Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.099
Filter
1.
J Cell Mol Med ; 28(14): e18565, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39044287

ABSTRACT

Cisplatin (CIS) is a platinum-derived chemotherapeutic agent commonly utilized in the treatment of various malignant tumours. However, anticancer doses of the drug cause serious damage to the brain. This study aimed to determine the potential protective effects of tangeretin, which has antioxidant and anti-inflammatory properties, in cisplatin-induced neurotoxicity on BALB/c mice brains. Male BALB/c mice were randomized and separated into four groups. Tangeretin was given for 10 days by gavage. CIS was injected as a single dose of 10 mg/kg intraperitoneally (ip) on the 10th day. Brain tissues, malondialdehyde (MDA), total glutathione (tGSH), glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT) and nitric oxide (NO) levels were measured to determine oxidative damage and myeloperoxidase, tumour necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1ß), IL-6 and IL-10 were measured to determine inflammatory activity. In addition, 8-OHdG and caspase-3 were analysed by immunofluorescence methods. While CIS administration remarkably elevated reactive oxygen species, MDA, and NO levels in brain tissue compared to the control, tGSH, GPx, SOD and CAT levels were significantly decreased. Also, it has been detected that TNF-α, IL-1ß and IL-6 obtained in CIS-treated groups increased as well as IL-10 decreased, thereby elevating the inflammatory response. In addition, 8-OHdG and caspase-3 immunoreactivity in neurons increased with CIS administration. Treatment with tangeretin ameliorated the deterioration in oxidant/antioxidant status, overpowered neuroinflammation and ameliorated neurotoxicity-induced apoptosis. This study shows that tangeretin has beneficial effects on CIS-induced neurodegeneration. Possible mechanisms underlying these beneficial effects include the antioxidant and anti-inflammatory properties of tangeretin.


Subject(s)
Brain , Cisplatin , Flavones , Mice, Inbred BALB C , Oxidative Stress , Animals , Cisplatin/adverse effects , Cisplatin/pharmacology , Male , Oxidative Stress/drug effects , Brain/metabolism , Brain/drug effects , Brain/pathology , Flavones/pharmacology , Antioxidants/pharmacology , Antioxidants/metabolism , Mice , Rats , Reactive Oxygen Species/metabolism , Anti-Inflammatory Agents/pharmacology , Malondialdehyde/metabolism , Glutathione Peroxidase/metabolism , Nitric Oxide/metabolism , Superoxide Dismutase/metabolism , Cytokines/metabolism , Glutathione/metabolism
2.
Mol Med ; 30(1): 113, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095693

ABSTRACT

BACKGROUND: To explore whether nobiletin has a protective effect on high-fat diet (HFD)-induced enteric nerve injury and its underlying mechanism. METHODS: An obesity model was induced by a HFD. Nobiletin (100 mg/kg and 200 mg/kg) and vehicle were administered by gastric gavage for 4 weeks. Lee's index, body weight, OGTT and intestinal propulsion assays were performed before sacrifice. After sampling, lipids were detected using Bodipy 493/503; lipid peroxidation was detected using MDA and SOD kits and the expression of PGP 9.5, Trem2, GFAP, ß-tubulin 3, Bax, Bcl2, Nestin, P75 NTR, SOX10 and EDU was detected using immunofluorescence. The GDNF, p-AKT, AKT, p-FOXO3a, FOXO3a and P21 proteins were detected using western blotting. The relative mRNA expression levels of NOS2 were detected via qPCR. Primary enteric neural stem cells (ENSCs) were cultured. After ENSCs were treated with palmitic acid (PA) and nobiletin, CCK-8 and caspase-3/7 activity assays were performed to evaluate proliferation and apoptosis. RESULTS: HFD consumption caused colon lipid accumulation and peroxidation, induced enteric nerve damage and caused intestinal motor dysfunction. However, nobiletin reduced lipid accumulation and peroxidation in the colon; promoted Trem2, ß-tubulin 3, Nestin, P75NTR, SOX10 and Bcl2 expression; inhibited Bax and GFAP expression; reduced NOS2 mRNA transcription; and regulated the GDNF/AKT/FOXO3a/P21 pathway. Nobiletin also promoted PA-induced impairment of ENSCs. CONCLUSIONS: Nobiletin restored HFD-induced enteric nerve injury, which may be associated with inhibiting enteric nerve apoptosis, promoting enteric nerve survival and regulating the GDNF/AKT/FOXO3a/P21 pathway.


Subject(s)
Diet, High-Fat , Enteric Nervous System , Flavones , Forkhead Box Protein O3 , Glial Cell Line-Derived Neurotrophic Factor , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Forkhead Box Protein O3/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Diet, High-Fat/adverse effects , Signal Transduction/drug effects , Male , Flavones/pharmacology , Flavones/therapeutic use , Enteric Nervous System/metabolism , Enteric Nervous System/drug effects , Neuroglia/metabolism , Neuroglia/drug effects , Mice , Disease Models, Animal , Rats , Obesity/metabolism , Obesity/drug therapy , Apoptosis/drug effects
3.
Biochem Biophys Res Commun ; 724: 150217, 2024 09 10.
Article in English | MEDLINE | ID: mdl-38865809

ABSTRACT

Neuropathy is a disturbance of function or a pathological change in nerves causing poor health and quality of life. A proportion of chronic pain patients in the community suffer persistent neuropathic pain symptoms because current drug therapies may be suboptimal so there is a need for new therapeutic modalities. This study investigated the neuroprotective flavonoid, 6-methoxyflavone (6MF), as a potential therapeutic agent and gabapentin as the standard comparator, against neuropathic models. Thus, neuropathic-like states were induced in Sprague-Dawley rats using sciatic nerve chronic constriction injury (CCI) mononeuropathy and systemic administration of streptozotocin (STZ) to induce polyneuropathy. Subsequent behaviors reflecting allodynia, hyperalgesia, and vulvodynia were assessed and any possible motoric side-effects were evaluated including locomotor activity, as well as rotarod discoordination and gait disruption. 6MF (25-75 mg/kg) antagonized neuropathic-like nociceptive behaviors including static- (pressure) and dynamic- (light brushing) hindpaw allodynia plus heat/cold and pressure hyperalgesia in the CCI and STZ models. 6MF also reduced static and dynamic components of vulvodynia in the STZ induced polyneuropathy model. Additionally, 6MF reversed CCI and STZ suppression of locomotor activity and rotarod discoordination, suggesting a beneficial activity on motor side effects, in contrast to gabapentin. Hence, 6MF possesses anti-neuropathic-like activity not only against different nociceptive modalities but also impairment of motoric side effects.


Subject(s)
Flavones , Hyperalgesia , Neuralgia , Rats, Sprague-Dawley , Animals , Rats , Neuralgia/drug therapy , Neuralgia/etiology , Flavones/pharmacology , Flavones/therapeutic use , Hyperalgesia/drug therapy , Male , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Gabapentin/pharmacology , Gabapentin/therapeutic use , Nociception/drug effects , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Female , gamma-Aminobutyric Acid/metabolism , Amines/pharmacology , Amines/therapeutic use , Sciatic Nerve/injuries , Sciatic Nerve/drug effects , Vulvodynia/drug therapy , Constriction , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Analgesics/pharmacology , Analgesics/therapeutic use
4.
Mol Carcinog ; 63(9): 1627-1642, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38888206

ABSTRACT

Flavonoids, constituting the most extensive category of polyphenols, founds in a variety of plants and comprise over 9000 compounds. Diosmetin, O-methylated flavone (3',5,7-trihydroxy-4'-methoxyflavone) of flavonoid aglycone diosmin have witnessed a significant surge in recent years. Many studies showed that flavonoids induced cytotoxicity in different organ specific cancer types. Thus, current review evaluates the anticancer potential of diosmetin and shed light on its mechanism of action such as cell cycle regulation, apoptosis via both intrinsic and extrinsic pathway, autophagy and tumour progression and metastasis. It also provides comprehensive analysis of different cancer targets and their role in breast, colon, hepatic, gliomas, leukemia, lung, prostate and skin cancer. Combination studies of diosmetin to improve drug sensitivity and reduce toxicity towards normal cells has been also discussed. Besides, in vitro studies, present review also discuss the anticancer potential of diosmetin on xenograft mice model. Different natural sources of diosmetin, limitations, pharmacokinetic analysis and toxicity study also summarized in current review. The emphasis on enhancing solubility and permeability for clinical utility has been thoroughly highlighted with particular attention given to the utilization of nano formulations to overcome existing barriers. At last, in-depth analysis of current challenges and a forward-looking perspective deliberated to address the existing gaps and position it as a promising lead compound for clinical applications in cancer treatment. This discussion is boosted by diosmetin's potential anticancer properties on different cancers, makes valuable candidates in the ongoing quest for effective therapeutic interventions against cancer.


Subject(s)
Flavonoids , Neoplasms , Signal Transduction , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Animals , Signal Transduction/drug effects , Flavonoids/pharmacology , Flavonoids/therapeutic use , Disease Progression , Flavones/pharmacology , Flavones/therapeutic use , Apoptosis/drug effects
5.
Cytokine ; 175: 156480, 2024 03.
Article in English | MEDLINE | ID: mdl-38232644

ABSTRACT

Cutaneous squamous cell carcinoma (CSCC) is the second most common malignant skin tumor and significantly affects patients' quality of life and health. The Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway activation is involved in CSCC development. Radix Tetrastigma hemsleyani flavone (RTHF) is an active Radix Tetrastigma extract (RTE), which was recently reported to have promising inhibitory effects on CSCC. However, the underlying functional mechanisms of this inhibition remain unknown. In the present study, A431 cells or SCL-1 cells were incubated with 1, 5, and 10 mg/mL RTHF for 48 h, respectively. A significantly increased wound closure rate, decreased number of migrated and invaded cells, decreased colony number, and elevated apoptotic rate were observed after treatment with 1, 5, and 10 mg/mL RTHF. Furthermore, after incubation with RTHF, p-JAK1/JAK1, p-JAK2/JAK2, and p-STAT3/STAT3 levels were drastically reduced. An A431 xenograft model was constructed, followed by oral administration of 15, 30, or 60 mg/kg RTHF for 21 consecutive days. A significantly lower increase in tumor volume and reduced tumor weight were observed in all RTHF-treated groups. In addition, JAK/STAT3 signaling was drastically repressed in tumor tissues. Collectively, RTHF inhibited CSCC progression, which may be associated with JAK/STAT3 pathway inactivation.


Subject(s)
Carcinoma, Squamous Cell , Flavones , Skin Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Janus Kinases/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Quality of Life , Cell Proliferation , Cell Line, Tumor , Flavones/pharmacology , Flavones/therapeutic use , Apoptosis
6.
Toxicol Appl Pharmacol ; 486: 116951, 2024 May.
Article in English | MEDLINE | ID: mdl-38705401

ABSTRACT

Cardiac lipotoxicity is a prevalent consequence of lipid metabolism disorders occurring in cardiomyocytes, which in turn precipitates the onset of heart failure. Mimetics of brain-derived neurotrophic factor (BDNF), such as 7,8-dihydroxyflavone (DHF) and 7,8,3'-trihydroxyflavone (THF), have demonstrated significant cardioprotective effects. However, it remains unclear whether these mimetics can protect cardiomyocytes against lipotoxicity. The aim of this study was to examine the impact of DHF and THF on the lipotoxic effects induced by palmitic acid (PA), as well as the concurrent mitochondrial dysfunction. H9c2 cells were subjected to treatment with PA alone or in conjunction with DHF or THF. Various factors such as cell viability, lactate dehydrogenase (LDH) release, death ratio, and mitochondrial function including mitochondrial membrane potential (MMP), mitochondrial-derived reactive oxygen species (mito-SOX) production, and mitochondrial respiration were assessed. PA dose-dependently reduced cell viability, which was restored by DHF or THF. Additionally, both DHF and THF decreased LDH content, death ratio, and mito-SOX production, while increasing MMP and regulating mitochondrial oxidative phosphorylation in cardiomyocytes. Moreover, DHF and THF specifically activated Akt signaling. The protective effects of DHF and THF were abolished when an Akt inhibitor was used. In conclusion, BDNF mimetics attenuate PA-induced injury in cardiomyocytes by alleviating mitochondrial impairments through the activation of Akt signaling.


Subject(s)
Brain-Derived Neurotrophic Factor , Flavones , Membrane Potential, Mitochondrial , Myocytes, Cardiac , Palmitic Acid , Proto-Oncogene Proteins c-akt , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Palmitic Acid/toxicity , Palmitic Acid/pharmacology , Animals , Proto-Oncogene Proteins c-akt/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Rats , Cell Line , Membrane Potential, Mitochondrial/drug effects , Flavones/pharmacology , Cell Survival/drug effects , Signal Transduction/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Reactive Oxygen Species/metabolism
7.
Neurochem Res ; 49(4): 980-997, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38170385

ABSTRACT

Diabetic neuropathic pain is one of the most devasting disorders of peripheral nervous system. The loss of GABAergic inhibition is associated with the development of painful diabetic neuropathy. The current study evaluated the potential of 3-Hydroxy-2-methoxy-6-methyl flavone (3-OH-2'MeO6MF), to ameliorate peripheral neuropathic pain using an STZ-induced hyperglycemia rat model. The pain threshold was assessed by tail flick, cold, mechanical allodynia, and formalin test on days 0, 14, 21, and 28 after STZ administration accompanied by evaluation of several biochemical parameters. Administration of 3-OH-2'-MeO6MF (1,10, 30, and 100 mg/kg, i.p) significantly enhanced the tail withdrawal threshold in tail-flick and tail cold allodynia tests. 3-OH-2'-MeO6MF also increased the paw withdrawal threshold in mechanical allodynia and decreased paw licking time in the formalin test. Additionally, 3-OH-2'-MeO6MF also attenuated the increase in concentrations of myeloperoxidase (MPO), thiobarbituric acid reactive substances (TBARS), nitrite, TNF-α, and IL 6 along with increases in glutathione (GSH). Pretreatment of pentylenetetrazole (PTZ) (40 mg/kg, i.p.) abolished the antinociceptive effect of 3-OH-2'-MeO6MF in mechanical allodynia. Besides, the STZ-induced alterations in the GABA concentration and GABA transaminase activity attenuated by 3-OH-2'-MeO6MF treatment suggest GABAergic mechanisms. Molecular docking also authenticates the involvement of α2ß2γ2L GABA-A receptors and GABA-T enzyme in the antinociceptive activities of 3-OH-2'-MeO6MF.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Flavones , Neuralgia , Rats , Animals , Hyperalgesia/drug therapy , Diabetic Neuropathies/drug therapy , Streptozocin , Molecular Docking Simulation , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/complications , Analgesics/pharmacology , gamma-Aminobutyric Acid/pharmacology , Flavones/pharmacology , Flavones/therapeutic use , Biomarkers
8.
Nutr Cancer ; 76(7): 638-655, 2024.
Article in English | MEDLINE | ID: mdl-38721626

ABSTRACT

Inhibiting breast cancer stem cell (BCSC) signaling pathways is a strategic method for successfully treating breast cancer. Nobiletin (NOB) is a compound widely found in orange peel that exhibits a toxic effect on various types of cancer cells, and inhibits the signaling pathways that regulate the properties of BCSCs; however, the effects of NOB on BCSCs remain elusive. The purpose of this study was to determine the target genes of NOB for inhibiting BCSCs using in vitro three-dimensional breast cancer cell culture (mammospheres) and in silico approaches. We combined in vitro experiments to develop mammospheres and conducted cytotoxicity, next-generation sequencing, and bioinformatics analyses, such as gene ontology, the Reactome pathway enrichment, network topology, gene set enrichment analysis, hub genes selection, genetic alterations, prognostic value related to the mRNA expression, and mRNA and protein expression of potential NOB target genes that inhibit BCSCs. Here, we show that NOB inhibited BCSCs in mammospheres from MCF-7 cells. We also identified CDC6, CHEK1, BRCA1, UCHL5, TOP2A, MTMR4, and EXO1 as potential NOB targets inhibiting BCSCs. NOB decreased G0/G1, but increased the G2/M cell population. These findings showed that NOB is a potential therapeutic candidate for BCSCs treatment by regulating cell cycle.


Subject(s)
Breast Neoplasms , Cell Cycle , Computational Biology , Flavones , Neoplastic Stem Cells , Flavones/pharmacology , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , MCF-7 Cells , Computational Biology/methods , Female , Cell Cycle/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects
9.
Photochem Photobiol Sci ; 23(9): 1771-1782, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39249169

ABSTRACT

Potent antioxidants, like 3-hydroxy flavones, attracted considerable attention due to their excited state intramolecular proton transfer (ESIPT)-based fluorescence behaviour. This article is an interesting demonstration of a series of synthetic 3-hydroxy flavone analogues having high antioxidant activity as molecular rotor-like viscosity probes. Among these flavone analogues, 4'-N,N-dimethylamino-3-hydroxy flavone (3) is the most potent one, showing the twisted intramolecular charge transfer (TICT)-dependent fluoroprobing activity toward the blood viscosity changes associated with diabetes and free fatty acids (FFA)-induced nuclear viscosity changes of MIN6 cells. The TICT dynamics of (3), which instigates its viscosity probing activity, was comprehended with the help of DFT-based computational studies. Abnormal cellular viscosity changes are the pathological traits for various diseases, and non-toxic flavone-based viscosity probes can be useful for diagnosing such pathological conditions.


Subject(s)
Antioxidants , Density Functional Theory , Flavones , Flavones/chemistry , Flavones/pharmacology , Viscosity , Antioxidants/chemistry , Antioxidants/pharmacology , Diabetes Mellitus/drug therapy , Animals , Protons , Mice , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Fluorescent Dyes/chemical synthesis , Fatty Acids, Nonesterified/chemistry , Fatty Acids, Nonesterified/metabolism , Humans
10.
Bioorg Med Chem Lett ; 98: 129577, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38065293

ABSTRACT

Transient receptor potential vanilloid 3 (TRPV3) channel is a temperature-sensitive and Ca2+-permeable nonselective cation channel, which is abundantly expressed in skin keratinocyte and plays an important role in skin homeostasis and repair. However, only a few TRPV3 inhibitors were reported. Few selective and potent modulators of the TRPV3 channel have hindered the progress of the investigation and clinical application. TRPV3 channel research still faces challenges and requires the new inhibitors. Flavonoids are a kind of natural compounds with various biological and pharmacological activities including anti-inflammatory and anti allergic effects, which is associated with some physiological effects mediated by TRPV3 channel. Herein, our group designed and synthesized a range of flavone derivatives, and investigated their inhibitory properties on the human TRPV3 channel by electrophysiology technique. Then, we identified a new potent TRPV3 antagonist 2d with IC50 of 0.62 µM. It also showed good selectivity on TRPV1, TRPV4, TRPA1 and TRPM8.


Subject(s)
Flavones , Transient Receptor Potential Channels , Humans , Flavones/pharmacology , Keratinocytes , Temperature , Transient Receptor Potential Channels/antagonists & inhibitors , TRPV Cation Channels
11.
Mol Biol Rep ; 51(1): 785, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951450

ABSTRACT

BACKGROUND: Kaempferia parviflora Wall. ex. Baker (KP) has been reported to exhibit anti-obesity effects. However, the detailed mechanism of the anti-obesity effect of KP extract (KPE) is yet to be clarified. Here, we investigated the effect of KPE and its component polymethoxyflavones (PMFs) on the adipogenic differentiation of human mesenchymal stem cells (MSCs). METHODS AND RESULTS: KPE and PMFs fraction (2.5 µg/mL) significantly inhibited lipid and triacylglyceride accumulation in MSCs; lipid accumulation in MSCs was suppressed during the early stages of differentiation (days 0-3) but not during the mid (days 3-7) or late (days 7-14) stages. Treatment with KPE and PMFs fractions significantly suppressed peroxisome proliferator-activated receptor-γ (PPARγ), CCAAT/enhancer binding protein α (C/EBPα), and various adipogenic metabolic factors. Treatment with KPE and PMFs fraction induced the activation of AMP-activated protein kinase (AMPK) signaling, and pretreatment with an AMPK signaling inhibitor significantly attenuated KPE- and PMFs fraction-induced suppression of lipid formation. CONCLUSIONS: Our findings demonstrate that KPE and PMFs fraction inhibit lipid formation by inhibiting the differentiation of undifferentiated MSCs into adipocyte lineages via AMPK signaling, and this may be the mechanism underlying the anti-obesity effects of KPE and PMFs. Our study lays the foundation for the elucidation of the anti-obesity mechanism of KPE and PMFs.


Subject(s)
AMP-Activated Protein Kinases , Adipogenesis , Cell Differentiation , Flavones , Mesenchymal Stem Cells , Plant Extracts , Signal Transduction , Zingiberaceae , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Adipogenesis/drug effects , Plant Extracts/pharmacology , Zingiberaceae/chemistry , AMP-Activated Protein Kinases/metabolism , Flavones/pharmacology , Cell Differentiation/drug effects , Signal Transduction/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Cells, Cultured
12.
J Gastroenterol Hepatol ; 39(8): 1586-1596, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38803139

ABSTRACT

BACKGROUND AND AIM: Drug therapy is the treatment of choice for Crohn's disease because it effectively controls or prevents intestinal inflammation. The purpose was to research the molecular mechanism of the total flavones of Abelmoschus manihot (TFA) on intestinal fibrosis in Crohn's disease. METHODS: A 2,4,6-Trinitrobenzenesulfonic acid (TNBS)-induced colitis model and IGF-1-treated intestinal fibroblasts were established. Then, TFA, 3-MA, and compound C were used treatments. Hematoxylin and eosin, Masson, and Picrosirius red staining were performed to observe the colon tissue. Immunohistochemical staining was used to detect α-SMA expression. Flow cytometry, CCK8, wound healing, and Transwell assays were conducted to determine apoptosis, proliferation, invasion, and migration. Col1a1 and Col3a1 levels were detected using quantitative polymerase chain reaction. Proteins related to autophagy and apoptosis were detected using western blotting. RESULTS: TFA treated intestinal fibrosis in chronic Crohn's disease. Colon length was the shortest in the ethanol + TNBS group, and TFA treatment significantly improved the situation. Intestinal fibrosis and the percentage of collagen area decreased after TFA treatment. TFA reduced fibrosis by enhancing autophagy stimulation, whereas an autophagy inhibitor reversed the TFA effect. TFA also inhibited migration, proliferation, and collagen synthesis in intestinal fibroblasts. Moreover, it enhanced autophagy and apoptosis of intestinal fibroblasts. TFA upregulated p-AMPK expression and decreases p-mTOR levels. Compound C partially rescued the effect of TFA, indicating that TFA affected intestinal fibroblasts via the AMPK/mTOR pathway in vitro and in vivo. TFA also downregulated Col1a1 and Col3a1 expression. CONCLUSION: TFA regulates autophagy through AMPK/mTOR signaling pathway to treat intestinal fibrosis, which may provide a new therapy for Crohn's disease treatment.


Subject(s)
AMP-Activated Protein Kinases , Abelmoschus , Autophagy , Crohn Disease , Fibrosis , Flavones , Signal Transduction , TOR Serine-Threonine Kinases , Crohn Disease/drug therapy , Crohn Disease/pathology , TOR Serine-Threonine Kinases/metabolism , Autophagy/drug effects , Abelmoschus/chemistry , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Flavones/pharmacology , Flavones/therapeutic use , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Male , Trinitrobenzenesulfonic Acid , Disease Models, Animal , Cell Proliferation/drug effects , Apoptosis/drug effects , Mice , Humans , Cells, Cultured
13.
J Biochem Mol Toxicol ; 38(11): e70003, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39392135

ABSTRACT

Citrus fruits are extensively cultivated and eaten both raw and in refined forms. Citrus fruit peels are highly concentrated in polyphenolic substances. This makes them useful resources. Polymethoxyflavones (PMFs), found in citrus peels, belong to a specific subclass of flavonoids where most or all hydroxyl groups are methylated. PMFs have been documented to possess chemopreventive actions, anticancer, anti-inflammatory, and anti-atherosclerosis properties, as well as neuroprotective effects. Sudachitin, a PMF, is primarily found in Citrus sudachi. Japan's Tokushima prefecture is home to this famous fruit. In recent years, there has been a growing interest among researchers in exploring the potential health benefits of sudachitin, spurred by its presence in traditional diets and its association with various positive health outcomes. Studies conducted over the past decade have revealed promising effects of sudachitin in multiple health conditions, including cancer, skin disorders, inflammatory conditions, diabetes, obesity, and neurodegenerative disorders. Although these promising results exist, there is still a need for thorough preclinical and clinical research to confirm sudachitin's effectiveness in treating chronic conditions. This review seeks to summarize animal and cell studies exploring sudachitin's pharmacological properties and the potential molecular pathways underlying its therapeutic effects. Through this, we aim to clarify the clinical potential of sudachitin across various disorders, paving the way for future research and the development of sudachitin-based therapies.


Subject(s)
Flavonoids , Humans , Animals , Flavonoids/therapeutic use , Citrus/chemistry , Neoplasms/drug therapy , Flavones/therapeutic use , Flavones/pharmacology
14.
Cell Biochem Funct ; 42(4): e4060, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816947

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a pervasive organic toxicant that damages body organs, including heart. Isosakuranetin (ISN) is a plant-based flavonoid that exhibits a broad range of pharmacological potentials. The current investigation was conducted to evaluate the potential role of ISN to counteract PFOS-induced cardiac damage in rats. Twenty-four albino rats (Rattus norvegicus) were distributed into four groups, including control, PFOS (10 mg/kg) intoxicated, PFOS + ISN (10 mg/kg + 20 mg/kg) treated, and ISN (20 mg/kg) alone supplemented group. It was revealed that PFOS intoxication reduced the expressions of Nrf-2 and its antioxidant genes while escalating the expression of Keap-1. Furthermore, PFOS exposure reduced the activities of glutathione reductase (GSR), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), Heme oxygenase-1 (HO-1) and glutathione (GSH) contents while upregulating the levels of reactive oxygen species (ROS) and malondialdehyde (MDA). Besides, PFOS administration upregulated the levels of creatine kinase-MB (CK-MB), troponin I, creatine phosphokinase (CPK), and lactate dehydrogenase (LDH). Moreover, the levels of tumor necrosis factor-alpha (TNF-α), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) were increased after PFOS intoxication. Additionally, PFOS exposure downregulated the expression of Bcl-2 while upregulating the expressions of Bax and Caspase-3. Furthermore, PFOS administration disrupted the normal architecture of cardiac tissues. Nonetheless, ISN treatment remarkably protected the cardiac tissues via regulating aforementioned dysregulations owing to its antioxidative, anti-inflammatory, and antiapoptotic properties.


Subject(s)
Alkanesulfonic Acids , Apoptosis , Cardiotoxicity , Flavonoids , Fluorocarbons , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Animals , Rats , Alkanesulfonic Acids/pharmacology , Alkanesulfonic Acids/toxicity , Apoptosis/drug effects , Flavones/pharmacology , Fluorocarbons/pharmacology , Fluorocarbons/toxicity , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Inflammation/pathology , Kelch-Like ECH-Associated Protein 1/drug effects , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Cardiotoxicity/metabolism , Flavonoids/pharmacology
15.
J Appl Toxicol ; 44(6): 833-845, 2024 06.
Article in English | MEDLINE | ID: mdl-38291015

ABSTRACT

Mulberrin, a naturally occurring flavone found in mulberry and Romulus Mori, exhibits diverse biological functions. Here, we showed that mulberrin extended both the lifespan and healthspan in C. elegans. Moreover, mulberrin increased the worms' resistance to toxicants and activated the expression of detoxification genes. The longevity-promoting effect of mulberrin was attenuated in nuclear hormone receptor (NHR) homologous nhr-8 and daf-12 mutants, indicating that the lifespan extending effects of mulberrin in C. elegans may depend on nuclear hormone receptors NHR-8/DAF-12. Further analyses revealed the potential associations between the longevity effects of mulberrin and the insulin/insulin-like growth factor signaling (IIS) and adenosine 5'-monophosphate-activated protein kinase (AMPK) pathways. Together, our findings suggest that mulberrin may prolong lifespan and healthspan by activating detoxification functions mediated by nuclear receptors.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Longevity , Receptors, Cytoplasmic and Nuclear , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Longevity/drug effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Inactivation, Metabolic , Flavones/pharmacology , Insulin/metabolism , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Mutation
16.
Phytother Res ; 38(2): 880-911, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38088265

ABSTRACT

Current pharmaceutical research is energetically excavating the pharmacotherapeutic role of herb-derived ingredients in multiple malignancies' targeting. Luteolin is one of the major phytochemical components that exist in various traditional Chinese medicine or medical herbs. Mounting evidence reveals that this phytoconstituent endows prominent therapeutic actions on diverse malignancies, with the underlying mechanisms, combined medication strategy, and pharmacokinetics elusive. Additionally, the clinical trial and pharmaceutical investigation of luteolin remain to be systematically delineated. The present review aimed to comprehensively summarize the updated information with regard to the anticancer mechanism, combined medication strategies, pharmacokinetics, clinical trials, and pharmaceutical researches of luteolin. The survey corroborates that luteolin executes multiple anticancer effects mainly by dampening proliferation and invasion, spurring apoptosis, intercepting cell cycle, regulating autophagy and immune, inhibiting inflammatory response, inducing ferroptosis, and pyroptosis, as well as epigenetic modification, and so on. Luteolin can be applied in combination with numerous clinical anticarcinogens and natural ingredients to synergistically enhance the therapeutic efficacy of malignancies while reducing adverse reactions. For pharmacokinetics, luteolin has an unfavorable oral bioavailability, it mainly persists in plasma as glucuronides and sulfate-conjugates after being metabolized, and is regarded as potent inhibitors of OATP1B1 and OATP2B1, which may be messed with the pharmacokinetic interactions of miscellaneous bioactive substances in vivo. Besides, pharmaceutical innovation of luteolin with leading-edge drug delivery systems such as host-guest complexes, nanoparticles, liposomes, nanoemulsion, microspheres, and hydrogels are beneficial to the exploitation of luteolin-based products. Moreover, some registered clinical trials on luteolin are being carried out, yet clinical research on anticancer effects should be continuously promoted.


Subject(s)
Flavones , Neoplasms , Humans , Luteolin/pharmacology , Luteolin/therapeutic use , Pharmaceutical Preparations , Flavones/pharmacology , Flavones/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Biological Availability
17.
Phytother Res ; 38(7): 3444-3458, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38685750

ABSTRACT

The escalating incidence of nonalcoholic fatty liver disease (NAFLD) is closely associated with a high-fat diet, leading to a decline in quality of life and significant health impairment. 7-Hydroxyflavone (7-HY) is a flavonoid known for its anti-inflammatory, anticarcinogenic, and antioxidant effects. This study aims to assess the ameliorative effects of 7-HY on NAFLD induced by a high-fat diet and elucidate underlying mechanisms. Oleic acid/palmitic acid-induced HepG2 cells and C57BL/6 mice on a high-fat diet were utilized as in vitro and in vivo models. In animal experiments, 7-HY was utilized as a dietary supplement. The 15-week in vivo experiment monitored body weight, body fat percentage, glucose tolerance, insulin tolerance, and metabolic indexes. Commercial kits assessed triglyceride (TG) and total cholesterol levels in cells, liver tissue, and blood. Discovery Studio identified potential targets of 7-HY, compared with NAFLD-associated targets in the GeneCards database. Results indicated 7-HY mitigated fat accumulation, hepatic steatosis, and oxidative stress induced by a high-fat diet. Furthermore, 7-HY showed potential efficacy in ameliorating abnormal glucose metabolism and promoting energy metabolism. Reverse target finding and molecular docking demonstrated a robust interaction between 7-HY and serine/threonine kinase 24 (STK24). Subsequent experimental results confirmed 7-HY's ability to inhibit TG deposition in HepG2 cells through interaction with STK24. In conclusion, 7-HY demonstrated the capacity to alleviate high-fat diet-induced NAFLD, presenting a novel strategy for the prevention and treatment of NAFLD.


Subject(s)
Diet, High-Fat , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Protein Serine-Threonine Kinases , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Humans , Hep G2 Cells , Mice , Diet, High-Fat/adverse effects , Male , Protein Serine-Threonine Kinases/metabolism , Oxidative Stress/drug effects , Flavones/pharmacology , Flavones/chemistry , Liver/drug effects , Liver/metabolism , Molecular Docking Simulation , Triglycerides/blood , Flavonoids/pharmacology
18.
Pestic Biochem Physiol ; 204: 106081, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277394

ABSTRACT

Xenobiotic response element (XRE) to flavone was the cis- regulatory elements that mediates the induction of the allelochemical-metabolizing CYP321A1 gene from Helicoverpa zea. However, it was unknown whether the XRE-Fla element existed in other species. Recently we have identified and cloned the CYP321A1 gene with promoter region in a related species, Helicoverpa armigera. Sequence similarity of two orthologous CYP321A1 genes was 97.27%, but the promoter sequence similarity was only 56.32%. Sequence alignment showed the XRE-Fla like element owns three mutations in H. armigera compared with H. zea. Progressive 5' deletions and internal mutation indicated that H. armigera XRE-Fla was the essential element of CYP321A1 gene in response to flavone. XRE-Fla mutations and EMSA analysis confirmed that the H. armigera XRE-Fla element binding factor was stronger than H. zea. The findings indicate the XRE element mutations mainly contribute to the differences between the flavone-induced expressions of two CYP321A1 genes, which improve the flexibility and adaptability for allelochemical response of H. armigera.


Subject(s)
Cytochrome P-450 Enzyme System , Flavones , Moths , Animals , Moths/genetics , Moths/drug effects , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Flavones/pharmacology , Insect Proteins/genetics , Insect Proteins/metabolism , Promoter Regions, Genetic , Base Sequence , Response Elements , Helicoverpa armigera
19.
Chem Biodivers ; 21(5): e202400518, 2024 May.
Article in English | MEDLINE | ID: mdl-38501574

ABSTRACT

In this study, two undescribed compounds (1 and 2), together with eight known compounds (3-10) were isolated from the aerial parts of Piper samentosum by various chromatography methods. Their chemical structures were determined to be 7'''-oxolyciumamide N (1), vitexin 2''-O-ß-D-(6'''-feruloyl)-glucopyranoside (2), 1,2-dihydro-6,8-dimethoxy-7-hydroxy-1-(3,4-dihydroxyphenyl)-N1,N2-bis-[2-(-hydroxyphenyl)ethyl]-2,3-napthalene dicarboamide (3), vitexin 6''-O-ß-D-glucopyranoside (4), vitexin 2''-O-α-L-rhamnopyranoside (5), methyl 2-hydroxybenzoate-2-O-ß-D-apiofuranosyl-(1→2)-O-ß-D-glucopyranoside (6), ficuside G (7), methyl 2-O-ß-D-glucopyranosylbenzoate (8), methyl 2,5-dihydroxybenzoate-5-O-ß-D-glucopyranoside (9), and 3,7-dimethyloct-1-ene-3,6,7-triol 6-O-ß-D-glucopyranoside (10) by spectroscopic data analysis including HR-ESI-MS, 1D-, and 2D-NMR spectra. Compounds 1-5 inhibited nitric oxide production in LPS-stimulated RAW264.7 macrophages with the IC50 values of 27.62, 74.03, 38.54, 70.39, and 44.95 µM, respectively. The NMR data of 9 were firstly reported herein.


Subject(s)
Flavones , Glucosides , Lipopolysaccharides , Nitric Oxide , Piper , Plant Components, Aerial , RAW 264.7 Cells , Mice , Animals , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Plant Components, Aerial/chemistry , Glucosides/isolation & purification , Glucosides/pharmacology , Glucosides/chemistry , Piper/chemistry , Flavones/isolation & purification , Flavones/pharmacology , Flavones/chemistry , Amides/chemistry , Amides/pharmacology , Amides/isolation & purification , Molecular Structure
20.
Immunopharmacol Immunotoxicol ; 46(4): 496-508, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951964

ABSTRACT

OBJECTIVE: This study aimed to investigate the use of 5,7,3',4'-tetramethoxyflavone (TMF) to treat pulmonary fibrosis (PF), a chronic and fatal lung disease. In vitro and in vivo models were used to examine the impact of TMF on PF. METHODS: NIH-3T3 (Mouse Embryonic Fibroblast) were exposed to transforming growth factor­ß1 (TGF-ß1) and treated with or without TMF. Cell growth was assessed using the MTT method, and cell migration was evaluated with the scratch wound assay. Protein and messenger ribonucleic acid (mRNA) levels of extracellular matrix (ECM) genes were analyzed by western blotting and quantitative reverse transcription-polymerase chain reaction (RT-PCR), respectively. Downstream molecules affected by TGF-ß1 were examined by western blotting. In vivo, mice with bleomycin-induced PF were treated with TMF, and lung tissues were analyzed with staining techniques. RESULTS: The in vitro results showed that TMF had no significant impact on cell growth or migration. However, it effectively inhibited myofibroblast activation and ECM production induced by TGF-ß1 in NIH-3T3 cells. This inhibition was achieved by suppressing various signaling pathways, including Smad, mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase/AKT (PI3K/AKT), and WNT/ß-catenin. The in vivo experiments demonstrated the therapeutic potential of TMF in reducing PF induced by bleomycin in mice, and there was no significant liver or kidney toxicity observed. CONCLUSION: These findings suggest that TMF has the potential to effectively inhibit myofibroblast activation and could be a promising treatment for PF. TMF achieves this inhibitory effect by targeting TGF-ß1/Smad and non-Smad pathways.


Subject(s)
Bleomycin , Fibroblasts , Pulmonary Fibrosis , Transforming Growth Factor beta1 , Animals , Mice , Transforming Growth Factor beta1/metabolism , NIH 3T3 Cells , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Bleomycin/toxicity , Fibroblasts/drug effects , Fibroblasts/metabolism , Flavones/pharmacology , Mice, Inbred C57BL , Cell Movement/drug effects , Male , Cell Proliferation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL