Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.433
Filtrer
1.
J Nutr Sci Vitaminol (Tokyo) ; 70(3): 280-287, 2024.
Article de Anglais | MEDLINE | ID: mdl-38945894

RÉSUMÉ

Excessive immune response and inflammation are associated with an increased risk of various diseases. In particular, excessive myeloperoxidase (MPO) activity in neutrophils causes inflammatory reactions and lifestyle-related diseases. Adlay has a long history of being used as a traditional Chinese medicine. Polyphenols present in adlay seeds are expected to have the effect of suppressing excessive immune and inflammatory responses. Here, we conducted a randomized, double-blind, parallel group, placebo-controlled study was conducted to evaluate the suppressing effects of adlay seeds extract on excessive immune responses. One hundred and twenty adults participated in the study and they were equally divided into an adlay tea intake group and a placebo group. MPO activity was significantly elevated in the placebo group after 8-wk ingestion, while no significant change was observed in the adlay group. Vascular endothelial functions improved in the adlay group, especially in subjects over 40 y old. These results indicate that adlay tea intake may suppress an excessive immune and inflammatory responses, and improve arterial stiffness. Since caffeic acid, p-coumaric acid, and ferulic acid detected in adlay tea are known to inhibit MPO activity, these polyphenols may be the major functional molecules. Collectively, adlay tea is considered to have a preventative effect against lifestyle-related diseases through improving vascular endothelial function by effects to maintain immune homeostasis of the contained polyphenols. This trial was registered at University Hospital Medical Information Network Clinical Trials Registry (UMIN000032263).


Sujet(s)
Endothélium vasculaire , Homéostasie , Myeloperoxidase , Polyphénols , Thé , Humains , Méthode en double aveugle , Mâle , Femelle , Adulte , Thé/composition chimique , Homéostasie/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Endothélium vasculaire/effets des médicaments et des substances chimiques , Polyphénols/pharmacologie , Polyphénols/administration et posologie , Myeloperoxidase/métabolisme , Graines/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie , Inflammation , Acides caféiques/pharmacologie , Médecine traditionnelle chinoise/méthodes
2.
Nutrients ; 16(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38931232

RÉSUMÉ

Abnormal glucose homeostasis is associated with metabolic syndromes including cardiovascular diseases, hypertension, type 2 diabetes mellitus, and obesity, highlighting the significance of maintaining a balanced glucose level for optimal biological function. This highlights the importance of maintaining normal glucose levels for proper biological functioning. Sulforaphane (SFN), the primary bioactive compound in broccoli from the Cruciferae or Brassicaceae family, has been shown to enhance glucose homeostasis effectively while exhibiting low cytotoxicity. This paper assesses the impact of SFN on glucose homeostasis in vitro, in vivo, and human trials, as well as the molecular mechanisms that drive its regulatory effects. New strategies have been proposed to enhance the bioavailability and targeted delivery of SFN in order to overcome inherent instability. The manuscript also covers the safety evaluations of SFN that have been documented for its production and utilization. Hence, a deeper understanding of the favorable influence and mechanism of SFN on glucose homeostasis, coupled with the fact that SFN is abundant in the human daily diet, may ultimately offer theoretical evidence to support its potential use in the food and pharmaceutical industries.


Sujet(s)
Homéostasie , Isothiocyanates , Sulfoxydes , Isothiocyanates/pharmacologie , Isothiocyanates/administration et posologie , Humains , Homéostasie/effets des médicaments et des substances chimiques , Animaux , Glucose/métabolisme , Brassica/composition chimique , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Biodisponibilité
3.
In Vivo ; 38(4): 1557-1570, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936927

RÉSUMÉ

BACKGROUND/AIM: This study examined the effects of tocotrienols (TT) in conjunction with statin on glucose homeostasis, bone microstructure, gut microbiome, and systemic and liver inflammatory markers in obese C57BL/6J mice. MATERIALS AND METHODS: Forty male C57BL/6J mice were fed a high-fat diet (HFD) and assigned into four groups in a 2 (no statin vs. 120 mg statin/kg diet)×2 (no TT vs. 400 mg TT/kg diet) factorial design for 14 weeks. RESULTS: Statin and TT improved glucose tolerance only when each was given alone, and only statin supplementation decreased insulin resistance. Consistently, only statin supplementation decreased serum insulin levels and HOMA-IR. Pancreatic insulin was also increased with statin treatment. Statin and TT, alone or in combination, reduced the levels of serum IL-6, but only TT attenuated the increased serum leptin levels induced by a HFD. Statin supplementation increased bone area/total area and connectivity density at LV-4, while TT supplementation increased bone area/total area and trabecular number, but decreased trabecular separation at the distal femur. Statin supplementation, but not TT, reduced hepatic inflammatory cytokine gene expression. Neither TT supplementation nor statin supplementation statistically altered microbiome species evenness or richness. However, they altered the relative abundance of certain microbiome species. Most notably, both TT and statin supplementation increased the relative abundance of Lachnospiraceae UCG-006. CONCLUSION: TT and statin collectively benefit bone microstructure, glucose homeostasis, and microbial ecology in obese mice. Such changes may be, in part, associated with suppression of inflammation in the host.


Sujet(s)
Os et tissu osseux , Alimentation riche en graisse , Compléments alimentaires , Microbiome gastro-intestinal , Homéostasie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Obésité , Tocotriénols , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Tocotriénols/pharmacologie , Tocotriénols/administration et posologie , Souris , Homéostasie/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Obésité/métabolisme , Mâle , Os et tissu osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/métabolisme , Os et tissu osseux/anatomopathologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Alimentation riche en graisse/effets indésirables , Bixaceae/composition chimique , Souris obèse , Extraits de plantes/pharmacologie , Extraits de plantes/administration et posologie , Glucose/métabolisme , Souris de lignée C57BL , Insulinorésistance , Glycémie , Modèles animaux de maladie humaine , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Marqueurs biologiques , Caroténoïdes
4.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38928030

RÉSUMÉ

Disruption of any stage of iron homeostasis, including uptake, utilization, efflux, and storage, can cause progressive damage to peripheral organs. The health hazards associated with occupational exposure to inhalation anesthetics (IA) in combination with chronic iron overload are not well documented. This study aimed to investigate changes in the concentration of essential metals in the peripheral organs of rats after iron overload in combination with IA. The aim was also to determine how iron overload in combination with IA affects tissue metal homeostasis, hepcidin-ferritin levels, and MMP levels according to physiological, functional, and tissue features. According to the obtained results, iron accumulation was most pronounced in the liver (19×), spleen (6.7×), lungs (3.1×), and kidneys (2.5×) compared to control. Iron accumulation is associated with elevated heavy metal levels and impaired essential metal concentrations due to oxidative stress (OS). Notably, the use of IA increases the iron overload toxicity, especially after Isoflurane exposure. The results show that the regulation of iron homeostasis is based on the interaction of hepcidin, ferritin, and other proteins regulated by inflammation, OS, free iron levels, erythropoiesis, and hypoxia. Long-term exposure to IA and iron leads to the development of numerous adaptation mechanisms in response to toxicity, OS, and inflammation. These adaptive mechanisms of iron regulation lead to the inhibition of MMP activity and reduction of oxidative stress, protecting the organism from possible damage.


Sujet(s)
Anesthésiques par inhalation , Hepcidines , Dextriferron , Fer , Stress oxydatif , Animaux , Rats , Hepcidines/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Fer/métabolisme , Mâle , Anesthésiques par inhalation/effets indésirables , Anesthésiques par inhalation/toxicité , Dextriferron/administration et posologie , Dextriferron/toxicité , Ferritines/métabolisme , Surcharge en fer/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/anatomopathologie , Rate/effets des médicaments et des substances chimiques , Rate/métabolisme , Rate/anatomopathologie , Rat Wistar , Homéostasie/effets des médicaments et des substances chimiques , Isoflurane/effets indésirables
5.
J Transl Med ; 22(1): 593, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918793

RÉSUMÉ

BACKGROUND: Sorafenib resistance is becoming increasingly common and disadvantageous for hepatocellular carcinoma (HCC) treatment. Ferroptosis is an iron dependent programmed cell death underlying the mechanism of sorafenib. Iron is crucial for synthesis of cofactors essential to mitochondrial enzymes and necessary for HCC proliferation, while mitochondrial iron overload and oxidative stress are associated with sorafenib induced ferroptosis. However, the crosstalk among iron homeostasis and sorafenib resistance is unclear. METHODS: We conducted bioinformatics analysis of sorafenib treated HCC datasets to analyze GCN5L1 and iron related gene expression with sorafenib resistance. GCN5L1 deleted HCC cell lines were generated by CRISPR technology. Sorafenib resistant HCC cell line was established to validate dataset analysis and evaluate the effect of potential target. RESULTS: We identified GCN5L1, a regulator of mitochondrial acetylation, as a modulator in sorafenib-induced ferroptosis via affecting mitochondrial iron homeostasis. GCN5L1 deficiency significantly increased sorafenib sensitivity in HCC cells by down-regulating mitochondrial iron transporters CISD1 expression to induce iron accumulation. Mitochondrial iron accumulation leads to an acceleration in cellular and lipid ROS. Sorafenib resistance is related to CISD1 overexpression to release mitochondrial iron and maintaining mitochondrial homeostasis. We combined CISD1 inhibitor NL-1 with sorafenib, which significantly enhanced sorafenib-induced ferroptosis by promoting mitochondrial iron accumulation and lipid peroxidation. The combination of NL-1 with sorafenib enhanced sorafenib efficacy in vitro and in vivo. CONCLUSIONS: Our findings demonstrate that GCN5L1/CISD1 axis is crucial for sorafenib resistance and would be a potential therapeutic strategy for sorafenib resistant HCC.


Sujet(s)
Carcinome hépatocellulaire , Résistance aux médicaments antinéoplasiques , Ferroptose , Homéostasie , Fer , Tumeurs du foie , Mitochondries , Sorafénib , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Fer/métabolisme , Humains , Homéostasie/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Souris nude , Espèces réactives de l'oxygène/métabolisme , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
6.
Theranostics ; 14(8): 3282-3299, 2024.
Article de Anglais | MEDLINE | ID: mdl-38855179

RÉSUMÉ

Rationale: Pharmacological targeting of mitochondrial ion channels is developing as a new direction in cancer therapy. The opening or closing of these channels can impact mitochondrial function and structure by interfering with intracellular ion homeostasis, thereby regulating cell fate. Nevertheless, their abnormal expression or regulation poses challenges in eliminating cancer cells, and further contributes to metastasis, recurrence, and drug resistance. Methods: We developed an engineered mitochondrial targeted delivery system with self-reinforcing potassium ion (K+) influx via amphiphilic mitochondrial targeting polymer (TMP) as carriers to co-deliver natural K+ channel agonists (Dinitrogen oxide, DZX) and artificial K+ channel molecules (5F8). Results: Using this method, DZX specifically activated natural K+ channels, whereas 5F8 assembled artificial K+ channels on the mitochondrial membrane, leading to mitochondrial K+ influx, as well as oxidative stress and activation of the mitochondrial apoptotic pathway. Conclusion: The synergistic effect of 5F8 and DZX presents greater effectiveness in killing cancer cells than DZX alone, and effectively inhibited tumor recurrence and lung metastasis following surgical resection of breast cancer tumors in animal models. This strategy innovatively integrates antihypertensive drugs with artificial ion channel molecules for the first time to effectively inhibit tumor recurrence and metastasis by disrupting intracellular ion homeostasis, which will provide a novel perspective for postoperative tumor therapy.


Sujet(s)
Homéostasie , Mitochondries , Animaux , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Humains , Homéostasie/effets des médicaments et des substances chimiques , Souris , Lignée cellulaire tumorale , Femelle , Récidive tumorale locale/prévention et contrôle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Potassium/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/secondaire , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Souris de lignée BALB C , Canaux ioniques/métabolisme , Canaux potassiques/métabolisme , Souris nude , Métastase tumorale
7.
ACS Appl Mater Interfaces ; 16(23): 29844-29855, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38829261

RÉSUMÉ

Copper plays critical roles as a metal active site cofactor and metalloallosteric signal for enzymes involved in cell proliferation and metabolism, making it an attractive target for cancer therapy. In this study, we investigated the efficacy of polydopamine nanoparticles (PDA NPs), classically applied for metal removal from water, as a therapeutic strategy for depleting intracellular labile copper pools in triple-negative breast cancer models through the metal-chelating groups present on the PDA surface. By using the activity-based sensing probe FCP-1, we could track the PDA-induced labile copper depletion while leaving total copper levels unchanged and link it to the selective MDA-MB-231 cell death. Further mechanistic investigations revealed that PDA NPs increased reactive oxygen species (ROS) levels, potentially through the inactivation of superoxide dismutase 1 (SOD1), a copper-dependent antioxidant enzyme. Additionally, PDA NPs were found to interact with the mitochondrial membrane, resulting in an increase in the mitochondrial membrane potential, which may contribute to enhanced ROS production. We employed an in vivo tumor model to validate the therapeutic efficacy of PDA NPs. Remarkably, in the absence of any additional treatment, the presence of PDA NPs alone led to a significant reduction in tumor volume by a factor of 1.66 after 22 days of tumor growth. Our findings highlight the potential of PDA NPs as a promising therapeutic approach for selectively targeting cancer by modulating copper levels and inducing oxidative stress, leading to tumor growth inhibition as shown in these triple-negative breast cancer models.


Sujet(s)
Cuivre , Indoles , Nanoparticules , Polymères , Espèces réactives de l'oxygène , Tumeurs du sein triple-négatives , Cuivre/composition chimique , Cuivre/pharmacologie , Polymères/composition chimique , Polymères/pharmacologie , Indoles/composition chimique , Indoles/pharmacologie , Humains , Animaux , Souris , Nanoparticules/composition chimique , Femelle , Espèces réactives de l'oxygène/métabolisme , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Lignée cellulaire tumorale , Oxydoréduction , Nanomédecine , Prolifération cellulaire/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Superoxide dismutase-1/métabolisme
8.
Cell Mol Life Sci ; 81(1): 269, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38884791

RÉSUMÉ

Betaine is an endogenous osmolyte that exhibits therapeutic potential by mitigating various neurological disorders. However, the underlying cellular and molecular mechanisms responsible for its neuroprotective effects remain puzzling.In this study, we describe a possible mechanism behind the positive impact of betaine in preserving neurons from excitotoxicity. Here we demonstrate that betaine at low concentration modulates the GABA uptake by GAT1 (slc6a1), the predominant GABA transporter in the central nervous system. This modulation occurs through the temporal inhibition of the transporter, wherein prolonged occupancy by betaine impedes the swift transition of the transporter to the inward conformation. Importantly, the modulatory effect of betaine on GAT1 is reversible, as the blocking of GAT1 disappears with increased extracellular GABA. Using electrophysiology, mass spectroscopy, radiolabelled cellular assay, and molecular dynamics simulation we demonstrate that betaine has a dual role in GAT1: at mM concentration acts as a slow substrate, and at µM as a temporal blocker of GABA, when it is below its K0.5. Given this unique modulatory characteristic and lack of any harmful side effects, betaine emerges as a promising neuromodulator of the inhibitory pathways improving GABA homeostasis via GAT1, thereby conferring neuroprotection against excitotoxicity.


Sujet(s)
Bétaïne , Transporteurs de GABA , Homéostasie , Acide gamma-amino-butyrique , Transporteurs de GABA/métabolisme , Bétaïne/pharmacologie , Bétaïne/métabolisme , Acide gamma-amino-butyrique/métabolisme , Animaux , Homéostasie/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Simulation de dynamique moléculaire , Humains , Rats , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/métabolisme , Cellules HEK293
9.
Inorg Chem ; 63(25): 11779-11787, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38850241

RÉSUMÉ

Cisplatin is a widely used anticancer drug. In addition to inducing DNA damage, increased levels of reactive oxygen species (ROS) play a significant role in cisplatin-induced cell death. Thioredoxin-1 (Trx1), a redox regulatory protein that can scavenge ROS, has been found to eliminate cisplatin-induced ROS, while elevated Trx1 levels are associated with cisplatin resistance. However, it is unknown whether the effect of Trx1 on the cellular response to cisplatin is due to its direct reaction and how this reaction influences the activity of Trx1. In this work, we performed detailed studies of the reaction between Trx1 and cisplatin. Trx1 is highly reactive to cisplatin, and the catalytic motif of Trx1 (CGPC) is the primary binding site of cisplatin. Trx1 can bind up to 6 platinum moieties, resulting in the structural alteration and oligomerization of Trx1 depending on the degree of platination. Platination of Trx1 inhibits its interaction with ASK1, a Trx1-binding protein that regulates cell apoptosis. Furthermore, the reaction with cisplatin suppresses drug-induced ROS generation, which could be associated with drug resistance. This study provides more insight into the mechanism of action of cisplatin.


Sujet(s)
Antinéoplasiques , Cisplatine , MAP Kinase Kinase Kinase 5 , Oxydoréduction , Espèces réactives de l'oxygène , Thiorédoxines , Cisplatine/pharmacologie , Cisplatine/composition chimique , Thiorédoxines/métabolisme , Thiorédoxines/composition chimique , Humains , Espèces réactives de l'oxygène/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , MAP Kinase Kinase Kinase 5/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
10.
Phytomedicine ; 131: 155782, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38851102

RÉSUMÉ

BACKGROUND: Asthma is a complex disease with mechanisms involving multiple factors, and there is still a lack of highly effective and low-side-effect drugs. Traditional Chinese medicine Fagopyrum Dibotrys Rhizoma (FDR) has been applied for the treatment of acute and chronic bronchitis as well as bronchial asthma due to its favorable pharmacological activity. However, the exact mechanism of FDR remains unclear. OBJECTIVE: A mouse model of asthma was created using OVA and HDM. To investigate the mechanism of FDR in asthma treatment, a combination of network pharmacology, lipidomics, and molecular biology approaches was employed. METHODS: To evaluate the therapeutic effects of FDR on asthma, we established two distinct models of asthma in C57BL/6 J mice using OVA and HDM, respectively. We then employed LC-MS to analyze the major chemical constituents in FDR. Next, the network pharmacology approach was used to predict the potential targets and mechanisms of FDR in asthma treatment. Additionally, lipidomics analysis of mouse serum was conducted using LC-MS. Finally, the impact of FDR on the ERK -cPLA2 signaling pathway was investigated through Western Blotting assay. RESULTS: FDR treatment has been shown to improve histomorphological changes, lung function and inflammation in models of OVA and HDM-induced asthma. Using UPLC/LTQ-Orbitrap-MS, we were able to identify 12 potential active components. Network pharmacology analysis revealed that FDR shares 75 targets with asthma. Further analysis using GO and KEGG pathways demonstrated the involvement of key pathways such as PI3K-Akt, TNF, and MAPK. Additionally, lipidomics analysis of the serum from OVA and HDM induced asthma mice showed disturbances in lipid metabolism, which were effectively ameliorated by FDR treatment. Mechanistically, FDR inhibits ERK1/2-cPLA2, leading to a reduction in lysophospholipids and restoration of lipid balance, thereby aiding in the treatment of asthma. CONCLUSION: FDR has been shown to improve lipid metabolism disorder in the serum of asthmatic mice, thereby potentially serving as a treatment for asthma. This can be achieved by regulating the activation levels of ERK1/2 and p38MAPK. Consequently, the production of lysophosphatide is reduced, thereby alleviating the disorder of lipid metabolism and achieving the desired therapeutic effect in asthma treatment.


Sujet(s)
Asthme , Modèles animaux de maladie humaine , Fagopyrum , Métabolisme lipidique , Souris de lignée C57BL , Rhizome , Animaux , Asthme/traitement médicamenteux , Métabolisme lipidique/effets des médicaments et des substances chimiques , Rhizome/composition chimique , Souris , Fagopyrum/composition chimique , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Femelle , Médicaments issus de plantes chinoises/pharmacologie , Homéostasie/effets des médicaments et des substances chimiques , Lipidomique , Transduction du signal/effets des médicaments et des substances chimiques , Pharmacologie des réseaux , Ovalbumine , Phospholipases A2/métabolisme
11.
Biomed Res Int ; 2024: 7747599, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884019

RÉSUMÉ

Introduction: PPIs, or proton pump inhibitors, are the most widely prescribed drugs. There is a debate regarding the relationship between long-term PPI use and the risk of type 2 diabetes mellitus (T2DM). A potential connection between T2DM and PPIs could be an elevated gastrin concentration. This study is aimed at investigating the long-term effects of PPI omeprazole (OZ) on glucose homeostasis and pancreatic gene expression profile in mice. Methods: Healthy adult male BALB/c mice were randomly divided into three equal groups (n = 10 in each one): (1) experimental mice that received OZ 20 mg/kg; (2) control mice that received 30 µl saline per os; (3) intact mice without any interventions. Mice were treated for 30 weeks. Glucose homeostasis was investigated by fasting blood glucose level, oral glucose tolerance test (GTT), insulin tolerance test (ITT), and basal insulin resistance (HOMA-IR). Serum gastrin and insulin concentration were determined by ELISA. Expressions of Sirt1, Pparg, Nfκb1 (p105), Nfe2l2, Cxcl5, Smad3, H2a.z, and H3f3b were measured by RT-PCR. Result: The ROC analysis revealed an increase in fasting blood glucose levels in OZ-treated mice in comparison with control and intact groups during the 30-week experiment. A slight but statistically significant increase in glucose tolerance and insulin sensitivity was observed in OZ-treated mice within 30 weeks of the experiment. The mice treated with OZ exhibited significant increases in serum insulin and gastrin levels, accompanied by a rise in the HOMA-IR level. These animals had a statistically significant increase in Sirt1, Pparg, and Cxcl5 mRNA expression. There were no differences in ß-cell numbers between groups. Conclusion: Long-term OZ treatment induced hypergastrin- and hyperinsulinemia and increased expression of Sirt1, Pparg, and Cxcl5 in mouse pancreatic tissues accompanied by specific changes in glucose metabolism. The mechanism of omeprazole-induced Cxcl5 mRNA expression and its association with pancreatic cancer risk should be investigated.


Sujet(s)
Glycémie , Gastrines , Homéostasie , Insulinorésistance , Souris de lignée BALB C , Oméprazole , Animaux , Oméprazole/pharmacologie , Oméprazole/effets indésirables , Gastrines/sang , Gastrines/métabolisme , Mâle , Souris , Homéostasie/effets des médicaments et des substances chimiques , Glycémie/métabolisme , Insuline/métabolisme , Insuline/sang , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Diabète de type 2/métabolisme , Diabète de type 2/génétique , Diabète de type 2/induit chimiquement , Hyperglycémie provoquée , Inhibiteurs de la pompe à protons/pharmacologie , Inhibiteurs de la pompe à protons/effets indésirables , Glucose/métabolisme
12.
Nutrients ; 16(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38892670

RÉSUMÉ

Tumor cells are characterized by a delicate balance between elevated oxidative stress and enhanced antioxidant capacity. This intricate equilibrium, maintained within a threshold known as redox homeostasis, offers a unique perspective for cancer treatment by modulating reactive oxygen species (ROS) levels beyond cellular tolerability, thereby disrupting this balance. However, currently used chemotherapy drugs require larger doses to increase ROS levels beyond the redox homeostasis threshold, which may cause serious side effects. How to disrupt redox homeostasis in cancer cells more effectively remains a challenge. In this study, we found that sodium selenite and docosahexaenoic acid (DHA), a polyunsaturated fatty acid extracted from marine fish, synergistically induced cytotoxic effects in colorectal cancer (CRC) cells. Physiological doses of DHA simultaneously upregulated oxidation and antioxidant levels within the threshold range without affecting cell viability. However, it rendered the cells more susceptible to reaching the upper limit of the threshold of redox homeostasis, facilitating the elevation of ROS levels beyond the threshold by combining with low doses of sodium selenite, thereby disrupting redox homeostasis and inducing MAPK-mediated paraptosis. This study highlights the synergistic anticancer effects of sodium selenite and DHA, which induce paraptosis by disrupting redox homeostasis in tumor cells. These findings offer a novel strategy for more targeted and less toxic cancer therapies for colorectal cancer treatment.


Sujet(s)
Tumeurs colorectales , Acide docosahexaénoïque , Homéostasie , Système de signalisation des MAP kinases , Oxydoréduction , Espèces réactives de l'oxygène , Sélénite de sodium , Acide docosahexaénoïque/pharmacologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Sélénite de sodium/pharmacologie , Humains , Oxydoréduction/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Stress oxydatif/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Synergie des médicaments , Antinéoplasiques/pharmacologie ,
13.
J Am Heart Assoc ; 13(13): e035990, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38934871

RÉSUMÉ

BACKGROUND: Experimental preeclampsia (ePE) has been shown to have worsened outcome from stroke. We investigated the effect of low-dose aspirin, known to prevent preeclampsia, on stroke hemodynamics and outcome, and the association between the vasoconstrictor and vasodilator cyclooxygenase products thromboxane A2 and prostacyclin. METHODS AND RESULTS: Middle cerebral artery occlusion was performed for 3 hours with 1 hour of reperfusion in normal pregnant rats on day 20 of gestation and compared with ePE treated with vehicle or low-dose aspirin (1.5 mg/kg per day). Multisite laser Doppler was used to measure changes in cerebral blood flow to the core middle cerebral artery and collateral vascular territories. After 30 minutes occlusion, phenylephrine was infused to increase blood pressure and assess cerebral blood flow autoregulation. Infarct and edema were measured using 2,3,5-triphenyltetrazolium chloride staining. Plasma levels of thromboxane A2, prostacyclin, and inflammatory markers in plasma and cyclooxygenase levels in cerebral arteries were measured. ePE had increased infarction compared with normal pregnant rats (P<0.05) that was reduced by aspirin (P<0.001). ePE also had intact cerebral blood flow autoregulation and reduced collateral perfusion during induced hypertension that was also prevented by aspirin. Aspirin increased prostacyclin in ePE (P<0.05) without reducing thromboxane B2, metabolite of thromboxane A2, or 8-isoprostane-prostaglandin-2α, a marker of lipid peroxidation. There were no differences in cyclooxygenase levels in cerebral arteries between groups. CONCLUSIONS: Low-dose aspirin in ePE reduced infarction that was associated with increased vasodilator prostacyclin and improved collateral perfusion during induced hypertension. The beneficial effect of aspirin on the brain and cerebral circulation is likely multifactorial and worth further study.


Sujet(s)
Acide acétylsalicylique , Circulation cérébrovasculaire , Circulation collatérale , Modèles animaux de maladie humaine , Homéostasie , Pré-éclampsie , Rat Sprague-Dawley , Animaux , Femelle , Grossesse , Acide acétylsalicylique/administration et posologie , Acide acétylsalicylique/pharmacologie , Circulation cérébrovasculaire/effets des médicaments et des substances chimiques , Pré-éclampsie/physiopathologie , Pré-éclampsie/métabolisme , Pré-éclampsie/traitement médicamenteux , Homéostasie/effets des médicaments et des substances chimiques , Circulation collatérale/effets des médicaments et des substances chimiques , Thromboxane A2/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/physiopathologie , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Vitesse du flux sanguin/effets des médicaments et des substances chimiques , Rats , Prostacycline/métabolisme , Fluxmétrie laser Doppler
14.
Molecules ; 29(12)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38930917

RÉSUMÉ

In the field of human health research, the homeostasis of copper (Cu) is receiving increased attention due to its connection to pathological conditions, including diabetes mellitus (DM). Recent studies have demonstrated that proteins associated with Cu homeostasis, such as ATOX1, FDX1, ATP7A, ATPB, SLC31A1, p53, and UPS, also contribute to DM. Cuproptosis, characterized by Cu homeostasis dysregulation and Cu overload, has been found to cause the oligomerization of lipoylated proteins in mitochondria, loss of iron-sulfur protein, depletion of glutathione, production of reactive oxygen species, and cell death. Further research into how cuproptosis affects DM is essential to uncover its mechanism of action and identify effective interventions. In this article, we review the molecular mechanism of Cu homeostasis and the role of cuproptosis in the pathogenesis of DM. The study of small-molecule drugs that affect these proteins offers the possibility of moving from symptomatic treatment to treating the underlying causes of DM.


Sujet(s)
Cuivre , Diabète , Conception de médicament , Homéostasie , Humains , Diabète/traitement médicamenteux , Diabète/métabolisme , Cuivre/composition chimique , Cuivre/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Animaux , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme
15.
Nutrients ; 16(9)2024 Apr 28.
Article de Anglais | MEDLINE | ID: mdl-38732567

RÉSUMÉ

Imbalances in lipid uptake and efflux and inflammation are major contributors to foam cell formation, which is considered a therapeutic target to protect against atherosclerosis. Naringin, a citrus flavonoid abundant in citrus fruits, has been reported to exert an antiatherogenic function, but its pharmacological mechanism is unclear. Naringin treatment effectively inhibits foam cell formation in THP-1 and RAW264.7 macrophages. In this study, mechanically, naringin maintained lipid homeostasis within macrophages through downregulation of the key genes for lipid uptake (MSR1 and CD36) and the upregulation of ABCA1, ABCG1 and SR-B1, which are responsible for cholesterol efflux. Meanwhile, naringin significantly decreased the cholesterol synthesis-related genes and increased the genes involved in cholesterol metabolism. Subsequently, the results showed that ox-LDL-induced macrophage inflammatory responses were inhibited by naringin by reducing the proinflammatory cytokines IL-1ß, IL-6 and TNF-α, and increasing the anti- inflammatory cytokine IL-10, which was further verified by the downregulation of pro-inflammatory and chemokine-related genes. Additionally, we found that naringin reprogrammed the metabolic phenotypes of macrophages by suppressing glycolysis and promoting lipid oxidation metabolism to restore macrophage phenotypes and functions. These results suggest that naringin is a potential drug for the treatment of AS as it inhibits macrophage foam cell formation by regulating metabolic phenotypes and inflammation.


Sujet(s)
Flavanones , Cellules spumeuses , Homéostasie , Métabolisme lipidique , Animaux , Humains , Souris , Cholestérol/métabolisme , Cytokines/métabolisme , Flavanones/pharmacologie , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Métabolisme lipidique/effets des médicaments et des substances chimiques , Lipoprotéines LDL/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Phénotype , Cellules RAW 264.7 , Cellules THP-1
16.
Ecotoxicol Environ Saf ; 279: 116451, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38759535

RÉSUMÉ

Bile acid homeostasis is critical to human health. Low-level exposure to antibiotics has been suggested to potentially disrupt bile acid homeostasis by affecting gut microbiota, but relevant data are still lacking in humans, especially for the level below human safety threshold. We conducted a cross-sectional study in 4247 Chinese adults by measuring 34 parent antibiotics and their metabolites from six common categories (i.e., tetracyclines, qinolones, macrolides, sulfonamides, phenicols, and lincosamides) and ten representative bile acids in fasting morning urine using liquid chromatography coupled to mass spectrometry. Daily exposure dose of antibiotics was estimated from urinary concentrations of parent antibiotics and their metabolites. Urinary bile acids and their ratios were used to reflect bile acid homeostasis. The estimated daily exposure doses (EDED) of five antibiotic categories with a high detection frequency (i.e., tetracyclines, qinolones, macrolides, sulfonamides, and phenicols) were significantly associated with urinary concentrations of bile acids and decreased bile acid ratios in all adults and the subset of 3898 adults with a cumulative ratio of antibiotic EDED to human safety threshold of less than one. Compared to a negative detection of antibiotics, the lowest EDED quartiles of five antibiotic categories and four individual antibiotics with a high detection frequency (i.e., ciprofloxacin, ofloxacin, trimethoprim, and florfenicol) in the adults with a positive detection of antibiotics had a decrease of bile acid ratio between 6.6% and 76.6%. Except for macrolides (1.2×102 ng/kg/day), the medians of the lowest EDED quartile of antibiotic categories and individual antibiotics ranged from 0.32 ng/kg/day to 10 ng/kg/day, which were well below human safety thresholds. These results suggested that low-level antibiotic exposure could disrupt bile acid homeostasis in adults and existing human safety thresholds may be inadequate in safeguarding against the potential adverse health effects of low-level exposure to antibiotics.


Sujet(s)
Antibactériens , Acides et sels biliaires , Homéostasie , Humains , Acides et sels biliaires/urine , Acides et sels biliaires/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Adulte , Mâle , Femelle , Études transversales , Adulte d'âge moyen , Chine , Exposition environnementale/analyse , Jeune adulte
17.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167210, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38704001

RÉSUMÉ

Oxaliplatin has been included as a basal drug in various chemotherapy regimens for colorectal cancer (CRC), a global health concern. However, acquired resistance to oxaliplatin affects the prognosis. This study aimed to determine whether the consumption of a KD increases the sensitivity of CRC cells to oxaliplatin via the inhibition of a classical stem cell marker, Krupple-like factor 5 (KLF5). KLF5 functions as a transcription factor for the leukemia inhibitory factor (LIF) and directly binds to its promoter region. LIF upregulation induces dephosphorylation of metal regulatory transcription factor 1 (MTF1), which is recruited to the promoter area of Ferroportin (FPN1), the only cellular iron exporter. FPN1 upregulation reduces the labile iron pool (LIP) and ferroptosis in CRC cells. KLF5 knockdown inhibits the LIF/MTF1/FPN1 axis and induces iron overload, thereby conferring sensitivity to oxaliplatin to CRC cells. KD mimicked KLF5 silencing and sensitized CRC cells to oxaliplatin via a similar mechanism. Thus, potential correlations were observed among ketogenesis, stemness, and iron homeostasis. This finding can be used to formulate a new strategy for overcoming oxaliplatin resistance in patients with CRC.


Sujet(s)
Transporteurs de cations , Tumeurs colorectales , Résistance aux médicaments antinéoplasiques , Homéostasie , Fer , Facteurs de transcription Krüppel-like , Facteur inhibiteur de la leucémie , Oxaliplatine , Humains , Oxaliplatine/pharmacologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Fer/métabolisme , Transporteurs de cations/métabolisme , Transporteurs de cations/génétique , Homéostasie/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Facteur inhibiteur de la leucémie/métabolisme , Facteur inhibiteur de la leucémie/génétique , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Animaux
18.
Eur J Histochem ; 68(2)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38742403

RÉSUMÉ

Chronic kidney disease (CKD) is a leading public health issue associated with high morbidity worldwide. However, there are only a few effective therapeutic strategies for CKD. Emodin, an anthraquinone compound from rhubarb, can inhibit fibrosis in tissues and cells. Our study aims to investigate the antifibrotic effect of emodin and the underlying molecular mechanism. A unilateral ureteral obstruction (UUO)-induced rat model was established to evaluate the effect of emodin on renal fibrosis development. Hematoxylin and eosin staining, Masson's trichrome staining, and immunohistochemistry staining were performed to analyze histopathological changes and fibrotic features after emodin treatment. Subsequently, a transforming growth factor-beta 1 (TGF-ß1)-induced cell model was used to assess the inhibition of emodin on cell fibrosis in vitro. Furthermore, Western blot analysis and real-time quantitative reverse transcription-polymerase chain reaction were performed to validate the regulatory mechanism of emodin on renal fibrosis progression. As a result, emodin significantly improved histopathological abnormalities in rats with UUO. The expression of fibrosis biomarkers and mitochondrial biogenesis-related proteins also decreased after emodin treatment. Moreover, emodin blocked TGF-ß1-induced fibrotic phenotype, lipid accumulation, and mitochondrial homeostasis in NRK-52E cells. Conversely, peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) silencing significantly reversed these features in emodin-treated cells. Collectively, emodin plays an important role in regulating PGC-1α-mediated mitochondria function and energy homeostasis. This indicates that emodin exhibits great inhibition against renal fibrosis and acts as a promising inhibitor of CKD.


Sujet(s)
Émodine , Fibrose , Mitochondries , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Insuffisance rénale chronique , Animaux , Émodine/pharmacologie , Émodine/usage thérapeutique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Fibrose/traitement médicamenteux , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mâle , Rats , Rat Sprague-Dawley , Homéostasie/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Obstruction urétérale/anatomopathologie , Obstruction urétérale/traitement médicamenteux , Facteur de croissance transformant bêta-1/métabolisme , Lignée cellulaire
19.
ACS Appl Bio Mater ; 7(5): 2836-2850, 2024 05 20.
Article de Anglais | MEDLINE | ID: mdl-38717017

RÉSUMÉ

High-altitude regions, cold deserts, permafrost regions, and the polar region have some of the severest cold conditions on earth and pose immense perils of cold injuries to exposed individuals. Accidental and unintended exposures to severe cold, either unintentionally or due to occupational risks, can greatly increase the risk of serious conditions including hypothermia, trench foot, and cold injuries like frostbite. Cold-induced vasoconstriction and intracellular/intravascular ice crystal formation lead to hypoxic conditions at the cellular level. The condition is exacerbated in individuals having inadequate and proper covering and layering, particularly when large area of the body are exposed to extremely cold environments. There is a paucity of preventive and therapeutic pharmacological modalities that have been explored for managing and treating cold injuries. Given this, an efficient modality that can potentiate the healing of frostbite was investigated by studying various complex pathophysiological changes that occur during severe cold injuries. In the current research, we report the effectiveness and healing properties of a standardized formulation, i.e., a herbosomal-loaded PEG-poloxamer topical formulation (n-HPTF), on frostbite. The intricate mechanistic pathways modulated by the novel formulation have been elucidated by studying the pathophysiological sequelae that occur following severe cold exposures leading to frostbite. The results indicate that n-HPTF ameliorates the outcome of frostbite, as it activates positive sensory nerves widely distributed in the epidermis transient receptor potential vanilloid 1 (TRPV1), significantly (p < 0.05) upregulates cytokeratin-14, promotes angiogenesis (VEGF-A), prominently represses the expression of thromboxane formation (TXA2), and significantly (p < 0.05) restores levels of enzymatic (glutathione reductase, superoxide dismutase, and catalase) and nonenzymatic antioxidants (glutathione). Additionally, n-HPTF attenuates oxidative stress and the expression of inflammatory proteins PGF-2α, NFκB-p65, TNF-α, IL-6, IL-1ß, malondialdehyde (MDA), advanced oxidative protein products (AOPP), and protein carbonylation (PCO). Masson's Trichrome staining showed that n-HPTF stimulates cellular proliferation, and increases collagen fiber deposition, which significantly (p < 0.05) promotes the healing of frostbitten tissue, as compared to control. We conclude that protection against severe cold injuries by n-HPTF is mediated via modulation of pathways involving TRPV1, VEGF-A, TXA2, redox homeostasis, and inflammatory cascades. The study is likely to have widespread implications for the prophylaxis and management of moderate-to-severe frostbite conditions.


Sujet(s)
Homéostasie , Poloxamère , Polyéthylène glycols , Canaux cationiques TRPV , Facteur de croissance endothéliale vasculaire de type A , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Canaux cationiques TRPV/métabolisme , Animaux , Poloxamère/composition chimique , Poloxamère/pharmacologie , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Homéostasie/effets des médicaments et des substances chimiques , Oxydoréduction , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Rats , Test de matériaux , Lésion due au froid/métabolisme , Lésion due au froid/traitement médicamenteux , Taille de particule , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Mâle , Liposomes/composition chimique , Humains , Administration par voie topique , Engelure/métabolisme , Engelure/traitement médicamenteux
20.
ACS Biomater Sci Eng ; 10(6): 3813-3824, 2024 06 10.
Article de Anglais | MEDLINE | ID: mdl-38779799

RÉSUMÉ

Photodynamic therapy (PDT) using aggregation-induced emission photosensitizer (AIE-PS) holds tremendous potential but is limited by its inherent disadvantages and the high concentrations of reduced glutathione (GSH) in tumor cells that can neutralize ROS to weaken PDT. Herein, we designed a nanodelivery system (CM-HSADSP@[PS-Sor]) in which albumin was utilized as a carrier for hydrophobic drug AIE-PS and Sorafenib, cross-linkers with disulfide bonds were introduced to form a nanogel core, and then cancer cell membranes were wrapped on its surface to confer homologous tumor targeting ability. A two-way strategy was employed to disturb redox-homeostasis through blocking GSH synthesis by Sorafenib and consuming excess GSH via abundant disulfide bonds, thereby promoting the depletion of GSH, which in turn increased the ROS levels in cancer cells to amplify the efficacy of ferroptosis and PDT, achieving an efficient in vivo antibreast cancer effect. This study brings a new strategy for ROS-based cancer therapy and expands the application of an albumin-based drug delivery system.


Sujet(s)
Ferroptose , Oxydoréduction , Photothérapie dynamique , Photosensibilisants , Ferroptose/effets des médicaments et des substances chimiques , Photothérapie dynamique/méthodes , Humains , Photosensibilisants/pharmacologie , Photosensibilisants/usage thérapeutique , Photosensibilisants/composition chimique , Animaux , Espèces réactives de l'oxygène/métabolisme , Souris , Lignée cellulaire tumorale , Glutathion/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Nanoparticules/usage thérapeutique , Tumeurs/traitement médicamenteux , Souris de lignée BALB C , Systèmes de délivrance de médicaments/méthodes , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Sorafénib/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...