Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.507
Filter
1.
Mar Drugs ; 22(9)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39330298

ABSTRACT

The role of oxidative stress in health and homeostasis has generated interest in the scientific community due to its association with cardiovascular and neurodegenerative diseases, cancer, and other diseases. Therefore, extensive research seeks to identify new exogenous antioxidant compounds for supplementation. Polysaccharides are recognized for their antioxidant properties. However, polysaccharide chemical modifications are often necessary to enhance these properties. Therefore, dextran was conjugated with gallic acid (Dex-Gal) and later combined with fucoidan A (FucA) to formulate blends aimed at achieving superior antioxidant activity compared to individual polysaccharides. A factorial design was employed to combine FucA and Dex-Gal in different proportions, resulting in five blends (BLD1, BLD2, BLD3, BLD4, and BLD5). An analysis of surface graphs from in vitro antioxidant tests, including total antioxidant capacity (TAC), reducing power, and hydroxyl radical scavenging, guided the selection of BLD4 as the optimal formulation. Tests on 3T3 fibroblasts under various conditions of oxidative stress induced by hydrogen peroxide revealed that BLD4 provided enhanced protection compared to its isolated components. The BLD4 formulation, resulting from the combination of Dex-Gal and FucA, showed promise as an antioxidant strategy, outperforming its individual components and suggesting its potential as a supplement to mitigate oxidative stress in adverse health conditions.


Subject(s)
Antioxidants , Dextrans , Gallic Acid , Oxidative Stress , Polysaccharides , Polysaccharides/pharmacology , Polysaccharides/chemistry , Gallic Acid/pharmacology , Gallic Acid/chemistry , Dextrans/chemistry , Dextrans/pharmacology , Antioxidants/pharmacology , Antioxidants/chemistry , Animals , Mice , Oxidative Stress/drug effects , 3T3 Cells , Hydrogen Peroxide , Fibroblasts/drug effects
2.
Anticancer Res ; 44(10): 4317-4326, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39348974

ABSTRACT

BACKGROUND/AIM: Methyl gallate (MG), a plant phenolic compound, has known anticancer properties. However, its effects on canine mammary gland tumors (CMTs) are unclear. This study evaluated the impact of MG on cell viability, migration, and apoptosis in two CMT cell lines. MATERIALS AND METHODS: CMT-U27 and CF41.mg cells were used. In vitro experiments included MTT and scratch assays, Annexin-V/propidium iodide double staining, immunocytochemistry, and western blot analyses. An in vivo CMT xenograft mouse model was also used to observe the effects of MG on tumor growth and vasculature. Immunohistochemistry was performed to analyze vessel density and apoptosis in tumor tissues. Cell migration and tube formation assays with canine aortic endothelial cells assessed the anti-angiogenic effects of MG. RESULTS: Data showed a significant decrease in cell viability and migration in both CMT cell lines after 24 h exposure to various MG concentrations. MG treatment induced dose-dependent apoptotic cell death and elevated cleaved caspase-3 expression. In vivo experiments confirmed tumor growth suppression 21 days post-treatment with 40 mg/kg MG. Tumor tissues displayed increased cleaved caspase-3 and reduced vessel density. MG also inhibited cell migration and disrupted tube formation in canine endothelial cells. CONCLUSION: MG has potential as an anticancer drug for CMTs by promoting apoptotic cell death and reducing angiogenesis, highlighting its therapeutic promise.


Subject(s)
Angiogenesis Inhibitors , Apoptosis , Cell Movement , Cell Survival , Gallic Acid , Neovascularization, Pathologic , Xenograft Model Antitumor Assays , Animals , Dogs , Apoptosis/drug effects , Female , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Cell Movement/drug effects , Angiogenesis Inhibitors/pharmacology , Cell Line, Tumor , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Cell Survival/drug effects , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Cell Proliferation/drug effects
3.
Chin J Nat Med ; 22(9): 797-807, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39326974

ABSTRACT

The Chinese herb Ephedra (also known as Mahuang) has been extensively utilized for the prevention and treatment of coronavirus-induced diseases, including coronavirus disease 2019 (COVID-19). However, the specific anti-SARS-CoV-2 compounds and mechanisms have not been fully elucidated. The main protease (Mpro) of SARS-CoV-2 is a highly conserved enzyme responsible for proteolytic processing during the viral life cycle, making it a critical target for the development of antiviral therapies. This study aimed to identify naturally occurring covalent inhibitors of SARS-CoV-2 Mpro from Ephedra and to investigate their covalent binding sites. The results demonstrated that the non-alkaloid fraction of Ephedra (ENA) exhibited a potent inhibitory effect against the SARS-CoV-2 Mpro effect, whereas the alkaloid fraction did not. Subsequently, the chemical constituents in ENA were identified, and the major constituents' anti-SARS-CoV-2 Mpro effects were evaluated. Among the tested constituents, herbacetin (HE) and gallic acid (GA) were found to inhibit SARS-CoV-2 Mpro in a time- and dose-dependent manner. Their combination displayed a significant synergistic effect on this key enzyme. Additionally, various techniques, including inhibition kinetic assays, chemoproteomic methods, and molecular dynamics simulations, were employed to further elucidate the synergistic anti-Mpro mechanisms of the combination of HE and GA. Overall, this study deciphers the naturally occurring covalent inhibitors of SARS-CoV-2 Mpro from Ephedra and characterizes their synergistic anti-Mpro synergistic effect, providing robust evidence to support the anti-coronavirus efficacy of Ephedra.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Ephedra , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Ephedra/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , Humans , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Gallic Acid/pharmacology , Gallic Acid/chemistry , COVID-19 Drug Treatment , Molecular Docking Simulation , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Flavonoids
4.
Int J Pharm ; 665: 124672, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39245084

ABSTRACT

Dasatinib (DAB) has been explored for repurposing in the treatment of breast cancer (BC) due to its known effectiveness in treating leukemia, in addition to its role as a tyrosine kinase inhibitor. Gallic acid (GA) was chosen as a co-former due to its anticancer potential in BC, as demonstrated in several previous studies. DAB is a low-solubility drug, which is a significant hurdle for its oral bioavailability. To address this limitation, a DAB and GA co-amorphous (DAB-GA-CA) system was developed using liquid-assisted grinding and ball mill technology to enhance solubility, bioavailability, and anti-tumor efficacy. Physical characterization investigation revealed that the emergence of the halo diffractogram in PXRD, single glass transition temperature (Tg) value at 111.7 °C in DSC thermogram, and irregularly shaped blocks with loose, porous surfaces in SEM analysis indicated the formation of the DAB-GA-CA system at 1:1 M ratio. Furthermore, FTIR, Raman spectroscopy, in-silico molecular docking, and molecular dynamic studies confirmed the intermolecular hydrogen connections between DAB and GA. Moreover, the outcomes of the ligands (DAB and GA) and receptors (BCL-2, mTOR, estrogen receptor, and HER-2) docking studies demonstrated that both DAB and GA could interact with those receptors, leading to preventive action on BC cells. Additionally, the solubility and dissolution rate significantly improved at pH 6.8, and the permeability study indicated that DAB-GA-CA showed 1.9 times higher apparent permeability compared to crystalline DAB. Furthermore, in vitro cytotoxicity assessments of the DAB-GA-CA system revealed 3.42 times lower IC50 than free DAB. The mitochondrial membrane depolarization, apoptotic index, and reactive oxygen species formation in MCF-7 cells were also notably higher in the DAB-GA-CA system than in free DAB. Hence, this research suggests that the DAB-GA-CA system could substantially enhance oral delivery, solubility, and therapeutic efficacy.


Subject(s)
Antineoplastic Agents , Dasatinib , Gallic Acid , Molecular Docking Simulation , Solubility , Gallic Acid/chemistry , Gallic Acid/pharmacology , Gallic Acid/administration & dosage , Dasatinib/pharmacology , Dasatinib/chemistry , Dasatinib/administration & dosage , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , MCF-7 Cells , Permeability , Drug Liberation , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Survival/drug effects , Biological Availability , Computer Simulation , Female
5.
J Mater Chem B ; 12(36): 9074-9097, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39171375

ABSTRACT

In this study, we employed a combination of electrospinning and electrospray techniques to fabricate wound dressings with a particle-fiber structure, providing dual characteristics of oxygen-releasing and intrinsic antioxidant properties, simultaneously. The electrospun part of the dressing was prepared from a blend of polycaprolactone/gallic acid-grafted-gelatin (GA-g-GE), enabling intrinsic ROS scavenging. To the best of our knowledge, this is the first time that PCL/GA-g-GE was fabricated by electrospinning. Furthermore, polyvinyl pyrrolidone (PVP) microparticles, containing calcium peroxide nanoparticles (CNPs), were considered as the oxygen production agent through the electrospray part. The CNP content was 1% and 3% w/w of PVP while biopolymer:PCL was 10% w/w. The fabricated structures were characterized in terms of fiber/particle morphology, elemental analysis, oxygen release behavior, ROS inhibition capacity, and water contact angle assessments. The covalent bonding of gallic acid to gelatin was confirmed by 1H-NMR, UV spectroscopy, and FTIR. According to the SEM results, the morphology of the prepared PCL/biopolymer fibers was bead-free and with a uniform average diameter. The analysis of released oxygen showed that by increasing the weight percentage of CNPs from 1 to 3 wt%, the amount of released oxygen increased from 120 mmHg to 195 mmHg in 24 h, which remained almost constant until 72 h. The obtained DPPH assay results revealed that the introduction of GA-g-GE into the fibrous structure could significantly improve the antioxidant properties of wound dressing compared to the control group without CNPs and modified gelatine. In vitro, the fabricated wound dressings were evaluated in terms of biocompatibility and the potential of the dressing to protect human dermal fibroblasts under oxidative stress and hypoxia conditions by an MTT assay. The presence of GA-g-GE led to remarkable protection of the cells against oxidative stress and hypoxia conditions. In vivo studies revealed that the incorporation of intrinsic ROS inhibition and oxygen-releasing properties could significantly accelerate the wound closure rate during the experimental period (7, 14, and 21 days). Additionally, histopathological investigations in terms of H&E and Masson's trichrome staining showed that the incorporation of the two mentioned capabilities remarkably facilitated the wound-healing process.


Subject(s)
Antioxidants , Bandages , Oxygen , Polyesters , Antioxidants/chemistry , Antioxidants/pharmacology , Oxygen/chemistry , Animals , Polyesters/chemistry , Gallic Acid/chemistry , Gallic Acid/pharmacology , Gelatin/chemistry , Particle Size , Wound Healing/drug effects , Rats , Reactive Oxygen Species/metabolism , Humans , Nanoparticles/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemical synthesis
6.
Ultrastruct Pathol ; 48(5): 351-366, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39105544

ABSTRACT

Tebuconazole (TEB) is a common triazole sterol demethylation inhibitor fungicide utilized to manage a variety of diseases in crops like cereals, fruits, and vegetables. The aim of this work was to assess the effects of TEB on the structure of the cerebellum in adult albino rats and possible protective impact of co-administration of Gallic acid (GA). Four groups of forty adult male albino rats were randomly selected, and the rats in group I received corn oil through daily gavage for 4 weeks. Group II received GA dissolved in the normal saline at a dose of 100 mg/kg through daily gavage for 4 weeks, group III administered with TEB dissolved in corn oil at its acceptable daily intake dose (0.02 mg/kg body weight) through daily gavage for 4 weeks, group IV rats received both TEB and GA. For light microscopic, ultrastructural, and immunohistochemical investigations, cerebellar specimens were prepared. TEB exposure led to neuronal damage in the form of degenerated Purkinje cells with vacuolated cytoplasm, areas of lost Purkinje cells, the basket cells appeared vacuolated with degenerated neuropil, the granule cells clumped with congested areas between them, dilated cerebellar islands, weak positive bcl2 immunoreactions in the Purkinje cells, and numerous GFAP-positive astrocytes. GA mitigated TEB-mediated histological changes in the cerebellar cortex. We concluded that TEB caused Purkinje neurons in the rat cerebellar cortex to degenerate and undergo apoptosis. GA had a neuroprotective benefit against TEB toxicity in the rat cerebellar cortex.


Subject(s)
Cerebellum , Fungicides, Industrial , Gallic Acid , Triazoles , Animals , Male , Rats , Cerebellum/drug effects , Cerebellum/pathology , Gallic Acid/pharmacology , Triazoles/pharmacology , Triazoles/toxicity , Fungicides, Industrial/toxicity , Immunohistochemistry , Neuroprotective Agents/pharmacology
7.
Biomacromolecules ; 25(9): 6026-6037, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39137337

ABSTRACT

Intracellular pathogens can survive inside the macrophages to protect themselves from eradication by the innate immune system and conventional antibiotics, resulting in severe bacterial infections. In this work, an antibiotic-free nanocomplex (HA/GA-Fe@NO-DON), exhibiting macrophage-targeted synergistic gas therapy (nitric oxide, NO)/chemodynamic therapy/immunotherapy, was reported. HA/GA-Fe nanoparticles were synthesized by the strong coordination interactions among carboxyl groups of hyaluronic acid (HA), polyphenol groups of gallic acid (GA), and Fe(II) ions. The hydrophobic glutathione (GSH)-responsive NO donor (NO-DON) was encapsulated in HA/GA-Fe nanoparticles to form the final nanocomplexes (HA/GA-Fe@NO-DON). HA on the nanocomplexes guides the macrophage-specific uptake and intracellular accumulation. After the uptake, HA/GA-Fe@NO-DON nanocomplexes could not only generate highly toxic hydroxyl radicals (•OH) by the Fenton reaction and GSH depletion but also release NO when stimulated by intracellular GSH. Meanwhile, the nanocomplexes could trigger an efficient proinflammation immune response to reinforce the antibacterial activity. This work presents the development of antibiotic-free macrophage-targeted HA/GA-Fe@NO-DON nanocomplexes as an effective adjuvant nanomedicine with synergistic gas therapy/chemodynamic therapy/immunotherapy for eliminating intracellular bacterial infection.


Subject(s)
Gallic Acid , Glutathione , Macrophages , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Mice , Animals , Glutathione/chemistry , Glutathione/metabolism , RAW 264.7 Cells , Gallic Acid/chemistry , Gallic Acid/pharmacology , Immunotherapy/methods , Nitric Oxide/metabolism , Nitric Oxide/chemistry , Hyaluronic Acid/chemistry , Bacterial Infections/drug therapy , Nanoparticles/chemistry , Iron/chemistry
8.
Phytomedicine ; 134: 155939, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39214016

ABSTRACT

BACKGROUND: Atherosclerosis (AS) is the most prevalent cardiovascular disease and remains the major contributor to death and mortality globally. Leonurine (LEO) is a unique alkaloid compound with protective effects on the cardiovascular system. However, the exact mechanisms underlying its cardiovascular-protecting action are still not fully elucidated. The methyltransferase 3 (METTL3), the catalytic core of the N6-methyladenosine modification (m6A) methyltransferase complex, has been shown to inhibit autophagy and exacerbate the process of AS via regulation of m6A modification of mRNA. PURPOSE: We aimed to determine whether the inhibited effect of LEO on AS is related to METTL3-mediated AKT1S1 stability. METHODS: The apolipoprotein E (ApoE) knockout mice was subjected to a high-fat diet (HFD), and THP-1 derived macrophages was exposed to oxidized low-density lipoprotein (ox-LDL), to establish the animal and cellular models of AS, respectively. RESULTS: We found that LEO effectively improved AS and reduced the plaque area and inflammation via diminishing macrophage lipid accumulation and remodeling the lipid metabolism profile. LEO activated ox-LDL-induced macrophage autophagy, enhancing lipid metabolism decrease, according to the lipidomic and molecular biology analyses. Additionally, LEO caused a marked increase in autophagy marker levels in mouse models with advanced AS. Furthermore, we found that LEO reactivated autophagy and reversed lipid accumulation by suppressing METTL3 expression. The m6A-seq from ox-LDL-induced macrophages showed that a total of five autophagy-related mRNA transcripts (AKT1S1, AKT1, RB1CC1, CFLAR, and MTMR4) were altered, and AKT1S1 was significantly upregulated by LEO. Mechanistically, LEO-mediated regulation of METTL3 decreased AKT1S1 expression by attenuating its mRNA stability. Silencing AKT1S1 inhibited LEO-METTL3 axis-mediated autophagy and enhanced lipid accumulation in ox-LDL-induced macrophages. CONCLUSION: The study first revealed that LEO exerts anti-atherosclerotic effect by activating METTL3-mediated macrophage autophagy in vivo and in vitro. The mechanism of LEO was further found to be the enhancement of METTL3-mediated AKT1S1 stability to activate autophagy thereby reducing lipid accumulation. This study provides a new perspective of natural medicines on the treatment of AS via an epigenetic manner.


Subject(s)
Atherosclerosis , Autophagy , Foam Cells , Gallic Acid , Lipoproteins, LDL , Methyltransferases , Proto-Oncogene Proteins c-akt , Animals , Methyltransferases/metabolism , Atherosclerosis/drug therapy , Autophagy/drug effects , Mice , Humans , Male , Proto-Oncogene Proteins c-akt/metabolism , Foam Cells/drug effects , Gallic Acid/pharmacology , Gallic Acid/analogs & derivatives , RNA Stability/drug effects , Lipid Metabolism/drug effects , Diet, High-Fat , Mice, Inbred C57BL , Mice, Knockout, ApoE , Disease Models, Animal , THP-1 Cells , RNA, Messenger/metabolism
9.
Nanomedicine (Lond) ; 19(25): 2065-2083, 2024.
Article in English | MEDLINE | ID: mdl-39143900

ABSTRACT

Aim: To developed and investigate gallic acid (GA) loaded self-nanoemulsifying drug delivery systems (SNEDDS) for treating onychomycosis via transungual route.Materials & methods: The SNEDDS were prepared by direct dispersion technique and were evaluated for characteristics parameters using Fourier transform infrared, differential scanning calorimetry, confocal microscopy, transmission electron microscopy and zeta sizer. Furthermore, the safety of prepared formulation was evaluated via Hen's egg test-chorioallantoic membrane study and stability was confirmed using different parameters. Also, its effectiveness was evaluated against fungal strain Trichophyton mentagrophytes.Results: The SNEDDS displayed a particle size of 199.8 ± 4.21 nm and a zeta potential; of -22.75 ± 2.09 mV. Drug release study illustrated a sustained release pattern with a release of 70.34 ± 0.20% over a period of 24 h. The penetration across the nail plate was found to be 1.59 ± 0.002 µg/mg and 0.97 ± 0.001 µg/mg for GA loaded SNEDDS and GA solution respectively. An irritation score of 0.52 ± 0.005 and 3.84 ± 0.001 was reported for GA loaded SNEDDS hydrogel and GA solution, indicating a decrease in the drug's irritation potential from slightly irritating to non irritating due to its entrapment within the SNEDDS.Conclusion: GA loaded SNEDDS has potential to address limitations of conventional treatments, enhancing the drug's efficacy and reducing the likelihood of resistance in the treatment of Onychomycosis.


[Box: see text].


Subject(s)
Antifungal Agents , Drug Delivery Systems , Drug Liberation , Emulsions , Gallic Acid , Hydrogels , Onychomycosis , Particle Size , Onychomycosis/drug therapy , Gallic Acid/chemistry , Gallic Acid/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/administration & dosage , Hydrogels/chemistry , Humans , Emulsions/chemistry , Animals , Trichophyton/drug effects , Nanoparticles/chemistry , Drug Carriers/chemistry
10.
J Control Release ; 374: 525-537, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39173954

ABSTRACT

Inflammation-related diseases are often marked by elevated levels of nitric oxide (NO) and reactive oxygen species (ROS), which play important roles in the modulation of inflammation. However, the development of organic materials effective in managing NO/ROS levels has remained a challenge. This study introduces a novel organic compound, NmeGA, engineered to scavenge both NO and ROS. NmeGA ingeniously integrates N-methyl-1,2,-phenylenediamine (Nme), a NO scavenger, with gallic acid (GA), a ROS scavenger, through an amide bond, endowing it with enhanced scavenging capabilities over its individual component. This compound exhibits reduced toxicity and increased lipophilicity value, underlining its increased biological applicability and highlighting its potential as an inflammation management tool. Through in vitro studies on lipopolysaccharide (LPS)-stimulated RAW 264.7 cells, NmeGA displayed remarkable scavenging efficiency for NO and ROS, coupled with significant anti-inflammatory effects. In an LPS-induced peritonitis model, administration of NmeGA substantially decreased mortality rates, NO and ROS levels, and inflammatory cytokine concentrations. These findings highlight NmeGA's versatility as a therapeutic agent against various inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents , Free Radical Scavengers , Inflammation , Lipopolysaccharides , Nitric Oxide , Reactive Oxygen Species , Animals , Reactive Oxygen Species/metabolism , Nitric Oxide/metabolism , Mice , RAW 264.7 Cells , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Free Radical Scavengers/pharmacology , Free Radical Scavengers/administration & dosage , Free Radical Scavengers/therapeutic use , Phenylenediamines/pharmacology , Phenylenediamines/chemistry , Gallic Acid/pharmacology , Gallic Acid/chemistry , Gallic Acid/therapeutic use , Peritonitis/drug therapy , Peritonitis/chemically induced , Male , Cytokines/metabolism
11.
J Ovarian Res ; 17(1): 178, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39217393

ABSTRACT

Oocyte aging is a key constraint on oocyte quality, leading to fertilization failure and abnormal embryonic development. In addition, it is likely to generate unfavorable assisted reproductive technology (ART) outcomes. SCM-198, a synthetic form of leonurine, was found to rescue the rate of oocyte fragmentation caused by postovulatory aging. Therefore, the aim of this study was to conduct a more in-depth investigation of SCM-198 by exploring its relationship with aged oocytes after ovulation or maternal aging and clarifying whether it affects cell quality. The results indicate that, compared to the postovulatory aged group, the 50 µM SCM-198 group significantly improved sperm-egg binding and increased fertilization of aged oocytes, restoring the spindle apparatus/chromosome structure, cortical granule distribution, and ovastacin and Juno protein distribution. The 50 µM SCM-198 group showed significantly normal mitochondrial distribution, low levels of reactive oxygen species (ROS), and a small quantity of early oocyte apoptosis compared to the postovulatory aged group. Above all, in vivo supplementation with SCM-198 effectively eliminated excess ROS and reduced the spindle/chromosome structural defects in aged mouse oocytes. In summary, these findings indicate that SCM-198 inhibits excessive oxidative stress in oocytes and alters oocyte quality both in vitro and in vivo.


Subject(s)
Gallic Acid , Oocytes , Ovulation , Oxidative Stress , Reactive Oxygen Species , Oocytes/metabolism , Oocytes/drug effects , Animals , Oxidative Stress/drug effects , Female , Mice , Reactive Oxygen Species/metabolism , Ovulation/drug effects , Gallic Acid/pharmacology , Gallic Acid/analogs & derivatives , Cellular Senescence/drug effects , Apoptosis , Male
12.
Int J Biol Macromol ; 277(Pt 3): 134343, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39097059

ABSTRACT

Gallic acid is a vegetable-derived and highly bioactive phenolic acid, but its antioxidant capacity is sensitive to environmental conditions. Chitosan is a biopolymer capable of exerting significant protection to various molecules, including phenolic compounds. A chitosan derivative that extends the antioxidant activity of gallic acid was synthesized by click chemistry and characterized by FT-IR, 1H NMR, and antioxidant capacity assays. Our results show that synthesized polymeric solutions and nanoparticles of N-(gallic acid) chitosan were both internalized by rat brain cells, processes that were modulated by extracellular Ca2+ and Na+. Their internalization was supported by dynamic light scattering and ζ-potential analyses, while Ca2+ imaging recordings performed in brain cells revealed the potential biological effect of N-(gallic acid) chitosan. We conclude that the synthesis of an N-(gallic acid) chitosan derivative through click chemistry is viable and may serve as strategy to prolong its antioxidant activity and to study its biological effects in vivo.


Subject(s)
Antioxidants , Brain , Calcium , Chitosan , Gallic Acid , Chitosan/chemistry , Chitosan/pharmacology , Animals , Gallic Acid/chemistry , Gallic Acid/pharmacology , Gallic Acid/analogs & derivatives , Rats , Antioxidants/pharmacology , Antioxidants/chemistry , Brain/metabolism , Brain/drug effects , Calcium/metabolism , Nanoparticles/chemistry
13.
Int J Nanomedicine ; 19: 8541-8553, 2024.
Article in English | MEDLINE | ID: mdl-39185347

ABSTRACT

Background: Chemodynamic therapy (CDT) faces challenges of low catalytic ion efficiency and ROS production. We developed a ROS nano-bomb, Cu/ZIF-8@GA-Fe, to address these issues. Methods: The nano-bomb was synthesized by doping copper into ZIF-8 and assembling Fe3+ and gallic acid (GA). It was tested for reactive oxygen species (ROS) generation in acidic conditions and its photothermal properties. Results: In an acidic micro environment, Cu/ZIF-8@GA-Fe effectively released Fe3+ and Cu2+, depleting GSH and generating ROS. The GA-Fe coating provided photothermal heat and was used to enhance Fenton reactions via dual ions for increasing ROS production. In vivo and in vitro experiments, Cu/ZIF-8@GA-Fe inhibited tumor growth with minimal side effects. Conclusion: Cu/ZIF-8@GA-Fe shows promise for safe and effective CDT, offering a synergistic approach to tumor therapy.


Subject(s)
Copper , Gallic Acid , Glutathione , Reactive Oxygen Species , Gallic Acid/chemistry , Gallic Acid/pharmacology , Copper/chemistry , Copper/pharmacology , Animals , Glutathione/chemistry , Reactive Oxygen Species/metabolism , Humans , Mice , Cell Line, Tumor , Photothermal Therapy/methods , Iron/chemistry , Neoplasms/therapy , Neoplasms/drug therapy , Mice, Inbred BALB C , Metal Nanoparticles/chemistry , Combined Modality Therapy , Female , Xenograft Model Antitumor Assays
14.
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
15.
Nutrients ; 16(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125398

ABSTRACT

Persimmon fruit processing-derived waste and by-products, such as peels and pomace, are important sources of dietary fiber and phytochemicals. Revalorizing these by-products could help promote circular nutrition and agricultural sustainability while tackling dietary deficiencies and chronic diseases. In this study, fiber-rich fractions were prepared from the by-products of Sharoni and Brilliant Red persimmon varieties. These fractions were quantified for their phenolic composition and assessed for their ability to promote the growth of beneficial human colonic Firmicutes species and for their in vitro anti-inflammatory potential. Gallic and protocatechuic acids, delphinidin, and cyanidin were the main phenolics identified. Faecalibacterium prausnitzii strains showed significantly higher growth rates in the presence of the Brilliant Red fraction, generating more than double butyrate as a proportion of the total short-chain fatty acids (39.5% vs. 17.8%) when compared to glucose. The fiber-rich fractions significantly decreased the inflammatory effect of interleukin-1ß in Caco-2 cells, and the fermented fractions (both from Sharoni and Brilliant Red) significantly decreased the inflammatory effect of interleukin-6 and tumor necrosis factor-α in the RAW 264.7 cells. Therefore, fiber-rich fractions from persimmon by-products could be part of nutritional therapies as they reduce systemic inflammation, promote the growth of beneficial human gut bacteria, and increase the production of beneficial microbial metabolites such as butyrate.


Subject(s)
Anti-Inflammatory Agents , Colon , Dietary Fiber , Diospyros , Humans , Dietary Fiber/pharmacology , Dietary Fiber/analysis , Diospyros/chemistry , Mice , Anti-Inflammatory Agents/pharmacology , Colon/microbiology , Colon/drug effects , Colon/metabolism , Animals , RAW 264.7 Cells , Caco-2 Cells , Gastrointestinal Microbiome/drug effects , Firmicutes , Faecalibacterium prausnitzii , Fruit/chemistry , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Hydroxybenzoates/pharmacology , Hydroxybenzoates/analysis , Phenols/pharmacology , Phenols/analysis , Fermentation , Gallic Acid/pharmacology , Anthocyanins/pharmacology , Anthocyanins/analysis
16.
Microb Pathog ; 194: 106830, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084307

ABSTRACT

Pseudomonas aeruginosa infections have become a serious threat to public health due to the increasing emergence of extensively antibiotic-resistant strains and high mortality rates. Therefore, the search for new therapeutic alternatives has become crucial. In this study, the antivirulence and antibacterial activity of methyl gallate was evaluated against six clinical isolates of extensively antibiotic-resistant P. aeruginosa. Methyl gallate exhibited minimal inhibitory concentrations of 256-384 µg/mL; moreover, the use of subinhibitory concentrations of the compound inhibited biofilm formation, swimming, swarming, proteolytic activity, and pyocyanin production. Methyl gallate plus antipseudomonal antibiotics showed a synergistic effect by reduced the MICs of ceftazidime, gentamicin and meropenem. Furthermore, the potential therapeutic effect of methyl gallate was demonstrated in an infection model. This study evidenced the antivirulence and antimicrobial activity of methyl gallate as a therapeutic alternative against P. aeruginosa.


Subject(s)
Anti-Bacterial Agents , Biofilms , Drug Synergism , Gallic Acid , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Virulence/drug effects , Humans , Animals , Drug Resistance, Multiple, Bacterial/drug effects , Pyocyanine/metabolism , Meropenem/pharmacology , Ceftazidime/pharmacology , Mice , Gentamicins/pharmacology , Disease Models, Animal
17.
Ecotoxicol Environ Saf ; 283: 116790, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39083864

ABSTRACT

Terrestrial dissolved organic matter (tDOM) holds great promise for controlling cyanobacteria blooms through watershed management. To identify tDOM that could inhibit the growth, photosynthesis and colony formation, unicellular Microcystis aeruginosa Kützing (FACHB-469) was cultivated and treated with varying concentrations of gallic acid, proline and tea polyphenols at different levels of iron. The results indicated that gallic acid and tea polyphenols could inhibit Microcystis growth by suppressing photosynthesis and colony formation by reducing extracellular polysaccharides (EPS) secretion. However, proline had no significant effect on the growth, photosynthesis, colony size and EPS content of Microcystis. Transcriptome analysis showed Microcystis may optimize the internal energy transfer mode of photosynthesis through the change of phycobilisome at different levels of iron. In addition, Microcystis adapted to different iron concentration environments by regulating the expression of genes associated with iron uptake and transport. These findings suggest that the effects of plant species on algal blooms should be considered in reforestation of watershed. This consideration necessitates finding a balance between the costs and benefits of controlling cyanobacteria blooms using tDOM.


Subject(s)
Iron , Microcystis , Photosynthesis , Microcystis/drug effects , Microcystis/growth & development , Photosynthesis/drug effects , Gallic Acid/pharmacology , Proline/metabolism , Polyphenols , Eutrophication , Tea/chemistry
18.
J Photochem Photobiol B ; 258: 112978, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002192

ABSTRACT

Staphylococcus aureus is characterized by its high resistance to conventional antibiotics, particularly methicillin-resistant (MRSA) strains, making it a predominant pathogen in acute and chronic wound infections. The persistence of acute S. aureus wound infections poses a threat by increasing the incidence of their chronicity. This study investigated the potential of photodynamic activation using phytochemical-antibiotic combinations to eliminate S. aureus under conditions representative of acute wound infections, aiming to mitigate the risk of chronicity. The strategy applied takes advantage of the promising antibacterial and photosensitising properties of phytochemicals, and their ability to act as antibiotic adjuvants. The antibacterial activity of selected phytochemicals (berberine, curcumin, farnesol, gallic acid, and quercetin; 6.25-1000 µg/mL) and antibiotics (ciprofloxacin, tetracycline, fusidic acid, oxacillin, gentamicin, mupirocin, methicillin, and tobramycin; 0.0625-1024 µg/mL) was screened individually and in combination against two S. aureus clinical strains (methicillin-resistant and -susceptible-MRSA and MSSA). The photodynamic activity of the phytochemicals was assessed using a light-emitting diode (LED) system with blue (420 nm) or UV-A (365 nm) variants, at 30 mW/cm2 (light doses of 9, 18, 27 J/cm2) and 5.5 mW/cm2 (light doses of 1.5, 3.3 and 5.0 J/cm2), respectively. Notably, all phytochemicals restored antibiotic activity, with 9 and 13 combinations exhibiting potentiating effects on MSSA and MRSA, respectively. Photodynamic activation with blue light (420 nm) resulted in an 8- to 80-fold reduction in the bactericidal concentration of berberine against MSSA and MRSA, while curcumin caused 80-fold reduction for both strains at the light dose of 18 J/cm2. Berberine and curcumin-antibiotic combinations when subjected to photodynamic activation (420 nm light, 10 min, 18 J/cm2) reduced S. aureus culturability by ≈9 log CFU/mL. These combinations lowered the bactericidal concentration of antibiotics, achieving a 2048-fold reduction for gentamicin and 512-fold reduction for tobramycin. Overall, the dual approach involving antimicrobial photodynamic inactivation and selected phytochemical-antibiotic combinations demonstrated a synergistic effect, drastically reducing the culturability of S. aureus and restoring the activity of gentamicin and tobramycin.


Subject(s)
Anti-Bacterial Agents , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Photosensitizing Agents , Phytochemicals , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcus aureus/drug effects , Phytochemicals/pharmacology , Phytochemicals/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/radiation effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Wound Infection/drug therapy , Wound Infection/microbiology , Curcumin/pharmacology , Curcumin/chemistry , Gallic Acid/chemistry , Gallic Acid/pharmacology , Berberine/pharmacology , Berberine/chemistry , Light , Photochemotherapy , Staphylococcal Infections/drug therapy , Quercetin/pharmacology , Quercetin/chemistry , Humans
19.
Sci Rep ; 14(1): 17220, 2024 07 26.
Article in English | MEDLINE | ID: mdl-39060363

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a serious threat to patients with nosocomial infections, and infection is strongly associated with biofilm formation. Gallic acid (GA) is a natural bioactive compound found in traditional Chinese medicines that exerts potent antimicrobial activity. However, the anti-MRSA biofilm efficacy of GA remained to be determined. This study investigated the antimicrobial activities of GA against MRSA and the mechanisms involved. The results revealed the significant antibacterial and antibiofilm activities of GA. The minimal inhibitory concentration of GA against MRSA was 32 µg/mL and a growth curve assay confirmed the significant inhibitory effect of GA on planktonic MRSA. Crystal violet and XTT assays showed that 8 µg/mL GA effectively inhibited the formation of new biofilms and disrupted existing biofilms by reducing both biofilm biomass and metabolic activities. Alkaline phosphatase and ß-galactosidase leakage assays and live/dead staining provided evidence that GA disrupted the integrity of bacterial cell walls and membranes within the biofilm. Scanning electron microscopy observations showed that GA significantly inhibited bacterial adhesion and aggregation, affecting the overall structure of the biofilm. Bacterial adhesion, polysaccharide intercellular adhesion (PIA) production and real-time quantitative PCR assay confirmed that GA inhibited bacterial adhesion, PIA synthesis, and the expression of icaAD and sarA. These results suggested that GA inhibited biofilm formation by inhibiting the expression of sarA, then downregulating the expression of icaA and icaD, thereby reducing the synthesis of PIA to attenuate the adhesion capacity of MRSA. GA is therefore a promising candidate for development as a pharmaceutical agent for the prevention and treatment of bacterial infections caused by MRSA.


Subject(s)
Anti-Bacterial Agents , Bacterial Adhesion , Biofilms , Gallic Acid , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Methicillin-Resistant Staphylococcus aureus/drug effects , Biofilms/drug effects , Gallic Acid/pharmacology , Bacterial Adhesion/drug effects , Anti-Bacterial Agents/pharmacology , Polysaccharides, Bacterial/pharmacology , Polysaccharides, Bacterial/metabolism , Humans
20.
Int J Biol Macromol ; 277(Pt 1): 134008, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39032879

ABSTRACT

In this study, an active film composed of gallic acid (GA), chitosan (CS), and cellulose nanocrystals (CNC) was prepared using a solution casting method and synergistic photodynamic inactivation (PDI) technology. Characterization of the film showed that the CS-CNC-GA composite film had high transparency and UV-blocking ability. The addition of GA (0.2 %-1.0 %) significantly enhanced the mechanical properties, water resistance, and thermal stability of the film. The tensile strength increased up to 46.30 MPa, and the lowest water vapor permeability was 1.16 × e-12 g/(cm·s·Pa). The PDI-treated CS-CNC-GA1.0 composite film exhibited significantly enhanced antibacterial activity, with inhibition zone diameters of 31.83 mm against Staphylococcus aureus and 21.82 mm against Escherichia coli. The CS-CNC-GA composite film also showed good antioxidant activity. Additionally, the CS-CNC-GA1.0 composite film generated a large amount of singlet oxygen under UV-C light irradiation. It was found that using the CS-CNC-GA1.0 composite film for packaging and storage of oysters at 4 °C effectively delayed the increase in pH, total colony count, and lipid oxidation in oysters. In conclusion, the CS-CNC-GA composite film based on PDI technology has great potential for applications in the preservation of aquatic products.


Subject(s)
Anti-Bacterial Agents , Cellulose , Chitosan , Gallic Acid , Gallic Acid/chemistry , Gallic Acid/pharmacology , Chitosan/chemistry , Cellulose/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Escherichia coli/drug effects , Food Packaging/methods , Staphylococcus aureus/drug effects , Nanoparticles/chemistry , Permeability , Nanocomposites/chemistry , Tensile Strength , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL