Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.572
Filter
1.
Mol Cancer ; 23(1): 196, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272040

ABSTRACT

Colitis-associated colorectal cancer (CAC) frequently develops in patients with inflammatory bowel disease (IBD) who have been exposed to a prolonged state of chronic inflammation. The investigation of pharmacological agents and their mechanisms to prevent precancerous lesions and inhibit their progression remains a significant focus and challenge in CAC research. Previous studies have demonstrated that vitexin effectively mitigates CAC, however, its precise mechanism of action warrants further exploration. This study reveals that the absence of the Vitamin D receptor (VDR) accelerates the progression from chronic colitis to colorectal cancer. Our findings indicate that vitexin can specifically target the VDR protein, facilitating its translocation into the cell nucleus to exert transcriptional activity. Additionally, through a co-culture model of macrophages and cancer cells, we observed that vitexin promotes the polarization of macrophages towards the M1 phenotype, a process that is dependent on VDR. Furthermore, ChIP-seq analysis revealed that vitexin regulates the transcriptional activation of phenazine biosynthesis-like domain protein (PBLD) via VDR. ChIP assays and dual luciferase reporter assays were employed to identify the functional PBLD regulatory region, confirming that the VDR/PBLD pathway is critical for vitexin-mediated regulation of macrophage polarization. Finally, in a mouse model with myeloid VDR gene knockout, we found that the protective effects of vitexin were abolished in mid-stage CAC. In summary, our study establishes that vitexin targets VDR and modulates macrophage polarization through the VDR/PBLD pathway, thereby alleviating the transition from chronic colitis to colorectal cancer.


Subject(s)
Apigenin , Colorectal Neoplasms , Macrophages , Receptors, Calcitriol , Apigenin/pharmacology , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/agonists , Receptors, Calcitriol/genetics , Animals , Mice , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Macrophages/metabolism , Macrophages/drug effects , Disease Models, Animal , Colitis/drug therapy , Colitis/pathology , Colitis/metabolism , Colitis/chemically induced , Disease Progression , RAW 264.7 Cells , Mice, Inbred C57BL
2.
Cancer Med ; 13(17): e70171, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39254067

ABSTRACT

PURPOSE: Therapeutic management of colorectal cancer (CRC) does not yet yield promising long-term results. Therefore, there is a need for further investigation of possible therapeutic options. Various experiments have studied the effects of apigenin on CRC and have shown conflicting results. This systematic review and meta-analysis investigates the currently existing evidence on the effect of apigenin on CRC. METHODS: Medline, Embase, Scopus, and Web of Science databases were searched for articles related to apigenin and its effect on CRC in the preclinical setting. Cell viability, growth inhibition, apoptosis, and cell cycle arrest for in-vitro, and body weight, tumor size, and mortality in in-vivo studies were extracted as outcomes. RESULTS: Thirty-nine articles investigating colorectal adenocarcinoma were included in this meta-analysis. Thirty-seven of these studies had data for in vitro experiments, with eight studies having data for in vivo experiments. Six articles had both in vitro and in vivo assessments. Our analysis showed apigenin reduces cell viability and induces growth inhibition, apoptosis, and cell cycle arrest in in vitro studies. The few in vivo studies indicate that apigenin decreases tumor size while showing no effects on the body weight of animal colorectal adenocarcinoma models. CONCLUSION: Our results demonstrated that apigenin, through reducing cell viability, inducing growth inhibition, apoptosis, and cell cycle arrest, and also by decreasing the tumor size, can be considered as a possible adjuvant agent in the management of colorectal adenocarcinoma. However, further in vivo studies are needed before any efforts to translate the current evidence into clinical studies.


Subject(s)
Adenocarcinoma , Apigenin , Apoptosis , Colorectal Neoplasms , Apigenin/pharmacology , Apigenin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Humans , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Apoptosis/drug effects , Animals , Cell Survival/drug effects , Cell Proliferation/drug effects , Cell Cycle Checkpoints/drug effects
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167488, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39218272

ABSTRACT

BACKGROUND: Obesity-related hypertension is a major cardiovascular risk factor. Apigenin, a natural flavonoid in celery, induces vascular dilation via endothelial transient receptor potential channel vanilla 4 (TRPV4) channels. This study aimed to explore apigenin's potential to alleviate obesity-related hypertension in mice and its underlying mechanisms. METHODS: The C57BL/6 and TRPV4 knockout mice were fed a high-fat diet and subjected to dietary intervention with apigenin. Body weight and tail blood pressure of the mice were measured during the feeding. Vascular reactivity was assessed through a DMT wire myograph systems in vitro. The distribution and expression of adiponectin and pro-inflammatory markers in brown fat were detected. Injecting adeno-associated eight (AAV8) viruses into brown adipose tissue (BAT) to determine whether adiponectin is indispensable for the therapeutic effect of apigenin. Palmitic acid (PA) was used in mouse brown adipocytes to examine the detailed mechanisms regulating adiponectin secretion. RESULTS: Apigenin improved vasodilation and reduced blood pressure in obese mice, effects partly blocked in TRPV4 knockout. It also reduced weight gain independently of TRPV4. Apigenin increased adiponectin secretion from BAT; knockdown of adiponectin weakened its benefits. Apigenin downregulated Cluster of differentiation 38 (CD38), restoring Nicotinamide adenine dinucleotide+ (NAD+) levels and activating the NAD+/Sirtuin 1 (SIRT1) pathway, enhancing adiponectin expression. CONCLUSIONS: Our study indicates that dietary apigenin is suitable as a nonpharmaceutical intervention for obesity-related hypertension. In mechanism, in addition to improving vascular relaxation through the activation of endothelial TRPV4 channels, apigenin also directly alleviated adipose inflammation and increased adiponectin levels by inhibiting CD38.


Subject(s)
Adiponectin , Apigenin , Diet, High-Fat , Hypertension , Mice, Inbred C57BL , Mice, Knockout , Obesity , TRPV Cation Channels , Vasodilation , Animals , Adiponectin/metabolism , Adiponectin/genetics , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Obesity/metabolism , Obesity/drug therapy , Obesity/pathology , Apigenin/pharmacology , Mice , Hypertension/metabolism , Hypertension/drug therapy , Hypertension/pathology , Vasodilation/drug effects , Male , Diet, High-Fat/adverse effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Blood Pressure/drug effects
4.
Drug Des Devel Ther ; 18: 3841-3851, 2024.
Article in English | MEDLINE | ID: mdl-39219698

ABSTRACT

Introduction: Apigenin is a natural flavonoid compound with promising potential for the attenuation of myocardial hypertrophy (MH). The compound can also modulate the expression of miR-185-5p that both promote MH and suppress autophagy. The current attempts to explain the anti-MH effect of apigenin by focusing on changes in miR-185-5p-mediated autophagy. Methods: Hypertrophic symptoms were induced in rats using transverse aortic constriction (TAC) method and in cardiomyocytes using Ang II and then handled with apigenin. Changes in myocardial function and structure and cell viability and surface area were measured. The role of miR-185-5p in the anti-MH function of apigenin was explored by detecting changes in autophagic processes and miR-185-5p/SREBP2 axis. Results: TAC surgery induced weight increase, structure destruction, and collagen deposition in hearts of model rats. Ang II suppresses cardiomyocyte viability and increased cell surface area. All these impairments were attenuated by apigenin and were associated with the restored level of autophagy. At the molecular level, the expression of miR-185-5p was up-regulated by TAC, while the expression of SREBP2 was down-regulated, which was reserved by apigenin both in vivo and in vitro. The induction of miR-185-5p in cardiomyocytes could counteracted the protective effects of apigenin. Discussion: Collectively, the findings outlined in the current study highlighted that apigenin showed anti-MH effects. The effects were related to the inhibition of miR-185-5p and activation of SREBP, which contributed to the increased autophagy.


Subject(s)
Apigenin , Autophagy , Cardiomegaly , MicroRNAs , Rats, Sprague-Dawley , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Apigenin/pharmacology , Autophagy/drug effects , Rats , Male , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Cell Survival/drug effects
5.
Nutrients ; 16(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39275332

ABSTRACT

Exceeding a healthy weight significantly elevates the likelihood of developing type 2 diabetes (T2DM). A commercially available singular constituent, available as either purified vitexin or iso-vitexin, has been associated with a decreased risk of T2DM, but its synergistic effect has not been reported yet. Vitexin and iso-vitexin were extracted using an ethanol-based solvent from mung bean seed coat (MBCE) and subsequently purified using preparative liquid chromatography (Prep-LC). Eleven mixture ratios of vitexin and/or iso-vitexin were determined for their antioxidant and antihyperglycemic activities. The 1:1.5 ratio of vitexin to iso-vitexin from MBCE demonstrated the most synergistic effects for enzyme inhibition and glucose uptake in HepG2 cells within an insulin-resistant system, while these ratios exhibited a significantly lower antioxidant capacity than that of each individual component. In a gut model system, the ratio of 1:1.5 (vitexin and iso-vitexin) regulated the gut microbiota composition in overweight individuals by decreasing the growth of Enterobacteriaceae and Enterococcaceae, while increasing in Ruminococcaceae and Lachnospiraceae. The application of vitexin/iso-vitexin for 24 h fermentation enhanced a high variety of abundances of 21 genera resulting in five genera of Parabacteroides, Ruminococcus, Roseburia, Enterocloster, and Peptacetobacter, which belonged to the phylum Firmicutes, exhibiting high abundant changes of more than 5%. Only two genera of Proteus and Butyricicoccus belonging to Proteobacteria and Firmicutes decreased. The findings suggest that these phytochemicals interactions could have synergistic effects in regulating glycemia, through changes in antihyperglycemic activity and in the gut microbiota in overweight individuals. This optimal ratio can be utilized by industries to formulate more potent functional ingredients for functional foods and to create nutraceutical supplements aimed at reducing the risk of T2DM in overweight individuals.


Subject(s)
Apigenin , Gastrointestinal Microbiome , Hypoglycemic Agents , Overweight , Seeds , Vigna , Apigenin/pharmacology , Gastrointestinal Microbiome/drug effects , Humans , Hypoglycemic Agents/pharmacology , Seeds/chemistry , Male , Hep G2 Cells , Diabetes Mellitus, Type 2 , Antioxidants/pharmacology , Plant Extracts/pharmacology , Female
6.
Ther Deliv ; 15(9): 717-735, 2024.
Article in English | MEDLINE | ID: mdl-39259258

ABSTRACT

Apigenin, a potent natural flavonoid, has emerged as a key therapeutic agent due to its multifaceted medicinal properties in combating various diseases. However, apigenin's clinical utility is greatly limited by its poor water solubility, low bioavailability and stability issues. To address these challenges, this review paper explores the innovative field of nanotechnology-based delivery systems, which have shown significant promise in improving the delivery and effectiveness of apigenin. This paper also explores the synergistic potential of co-delivering apigenin with conventional therapeutic agents. Despite the advantageous properties of these nanoformulations, critical challenges such as scalable production, regulatory approvals and comprehensive long-term safety assessments remain key hurdles in their clinical adoption which must be addressed for commercialization of apigenin-based formulations.


Apigenin is a natural substance found in plants that might help treat illnesses like cancer, diabetes, heart problems and brain disorders. But it doesn't work very well because it doesn't dissolve in water, is hard for the body to use and isn't very stable. To fix this, scientists are putting apigenin inside tiny carriers called nanocarriers. These tiny carriers help apigenin dissolve better, be absorbed by the body more easily and work better.There are different kinds of nanocarriers, like tiny fat bubbles, tiny solid particles and tiny gels. These can be made to target specific parts of the body, which helps reduce side effects. Apigenin can also be mixed with other medicines in these carriers to work even better.However, there are big challenges in making these treatments widely available, like making enough of them, getting permission from health authorities and making sure they are safe for a long time. This review talks about the latest progress and future possibilities in using nanotechnology to deliver apigenin, aiming to make it better for treating diseases.


Subject(s)
Apigenin , Apigenin/administration & dosage , Apigenin/chemistry , Apigenin/pharmacokinetics , Apigenin/pharmacology , Humans , Animals , Biological Availability , Solubility , Drug Delivery Systems/methods , Nanoparticles/chemistry , Nanotechnology/methods , Nanoparticle Drug Delivery System/chemistry
7.
Microb Pathog ; 195: 106851, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39197693

ABSTRACT

Mutations in glucokinase (GCK) can either enhance or inhibit insulin secretion, leading to different forms of diabetes, including gestational diabetes. While many glucokinase activators (GKAs) have been explored as treatments, their long-term effectiveness has often been unsatisfactory. However, recent interest has surged with the introduction of dorzagliatin and TTP399. This study investigates the efficacy of four previously studied compounds (Swertiamarin, Apigenin, Mangiferin, and Tatanan A) in activating GCK using computational methods. Initial molecular docking revealed binding affinities ranging from -6.7 to -8.6 kcal/mol. The compounds were then evaluated for drug-likeness and pharmacokinetic properties. Re-docking studies were performed for validation. Based on their favorable binding affinities and compliance with Lipinski's rule and ADMET criteria, three compounds (Swertiamarin, Apigenin, and Tatanan A) were selected for molecular dynamics (MD) simulations. MD simulations demonstrated that Swertiamarin showed excellent stability, as indicated by analyses of RMSD, RMSF, radius of gyration (Rg), hydrogen bonding, and principal component analysis (PCA). These results suggest that Swertiamarin holds promise for further investigation in in vivo and clinical settings to evaluate its potential in enhancing GCK activity and treating diabetes. This study assessed the potential of four compounds as GCK activators using molecular docking, pharmacokinetic profiling, and MD simulations. Swertiamarin, in particular, showed significant stability and adherence to drug-likeness criteria, making it a promising candidate for further research in combating diabetes.


Subject(s)
Glucokinase , Molecular Docking Simulation , Molecular Dynamics Simulation , Glucokinase/metabolism , Glucokinase/chemistry , Glucokinase/genetics , Humans , Medicine, Chinese Traditional , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry , Hydrogen Bonding , Enzyme Activators/pharmacology , Enzyme Activators/chemistry , Computer Simulation , Apigenin/pharmacology , Apigenin/chemistry
8.
Eur J Pharmacol ; 981: 176848, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39094925

ABSTRACT

Neuropathic pain (NP) is associated with astrocytes activation induced by nerve injury. Reactive astrocytes, strongly induced by central nervous system damage, can be classified into A1 and A2 types. Vitexin, a renowned flavonoid compound, is known for its anti-inflammatory and analgesic properties. However, its role in NP remains unexplored. This study aims to investigate the effects of vitexin on astrocyte polarization and its underlying mechanisms. A mouse model of NP was established, and primary astrocytes were stimulated with sphingosine-1-phosphate (S1P) to construct a cellular model. The results demonstrated significant activation of spinal astrocytes on days 14 and 21. Concurrently, reactive astrocytes predominantly differentiated into the A1 type. Western blot analysis revealed an increase in A1 astrocyte-associated protein (C3) and a decrease in A2 astrocyte-associated protein (S100A10). Serum S1P levels increased on days 14 and 21, alongside a significant upregulation of Sphingosine-1-phosphate receptor 1 (S1PR1) mRNA expression and elevated expression of chemokines. In vitro, stimulation with S1P inhibited the Phosphatidylinositol 3-kinase and protein kinase B (PI3K/Akt) signaling pathway and autophagy flux, promoting polarization of astrocytes towards the A1 phenotype while suppressing the polarization of A2 astrocytes. Our findings suggest that vitexin, acting on astrocytes but not microglia, attenuates S1P-induced downregulation of PI3K/Akt signaling, restores autophagy flux in astrocytes, regulates A1/A2 astrocyte ratio, and reduces chemokine and S1P secretion, thereby alleviating neuropathic pain caused by nerve injury.


Subject(s)
Apigenin , Astrocytes , Autophagy , Lysophospholipids , Neuralgia , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Sphingosine-1-Phosphate Receptors , Sphingosine , Animals , Apigenin/pharmacology , Apigenin/therapeutic use , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Neuralgia/drug therapy , Neuralgia/metabolism , Neuralgia/pathology , Sphingosine-1-Phosphate Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Signal Transduction/drug effects , Autophagy/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Male , Mice, Inbred C57BL , Cell Polarity/drug effects
9.
Bioorg Chem ; 151: 107704, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39126870

ABSTRACT

A series of scutellarein 7-l-amino acid carbamate-4'-cycloalkylamine propyl ether conjugates were designed and synthesized for the first time as multifunctional agents for Alzheimer's disease (AD) therapy. The designed compounds exhibited more balanced and effective multi-target potency. Among them, compound 11l, l-Valine carbamate derivative of scutellarein cycloheptylamine ether, exhibited the most potent inhibition of electric eel AChE enzymes and human AChE enzymes, with an IC50 values of 7.04 µM and 9.73 µM, respectively. Moreover, 11l exhibited more potent H3R antagonistic activities than clobenpropit, with an IC50 value of 1.09 nM. Compound 11l not only displayed excellent inhibition of self- and Cu2+-induced Aß1-42 aggregation (95.48 % and 88.63 % inhibition, respectively) but also induced the disassembly of self- and Cu2+-induced Aß fibrils (80.16 % and 89.30 % disaggregation, respectively). Moreover, 11l significantly reduced tau protein hyperphosphorylation induced by Aß25-35. It exhibited effective antioxidant activity and neuroprotective potency, and inhibited RSL3-induced PC12 cell ferroptosis. Assays of hCMEC/D3 and hPepT1-MDCK cell line permeability indicated that 11l would have optimal blood-brain barrier permeability and intestinal absorption characteristics. In addition, in vivo studies revealed that compound 11l significantly attenuated learning and memory impairment in an AD mouse model. Finally, a pharmacokinetic characterization of 11l indicated favorable druggability and pharmacokinetic properties. Taken together, our results suggest that 11l is a potential candidate for AD treatment and merits further investigation.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Apigenin , Cholinesterase Inhibitors , Histamine H3 Antagonists , Receptors, Histamine H3 , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/chemical synthesis , Humans , Animals , Structure-Activity Relationship , Acetylcholinesterase/metabolism , Mice , Histamine H3 Antagonists/pharmacology , Histamine H3 Antagonists/chemistry , Histamine H3 Antagonists/chemical synthesis , Ligands , Apigenin/pharmacology , Apigenin/chemistry , Apigenin/chemical synthesis , Receptors, Histamine H3/metabolism , Molecular Structure , Dose-Response Relationship, Drug , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/chemical synthesis , Electrophorus , Rats , Peptide Fragments/metabolism , Male , PC12 Cells
10.
Chem Pharm Bull (Tokyo) ; 72(8): 751-761, 2024.
Article in English | MEDLINE | ID: mdl-39143008

ABSTRACT

Gout is the second largest metabolic disease worldwide after diabetes, with acute gouty arthritis as most common symptom. Xanthine oxidase (XOD) and the NOD like receptor-3 (NLRP3) inflammasome are the key targets for acute gout treatment. Chlorogenic acid has been reported with a good anti-inflammatory activity, and Apigenin showed an excellent potential in XOD inhibition. Therefore, a series of chlorogenic acid-apigenin (CA) conjugates with varying linkers were designed and synthesized as dual XOD/NLRP3 inhibitors, and their activities both in XOD and NLRP3 inhibition were evaluated. An in vitro study of XOD inhibitory activity revealed that the majority of CA conjugates exhibited favorable XOD inhibitory activity. Particularly, the effects of compounds 10c and 10d, with an alkyl linker on the apigenin moiety, were stronger than that of allopurinol. The selected CA conjugates also demonstrated a favorable anti-inflammatory activity in RAW264.7 cells. Furthermore, compound 10d, which showed the optimal activity both in XOD inhibition and anti-inflammatory, was chosen and its inhibitory ability on NLRP3 and related proinflammatory cytokines was further tested. Compound 10d effectively reduced NLRP3 expression and the secretion of interluekin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) with an activity stronger than the positive control isoliquiritigenin (ISL). Based on these findings, compound 10d exhibits dual XOD/NLRP3 inhibitory activity and, therefore, the therapeutic effects on acute gout is worthy of further study.


Subject(s)
Apigenin , Chlorogenic Acid , Gout Suppressants , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Mice , Apigenin/pharmacology , Apigenin/chemistry , Apigenin/chemical synthesis , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RAW 264.7 Cells , Chlorogenic Acid/pharmacology , Chlorogenic Acid/chemistry , Chlorogenic Acid/chemical synthesis , Gout Suppressants/pharmacology , Gout Suppressants/chemical synthesis , Gout Suppressants/chemistry , Gout Suppressants/therapeutic use , Structure-Activity Relationship , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism , Molecular Structure , Gout/drug therapy , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry
11.
Arch Environ Occup Health ; 79(3-4): 143-151, 2024.
Article in English | MEDLINE | ID: mdl-39169800

ABSTRACT

Petrol vapors as important occupational and environmental pollutants can cause oxidative stress and may play a role in the development of neurodegenerative diseases along with the risk factors involved. This research is designed as a preliminary study to evaluate the protective effects of apigenin (APG) on oxidative stress caused by petrol vapors inhalation in rats. A total of 24 male Wistar rats were randomly divided into four groups inside the inhalation chamber. Body weight changes and oxidative stress markers were investigated. The average body weight of the group exposed to petrol vapors was significantly lower compared to the other groups. The level of reactive oxygen species (ROS), content of oxidized-glutathione (GSSG), and Malondialdehyde were found to be higher in the petrol-inhaled group, while the content of reduced-glutathione (GSH) was lower compared to the other groups. APG administration did result in any significant improvement in these toxicities induced by petrol vapor. APG administration may ameliorate the petrol-induced oxidative stress. In chronic exposures, in addition to personal protection and engineering control, the use of compounds of natural origin may help in reducing the side effects (such as CNS) caused by exposure to petrol vapors.


Subject(s)
Apigenin , Oxidative Stress , Rats, Wistar , Reactive Oxygen Species , Animals , Oxidative Stress/drug effects , Male , Rats , Reactive Oxygen Species/metabolism , Apigenin/pharmacology , Glutathione/metabolism , Gasoline/toxicity , Environmental Pollutants/toxicity , Malondialdehyde/metabolism , Inhalation Exposure/adverse effects , Body Weight/drug effects , Air Pollutants, Occupational
12.
Kidney Blood Press Res ; 49(1): 753-762, 2024.
Article in English | MEDLINE | ID: mdl-39079512

ABSTRACT

INTRODUCTION: Vitexin is a natural flavonoid compound extracted from Vitex leaves or seeds, exhibiting various pharmacological activities including anticancer, antihypertensive, anti-inflammatory, and spasmolytic effects. However, its protective effects on hypertensive nephropathy (HN) and the underlying mechanisms remain unclear. METHODS: Spontaneous hypertension rats were fed a high-sugar and high-fat diet for 8 weeks to induce the disease HN model. From the 5th week, the rats were administered vitexin via gavage. Blood pressure was measured biweekly using the tail-cuff method. Histopathological changes were assessed using HE staining, and biochemical analyses were performed to evaluate the effects of vitexin on HN rats. The underlying mechanisms of vitexin treatment were investigated through western blotting. RESULTS: The data demonstrated that vitexin significantly lowered systolic, diastolic, and mean arterial pressures and ameliorated histopathological changes in HN rats. Biochemical analyses revealed that vitexin reduced the levels of creatinine (Cr), blood urea nitrogen (BUN), total cholesterol (TC), triglycerides (TG), total protein (TP), low-density lipoprotein cholesterol (LDL-C), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), malondialdehyde (MDA), and advanced glycation end products (AGEs), while increasing the levels of albumin (ALB) and superoxide dismutase (SOD). Western blotting results indicated that vitexin treatment decreased the expression of TNF-α, IL-6, and nuclear factor kappa-B (NF-κB), while increasing the expression of SOD. CONCLUSION: The findings of this study suggest that vitexin exerts protective effects against HN, providing pharmacological evidence for its potential use in HN treatment.


Subject(s)
Apigenin , Hypertension, Renal , Animals , Apigenin/pharmacology , Apigenin/therapeutic use , Rats , Hypertension, Renal/drug therapy , Hypertension, Renal/pathology , Male , Rats, Inbred SHR , Nephritis/drug therapy , Nephritis/prevention & control , Nephritis/pathology , Blood Pressure/drug effects , NF-kappa B/metabolism
13.
J Pharm Biomed Anal ; 248: 116325, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38959755

ABSTRACT

The high prevalence of cancer and detrimental side effects associated with many cancer treatments necessitate the search for effective alternative therapies. Natural products are increasingly being recognized and investigated for their potential therapeutic benefits. Scutellaria barbata D. Don (SBD), a plant with potent antitumor properties, has attracted significant interest from oncology researchers. Its primary flavonoid components-scutellarin and luteolin-which have limited oral bioavailability due to poor absorption. This hinders its application for cancer treatment. The gut microbiota, which is considered a metabolic organ, can modulate the biotransformation of compounds, thereby altering their bioavailability and efficacy. In this study, we employed liquid chromatography tandem mass spectrometry (LC-MS/MS 8060) and ion trap-time of flight (LC-MSn-IT-TOF) analysis to investigate the ex vivo metabolism of scutellarin and luteolin by the gut microbiota. Five metabolites and one potential metabolite were identified. We summarized previous studies on their antitumor effects and performed in vitro tumor cell line studies to prove their antitumor activities. The possible key pathway of gut microbiota metabolism in vitro was validated using molecular docking and pure enzyme metabolic experiments. In addition, we explored the antitumor mechanisms of the two components of SBD through network pharmacology, providing a basis for subsequent target identification. These findings expand our understanding of the antitumor mechanisms of SBD. Notably, this study contributes to the existing body of knowledge regarding flavonoid biotransformation by the gut microbiota, highlighting the therapeutic potential of SBD in cancer treatment. Moreover, our results provide a theoretical basis for future in vivo pharmacokinetic studies, aiming to optimize the clinical efficacy of SBD in oncological applications.


Subject(s)
Apigenin , Gastrointestinal Microbiome , Glucuronates , Luteolin , Scutellaria , Tandem Mass Spectrometry , Gastrointestinal Microbiome/drug effects , Luteolin/pharmacology , Luteolin/metabolism , Luteolin/pharmacokinetics , Scutellaria/chemistry , Apigenin/pharmacology , Glucuronates/metabolism , Humans , Tandem Mass Spectrometry/methods , Cell Line, Tumor , Animals , Molecular Docking Simulation , Plant Extracts/pharmacology , Chromatography, Liquid/methods , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biological Availability , Male , Biotransformation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics
14.
J Ethnopharmacol ; 334: 118518, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38964628

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Salvia miltiorrhiza Bunge (S. miltiorrhiza) is an important Traditional Chinese herbal Medicine (TCM) used to treat cardio-cerebrovascular diseases. Based on the pharmacodynamic substance of S. miltiorrhiza, the aim of present study was to investigate the underlying mechanism of S. miltiorrhiza against cardiac fibrosis (CF) through a systematic network pharmacology approach, molecular docking and dynamics simulation as well as experimental investigation in vitro. MATERIALS AND METHODS: A systematic pharmacological analysis was conducted using the Traditional Chinese Medicine Pharmacology (TCMSP) database to screen the effective chemical components of S. miltiorrhiza, then the corresponding potential target genes of the compounds were obtained by the Swiss Target Prediction and TCMSP databases. Meanwhile, GeneCards, DisGeNET, OMIM, and TTD disease databases were used to screen CF targets, and a protein-protein interaction (PPI) network of drug-disease targets was constructed on S. miltiorrhiza/CF targets by Search Tool for the Retrieval of Interacting Genes/Proteins (STING) database. After that, the component-disease-target network was constructed by software Cytoscape 3.7. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed for the intersection targets between drug and disease. The relationship between active ingredient of S. miltiorrhiza and disease targets of CF was assessed via molecular docking and molecular dynamics simulation. Subsequently, the underlying mechanism of the hub compound on CF was experimentally investigated in vitro. RESULTS: 206 corresponding targets to effective chemical components from S. miltiorrhiza were determined, and among them, there were 82 targets that overlapped with targets of CF. Further, through PPI analysis, AKT1 and GSK3ß were the hub targets, and which were both enriched in the PI3K/AKT signaling pathway, it was the sub-pathways of the lipid and atherosclerosis pathway. Subsequently, compound-disease-genes-pathways diagram is constructed, apigenin (APi) was a top ingredients and AKT1 (51) and GSK3ß (22) were the hub genes according to the degree value. The results of molecular docking and dynamics simulation showed that APi has strong affinities with AKT and GSK3ß. The results of cell experiments showed that APi inhibited cells viability, proliferation, proteins expression of α-SMA and collagen I/III, phosphorylation of AKT1 and GSK3ß in MCFs induced by TGFß1. CONCLUSION: Through a systematic network pharmacology approach, molecular docking and dynamics simulation, and confirmed by in vitro cell experiments, these results indicated that APi interacts with AKT and GSK3ß to disrupt the phosphorylation of AKT and GSK3ß, thereby inhibiting the proliferation and differentiation of MCFs induced by TGFß1, which providing new insights into the pharmacological mechanism of S. miltiorrhiza in the treatment of CF.


Subject(s)
Apigenin , Cell Differentiation , Cell Proliferation , Glycogen Synthase Kinase 3 beta , Molecular Docking Simulation , Proto-Oncogene Proteins c-akt , Signal Transduction , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Cell Proliferation/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Animals , Apigenin/pharmacology , Apigenin/chemistry , Cell Differentiation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Protein Interaction Maps , Rats , Network Pharmacology , Molecular Dynamics Simulation , Cell Line , Humans
15.
Int Immunopharmacol ; 139: 112710, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39029229

ABSTRACT

PANoptosis is manifested with simultaneous activation of biomarkers for both pyroptotic, apoptotic and necroptotic signaling via the molecular platform PANoptosome and it is involved in pathologies of various inflammatory diseases including hemophagocytic lymphohistiocytosis (HLH). Scutellarin is a flavonoid isolated from herbal Erigeron breviscapus (Vant.) Hand.-Mazz. and has been shown to possess multiple pharmacological effects, but it is unknown whether scutellarin has any effects on PANoptosis and related inflammatory diseases. In this study, we found that scutellarin inhibited cell death in bone marrow-derived macrophages (BMDMs) and J774A.1 cells treated with TGF-ß-activated kinase 1 (TAK1) inhibitor 5Z-7-oxozeaenol (OXO) plus lipopolysaccharide (LPS), which has been commonly used to induce PANoptosis. Western blotting showed that scutellarin dose-dependently inhibited the activation biomarkers for pyroptotic (Caspase-1p10 and GSDMD-NT), apoptotic (cleaved Casp3/8/9 and GSDME-NT), and necroptotic (phosphorylated MLKL) signaling. The inhibitory effect of scutellarin was unaffected by NLRP3 or Caspase-1 deletion. Interestingly, scutellarin blocked the assembly of PANoptosome that encompasses ASC, RIPK3, Caspase-8 and ZBP1, suggesting its action on upstream signaling. Consistent with this, scutellarin inhibited mitochondrial damage and mitochondrial reactive oxygen species (mtROS) generation in cells treated with OXO+LPS. Further, mito-TEMPO that can scavenge mtROS significantly inhibited OXO+LPS-induced PANoptotic cell death. In line with the in vitro results, scutellarin markedly alleviated systemic inflammation, multiple organ injury, and activation of PANoptotic biomarkers in mice with HLH. Collectively, our data suggest that scutellarin can inhibit PANoptosis by suppressing mitochondrial damage and mtROS generation and thereby mitigating multiple organ injury in mice with inflammatory disorders.


Subject(s)
Apigenin , Glucuronates , Lipopolysaccharides , Mice, Inbred C57BL , Mitochondria , Reactive Oxygen Species , Apigenin/pharmacology , Apigenin/therapeutic use , Glucuronates/pharmacology , Glucuronates/therapeutic use , Animals , Reactive Oxygen Species/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mice , Cell Line , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Necroptosis/drug effects , Male , MAP Kinase Kinase Kinases/metabolism , Inflammation/drug therapy , Signal Transduction/drug effects , Zearalenone/administration & dosage , Lactones , Resorcinols
16.
Eur J Pharmacol ; 980: 176865, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39084453

ABSTRACT

Vitexin is a natural flavonoid glycoside compound extracted from the leaves and seeds of Vitex negundo. It is widely distributed in the leaves and stems of numerous plants and exhibites remarkable anti-tumor, anti-inflammatory, and anti-hypertensive properties. However, whether vitexin presents the anti-aging and senescence prevention effect has not been fully elucidated. The purpose of this study is to investigate the effect of vitexin on progeria mice and cellular senescence, as well as its underlying molecular mechanisms. To generate a premature aging/senescence model in vivo and in vitro, we used D-galactose (D-gal), hydrogen peroxide (H2O2), and adriamycin (ADR), respectively. Our findings demonstrated that vitexin potentially delays D-gal-induced progeria mice; similar effects were observed in stress-induced premature senescent fibroblasts in culture. Interestingly, this effect of vitexin is closely correlated with the reduction of the senescence-associated secretory phenotype (SASP) and the inhibition of the SASP-related JAK2/STAT3 pathway. Furthermore, we determined that vitexin meets the pharmacological parameters using the freely available ADMET web tool. Collectively, our findings demonstrate that vitexin possesses anti-senescence and anti-aging properties due to the inhibition of SASP and suppression of JAK2/STAT3 signaling pathway.


Subject(s)
Apigenin , Cellular Senescence , Galactose , Janus Kinase 2 , Progeria , STAT3 Transcription Factor , Animals , Apigenin/pharmacology , Apigenin/therapeutic use , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Cellular Senescence/drug effects , Mice , Progeria/drug therapy , Progeria/pathology , Progeria/metabolism , Signal Transduction/drug effects , Male , Aging, Premature/chemically induced , Aging, Premature/drug therapy , Aging, Premature/metabolism , Aging, Premature/pathology , Disease Models, Animal , Senescence-Associated Secretory Phenotype/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism
17.
Int J Mol Sci ; 25(14)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39062932

ABSTRACT

Flavonoids, a class of natural compounds with anticancer activity, exhibit varying biological activities and potencies based on their structural differences. Acylation, including acetylation of flavonoids, generally increases their structural diversity, which is closely related to the diversity of bioactivity within this group of compounds. However, it remains largely unknown how acetylation affects the bioactivity of many flavonoids. Based on our previous findings that O-acetylation enhances quercetin's bioactivity against various cancer cells, we synthesized 12 acetylated flavonoids, including seven novel compounds, to investigate their anticancer activities in the MDA-MB-231, HCT-116, and HepG2 cell lines. Our results showed that acetylation notably enhanced the cell proliferation inhibitory effect of quercetin and kaempferol across all cancer cell lines tested. Interestingly, while the 5,7,4'-O-triacetate apigenin (3Ac-A) did not show an enhanced the effect of inhibition of cell proliferation through acetylation, it exhibited significantly strong anti-migration activity in MDA-MB-231 cells. In contrast, the 7,4'-O-diacetate apigenin (2Ac-Q), which lacks acetylation at the 5-position hydroxy group, showed enhanced cell proliferation inhibitory effect but had weaker anti-migration effects compared to 3Ac-A. These results indicated that acetylated flavonoids, especially quercetin, kaempferol, and apigenin derivatives, are promising for anticancer applications, with 3Ac-A potentially having unique anti-migration pathways independent of apoptosis induction. This study highlights the potential application of flavonoids in novel chemopreventive strategies for their anti-cancer activity.


Subject(s)
Cell Proliferation , Flavonoids , Humans , Acetylation/drug effects , Flavonoids/pharmacology , Flavonoids/chemistry , Cell Proliferation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Quercetin/pharmacology , Quercetin/chemistry , Kaempferols/pharmacology , Kaempferols/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Hep G2 Cells , Apigenin/pharmacology , Apigenin/chemistry
18.
Biomed Pharmacother ; 177: 117075, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964181

ABSTRACT

Obesity is a growing epidemic among reproductive-age men, which can cause and exacerbate male infertility by means of associated comorbidities, endocrine abnormalities, and direct effects on the fidelity and throughput of spermatogenesis. A prominent consequence of male obesity is a reduction in testosterone levels. Natural products have shown tremendous potential anti-obesity effects in metabolic diseases. This study aimed to investigate the potential of apigenin (AP) to alleviate testicular dysfunction induced by a high-fat diet (HFD) and to investigate the underlying mechanisms, focusing on endoplasmic reticulum stress (ERS) and testosterone synthesis. A murine model of obesity was established using HFD-fed mice. The effects of AP on obesity, lipid metabolism, testicular dysfunction, and ERS were assessed through various physiological, histological, and molecular techniques. Administration of AP (10 mg/kg) ameliorated HFD-induced obesity and testicular dysfunction in a mouse model, as evidenced by decreased body weight, improved lipid profiles and testicular pathology, and restored protein levels related to testosterone. Furthermore, in vitro studies demonstrated that AP relieved ERS and recovered testosterone synthesis in murine Leydig cells (TM3) treated with free fatty acids (FFAs). It was also observed that AP rescued testosterone synthesis enzymes in TM3 cells, similar to that observed with the inhibitor of the PERK pathway (GSK2606414). In addition, ChIP, qPCR, and gene silencing showed that the C/EBP homologous protein (CHOP) bound directly to the promoter region of steroidogenic STAR and negatively modulated its expression. Collectively, AP has remarkable potential to alleviate HFD-induced obesity and testicular dysfunction. Its protective effects are attributable partly to mitigating ERS and restoring testosterone synthesis in Leydig cells.


Subject(s)
Apigenin , Diet, High-Fat , Endoplasmic Reticulum Stress , Leydig Cells , Mice, Inbred C57BL , Obesity , Testis , Testosterone , Animals , Male , Endoplasmic Reticulum Stress/drug effects , Apigenin/pharmacology , Mice , Diet, High-Fat/adverse effects , Obesity/drug therapy , Obesity/metabolism , Leydig Cells/drug effects , Leydig Cells/metabolism , Testis/drug effects , Testis/metabolism , Testis/pathology , Cell Line , Lipid Metabolism/drug effects
19.
Exp Cell Res ; 441(1): 114150, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38971519

ABSTRACT

Despite significant advances in the treatment of colorectal cancer (CRC), identification of novel targets and treatment options are imperative for improving its prognosis and survival rates. The mitochondrial SIRT3 and SHMT2 have key roles in metabolic reprogramming and cell proliferation. This study investigated the potential use of the natural product apigenin in CRC treatment employing both in vivo and in vitro models and explored the role of SIRT3 and SHMT2 in apigenin-induced CRC apoptosis. The role of SHMT2 in CRC patients' survival was verified using TCGA database. In vivo, apigenin treatment restored the normal colon appearance. On the molecular level, apigenin augmented the immunohistochemical expression of cleaved caspase-3 and attenuated SIRT3 and SHMT2 mRNA expression CRC patients with decreased SHMT2 expression had improved overall and disease-free survival rates. In vitro, apigenin reduced the cell viability in a time-dependent manner, induced G0/G1 cell cycle arrest, and increased the apoptotic cell population compared to the untreated control. Mechanistically, apigenin treatment mitigated the expression of SHMT2, SIRT3, and its upstream long intergenic noncoding RNA LINC01234 in CRC cells. Conclusively, apigenin induces caspase-3-dependent apoptosis in CRC through modulation of SIRT3-triggered mitochondrial pathway suggesting it as a promising therapeutic agent to improve patient outcomes.


Subject(s)
Apigenin , Apoptosis , Cell Proliferation , Colorectal Neoplasms , Sirtuin 3 , Apigenin/pharmacology , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Sirtuin 3/metabolism , Sirtuin 3/genetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Animals , Mice , Gene Expression Regulation, Neoplastic/drug effects , Mice, Nude , Cell Line, Tumor , Signal Transduction/drug effects , Cell Survival/drug effects , Xenograft Model Antitumor Assays , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Glycine Hydroxymethyltransferase
20.
Eur J Pharmacol ; 978: 176800, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38950835

ABSTRACT

Adiponectin plays key roles in energy metabolism and ameliorates inflammation, oxidative stress, and mitochondrial dysfunction via its primary receptors, adiponectin receptors -1 and 2 (AdipoR1 and AdipoR2). Systemic depletion of adiponectin causes various metabolic disorders, including MASLD; however adiponectin supplementation is not yet achievable owing to its large size and oligomerization-associated complexities. Small-molecule AdipoR agonists, thus, may provide viable therapeutic options against metabolic disorders. Using a novel luciferase reporter-based assay here, we have identified Apigenin-6-C-glucoside (ACG), but not apigenin, as a specific agonist for the liver-rich AdipoR isoform, AdipoR2 (EC50: 384 pM) with >10000X preference over AdipoR1. Immunoblot analysis in HEK-293 overexpressing AdipoR2 or HepG2 and PLC/PRF/5 liver cell lines revealed rapid AMPK, p38 activation and induction of typical AdipoR targets PGC-1α and PPARα by ACG at a pharmacologically relevant concentration of 100 nM (reported cMax in mouse; 297 nM). ACG-mediated AdipoR2 activation culminated in a favorable modulation of key metabolic events, including decreased inflammation, oxidative stress, mitochondrial dysfunction, de novo lipogenesis, and increased fatty acid ß-oxidation as determined by immunoblotting, QRT-PCR and extracellular flux analysis. AdipoR2 depletion or AMPK/p38 inhibition dampened these effects. The in vitro results were recapitulated in two different murine models of MASLD, where ACG at 10 mg/kg body weight robustly reduced hepatic steatosis, fibrosis, proinflammatory macrophage numbers, and increased hepatic glycogen content. Together, using in vitro experiments and rodent models, we demonstrate a proof-of-concept for AdipoR2 as a therapeutic target for MASLD and provide novel chemicobiological insights for the generation of translation-worthy pharmacological agents.


Subject(s)
Apigenin , Glucosides , Receptors, Adiponectin , Receptors, Adiponectin/agonists , Receptors, Adiponectin/metabolism , Animals , Humans , Mice , Apigenin/pharmacology , Apigenin/therapeutic use , Glucosides/pharmacology , Glucosides/therapeutic use , Male , Hep G2 Cells , HEK293 Cells , Disease Models, Animal , Mice, Inbred C57BL , Oxidative Stress/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , AMP-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL