Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.037
Filter
Add more filters

Publication year range
1.
Nature ; 586(7827): 133-138, 2020 10.
Article in English | MEDLINE | ID: mdl-32728212

ABSTRACT

Somatic mutations in p53, which inactivate the tumour-suppressor function of p53 and often confer oncogenic gain-of-function properties, are very common in cancer1,2. Here we studied the effects of hotspot gain-of-function mutations in Trp53 (the gene that encodes p53 in mice) in mouse models of WNT-driven intestinal cancer caused by Csnk1a1 deletion3,4 or ApcMin mutation5. Cancer in these models is known to be facilitated by loss of p533,6. We found that mutant versions of p53 had contrasting effects in different segments of the gut: in the distal gut, mutant p53 had the expected oncogenic effect; however, in the proximal gut and in tumour organoids it had a pronounced tumour-suppressive effect. In the tumour-suppressive mode, mutant p53 eliminated dysplasia and tumorigenesis in Csnk1a1-deficient and ApcMin/+ mice, and promoted normal growth and differentiation of tumour organoids derived from these mice. In these settings, mutant p53 was more effective than wild-type p53 at inhibiting tumour formation. Mechanistically, the tumour-suppressive effects of mutant p53 were driven by disruption of the WNT pathway, through preventing the binding of TCF4 to chromatin. Notably, this tumour-suppressive effect was completely abolished by the gut microbiome. Moreover, a single metabolite derived from the gut microbiota-gallic acid-could reproduce the entire effect of the microbiome. Supplementing gut-sterilized p53-mutant mice and p53-mutant organoids with gallic acid reinstated the TCF4-chromatin interaction and the hyperactivation of WNT, thus conferring a malignant phenotype to the organoids and throughout the gut. Our study demonstrates the substantial plasticity of a cancer mutation and highlights the role of the microenvironment in determining its functional outcome.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Gastrointestinal Microbiome/genetics , Genes, Tumor Suppressor , Mutation , Oncogenes/genetics , Tumor Suppressor Protein p53/genetics , Animals , Anti-Bacterial Agents/pharmacology , Carcinogenesis/drug effects , Female , Gallic Acid/pharmacology , Gastrointestinal Microbiome/drug effects , Humans , Male , Mice , Organoids/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway/drug effects
2.
Nano Lett ; 24(26): 8179-8188, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38885447

ABSTRACT

The unique "Iron Addiction" feature of cancer stem cells (CSCs) with tumorigenicity and plasticity generally contributes to the tumor recurrence and metastasis after a lumpectomy. Herein, a novel "Ferroptosis Amplification" strategy is developed based on integrating gallic acid-modified FeOOH (GFP) and gallocyanine into Pluronic F-127 (F127) and carboxylated chitosan (CC)-based hydrogel for CSCs eradication. This "Ferroptosis Amplifier" hydrogel is thermally sensitive and achieves rapid gelation at the postsurgical wound in a breast tumor model. Specifically, gallocyanine, as the Dickkopf-1 (DKK1) inhibitor, can decrease the expression of SLC7A11 and GPX4 and synergistically induce ferroptosis of CSCs with GFP. Encouragingly, it is found that this combination suppresses the migratory and invasive capability of cancer cells via the downregulation of matrix metalloproteinase 7 (MMP7). The in vivo results further confirm that this "Ferroptosis Amplification" strategy is efficient in preventing tumor relapse and lung metastasis, manifesting an effective and promising postsurgical treatment for breast cancer.


Subject(s)
Breast Neoplasms , Ferroptosis , Hydrogels , Neoplastic Stem Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Hydrogels/chemistry , Humans , Animals , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Mice , Ferroptosis/drug effects , Cell Line, Tumor , Poloxamer/chemistry , Poloxamer/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Chitosan/analogs & derivatives , Gallic Acid/pharmacology , Gallic Acid/chemistry , Gallic Acid/therapeutic use
3.
J Cell Physiol ; 239(4): e31196, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38240115

ABSTRACT

The NOD-, LRR-, and Pyrin domain-containing protein 3 (NLRP3) inflammasome plays key roles in regulating inflammation. Numerous studies show that the abnormal activation of NLRP3 associates with the initiation and progression of various diseases. Hence, the NLRP3 inflammasome may be a promising therapeutic target for these diseases. Octyl gallate (OG) is a small molecule with antioxidant, antimicrobial, antifungal, and anti-inflammatory activities; however, the mechanism underlying its anti-inflammatory activity is still unclear. Here, we developed a screening system for NLRP3-inflammasome inhibitors. A total of 3287 small molecules were screened for inhibitors of nigericin-induced NLRP3 oligomerization. OG was identified as a novel inhibitor. We show that OG directly targets the LRR domain of NLRP3 and thereby blocks the inflammatory cascade of the NLRP3 inflammasome. This contrasts with the mode-of-action of other direct NLRP3 inhibitors, which all bind to the NACHT domain of NLRP3. Interestingly, OG also inhibits the priming step by downregulating the Raf-MEK1/2-ERK1/2 axis. Thus, OG inhibits the NLRP3 inflammasome by two distinct mechanisms. Importantly, OG injection ameliorated the inflammation in mouse models of foot gout and sepsis. Our study identifies OG as a potential therapeutic agent for NLRP3-associated diseases.


Subject(s)
Anti-Inflammatory Agents , Gallic Acid , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Gallic Acid/analogs & derivatives , Inflammasomes/drug effects , Inflammation/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice, Inbred C57BL , Male , Protein Domains
4.
Photosynth Res ; 159(2-3): 153-164, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37204684

ABSTRACT

Different light spectra from light-emitting diodes (LEDs) trigger species-specific adaptive responses in plants. We exposed Artemisia argyi (A. argyi) to four LED spectra: white (the control group), monochromatic red light (R), monochromatic blue light (B), or a mixture of R and B light of photon flux density ratio is 3 (RB), with equivalent photoperiod (14 h) and light intensity (160 µmol s-1 m-2). R light accelerated photomorphogenesis but decreased biomass, while B light significantly increased leaf area and short-term exposure (7 days) to B light increased total phenols and flavonoids. HPLC identified chlorogenic acid, 3,5-dicaffeoylquinic acid, gallic acid, jaceosidin, eupatilin, and taxol compounds, with RB and R light significantly accumulating chlorogenic acid, 3,5-dicaffeoylquinic acid, and gallic acid, and B light promoting jaceosidin, eupatilin, and taxol. OJIP measurements showed that B light had the least effect on the effective quantum yield ΦPSII, with higher rETR(II), Fv/Fm, qL and PIabs, followed by RB light. R light led to faster photomorphology but lower biomass than RB and B lights and produced the most inadaptability, as shown by reduced ΦPSII and enlarged ΦNPQ and ΦNO. Overall, short-term B light promoted secondary metabolite production while maintaining effective quantum yield and less energy dissipation.


Subject(s)
Artemisia , Chlorogenic Acid/analogs & derivatives , Artemisia/metabolism , Fluorescence , Gallic Acid , Chlorophyll/metabolism , Paclitaxel
5.
Toxicol Appl Pharmacol ; 490: 117033, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38997070

ABSTRACT

Gallic acid (GA) has been found by a large number of studies to have pharmacological effects such as antioxidant and anti-inflammatory properties. However, the underlying therapeutic mechanisms are not fully understood.. Studies have shown that altering the intestinal flora affects host metabolism and effectively mediates the development of synovitis. The aim of this study was to explore the pharmacological effects of GA in the treatment of synovial inflammation and anti-synovial fibrosis in knee osteoarthritis (KOA) and the underlying mechanisms by macrogenomics combined with off-target metabolomics. We established a synovitis model via in vivo and in vitro experiments to observe the effect of GA intervention on synovitis. Moreover, we collected serum and feces from rats and analyzed the changes in intestinal flora by macro-genome sequencing and the changes in metabolites in the serum by untargeted metabolomics. We found that GA reduced the levels of IL-1ß, IL-6, and TNF-α, and decreased the protein expression levels of α-SMA, TGF-ß, and Collagen I in synovial tissues and cells, and the composition and function of the intestinal flora were similarly altered. Combined with macrogenomic pathway enrichment analysis and metabolic pathway enrichment analysis, these findings revealed that GA impacts Bacteroidia and Muribaculaceae abundance, and via the following metabolic pathways: sphingolipid metabolism, glycerophospholipid metabolism, and arginine biology.to ameliorate synovial inflammation and fibrosis in KOA. The therapeutic effect of GA on KOA synovitis and fibrosis is partly attributed to the alleviation of metabolic disorder and the rebalancing of the intestinal flora. These results provides a rationale for the therapeutic application of GA in the treatment of synovitis.


Subject(s)
Fibrosis , Gallic Acid , Gastrointestinal Microbiome , Rats, Sprague-Dawley , Animals , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Gastrointestinal Microbiome/drug effects , Male , Rats , Synovitis/drug therapy , Synovitis/pathology , Synovitis/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Disease Models, Animal , Metabolomics
6.
Arch Biochem Biophys ; 756: 109978, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636693

ABSTRACT

A 2D-intestinal epithelial Caco-2/RAW 264.7 macrophage co-culture model was developed to demonstrate the relative efficacy of different phenolic acids to mitigate changes in Caco-2 epithelial cell redox state initiated both directly by autoxidation products, H2O2, and indirectly through cell communication events originating from cytokine stimulated macrophage. An inducer cocktail (lipopolysaccharide + interferon gamma) was used to activate RAW 264.7 cells in the 2D- Caco-2/RAW co-culture and intracellular changes in Caco-2 cell redox signaling occurred in response to positive changes (p < 0.05) in inflammatory biomarkers derived in macrophage that included IL-6, TNF-α, nitric oxide and peroxynitrite, respectively. Phenolic acids varied in relative capacity to reduce NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) in cocktail inflamed induced macrophage. This response in addition to the relative predisposition of gallic acid (GA) to undergo autoxidation to generate H2O2 activity (p < 0.05), culminated in downstream cell signaling in Caco-2 nuclear factor erythroid 2-related factor (Nrf2) activity (increase 26.9 %), altered monolayer integrity (increase 33.7 %), and release of interleukin 8 (IL-8) (decrease 80.5 %) (p < 0.05). It can be concluded that the co-culture model described herein was useful to assess the importance of communication between cytokine stimulated macrophage and intestinal cells. Moreover, the relative unique efficacy of GA, compared to other phenolic acids tested to protect against activated macrophage induced changes related to intestinal dysfunction were particularly relevant to epithelial redox signaling, intestinal permeability and regulation of tight junction proteins. This study concludes that phenolic acids are not equal in the capacity to protect against intestinal cell dysfunction despite some indication of biological activity.


Subject(s)
Coculture Techniques , Gallic Acid , Tight Junction Proteins , Caco-2 Cells , Gallic Acid/pharmacology , Humans , Mice , Animals , RAW 264.7 Cells , Tight Junction Proteins/metabolism , Inflammation/metabolism , Oxidation-Reduction/drug effects , Hydrogen Peroxide/metabolism , Macrophages/metabolism , Macrophages/drug effects , Intestines/cytology , Intestines/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects
7.
Mol Pharm ; 21(5): 2577-2589, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38647021

ABSTRACT

This study aimed to repurpose the antifungal drug flucytosine (FCN) for anticancer activity together with cocrystals of nutraceutical coformers sinapic acid (SNP) and syringic acid (SYA). The cocrystal screening experiments with SNP resulted in three cocrystal hydrate forms in which two are polymorphs, namely, FCN-SNP F-I and FCN-SNP F-II, and the third one with different stoichiometry in the asymmetric unit (1:2:1 ratio of FCN:SNP:H2O, FCN-SNP F-III). Cocrystallization with SYA resulted in two hydrated cocrystal polymorphs, namely, FCN-SYA F-I and FCN-SYA F-II. All the cocrystal polymorphs were obtained concomitantly during the slow evaporation method, and one of the polymorphs of each system was produced in bulk by the slurry method. The interaction energy and lattice energies of all cocrystal polymorphs were established using solid-state DFT calculations, and the outcomes correlated with the experimental results. Further, the in vitro cytotoxic activity of the cocrystals was determined against DU145 prostate cancer and the results showed that the FCN-based cocrystals (FCN-SNP F-III and FCN-SYA F-I) have excellent growth inhibitory activity at lower concentrations compared with parent FCN molecules. The prepared cocrystals induce apoptosis by generating oxidative stress and causing nuclear damage in prostate cancer cells. The Western blot analysis also depicted that the cocrystals downregulate the inflammatory markers such as NLRP3 and caspase-1 and upregulate the intrinsic apoptosis signaling pathway marker proteins, such as Bax, p53, and caspase-3. These findings suggest that the antifungal drug FCN can be repurposed for anticancer activity.


Subject(s)
Antifungal Agents , Antineoplastic Agents , Apoptosis , Drug Repositioning , Flucytosine , Prostatic Neoplasms , Signal Transduction , Apoptosis/drug effects , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Male , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Repositioning/methods , Flucytosine/pharmacology , Flucytosine/chemistry , Coumaric Acids/chemistry , Coumaric Acids/pharmacology , Gallic Acid/chemistry , Gallic Acid/pharmacology , Gallic Acid/analogs & derivatives , Crystallization , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
8.
Biomacromolecules ; 25(7): 4358-4373, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38924782

ABSTRACT

Chitosan (CS)-based photo-cross-linkable hydrogels have gained increasing attention in biomedical applications. In this study, we grafted CS with gallic acid (GA) by carbodiimide chemistry to prepare the GA-CS conjugate, which was subsequently modified with methacrylic anhydride (MA) modification to obtain the methacrylated GA-CS conjugate (GA-CS-MA). Our results demonstrated that the GA-CS-MA hydrogel not only exhibited improved physicochemical properties but also showed antibacterial, antioxidative, and anti-inflammatory capacity. It showed moderate antibacterial activity and especially showed a more powerful inhibitory effect against Gram-positive bacteria. It modulated macrophage polarization, downregulated pro-inflammatory gene expression, upregulated anti-inflammatory gene expression, and significantly reduced reactive oxygen species (ROS) and nitric oxide (NO) production under lipopolysaccharide (LPS) stimulation. Subcutaneously implanted GA-CS-MA hydrogels induced significantly lower inflammatory responses, as evidenced by less inflammatory cell infiltration, thinner fibrous capsule, and predominately promoted M2 polarization. This study provides a feasible strategy to prepare CS-based photo-cross-linkable hydrogels with improved physicochemical properties for biomedical applications.


Subject(s)
Anti-Bacterial Agents , Anti-Inflammatory Agents , Antioxidants , Chitosan , Gallic Acid , Hydrogels , Methacrylates , Chitosan/chemistry , Gallic Acid/chemistry , Gallic Acid/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/chemical synthesis , Mice , Antioxidants/chemistry , Antioxidants/pharmacology , Antioxidants/chemical synthesis , Methacrylates/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , RAW 264.7 Cells , Cross-Linking Reagents/chemistry , Macrophages/drug effects , Macrophages/metabolism , Nitric Oxide/metabolism
9.
Microb Cell Fact ; 23(1): 162, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824548

ABSTRACT

BACKGROUND: Syringic acid (SA) is a high-value natural compound with diverse biological activities and wide applications, commonly found in fruits, vegetables, and herbs. SA is primarily produced through chemical synthesis, nonetheless, these chemical methods have many drawbacks, such as considerable equipment requirements, harsh reaction conditions, expensive catalysts, and numerous by-products. Therefore, in this study, a novel biotransformation route for SA production was designed and developed by using engineered whole cells. RESULTS: An O-methyltransferase from Desulfuromonas acetoxidans (DesAOMT), which preferentially catalyzes a methyl transfer reaction on the meta-hydroxyl group of catechol analogues, was identified. The whole cells expressing DesAOMT can transform gallic acid (GA) into SA when S-adenosyl methionine (SAM) is used as a methyl donor. We constructed a multi-enzyme cascade reaction in Escherichia coli, containing an endogenous shikimate kinase (AroL) and a chorismate lyase (UbiC), along with a p-hydroxybenzoate hydroxylase mutant (PobA**) from Pseudomonas fluorescens, and DesAOMT; SA was biosynthesized from shikimic acid (SHA) by using whole cells catalysis. The metabolic system of chassis cells also affected the efficiency of SA biosynthesis, blocking the chorismate metabolism pathway improved SA production. When the supply of the cofactor NADPH was optimized, the titer of SA reached 133 µM (26.2 mg/L). CONCLUSION: Overall, we designed a multi-enzyme cascade in E. coli for SA biosynthesis by using resting or growing whole cells. This work identified an O-methyltransferase (DesAOMT), which can catalyze the methylation of GA to produce SA. The multi-enzyme cascade containing four enzymes expressed in an engineered E. coli for synthesizing of SA from SHA. The metabolic system of the strain and biotransformation conditions influenced catalytic efficiency. This study provides a new green route for SA biosynthesis.


Subject(s)
Biocatalysis , Escherichia coli , Gallic Acid , Metabolic Engineering , Gallic Acid/metabolism , Gallic Acid/analogs & derivatives , Escherichia coli/metabolism , Escherichia coli/genetics , Metabolic Engineering/methods , Methyltransferases/metabolism , Methyltransferases/genetics , Shikimic Acid/metabolism , Pseudomonas fluorescens/metabolism , Pseudomonas fluorescens/enzymology , Pseudomonas fluorescens/genetics , Biotransformation
10.
J Pharmacol Sci ; 155(4): 140-147, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880548

ABSTRACT

Previously, we have shown that pyrogallol alleviated nasal symptoms and suppressed IL-9 gene up-regulation in allergy model rats by inhibiting calcineurin/NFAT signaling. As pyrogallol has antioxidative activity, it may be responsible for inhibiting calcineurin/NFAT signaling-mediated IL-9 gene expression. However, the relationship between antioxidative activity and suppression of IL-9 gene expression has not been elucidated yet. Here, we conducted the structure-activity relationship studies of pyrogallol and its structurally related compounds to understand the mechanism of IL-9 gene suppression by pyrogallol. 2, 2-Diphenyl-1-picrylhydrazyl radical scavenging assay showed that the antioxidative activity of catechol, resorcinol, phloroglucinol, and gallic acid is 60.1%, 10.4%, 18.8%, and 113.5% of pyrogallol, respectively. Catechol, resorcinol, and phloroglucinol did not suppress NFAT dephosphorylation. Gallic acid suppressed dephosphorylation of NFAT. Gallic acid also suppressed ionomycin-induced up-regulation of IL-9 gene expression with the IC50 value of 82.6 µM. However, catechol, resorcinol and phloroglucinol showed no suppressive activity. In addition, using gallic acid-immobilized beads, we isolated and identified Poly(U)-binding-splicing factor 60 (PUF60) as a pyrogallol binding protein. These results suggest that the antioxidative activity of pyrogallol is not likely to be the mechanism of IL-9 gene suppression. Data also suggest that PUF60 is one of its target molecules responsible for the suppression of calcineurin/NFAT signaling by pyrogallol.


Subject(s)
Antioxidants , Calcineurin , NFATC Transcription Factors , Pyrogallol , Signal Transduction , Pyrogallol/pharmacology , Calcineurin/metabolism , Signal Transduction/drug effects , NFATC Transcription Factors/metabolism , Structure-Activity Relationship , Antioxidants/pharmacology , Humans , Gallic Acid/pharmacology , Gene Expression/drug effects , Animals , Phosphorylation/drug effects , Up-Regulation/drug effects , Rats
11.
J Periodontal Res ; 59(1): 204-219, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37957813

ABSTRACT

BACKGROUND AND OBJECTIVE: Gallic acid (GA) possesses various beneficial functions including antioxidant, anticancer, anti-inflammatory as well as inhibiting osteoclastogeneis. However, effects on osteogenic differentiation, especially in human ligament periodontal (hPDL) cells, remain unclear. Thus, the aim of this study was to evaluate the function of GA on osteogenesis and anti-inflammation in hPDL cells and to explore the involved underlying mechanism. METHODS: Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) treatment was used as a model for periodontitis. ROS production was determined by H2DCFDA staining. Trans-well and wound healing assays were performed for checking the migration effect of GA. Alizarin red and alkaline phosphatase activity (ALP) assays were performed to evaluate osteogenic differentiation. Osteogenesis and inflammatory-related genes and proteins were measured by real-time PCR and western blot. RESULTS: Our results showed that GA-treated hPDL cells had higher proliferation and migration effect. GA inhibited ROS production-induced by Pg-LPS. Besides, GA abolished Pg-LPS-induced inflammation cytokines (il-6, il-1ß) and inflammasome targets (Caspase-1, NLRP3). In addition, GA promoted ALP activity and mineralization in hPDL cells, lead to enhance osteoblast differentiation process. The effect of GA is related to G-protein-coupled receptor 35 (GPR35)/GSK3ß/ß-catenin signaling pathway. CONCLUSION: GA attenuated Pg-LPS-induced inflammatory responses and periodontitis in hPDL cells. Taken together, GA may be targeted for therapeutic interventions in periodontal diseases.


Subject(s)
Osteogenesis , Periodontitis , Humans , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/pharmacology , Periodontal Ligament , beta Catenin/metabolism , Gallic Acid/pharmacology , Gallic Acid/metabolism , Lipopolysaccharides/pharmacology , Reactive Oxygen Species/metabolism , Cells, Cultured , Signal Transduction , Cell Differentiation , Periodontitis/drug therapy , Periodontitis/metabolism , Anti-Inflammatory Agents/pharmacology , Receptors, G-Protein-Coupled/metabolism , Osteoblasts
12.
Mol Biol Rep ; 51(1): 344, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38400882

ABSTRACT

BACKGROUND: There is increasing evidence that honey has anti-inflammatory, antioxidant, and anti-cancer effects. This study aims to assess and contrast the cytotoxic, anti-metastatic, and apoptotic effects of Ziziphus jujube honey and commercial honey on MCF7 cells. METHODS AND RESULTS: Two honey samples, Ziziphus jujube (JH) and commercial honey (CH), were categorized into high and low groups based on their phenolic content, antioxidant capacity, and diastase activity (PAD score). The viability and migration ability of MCF-7 cells treated with JH and CH were evaluated. Also, quantitative polymerase chain reaction (Q-PCR) was performed to assess the effect of the two honey samples on the expression of Bax, p53, p21 and Bcl-2 genes. JH had a total phenolic content of 606.4 ± 0.1 µg gallic acid equivalent/mg, while CH had a value of 112.1 ± 0.09 µg gallic acid equivalent/mg. The total antioxidant capacity of the two samples was compared. It was 203.5 ± 10.5µM/l in JH and 4.6 ± 10.5 µM/l in CH. In addition, JH had a diastatic activity of 524.1 ± 0.25 U/l, while CH had a value of 209.7 ± 0.56 U/l. According to the results, JH had a high PAD value, while CH had a low PAD value. Cell viability was measured using the results of the MTT assay. The results showed that JH inhibited the growth of MCF-7 cells more strongly (IC50 of 170 ± 4.2 µg/ml) than CH (IC50 of 385.3 ± 4.5 µg/l). The scratch assay showed that treatment with JH decreased the migration rate of MCF-7 cells in a dose-dependent manner compared to the CH and control groups. In addition, the results of q-PCR analysis showed significant upregulation of Bax, p53 and p21 genes and downregulation of Bcl-2 gene in the JH-treated group compared to the CH and control groups. CONCLUSION: These results showed that honey with an increased content of phenolic compounds, antioxidant capacity, and diastatic activity has anticancer properties by effectively suppressing tumor development. This suppression occurs via several mechanisms, including suppression of proliferation and metastasis, and promotion of apoptosis.


Subject(s)
Breast Neoplasms , Honey , Ziziphus , Humans , Female , MCF-7 Cells , Breast Neoplasms/drug therapy , Antioxidants/pharmacology , bcl-2-Associated X Protein/genetics , Honey/analysis , Tumor Suppressor Protein p53/genetics , Phenols/pharmacology , Phenols/analysis , Gallic Acid
13.
J Biochem Mol Toxicol ; 38(9): e23809, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39148263

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is an alarming ailment that leads to severe liver damage and increases the risk of serious health conditions. The prevalence of NAFLD due to oxidative stress could be mitigated by plant-derived antioxidants. This study aims to investigate the effects of syringic acid (SA) on NAFLD in a high-fat diet (HFD) rat model. Twenty-four rats were randomly divided into four groups (n = 6): normal control, HFD, SA-administered HFD, and positive control SA on a normal diet. Rats in the normal control and positive control groups received a normal diet, and the remaining groups received an HFD for 8 weeks. SA (20 mg/kg b.w.) was orally (gavage) administered for 8 weeks. Lipid profiles were controlled by SA against HFD-fed rats (p < 0.05). SA reduced the serum aspartate aminotransferase and alanine aminotransferase levels by 70%-190%. SA also suppressed pro-inflammatory cytokines and attenuated histopathological and immunohistochemical changes against HFD-fed rats. SA reversed oxidative stress by suppressing the malondialdehyde formation by 82% and replenished the nonenzymatic and enzymatic antioxidant activities (p < 0.05). Gene expressions of nuclear factor-erythroid 2-related factor/heme oxygenase 1 (Nrf2/HO-1) were elevated in SA-treated rats. Ameliorative effects of SA on NAFLD induced by an HFD in rats were prominent through the reversal of oxidative stress and inflammation, regulated by an intrinsic mechanism of defense against oxidative stress, the Nrf2/HO-1 pathway.


Subject(s)
Gallic Acid , Heme Oxygenase (Decyclizing) , NF-E2-Related Factor 2 , Non-alcoholic Fatty Liver Disease , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Rats , Male , Signal Transduction/drug effects , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Oxidative Stress/drug effects , Heme Oxygenase-1/metabolism , Diet, High-Fat/adverse effects , Rats, Sprague-Dawley , Antioxidants/pharmacology , Liver/metabolism , Liver/drug effects , Liver/pathology
14.
J Appl Microbiol ; 135(1)2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38148145

ABSTRACT

AIMS: To evaluate the antifungal and antibiofilm activity of gallic acid derivatives TPP+-C10 and TPP+-C12 and their effects on mitochondrial function on two Candida albicans reference strains (ATCC 90029 and ATCC 10231). METHODS AND RESULTS: First, we determined minimal inhibitory concentration (MIC) using a microdilution assay. Both compounds exerted antifungal effects, and their MICs ranged from 3.9 to 13 µM, with no statistically significant differences between them (P > 0.05, t-test). These concentrations served as references for following assays. Subsequently, we measured oxygen consumption with a Clark electrode. Our observations revealed that both drugs inhibited oxygen consumption in both strains with TPP+-C12 exerting a more pronounced inhibitory effect. We then employed flow cytometry with TMRE as a probe to assess mitochondrial membrane potential. For each strain assayed, the compounds induced a decay in transmembrane potential by 75%-90% compared to the control condition (P < 0.05, ANOVA). Then, we measured ATP levels using a commercial kit. TPP+-C12 showed a 50% decrease of ATP content (P < 0.05 ANOVA), while TPP+-C10 exhibited a less pronounced effect. Finally, we assessed the antibiofilm effect using the MTT reduction assay. Both compounds were effective, but TPP+-C12 displayed a greater potency, requiring a lower concentration to inhibit 50% of biofilms viability (P < 0.05, t-test). CONCLUSIONS: Derivatives of gallic acid linked to a TPP+ group exert antifungal and antibiofilm activity through impairment of mitochondrial function in C. albicans.


Subject(s)
Antifungal Agents , Candida albicans , Antifungal Agents/pharmacology , Gallic Acid/pharmacology , Microbial Sensitivity Tests , Biofilms , Mitochondria , Adenosine Triphosphate
15.
Bioorg Chem ; 147: 107381, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669781

ABSTRACT

The development of XOD/URAT1 dual target inhibitors has emerged as a promising therapeutic strategy for the management of hyperuricemia. Here, through virtual screening, we have identified digallic acid as a novel dual target inhibitor of XOD/URAT1 and subsequently evaluated its pharmacological properties, pharmacokinetics, and toxicities. Digallic acid inhibited URAT1 with an IC50 of 5.34 ± 0.65 µM, which is less potent than benzbromarone (2.01 ± 0.36 µM) but more potent than lesinurad (10.36 ± 1.23 µM). Docking and mutation analysis indicated that residues S35, F241 and R477 of URAT1 confer a high affinity for digallic acid. Digallic acid inhibited XOD with an IC50 of 1.04 ± 0.23 µM. Its metabolic product, gallic acid, inhibited XOD with an IC50 of 0.91 ± 0.14 µM. Enzyme kinetic studies indicated that both digallic acid and gallic acid act as mixed-type XOD inhibitors. It shares the same binding mode as digallic acid, and residues E802, R880, F914, T1010, N768 and F1009 contribute to their high affinity. The anion group (carboxyl) of digallic acid contribute significantly to its inhibition activity on both XOD and URAT1 as indicated by docking analysis. Remarkably, at a dosage of 10 mg/kg in vivo, digallic acid exhibited a stronger urate-lowering and uricosuric effect compared to the positive drug benzbromarone and lesinurad. Pharmacokinetic study indicated that digallic acid can be hydrolyzed into gallic acid in vivo and has a t1/2 of 0.77 ± 0.10 h. Further toxicity evaluation indicated that digallic acid exhibited no obvious renal toxicity, as reflected by CCK-8, biochemical analysis (CR and BUN) and HE examination. The findings of our study can provide valuable insights for the development of XOD/URAT1 dual target inhibitors, and digallic acid deserves further investigation as a potential anti-hyperuricemic drug.


Subject(s)
Dose-Response Relationship, Drug , Enzyme Inhibitors , Hyperuricemia , Organic Anion Transporters , Organic Cation Transport Proteins , Hyperuricemia/drug therapy , Humans , Animals , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Structure-Activity Relationship , Molecular Structure , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Urate Oxidase/chemistry , Drug Discovery , Molecular Docking Simulation , Mice , Male , Gallic Acid/chemistry , Gallic Acid/pharmacology , Gallic Acid/analogs & derivatives , Rats, Sprague-Dawley
16.
Nutr Neurosci ; 27(3): 271-288, 2024 Mar.
Article in English | MEDLINE | ID: mdl-36947578

ABSTRACT

OBJECTIVES: In this study mice were fed a high-fat diet for 12 weeks to establish diet-induced obesity and syringic acid (SA) was assessed for anti-obese, neuroprotective, and neurogenesis. METHOD: Animals were given HFD for 12 weeks to measure metabolic characteristics and then put through the Barns-maze and T-maze tests to measure memory. Additionally, the physiology of the blood-brain barrier, oxidative stress parameters, the expression of inflammatory genes, neurogenesis, and histopathology was evaluated in the brain. RESULT: DIO raised body weight, BMI, and other metabolic parameters after 12 weeks of overfeeding. A reduced spontaneous alternation in behavior (working memory, reference memory, and total time to complete a task), decreased enzymatic and non-enzymatic antioxidants, oxidative biomarkers, increased neurogenesis, and impaired blood-brain barrier were all seen in DIO mice. SA (50 mg/kg) treatment of DIO mice (4 weeks after 8 weeks of HFD feeding) reduced diet-induced changes in lipid parameters associated with obesity, hepatological parameters, memory, blood-brain barrier, oxidative stress, neuroinflammation, and neurogenesis. SA also reduced the impact of malondialdehyde and enhanced the effects of antioxidants such as glutathione, superoxide dismutase (SOD), and total thiol (MDA). Syringic acid improved neurogenesis, cognition, and the blood-brain barrier while reducing neurodegeneration in the hippocampal area. DISCUSSION: According to the results of the study, syringic acid therapy prevented neurodegeneration, oxidative stress, DIO, and memory loss. Syringic acid administration may be a useful treatment for obesity, memory loss, and neurogenesis, but more research and clinical testing is needed.


Subject(s)
Gallic Acid/analogs & derivatives , Obesity , Overweight , Mice , Animals , Obesity/etiology , Obesity/prevention & control , Obesity/drug therapy , Brain/metabolism , Antioxidants/pharmacology , Diet, High-Fat/adverse effects , Oxidative Stress , Memory Disorders , Neurogenesis/physiology
17.
J Nanobiotechnology ; 22(1): 98, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38461231

ABSTRACT

Chemodynamic therapy (CDT) based on intracellular Fenton reaction to produce highly cytotoxic reactive oxygen species (ROS) has played an essential role in tumor therapy. However, this therapy still needs to be improved by weakly acidic pH and over-expression of glutathione (GSH) in tumor microenvironment (TEM), which hinders its future application. Herein, we reported a multifunctional bimetallic composite nanoparticle MnO2@GA-Fe@CAI based on a metal polyphenol network (MPN) structure, which could reduce intracellular pH and endogenous GSH by remodeling tumor microenvironment to improve Fenton activity. MnO2 nanoparticles were prepared first and MnO2@GA-Fe nanoparticles with Fe3+ as central ion and gallic acid (GA) as surface ligands were prepared by the chelation reaction. Then, carbonic anhydrase inhibitor (CAI) was coupled with GA to form MnO2@GA-Fe@CAI. The properties of the bimetallic composite nanoparticles were studied, and the results showed that CAI could reduce intracellular pH. At the same time, MnO2 could deplete intracellular GSH and produce Mn2+ via redox reactions, which re-established the TME with low pH and GSH. In addition, GA reduced Fe3+ to Fe2+. Mn2+ and Fe2+ catalyzed the endogenous H2O2 to produce high-lever ROS to kill tumor cells. Compared with MnO2, MnO2@GA-Fe@CAI could reduce the tumor weight and volume for the xenograft MDA-MB-231 tumor-bearing mice and the final tumor inhibition rate of 58.09 ± 5.77%, showing the improved therapeutic effect as well as the biological safety. Therefore, this study achieved the high-efficiency CDT effect catalyzed by bimetallic through reshaping the tumor microenvironment.


Subject(s)
Nanoparticles , Neoplasms , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Hydrogen Peroxide , Manganese Compounds/pharmacology , Reactive Oxygen Species , Oxides , Gallic Acid , Glutathione , Hydrogen-Ion Concentration , Cell Line, Tumor , Tumor Microenvironment
18.
Biochemistry (Mosc) ; 89(1): 173-183, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38467553

ABSTRACT

Natural polyphenols are promising compounds for the pharmacological control of oxidative stress in various diseases. However, low bioavailability and rapid metabolism of polyphenols in a form of glycosides or aglycones have stimulated the search for the vehicles that would provide their efficient delivery to the systemic circulation. Conjugation of polyphenols with cationic amphiphilic peptides yields compounds with a strong antioxidant activity and ability to pass through biological barriers. Due to a broad range of biological activities characteristic of polyphenols and peptides, their conjugates can be used in the antioxidant therapy, including the treatment of viral, oncological, and neurodegenerative diseases. In this work, we synthesized linear and dendrimeric cationic amphiphilic peptides that were then conjugated with gallic acid (GA). GA is a non-toxic natural phenolic acid and an important functional element of many flavonoids with a high antioxidant activity. The obtained GA-peptide conjugates showed the antioxidant (antiradical) activity that exceeded 2-3 times the antioxidant activity of ascorbic acid. GA attachment had no effect on the toxicity and hemolytic activity of the peptides. GA-modified peptides stimulated the transmembrane transfer of the pGL3 plasmid encoding luciferase reporter gene, although GA attachment at the N-terminus of peptides reduced their transfection activity. Several synthesized conjugates demonstrated the antibacterial activity in the model of Escherichia coli Dh5α growth inhibition.


Subject(s)
Antioxidants , Polyphenols , Antioxidants/pharmacology , Antioxidants/chemistry , Polyphenols/pharmacology , Polyphenols/chemistry , Peptides/pharmacology , Peptides/chemistry , Gallic Acid/pharmacology , Gallic Acid/chemistry , Anti-Bacterial Agents/chemistry
19.
J Infect Chemother ; 30(9): 867-875, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38462174

ABSTRACT

INTRODUCTION: Gallic acid (GA) has a good therapeutic effect in bacteriological inhibition and plays a variety of functions in maintaining the stability of the immune system. The aim of the present study was to investigate the effect of GA on the bactericidal activity of macrophages against Vibrio vulnificus (Vv). METHODS: A cell counting kit-8 (CCK-8) assay was carried out to test the cytotoxicity of GA on J774A.1 cells. Concentration of proinflammatory cytokines in J774A.1 cells were evaluated by ELISA. The internalization and degradation of Vv in the phagosomes were observed by transmission electron microscopy (TEM). The phagosome acidification and phagolysosome formation were detected to evaluate the bacteria-clearing function of J774A.1 cells. The bactericidal activity of GA in vivo was also investigated by collecting the survival time of Vv infected mice and observing the inflammatory infiltration of organs. RESULTS: Our results demonstrated that GA at 50 µM significantly inhibited the proinflammatory cytokines levels, promoted phagosome acidification and phagolysosome formation in J774A.1 cells with Vv infection. This may be related to the activation of NLRP3/mTOR signaling pathway. Additionally, GA treatment improves the survival and bactericidal activity of mice infected with Vv. CONCLUSIONS: In summary, GA exerts bactericidal activity against Vv infection by regulating the formation and acidification of phagocytic lysosomes in macrophages.


Subject(s)
Gallic Acid , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Phagosomes , Signal Transduction , TOR Serine-Threonine Kinases , Vibrio vulnificus , Gallic Acid/pharmacology , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Mice , Signal Transduction/drug effects , Macrophages/drug effects , Macrophages/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Vibrio vulnificus/drug effects , Cell Line , Cytokines/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Female
20.
Biofouling ; 40(1): 64-75, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38373897

ABSTRACT

Aeromonas hydrophila, a Gram-negative zoonotic bacterium, causes high mortality in fish farming and immunocompromised patients. This study aimed to extract methyl gallate (MG) from the flowers of Camellia nitidissima Chi and evaluate its potential as a quorum sensing inhibitor (QSI) against Aeromonas hydrophila SHAe 115. MG reduced QS-associated virulence factors, including hemolysis, protease, and lipase, while impairing swimming motility and biofilm formation. Additionally, MG down-regulated positive regulatory genes (ahyR, fleQ) and up-regulated negative regulators (litR, fleN). This highlights MG's promise as a potent QSI for A. hydrophila SHAe 115, advancing strategies against infections in aquaculture and human health.


Subject(s)
Biofilms , Gallic Acid/analogs & derivatives , Quorum Sensing , Animals , Humans , Quorum Sensing/genetics , Virulence/genetics , Aeromonas hydrophila/genetics , Virulence Factors/genetics , Bacterial Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL