Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.414
Filter
1.
J Nanobiotechnology ; 22(1): 471, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118143

ABSTRACT

Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease, while there is a lack of pharmaceutical interventions to halt AAA progression presently. To address the multifaceted pathology of AAA, this work develops a novel multifunctional gene delivery system to simultaneously deliver two siRNAs targeting MMP-2 and MMP-9. The system (TPNs-siRNA), formed through the oxidative polymerization and self-assembly of epigallocatechin gallate (EGCG), efficiently encapsulates siRNAs during self-assembly. TPNs-siRNA safeguards siRNAs from biological degradation, facilitates intracellular siRNA transfection, promotes lysosomal escape, and releases siRNAs to silence MMP-2 and MMP-9. Additionally, TPNs, serving as a multi-bioactive material, mitigates oxidative stress and inflammation, fosters M1-to-M2 repolarization of macrophages, and inhibits cell calcification and apoptosis. In experiments with AAA mice, TPNs-siRNA accumulated and persisted in aneurysmal tissue after intravenous delivery, demonstrating that TPNs-siRNA can be significantly distributed in macrophages and VSMCs relevant to AAA pathogenesis. Leveraging the carrier's intrinsic multi-bioactive properties, the targeted siRNA delivery by TPNs exhibits a synergistic effect for enhanced AAA therapy. Furthermore, TPNs-siRNA is gradually metabolized and excreted from the body, resulting in excellent biocompatibility. Consequently, TPNs emerges as a promising multi-bioactive nanotherapy and a targeted delivery nanocarrier for effective AAA therapy.


Subject(s)
Aortic Aneurysm, Abdominal , Matrix Metalloproteinase 9 , Mice, Inbred C57BL , Nanoparticles , RNA, Small Interfering , Aortic Aneurysm, Abdominal/drug therapy , Animals , Mice , Nanoparticles/chemistry , Male , Matrix Metalloproteinase 9/metabolism , Polyphenols/chemistry , Polyphenols/pharmacology , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Tea/chemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Humans , Macrophages/metabolism , Macrophages/drug effects , Gene Transfer Techniques , Oxidative Stress/drug effects , RAW 264.7 Cells , Apoptosis/drug effects
2.
Anim Biotechnol ; 35(1): 2381080, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39087503

ABSTRACT

Mastitis, a serious threat to the health and milk production function of dairy cows decreases milk quality. Blood from three healthy cows and three mastitis cows were collected in this study and their transcriptome was sequenced using the Illumina HiSeq platform. Differentially expressed genes (DEGs) were screened according to the |log2FoldChange| > 1 and P-value < 0.05 criteria. Pathway enrichment and functional annotation were performed through KEGG and GO analyses. Finally, the mechanism of the AMP-activated protein kinase (AMPK) mediation of (-)-epigallocatechin-3-gallate (EGCG) to promote lipid metabolism in mastitis cows was analyzed in bovine mammary epithelial cells (BMECs). Transcriptome analysis revealed a total of 825 DEGs, with 474 genes showing increased expression and 351 genes showing decreased expression. The KEGG analysis of DEGs revealed that they were mainly linked to tumour necrosis factor, nuclear factor-κB signalling pathway, and lipid metabolism-related signalling pathway, whereas GO functional annotation found that DEGs were enriched in threonine and methionine kinase activity, cellular metabolic processes, and cytoplasm. AMPK expression, which is involved in several lipid metabolism pathways, was downregulated in mastitis cows. The results of in vitro experiments showed that the inhibition of AMPK promoted the expression of lipid synthesis genes in lipopolysaccharide-induced BMECs and that EGCG could promote lipid synthesis by decreasing the expression of AMPK and downregulating the expression of inflammatory factors in inflammatory BMECs. In conclusion, our study demonstrated that AMPK mediated EGCG to inhabit of inflammatory responses and promote of lipid synthesis in inflammatory BMECs.


Subject(s)
AMP-Activated Protein Kinases , Catechin , Lipid Metabolism , Mammary Glands, Animal , Mastitis, Bovine , Animals , Cattle , Catechin/analogs & derivatives , Catechin/pharmacology , Female , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Mastitis, Bovine/genetics , Lipid Metabolism/drug effects , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling/veterinary , Transcriptome/drug effects
3.
Food Res Int ; 192: 114833, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39147522

ABSTRACT

This study examined the suppressive effects of 16 selected plant-based foods on α-glucosidase and pancreatic lipase and their antioxidant properties. Among these, the bark of Cinnamomum cassia (Cinnamon, WLN-FM 15) showed the highest inhibitory activity against α-glucosidase and the highest antioxidant activity. Additionally, WLN-FM 15 showed promising results in the other tests. To further identify the bioactive constituents of WLN-FM 15, a multi-bioactivity-labeled molecular networking approach was used through a combination of GNPS-based molecular networking, DPPH-HPLC, and affinity-based ultrafiltration-HPLC. A total of nine procyanidins were identified as antioxidants and inhibitors of α-glucosidase and pancreatic lipase in WLN-FM 15. Subsequently, procyanidins A1, A2, B1, and C1 were isolated, and their efficacy was confirmed through functional assays. In summary, WLN-FM 15 has the potential to serve as a functional food ingredient with the procyanidins as its bioactive constituents. These results also suggest that the multi-bioactivity-labeled molecular networking approach is reliable for identifying bioactive constituents in plant-based foods.


Subject(s)
Antioxidants , Biflavonoids , Catechin , Cinnamomum aromaticum , Glycoside Hydrolase Inhibitors , Lipase , Plant Bark , Proanthocyanidins , Proanthocyanidins/pharmacology , Proanthocyanidins/chemistry , Proanthocyanidins/analysis , Lipase/antagonists & inhibitors , Lipase/metabolism , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/analysis , Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolase Inhibitors/chemistry , Plant Bark/chemistry , Cinnamomum aromaticum/chemistry , Biflavonoids/pharmacology , Biflavonoids/analysis , Biflavonoids/chemistry , Catechin/analysis , Catechin/chemistry , Catechin/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Chromatography, High Pressure Liquid , Pancreas/enzymology , alpha-Glucosidases/metabolism , Network Pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry
4.
J Neurosci Res ; 102(8): e25372, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39086264

ABSTRACT

The objective of this study was to investigate the potential mechanisms by which (+)-catechin alleviates neuropathic pain. Thirty-two male Sprague-Dawley rats were divided into four groups: the sham group, the chronic constriction injury (CCI)group, the CCI+ ibuprofen group, and the CCI+ (+)-catechin group. CCI surgery induces thermal hyperalgesia in rats and (+)-catechin ameliorated CCI-induced thermal hyperalgesia and repaired damaged sciatic nerve in rats. CCI decreased SOD levels in male rat spinal cord dorsal horn and promoted MDA production, induced oxidative stress by increasing NOX4 levels and decreasing antioxidant enzyme HO-1 levels, and also increased protein levels of TLR4, p-NF-κB, NLRP3 inflammasome components, and IL-1ß. In contrast, (+)-catechin reversed the above results. In i vitro experiments, (+)-catechin reduced the generation of reactive oxygen species (ROS) in GMI-R1 cells after LPS stimulation and attenuated the co-expression of IBA-1 and NLRP3. It also showed significant inhibition of the NF-κB and NLRP3 inflammatory pathways and activation of the Nrf2-mediated antioxidant system. Overall, these findings suggest that (+)-catechin inhibits the activation of the NLRP3 inflammasome through the triggering of the Nrf2-induced antioxidant system, the inhibition of the TLR4/NF-κB pathway, and the production of ROS to alleviate CCI-induced neuropathic pain in male rats.


Subject(s)
Antioxidants , Catechin , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Neuralgia , Signal Transduction , Animals , Male , Rats , Antioxidants/pharmacology , Catechin/pharmacology , Hyperalgesia/metabolism , Hyperalgesia/drug therapy , Inflammasomes/metabolism , Inflammasomes/drug effects , Neuralgia/metabolism , Neuralgia/drug therapy , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/drug effects
5.
ACS Nano ; 18(32): 21512-21522, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39096486

ABSTRACT

Although minimally invasive interventional occluders can effectively seal heart defect tissue, they still have some limitations, including poor endothelial healing, intense inflammatory response, and thrombosis formation. Herein, a polyphenol-reinforced medicine/peptide glycocalyx-like coating was prepared on cardiac occluders. A coating consisting of carboxylated chitosan, epigallocatechin-3-gallate (EGCG), tanshinone IIA sulfonic sodium (TSS), and hyaluronic acid grafted with 3-aminophenylboronic acid was prepared. Subsequently, the mercaptopropionic acid-GGGGG-Arg-Glu-Asp-Val peptide was grafted by the thiol-ene "click" reaction. The coating showed good hydrophilicity and free radical-scavenging ability and could release EGCG-TSS. The results of biological experiments suggested that the coating could reduce thrombosis by promoting endothelialization, and promote myocardial repair by regulating the inflammatory response. The functions of regulating cardiomyocyte apoptosis and metabolism were confirmed, and the inflammatory regulatory functions of the coating were mainly dependent on the NF-kappa B and TNF signaling pathway.


Subject(s)
Glycocalyx , Hydrogels , Polyphenols , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Polyphenols/chemistry , Polyphenols/pharmacology , Glycocalyx/metabolism , Glycocalyx/chemistry , Glycocalyx/drug effects , Immunomodulation/drug effects , Regeneration/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Apoptosis/drug effects , Mice , Myocardium/metabolism , Catechin/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Rats, Sprague-Dawley , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Male
6.
Food Chem Toxicol ; 191: 114906, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39095006

ABSTRACT

The study aimed to examine effects of (-)-epigallocatechin-3-gallate (EGCG) on energy metabolism and mitochondrial dynamics in mouse model of renal injury caused by doxorubicin (DOX). Here, mice were divided into Control group, EGCG-only treated group, DOX group, and three doses of EGCG plus DOX groups. Our results showed that EGCG behaved beneficial effects against kidney injury via attenuation of pathological changes in kidney tissue, which was confirmed by reducing serum creatinine (SCr), blood urea nitrogen (BUN), and apoptosis. Subsequently, changes in reactive oxygen species generation, malondialdehyde content, and activities of antioxidant enzymes were considerably ameliorated in EGCG + DOX groups when compared to DOX group. Furthermore, EGCG-evoked renal protection was associated with increases of mitochondrial membrane potential and decreases of mitochondrial fission protein Dynamin-related protein 1 (Drp1). Moreover, changing glycolysis into mitochondrial oxidative phosphorylation was observed, evidenced by controlling activities of malate dehydrogenase (MDH) and hexokinase (HK) in EGCG + DOX groups when compared to DOX group, indicating that reprogramming energy metabolism was linked to EGCG-induced renal protection in mice. Therefore, EGCG was demonstrated to have a protective effect against kidney injury by reducing oxidative damage, metabolic disorders, and mitochondrial dysfunction, suggesting that EGCG has potential as a feasible strategy to prevent kidney injury.


Subject(s)
Catechin , Doxorubicin , Dynamins , Mitochondrial Dynamics , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Mice , Mitochondrial Dynamics/drug effects , Male , Doxorubicin/toxicity , Dynamins/metabolism , Kidney/drug effects , Kidney/metabolism , Homeostasis/drug effects , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Acute Kidney Injury/drug therapy , Acute Kidney Injury/chemically induced , Mitochondria/drug effects , Mitochondria/metabolism , Energy Metabolism/drug effects , Oxidative Stress/drug effects , Antioxidants/pharmacology
7.
Nutrients ; 16(14)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39064787

ABSTRACT

Chronic obesity is an alarmingly growing global public health concern, posing substantial challenges for the prevention of chronic diseases, including hyperinsulinemia, type 2 diabetes, hyperlipidemia, hypertension, and coronary artery disease, and there is an urgent need for early mitigation strategies. We previously reported the obesity-reducing effects of green tea and ß-cryptoxanthin intake. However, since tea has a complex mixture of compounds, it remained unclear which component contributed the most to this effect. Using high-performance liquid chromatography, we analyzed the components of tea in this study to determine if consumption of any combination of these compounds with ß-cryptoxanthin had an obesity-reducing effect. Consuming epigallocatechin gallate (EGCG), a component of green tea, and ß-cryptoxanthin for 4 weeks led to a decrease in body weight. Moreover, the weight and size of the white adipose tissues were significantly reduced, and blood biochemistry test results were comparable to normal values, with particular improvement in liver function. This indicated that intake of EGCG and ß-cryptoxanthin reduces obesity in both subcutaneous and visceral fat. These findings suggest that simultaneous intake of EGCG and ß-cryptoxanthin not only reduces obesity but also has a systemic beneficial effect on the body's normal physiological function.


Subject(s)
Beta-Cryptoxanthin , Catechin , Obesity , Catechin/analogs & derivatives , Catechin/pharmacology , Obesity/drug therapy , Beta-Cryptoxanthin/pharmacology , Male , Animals , Tea/chemistry , Drug Synergism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Anti-Obesity Agents/pharmacology , Mice, Inbred C57BL , Weight Loss/drug effects
8.
Nat Commun ; 15(1): 6256, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39048544

ABSTRACT

Maintenance of NAD pools is critical for neuronal survival. The capacity to maintain NAD pools declines in neurodegenerative disease. We identify that low NMNAT2, the critical neuronal NAD producing enzyme, drives retinal susceptibility to neurodegenerative insults. As proof of concept, gene therapy over-expressing full length human NMNAT2 is neuroprotective. To pharmacologically target NMNAT2, we identify that epigallocatechin gallate (EGCG) can drive NAD production in neurons through an NMNAT2 and NMN dependent mechanism. We confirm this by pharmacological and genetic inhibition of the NAD-salvage pathway. EGCG is neuroprotective in rodent (mixed sex) and human models of retinal neurodegeneration. As EGCG has poor drug-like qualities, we use it as a tool compound to generate novel small molecules which drive neuronal NAD production and provide neuroprotection. This class of NMNAT2 targeted small molecules could have an important therapeutic impact for neurodegenerative disease following further drug development.


Subject(s)
Catechin , NAD , Neurons , Neuroprotective Agents , Nicotinamide-Nucleotide Adenylyltransferase , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , NAD/metabolism , Humans , Animals , Neurons/metabolism , Neurons/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Neuroprotective Agents/pharmacology , Male , Mice , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics , Female , Retina/metabolism , Retina/drug effects , Mice, Inbred C57BL , Rats , Disease Models, Animal , Genetic Therapy/methods
9.
FASEB J ; 38(13): e23749, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38953707

ABSTRACT

Pulmonary fibrosis is a formidable challenge in chronic and age-related lung diseases. Myofibroblasts secrete large amounts of extracellular matrix and induce pro-repair responses during normal wound healing. Successful tissue repair results in termination of myofibroblast activity via apoptosis; however, some myofibroblasts exhibit a senescent phenotype and escape apoptosis, causing over-repair that is characterized by pathological fibrotic scarring. Therefore, the removal of senescent myofibroblasts using senolytics is an important method for the treatment of pulmonary fibrosis. Procyanidin C1 (PCC1) has recently been discovered as a senolytic compound with very low toxicity and few side effects. This study aimed to determine whether PCC1 could improve lung fibrosis by promoting apoptosis in senescent myofibroblasts and to investigate the mechanisms involved. The results showed that PCC1 attenuates bleomycin (BLM)-induced pulmonary fibrosis in mice. In addition, we found that PCC1 inhibited extracellular matrix deposition and promoted the apoptosis of senescent myofibroblasts by increasing PUMA expression and activating the BAX signaling pathway. Our findings represent a new method of pulmonary fibrosis management and emphasize the potential of PCC1 as a senotherapeutic agent for the treatment of pulmonary fibrosis, providing hope for patients with pulmonary fibrosis worldwide. Our results advance our understanding of age-related diseases and highlight the importance of addressing cellular senescence in treatment.


Subject(s)
Bleomycin , Catechin , Cellular Senescence , Mice, Inbred C57BL , Myofibroblasts , Pulmonary Fibrosis , Animals , Bleomycin/toxicity , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Mice , Cellular Senescence/drug effects , Catechin/pharmacology , Catechin/analogs & derivatives , Proanthocyanidins/pharmacology , Apoptosis/drug effects , Male , Biflavonoids/pharmacology , Signal Transduction/drug effects
10.
Bull Exp Biol Med ; 177(1): 88-92, 2024 May.
Article in English | MEDLINE | ID: mdl-38960956

ABSTRACT

We studied antimicrobial activity of epigallocatechin-3-gallate (EGCG), a green tea polyphenolic catechin, and its combined use with ceftazidime (CAZ) against bacterial strains of Klebsiella pneumoniae. EGCG exhibited no activity against strains of K. pneumoniae with a different sensitivity to CAZ. However, for a "sensitive" strain, a decrease in minimum inhibitory concentration (MIC) of CAZ (from 0.064 to 0.023 mg/liter) was revealed when CAZ was co-administered with EGCG. For a "resistant" stain, MIC of CAZ remained high, but activation of EGCG at its high concentrations was observed. Indirect evidence of antimicrobial effect of EGCG co-administered with CAZ on Klebsiella was obtained.


Subject(s)
Anti-Bacterial Agents , Catechin , Ceftazidime , Klebsiella pneumoniae , Microbial Sensitivity Tests , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/chemistry , Klebsiella pneumoniae/drug effects , Ceftazidime/pharmacology , Anti-Bacterial Agents/pharmacology , Tea/chemistry
11.
Sci Adv ; 10(28): eadn1745, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996026

ABSTRACT

Rapid drug clearance and off-target effects of therapeutic drugs can induce low bioavailability and systemic side effects and gravely restrict the therapeutic effects of inflammatory bowel diseases (IBDs). Here, we propose an amplifying targeting strategy based on orally administered gallium (Ga)-based liquid metal (LM) nano-agents to efficiently eliminate reactive oxygen and nitrogen species (RONS) and modulate the dysregulated microbiome for remission of IBDs. Taking advantage of the favorable adhesive activity and coordination ability of polyphenol structure, epigallocatechin gallate (EGCG) is applied to encapsulate LM to construct the formulations (LM-EGCG). After adhering to the inflamed tissue, EGCG not only eliminates RONS but also captures the dissociated Ga to form EGCG-Ga complexes for enhancive accumulation. The detained composites protect the intestinal barrier and modulate gut microbiota for restoring the disordered enteral microenvironment, thereby relieving IBDs. Unexpectedly, LM-EGCG markedly decreases the Escherichia_Shigella populations while augmenting the abundance of Akkermansia and Bifidobacterium, resulting in favorable therapeutic effects against the dextran sulfate sodium-induced colitis.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Animals , Inflammatory Bowel Diseases/drug therapy , Administration, Oral , Gastrointestinal Microbiome/drug effects , Mice , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/administration & dosage , Catechin/pharmacology , Gallium/chemistry , Gallium/pharmacology , Disease Models, Animal , Inflammation/drug therapy , Reactive Oxygen Species/metabolism , Colitis/drug therapy , Humans , Reactive Nitrogen Species/metabolism
12.
Nutrients ; 16(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38999821

ABSTRACT

Atherosclerosis (AS) is a common clinical sickness and the major pathological basis of ischemic cardiocerebrovascular diseases (CCVDs). The pathogenesis of AS involves a variety of risk factors, and there is a lack of effective preventive and curative drugs that can completely treat AS. In recent years, with the improvement of people's living standards and changes in dietary habits, the morbidity and mortality rates of AS are on the rise, and the age of onset tends to be younger. The formation of AS is closely related to a variety of factors, and the main factors include lipid metabolism disorders, endothelial damage, inflammation, unstable plaques, etc. Epigallocatechin gallate (EGCG), as one of the main components of catechins, has a variety of pharmacological effects, and its role in the prevention of AS and the protection of cardiovascular and cerebral blood vessels has been highly valued. Recent epidemiological investigations and various in vivo and ex vivo experiments have shown that EGCG is capable of resisting atherosclerosis and reducing the morbidity and mortality of AS. In this paper, we reviewed the anti-AS effects of EGCG and its mechanisms in recent years, including the regulation of lipid metabolism, regulation of intestinal flora disorders, improvement of vascular endothelial cell functions, inhibition of inflammatory factors expression, regulation of inflammatory signaling pathways, inhibition of matrix metalloproteinase (MMP) expression, and inhibition of platelet aggregation, which are helpful for the prevention of cardiocerebrovascular diseases.


Subject(s)
Atherosclerosis , Catechin , Lipid Metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/therapeutic use , Humans , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Lipid Metabolism/drug effects , Animals , Gastrointestinal Microbiome/drug effects
13.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000465

ABSTRACT

The complexification of in vitro models requires the compatibility of cells with the same medium. Since immune cells are the most sensitive to growth conditions, growing intestinal epithelial cells in their usual medium seems to be necessary. This work was aimed at comparing the sensitivity of these epithelial cells to pro-inflammatory stimuli but also to dietary polyphenols in both DMEM and RPMI-1640 media. Co-cultures of Caco-2 and HT29-MTX cells were grown for 21 days in the two media before their stimulation with a cocktail of TNF-α (20 ng/mL), IL-1ß (1 ng/mL), and IFN-γ (10 ng/mL) or with LPS (10 ng/mL) from E. coli (O111:B4). The role of catechins (15 µM), a dietary polyphenol, was evaluated after its incubation with the cells before their stimulation for 6 h. The RPMI-1640 medium did not alter the intensity of the inflammatory response observed with the cytokines. By contrast, LPS failed to stimulate the co-culture in inserts regardless of the medium used. Lastly, catechins were unable to prevent the pro-inflammatory response observed with the cytokines in the two media. The preservation of the response of this model of intestinal epithelium in RPMI-1640 medium is promising when considering its complexification to evaluate the complex cellular crosstalk leading to intestinal homeostasis.


Subject(s)
Coculture Techniques , Intestinal Mucosa , Lipopolysaccharides , Polyphenols , Humans , Coculture Techniques/methods , Polyphenols/pharmacology , Caco-2 Cells , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Lipopolysaccharides/pharmacology , HT29 Cells , Culture Media/chemistry , Culture Media/pharmacology , Cytokines/metabolism , Catechin/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Inflammation/metabolism , Inflammation/pathology
14.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000554

ABSTRACT

Triple-negative breast cancer (TNBC) accounts for 15% of all breast cancers and is highly aggressive. Despite an initial positive response to chemotherapy, most patients experience rapid disease progression leading to relapse and metastasis. This is attributed to the presence of breast cancer stem cells (BCSCs) within the tumor, which are characterized by self-renewal, pluripotency, and resistance mechanisms. Targeting BCSCs has become critical as conventional therapies fail to eradicate them due to a lack of specific targets. Curcumin, a polyphenol derived from turmeric (Curcuma longa), exhibits anticancer effects against breast cancer cells and BCSCs. The use of curcumin derivatives has been suggested as an approach to overcome the bioavailability and solubility problems of curcumin in humans, thereby increasing its anticancer effects. The aim of this study was to evaluate the cellular and molecular effects of six synthetic compounds derived from the natural polyphenol epigallocatechin gallate (EGCG) (TL1, TL2) and curcumin derivatives (TL3, TL4, TL5, and TL6) on a TNBC mesenchymal stem-like cell line. The activity of the compounds against BCSCs was also determined by a mammosphere inhibition assay and studying different BCSC markers by Western blotting. Finally, a drug combination assay was performed with the most promising compounds to evaluate their potential synergistic effects with the chemotherapeutic agents doxorubicin, cisplatin, and paclitaxel. The results showed that compounds exhibited specific cytotoxicity against the TNBC cell line and BCSCs. Interestingly, the combination of the curcumin derivative TL3 with doxorubicin and cisplatin displayed a synergistic effect in TNBC cells.


Subject(s)
Curcumin , Neoplastic Stem Cells , Polyphenols , Triple Negative Breast Neoplasms , Humans , Curcumin/pharmacology , Curcumin/analogs & derivatives , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Polyphenols/pharmacology , Polyphenols/chemistry , Cell Line, Tumor , Female , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/chemistry
15.
Physiol Rep ; 12(13): e16095, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38946616

ABSTRACT

The present study aimed to investigate the effect of catechin-loaded Chitosan-Alginate nanoparticles (NPs) on cognitive function in an aluminum chloride (AlCl3)-induced rat model of Alzheimer's disease (AD). The Catechin-loaded Chitosan-Alginate nanocarriers were synthesized through ionotropic gelation (IG) method. Physio-chemical characterization was conducted with the Zetasizer Nano system, the scanning electron microscope, and the Fourier transform infrared spectroscopy. The experiments were performed over 21 days on six groups of male Wistar rats. The control group, AlCl3 treated group, Catechin group, nanocarrier group, treatment group 1 (AlCl3 + Catechin), and treatment group 2 (AlCl3 + nanocarrier). A behavioral study was done by the Morris water maze (MWM) test. In addition, the level of oxidative indices and acetylcholine esterase (AChE) activity was determined by standard procedures at the end of the study. AlCl3 induced a significant increase in AChE activity, along with a significant decrease in the level of Catalase (CAT) and total antioxidant capacity (TAC) in the hippocampus. Moreover, the significant effect of AlCl3 was observed on the behavioral parameters of the MWM test. Both forms of Catechin markedly improved AChE activity, oxidative biomarkers, spatial memory, and learning. The present study indicated that the administration of Catechin-loaded Chitosan-Alginate NPs is a beneficial therapeutic option against behavioral and chemical alteration of AD in male Wistar rats.


Subject(s)
Alginates , Aluminum Chloride , Alzheimer Disease , Catechin , Chitosan , Nanoparticles , Rats, Wistar , Animals , Catechin/administration & dosage , Catechin/pharmacology , Aluminum Chloride/toxicity , Chitosan/chemistry , Chitosan/administration & dosage , Alginates/chemistry , Alginates/administration & dosage , Male , Alzheimer Disease/drug therapy , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Rats , Administration, Oral , Cognition/drug effects , Acetylcholinesterase/metabolism , Maze Learning/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Disease Models, Animal , Antioxidants/pharmacology , Antioxidants/administration & dosage , Oxidative Stress/drug effects , Drug Carriers/chemistry
16.
Breast Cancer Res ; 26(1): 114, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38978121

ABSTRACT

The protein Bcl-2, well-known for its anti-apoptotic properties, has been implicated in cancer pathogenesis. Identifying the primary gene responsible for promoting improved cell survival and development has provided compelling evidence for preventing cellular death in the progression of malignancies. Numerous research studies have provided evidence that the abundance of Bcl-2 is higher in malignant cells, suggesting that suppressing Bcl-2 expression could be a viable therapeutic approach for cancer treatment. In this study, we acquired a compound collection using a database that includes constituents from Traditional Chinese Medicine (TCM). Initially, we established a pharmacophore model and utilized it to search the TCM database for potential compounds. Compounds with a fitness score exceeding 0.75 were selected for further analysis. The Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) analysis identified six compounds with favorable therapeutic characteristics. The compounds that successfully passed the initial screening process based on the pharmacodynamic model were subjected to further evaluation. Extra-precision (XP) docking was employed to identify the compounds with the most favorable XP docking scores. Further analysis using the Molecular Mechanics Generalized Born Surface Area (MM-GBSA) method to calculate the overall free binding energy. The binding energy between the prospective ligand molecule and the target protein Bcl-2 was assessed by a 100 ns molecular dynamics simulation for curcumin and Epigallocatechin gallate (EGCG). The findings of this investigation demonstrate the identification of a molecular structure that effectively inhibits the functionality of the Bcl-2 when bound to the ligand EGCG. Consequently, this finding presents a novel avenue for the development of pharmaceuticals capable of effectively addressing both inflammatory and tumorous conditions.


Subject(s)
Catechin , Curcumin , Molecular Docking Simulation , Proto-Oncogene Proteins c-bcl-2 , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/chemistry , Catechin/therapeutic use , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Humans , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Protein Binding , Pharmacophore
17.
Brain Res Bull ; 215: 111033, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032586

ABSTRACT

Brain Muscle ARNT-Like Protein 1 (BMAL1) suppresses oxidative stress in brain injury during surgery. Epigallocatechin-3-gallate (EGCG), a monomer in green tea, has been identified as an antioxidant and a potential agonist for BMAL1. In this work, the mechanism by which BMAL1 is regulated was investigated, as well as the therapeutic effect of EGCG on surgically injured rats. The pathological environment after brain injury during surgery was simulated by excising the right frontal lobe of rats. Rats received an intraperitoneal injection of EGCG immediately after surgery. Neurological scores and cerebral edema were recorded after surgery. Fluoro-Jade C staining, TUNEL staining, western blot, and lipid peroxidation analyses were conducted 3 days later. Here we show that the endogenous BMAL1 level decreased after brain injury. Postoperative administration of EGCG up-regulated the content of BMAL1 around the cerebral cortex, reduced the oxidative stress level, reduced neuronal apoptosis and the number of degenerated neurons, alleviated cerebral edema, and improved neurological scores in rats. This suggests that BMAL1 is an effective target for treating surgical brain injury, as well as that EGCG may be a promising agent for alleviating postoperative brain injury.


Subject(s)
ARNTL Transcription Factors , Catechin , Rats, Sprague-Dawley , Up-Regulation , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , ARNTL Transcription Factors/metabolism , Male , Up-Regulation/drug effects , Rats , Oxidative Stress/drug effects , Neuroprotective Agents/pharmacology , Disease Models, Animal , Brain Injuries/metabolism , Brain Injuries/drug therapy , Brain Edema/metabolism , Brain Edema/drug therapy , Apoptosis/drug effects , Antioxidants/pharmacology
18.
Biol Pharm Bull ; 47(7): 1331-1337, 2024.
Article in English | MEDLINE | ID: mdl-39048354

ABSTRACT

Green tea (GT) catechins exhibit antiviral effects in experimental studies. However, we lack clinical evidence on the preventive effects of catechin concentrations in gargling against acute upper respiratory tract infections (URTIs). Therefore, we aimed to investigate the concentration-dependence of GT catechins in gargling on the incidence of URTIs. We conducted an open-label randomized study. The target population consisted of 209 students from the University of Shizuoka and Meiji University, who were randomly assigned to high-catechin (approximate catechin concentration: 76.4 mg/dL), low-catechin (approximate catechin concentration: 30.8 mg/dL), and a control water gargling (catechin concentration: 0 mg/dL) group. All participants gargled water or GT daily for 12 weeks. The symptoms of URTIs were recorded on a daily survey form by participants. The incidences of URTIs occurred in 6 (9.1%), 7 (10.8%), and 11 (15.7%) participants in the high-catechin, low-catechin, and water groups, respectively. Cox proportional hazards analysis, using background factors and prevention status as covariates, revealed a hazard ratio of 0.57 (95% Confidence Interval (CI): 0.21-1.55, p = 0.261) for the high-catechin vs. water group and 0.54 (95% CI: 0.20-1.50, p = 0.341) for the low-catechin vs. water group. Our findings showed the incidence of URTIs in a concentration-dependent GT gargling was not significantly different, partly owing to the low event rates caused by intense precautions against the coronavirus disease 2019 pandemic. Our study would serve as a foundation for the development of an advanced protocol with optimal concentrations and a larger number of participants.


Subject(s)
Catechin , Respiratory Tract Infections , Tea , Catechin/pharmacology , Catechin/therapeutic use , Catechin/administration & dosage , Humans , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/epidemiology , Male , Female , Tea/chemistry , Young Adult , Adult , Dose-Response Relationship, Drug , Acute Disease , Incidence , Antiviral Agents/therapeutic use
19.
Toxicon ; 247: 107857, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38996976

ABSTRACT

Fluoride is a double-edged sword. It was widely used for early caries prevention while excessive intake caused a toxicology effect, affected enamel development, and resulted in dental fluorosis. The study aimed to evaluate the protective effect and mechanism of Epigallocatechin-3-gallate (EGCG) on the apoptosis induced by fluoride in ameloblast-like cells. We observed that NaF triggered apoptotic alterations in cell morphology, excessive NaF arrested cell cycle at the G1, and induced apoptosis by up-regulating Bax and down-regulating Bcl-2. NaF activated the insulin-like growth factor receptor (IGFR), and phosphatidylinositol-3-hydroxylase (p-PI3K), while dose-dependently down-regulating the expression of Forkhead box O1 (FoxO1). EGCG supplements reversed the changes in LS8 morphology, the cell cycle, and apoptosis induced by fluoride. These results indicated that EGCG possesses a protective effect against fluoride toxicity. Furthermore, EGCG suppressed the activation of p-PI3K and the down-regulation of FoxO1 caused by fluoride. Collectively, our findings suggested that EGCG attenuated fluoride-induced apoptosis by inhibiting the PI3K/FoxO1 signaling pathway. EGCG may serve as a new alternative method for dental fluorosis prevention, control, and treatment.


Subject(s)
Ameloblasts , Apoptosis , Catechin , Fluorides , Phosphatidylinositol 3-Kinases , Signal Transduction , Catechin/analogs & derivatives , Catechin/pharmacology , Apoptosis/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Animals , Fluorides/toxicity , Fluorides/pharmacology , Ameloblasts/drug effects , Ameloblasts/metabolism , Signal Transduction/drug effects , Forkhead Box Protein O1/metabolism , Cell Line , Mice , Sodium Fluoride/toxicity , Fluorosis, Dental
20.
Sci Rep ; 14(1): 16809, 2024 07 22.
Article in English | MEDLINE | ID: mdl-39039202

ABSTRACT

Long-term exposure to hyperglycemic conditions leads to ß-cell dysfunction, particularly mitochondrial dysfunction, and inflammatory and oxidative stress responses, which are considered the primary causes of ß-cell death and the hallmarks of diabetes. Plant-active ingredients may play a key role in glycemic control. Epigallocatechin gallate (EGCG) is a characteristic catechin derived from tea that possesses anti-diabetic properties. Nonetheless, its underlying mechanisms remain elusive. Herein, the protective role of EGCG on high glucose (33 mM)-induced pancreatic beta cell dysfunction and its possible molecular mechanisms were investigated. Briefly, MIN6 cells were treated with glucose and EGCG (10 µM, 20 µM, and 40 µM) for 48 h. Our results revealed that EGCG dose-dependently restored mitochondrial membrane potential and concomitantly alleviated cell apoptosis. Mechanistically, the expression level of apoptotic protein BAX and Dynamic related protein 1 (DRP1) was significantly downregulated following EGCG treatment, whereas that of the anti-apoptotic protein BCL-2 was significantly upregulated. Taken together, EGCG alleviated high glucose-induced pancreatic beta cell dysfunction by targeting the DRP1-related mitochondrial apoptosis pathway and thus can serve as a nutritional intervention for the preservation of beta cell dysfunction in patients with type 2 diabetes mellitus.


Subject(s)
Apoptosis , Catechin , Dynamins , Glucose , Insulin-Secreting Cells , Mitochondria , Catechin/analogs & derivatives , Catechin/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Apoptosis/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Glucose/metabolism , Dynamins/metabolism , Dynamins/genetics , Animals , Mice , Cell Line , Membrane Potential, Mitochondrial/drug effects , bcl-2-Associated X Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL