced therapeutic effects on AD. CONCLUSIONS: Our study highlights the importance of lysosomal dysfunction in AD etiology and points to the high translational potential of UA. HIGHLIGHTS: Long-term urolithin A (UA) treatment improved learning, memory, and olfactory function in Alzheimer's disease (AD) mice. UA restored lysosomal functions in part by regulating cathepsin Z (Ctsz) protein. UA modulates immune responses and AD-specific pathophysiological pathways.


Subject(s)
Alzheimer Disease , Coumarins , Disease Models, Animal , Lysosomes , Mice, Transgenic , Mitophagy , Alzheimer Disease/drug therapy , Animals , Coumarins/pharmacology , Coumarins/therapeutic use , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mitophagy/drug effects , Amyloid beta-Peptides/metabolism , Cognition/drug effects
19.
PeerJ ; 12: e17380, 2024.
Article in English | MEDLINE | ID: mdl-38799063

ABSTRACT

As the inflammatory subtype of nonalcoholic fatty liver disease (NAFLD), the progression of nonalcoholic steatohepatitis (NASH) is associated with disorders of glycerophospholipid metabolism. Scoparone is the major bioactive component in Artemisia capillaris which has been widely used to treat NASH in traditional Chinese medicine. However, the underlying mechanisms of scoparone against NASH are not yet fully understood, which hinders the development of effective therapeutic agents for NASH. Given the crucial role of glycerophospholipid metabolism in NASH progression, this study aimed to characterize the differential expression of glycerophospholipids that is responsible for scoparone's pharmacological effects and assess its efficacy against NASH. Liquid chromatography-multiple reaction monitoring-mass spectrometry (LC-MRM-MS) was performed to get the concentrations of glycerophospholipids, clarify mechanisms of disease, and highlight insights into drug discovery. Additionally, pathologic findings also presented consistent changes in high-fat diet-induced NASH model, and after scoparone treatment, both the levels of glycerophospholipids and histopathology were similar to normal levels, indicating a beneficial effect during the observation time. Altogether, these results refined the insights on the mechanisms of scoparone against NASH and suggested a route to relieve NASH with glycerophospholipid metabolism. In addition, the current work demonstrated that a pseudotargeted lipidomic platform provided a novel insight into the potential mechanism of scoparone action.


Subject(s)
Coumarins , Glycerophospholipids , Lipidomics , Non-alcoholic Fatty Liver Disease , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Glycerophospholipids/metabolism , Coumarins/pharmacology , Coumarins/therapeutic use , Lipidomics/methods , Mice , Chromatography, Liquid/methods , Male , Disease Models, Animal , Mice, Inbred C57BL , Diet, High-Fat/adverse effects , Mass Spectrometry/methods , Lipid Metabolism/drug effects
20.
Int Immunopharmacol ; 133: 112131, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38669945

ABSTRACT

BACKGROUND: Osthole is a natural active ingredient extracted from the traditional Chinese medicine Cnidium monnieri. It has been demonstrated to have anti-inflammatory, anti-fibrotic, and anti-hyperglycemic properties. However, its effect on diabetic kidney disease (DKD) remains uncertain. This study aims to assess the preventive and therapeutic effects of osthole on DKD and investigate its underlying mechanisms. METHODS: A streptozotocin/high-fat and high-sucrose diet induced Type 2 diabetic rat model was established. Metformin served as the positive drug control. Diabetic rats were treated with metformin or three different doses of osthole for 8 weeks. Throughout the treatment period, the progression of DKD was assessed by monitoring increases in urinary protein, serum creatinine, urea nitrogen, and uric acid, along with scrutinizing kidney pathology. Enzyme-linked immunosorbent assay (ELISA) was employed to detect inflammatory factors and oxidative stress levels. At the same time, immunohistochemical staining was utilized to evaluate changes in alpha-smooth muscle actin, fibronectin, E-cadherin, and apoptosis. The alterations in TGF-ß1/Smads signaling pathway were ascertained through western blot and immunofluorescence. Furthermore, we constructed a high glucose-stimulated HBZY-1 cells model to uncover its molecular protective mechanism. RESULTS: Osthole significantly reduced fasting blood glucose, insulin resistance, serum creatinine, uric acid, blood urea nitrogen, urinary protein excretion, and glomerular mesangial matrix deposition in diabetic rats. Additionally, significant improvements were observed in inflammation, oxidative stress, apoptosis, and fibrosis levels. The increase of ROS, apoptosis and hypertrophy in HBZY-1 cells induced by high glucose was reduced by osthole. Immunofluorescence and western blot results demonstrated that osthole down-regulated the TGF-ß1/Smads signaling pathway and related protein expression. CONCLUSION: Our findings indicate that osthole exhibits potential preventive and therapeutic effects on DKD. It deserves further investigation as a promising drug for preventing and treating DKD.


Subject(s)
Coumarins , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Oxidative Stress , Signal Transduction , Animals , Humans , Male , Rats , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Cell Line , Coumarins/pharmacology , Coumarins/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Inflammation/drug therapy , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL