Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30.409
Filtrer
1.
Acta Neurobiol Exp (Wars) ; 84(2): 218-229, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39087834

RÉSUMÉ

The present study was assumed that N­acetylcysteine (AC) might improve cognitive function in adolescent rats with hypothyroidism through various mechanisms. Sixty adolescent rats were randomly divided into the following groups: Vehicle (received normal saline intraperitoneally (IP)); Propylthiouracil (PTU)­induced hypothyroidism (0.05%, dissolved in drinking water); Hypothyroid rats were IP treated with different doses of AC (50, 100, and 150 mg/kg/day) for a period of six weeks; Normal rats treated with the highest doses of AC (150 mg/kg/day). Behavioral and biochemical analyses were studied for all groups. In the Morris water maze test, AC significantly reduced both the time to find the hidden platform and the distance travelled as compared to non­treated hypothyroid rats. In the passive avoidance test, the latency of entering the dark chamber was significantly increased by AC, whereas decreased the time spent in the darkroom of the chamber compared to the hypothyroid rats. In biochemical results, AC reduced both malondialdehyde content and nitrite while increased the thiol content, catalase and superoxide dismutase enzymes activity in both the cortex and the hippocampus, and a notable improvement in brain­derived neurotrophic factor (BDNF) levels in hippocampal tissues of the hypothyroid rats, while decreasing the level of interleukin­6 in rat hippocampal region. Therefore, based on the results, the beneficial effects of AC on cognitive impairment in adolescent hypothyroid rats are probably related to its anti­oxidant properties and notable improvement in BDNF levels.


Sujet(s)
Acétylcystéine , Facteur neurotrophique dérivé du cerveau , Hippocampe , Hypothyroïdie , Stress oxydatif , Animaux , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Mâle , Hypothyroïdie/métabolisme , Hypothyroïdie/induit chimiquement , Hypothyroïdie/complications , Acétylcystéine/pharmacologie , Facteur neurotrophique dérivé du cerveau/métabolisme , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Rat Wistar , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/prévention et contrôle , Troubles de la mémoire/étiologie , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Mémoire/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Antioxydants/pharmacologie
2.
Chem Biol Drug Des ; 104(2): e14598, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39090783

RÉSUMÉ

Acne caused by inflammation of hair follicles and sebaceous glands is a common chronic skin disease. Arctigenin (ATG) is an extract of Arctium lappa L., which has significant anti-inflammatory effects. However, the effect and mechanism of ATG in cutaneous inflammation mediated by Cutibacterium acnes (C. acnes) has not been fully evaluated. The purpose of this study was to explore the effect and potential mechanism of ATG in the treatment of acne through network pharmacology and experimental confirmation. An acne model was established by injected live C. acnes into living mice and treated with ATG. Our data showed that ATG effectively improved acne induced by live C. acnes, which was confirmed by determining ear swelling rate, estradiol concentration and hematoxylin and eosin (H&E) staining. In addition, ATG inhibited the NLRP3 inflammasome signaling pathway in mice ear tissues and reduced the secretion of pro-inflammatory cytokines IL-1ß to relieve inflammation. The results of network pharmacology and molecular docking confirmed that ATG can regulate 17ß-Estradiol (E2) levels through targeted to CYP19A1, and finally inhibited skin inflammation. Taken together, our results confirmed that ATG regulated E2 secretion by targeting CYP19A1, thereby inhibiting the NLRP3 inflammasome signaling pathway and improving inflammation levels in acne mice. This study provides a basis for the feasibility of ATG in treating acne in clinical practice.


Sujet(s)
Acné juvénile , Aromatase , Furanes , Lignanes , Simulation de docking moléculaire , Pharmacologie des réseaux , Animaux , Furanes/composition chimique , Furanes/pharmacologie , Souris , Lignanes/pharmacologie , Lignanes/composition chimique , Lignanes/usage thérapeutique , Acné juvénile/traitement médicamenteux , Acné juvénile/microbiologie , Aromatase/métabolisme , Aromatase/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/antagonistes et inhibiteurs , Inflammasomes/métabolisme , Humains , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Anti-inflammatoires/usage thérapeutique , Propionibacterium acnes/effets des médicaments et des substances chimiques , Interleukine-1 bêta/métabolisme , Modèles animaux de maladie humaine
3.
Bull Exp Biol Med ; 177(2): 207-211, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39090471

RÉSUMÉ

In an experimental model of Alzheimer's disease in mice, oral administration of trehalose disaccharide reduces neuroinflammation assessed by the expression level of microglia activation marker Iba1 and affects the neutrophil degranulation activity. A potential anti-inflammatory effect of 4% trehalose solution associated with a decrease in the activity of leukocyte elastase in plasma was revealed.


Sujet(s)
Maladie d'Alzheimer , Modèles animaux de maladie humaine , Microglie , Tréhalose , Animaux , Tréhalose/pharmacologie , Tréhalose/usage thérapeutique , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Souris , Protéines des microfilaments/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Mâle , Protéines de liaison au calcium/métabolisme , Leukocyte elastase/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Diholoside/pharmacologie , Anti-inflammatoires/pharmacologie
4.
Bull Exp Biol Med ; 177(2): 217-220, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39093473

RÉSUMÉ

PT1 peptide isolated from the venom of spider Geolycosa sp. is a modulator of P2X3 receptors that play a role in the development of inflammation and the transmission of pain impulses. The anti-inflammatory and analgesic efficacy of the PT1 peptide was studied in a model of complete Freund's adjuvant-induced paw inflammation in CD-1 mice. The analgesic activity of PT1 peptide was maximum after intramuscular injection at a dose of 0.01 mg/kg, which surpassed the analgesic effect of diclofenac at a dose of 1 mg/kg. The anti-inflammatory activity was maximum after intramuscular injection at a dose of 0.0001 mg/kg; a decrease in paw thickness was observed as soon as 2 h after the administration of the PT1 peptide against the background of inflammation development. All tested doses of PT1 peptide showed high anti-inflammatory activity 4 and 24 h after administration. PT1 peptide at a dose of 0.01 mg/kg when injected intramuscularly simultaneously produced high anti-inflammatory and analgesic effects compared to other doses of the peptide. Increasing the dose of PT1 peptide led to a gradual decrease in its analgesic and anti-inflammatory activity; increasing the dose of intramuscular injection to 0.1 and 1 mg/kg is inappropriate.


Sujet(s)
Analgésiques , Anti-inflammatoires , Inflammation , Peptides , Animaux , Souris , Analgésiques/pharmacologie , Analgésiques/usage thérapeutique , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/administration et posologie , Mâle , Peptides/pharmacologie , Peptides/administration et posologie , Peptides/usage thérapeutique , Injections musculaires , Adjuvant Freund , Venins d'araignée/pharmacologie , Diclofenac/pharmacologie , Diclofenac/usage thérapeutique , Diclofenac/administration et posologie , Modèles animaux de maladie humaine , Douleur/traitement médicamenteux
5.
Immun Inflamm Dis ; 12(8): e1370, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39110084

RÉSUMÉ

BACKGROUND: Endometriosis is associated with a wide variety of signs and symptoms and can lead to infertility, embryo death, and even miscarriage. Although the exact pathogenesis and etiology of endometriosis is still unclear, it has been shown that it has a chronic inflammatory nature and angiogenesis is also involved in it. OBJECTIVE: This review aims to explore the role of inflammation and angiogenesis in endometriosis and suggest a potential treatment targeting these pathways. FINDINGS: Among the pro-inflammatory cytokines, studies have shown solid roles for interleukin 1ß (IL-ß), IL-6, and tumor necrosis factor α (TNF-α) in the pathogenesis of this condition. Other than inflammation, angiogenesis, the formation of new blood vessels from pre-existing capillaries, is also involved in the pathogenesis of endometriosis. Among angiogenic factors, vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1α (HIF-1α), transforming growth factor ß1 (TGF-ß1), and matrix metalloproteinases (MMPs) are more essential in the pathogenesis of endometriosis. Interestingly, it has been shown that inflammation and angiogenesis share some similar pathways with each other that could be potentially targeted for treatment of diseases caused by these two processes. Cannabidiol (CBD) is a non-psychoactive member of cannabinoids which has well-known and notable anti-inflammatory and antiangiogenic properties. This agent has been shown to decrease IL-1ß, IL-6, TNF-α, VEGF, TGFß, and MMPs in different animal models of diseases. CONCLUSION: It seems that CBD could be a possible treatment for endometriosis due to its anti-inflammatory and antiangiogenic activity, however, further studies are needed.


Sujet(s)
Cannabidiol , Endométriose , Inflammation , Néovascularisation pathologique , Endométriose/traitement médicamenteux , Endométriose/anatomopathologie , Femelle , Humains , Cannabidiol/usage thérapeutique , Cannabidiol/pharmacologie , Néovascularisation pathologique/traitement médicamenteux , Inflammation/traitement médicamenteux , Animaux , Cytokines/métabolisme , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Inhibiteurs de l'angiogenèse/pharmacologie ,
6.
Behav Brain Res ; 472: 115178, 2024 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-39098396

RÉSUMÉ

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disorder that seriously affects the quality of the elderly's lives worldwide. The main pathological features of AD are amyloid plaques formed by ß-amyloid (Aß) and neuronal fibrillary tangls (NFTs) formed by hyperphosphorylated Tau protein. The formation process of these pathological features is closely related to inflammatory response, so anti-inflammatory treatment has become a potential treatment for AD. In recent years, more and more research has shown that the anti-inflammatory therapy can relieve the symptoms of AD and improve cognitive function, which provides a valuable research direction for the treatment of AD strategy. Therefore, a comprehensive understanding of the hotspots and development trends of AD anti-inflammatory research is important for promoting the further development of this field and improving the quality of life of patients. METHODS: This study used bibliometric methods, with AD and anti-inflammatory as key words, collected 7638 AD anti-inflammatory studies collected in Web of Science Core Collection (WoSCC) literature database since 2000, and conducted an in-depth analysis of the research hotspots and potential trends in this field. RESULTS: The depth and breadth of AD anti-inflammatory research are in the stage of rapid development, and the hot focus is on exploring the role of inflammation in the pathogenesis of AD, especially the interaction of microglia in the neuroinflammatory mechanism. Secondly, the treatment effect and potential risks of anti-inflammatory drugs such as non-steroidal anti-inflammatory drugs (NSAIDs) on AD are also the focus of research. Therefore, researchers have carried out a series of animal experiments and prospective clinical studies on anti-inflammatory drugs for the treatment of AD, forming a comprehensive research system from basic research to clinical research. As for the future development trend, we believe that the further exploration of inflammation in the pathogenesis of AD will still be one of the key directions, and the application of big data and artificial intelligence technology is expected to provide strong support for the association between inflammation and AD progression. Moreover, the development of novel anti-inflammatory drugs for the inflammatory mechanism of AD will be another major trend for future research. At the same time, personalized treatment strategies and alternative supplements of medicine will also become one of the hotspots of future research. Through the comprehensive use of anti-inflammatory drugs, nutritional supplements, lifestyle intervention and other means, more comprehensive and effective treatment plans for AD patients are expected. CONCLUSION: This research analyzes the overall development trend of AD anti-inflammatory research field since 2000, and provides a comprehensive perspective for the progress of AD anti-inflammatory research. Overall, the field of AD anti-inflammatory research is facing a broad development prospect. In the future, with further research and technological advances, we have resason to expect more effective and safer treatment options for AD patients to help them improve their quality of life and delay disease progression.


Sujet(s)
Maladie d'Alzheimer , Anti-inflammatoires , Inflammation , Maladie d'Alzheimer/traitement médicamenteux , Humains , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Inflammation/traitement médicamenteux , Animaux , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/immunologie
7.
Neurosci Lett ; 837: 137923, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39106918

RÉSUMÉ

Caffeine, a nonselective adenosine receptor antagonist, is the major component of coffee and the most consumed psychostimulant at nontoxic doses in the world. It has been identified that caffeine consumption reduces the risk of several neurological diseases. However, the mechanisms by which it impacts the pathophysiology of neurological diseases remain to be elucidated. In this study, we investigated whether caffeine exerts anti-inflammatory effects on lipopolysaccharide (LPS)-induced inflammation and depression in vivo and explored the potential mechanism of caffeine through LPS-induced brain injury. Adult male Sprague-Dawley (SD) rats were intraperitoneal injected with various concentrations of LPS to induce the neuroinflammation and depressive-like behavior. Then SD rats were treated with caffeine in the presence or absence of LPS. Open-filed and closed-field tests were applied to detect the behaviors of SD rats, while western blot was performed to measure the phosphorylation level of protein kinase B (p-AKT) and nuclear factor κB (NF-κB) in the cortex after caffeine was orally administered. Our findings indicated that caffeine markedly improved the neuroinflammation and depressive-like behavior of LPS-treated SD rats. Mechanistic investigations demonstrated that caffeine down-regulated the expression of p-AKT and NF-κB in LPS-induced SD rats cortex. Taken together, these results indicated that caffeine, a potential agent for preventing inflammatory diseases, may suppress LPS-induced inflammatory and depressive responses by regulating AKT phosphorylation and NF-κB.


Sujet(s)
Caféine , Dépression , Lipopolysaccharides , Facteur de transcription NF-kappa B , Maladies neuro-inflammatoires , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Animaux , Facteur de transcription NF-kappa B/métabolisme , Mâle , Caféine/pharmacologie , Caféine/usage thérapeutique , Protéines proto-oncogènes c-akt/métabolisme , Dépression/traitement médicamenteux , Dépression/induit chimiquement , Dépression/métabolisme , Rats , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/induit chimiquement , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Phosphorylation/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/induit chimiquement
8.
BMC Pharmacol Toxicol ; 25(1): 46, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39123263

RÉSUMÉ

BACKGROUND: Echis ocellatus envenoming is potentially toxic initiating clinical damages on male reproductive system. Kaempferol is a therapeutic agent with neutralizing potentials on snake venom toxins. This study investigated the antagonistic effect of kaempferol on E. ocellatus venom (EoV)-induced reproductive toxicities. METHODS: Fifty adult male rats were sorted at random into five groups of ten rats for this study. The control rats were allotted to group 1, while rats in groups 2-5 were injected with 0.22 mg/kg bw (LD50) of EoV intraperitoneally. Rats in group 2 were not treated while groups 3-5 rats were treated with serum antivenom (0.2 ml), and 4 and 8 mg/kg bw of kaempferol post envenoming, respectively. RESULTS: EoV actuated reproductive toxicity, significantly decreased sperm parameters, and enhanced inflammatory, oxidative stress, and apoptotic biomarkers in reproductive organs of untreated envenomed rats. However, treatment with kaempferol alleviated the venom-induced reproductive disorders with a dose dependent effect. Kaempferol significantly increased the testicular weight, organo-somatic index, sperm parameters, and normalized the levels of serum luteinizing hormone, testosterone, and follicle stimulating hormone. Kaempferol ameliorated testicular and epididymal oxidative stress as evidenced by significant decrease in malondialdehyde (MDA) levels, enhancement of reduced glutathione (GSH) levels, superoxide dismutase (SOD) and glutathione peroxidase (GPX) activities. The inflammatory biomarkers; nitric oxide (NO) levels and myeloperoxidase activity (MPO), and apoptotic biomarkers; caspase 3 and caspase 9 activities were substantially suppressed in the testis and epididymis of envenomed rats treated with kaempferol. CONCLUSION: Results revealed kaempferol as a potential remedial agent against reproductive toxicity that could manifest post-viper envenoming.


Sujet(s)
Apoptose , Kaempférols , Spermatozoïdes , Testicule , Animaux , Mâle , Rats , Apoptose/effets des médicaments et des substances chimiques , Echis , Inflammation/traitement médicamenteux , Inflammation/induit chimiquement , Kaempférols/pharmacologie , Kaempférols/usage thérapeutique , Stress oxydatif/effets des médicaments et des substances chimiques , Rat Wistar , Spermatozoïdes/effets des médicaments et des substances chimiques , Testicule/effets des médicaments et des substances chimiques , Testicule/anatomopathologie , Testicule/métabolisme , Venins de vipère/toxicité
9.
J Am Chem Soc ; 146(32): 22747-22758, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39086108

RÉSUMÉ

Nanomedicine has promising applications in disease treatment, given the remarkable safety concerns (e.g., nanotoxicity and inflammation) of nanomaterials, and realizing the trade-off between the immune response and organ burden of NPs and deeply understanding the interactions of the organism-nano systems are crucial to facilitate the biological applications of NPs. Here, we propose an interpretable causal system optimization (ICSO) framework and construct the upstream and downstream tasks of accurate prediction and intelligent NP optimization. ICSO framework screens the key drivers (recovery duration, specific surface area, and nanomaterial size) and potential causal information for immune responses and organ burden, revealing the hidden priming/constraint effects in bionano interactions. ICSO can be used to quantify the thresholds of biological responses to multiple properties (e.g., the specific surface area, diameter, and zeta potential). ICSO provides quantitative information and constraint conditions for the design of highly biocompatible and targeted organ delivery nanomaterials. For example, negative inflammation is reduced by 36.19%, and positive lung accumulation is promoted by 40.14% by optimizing the specific surface areas and shape and increasing the diameter-to-length ratio. ICSO overcomes the limitations of experience-dependent approaches and provides powerful and automated solutions for decision-makers during nanomaterial design.


Sujet(s)
Nanoparticules , Nanoparticules/composition chimique , Nanomédecine , Humains , Taille de particule , Propriétés de surface , Inflammation/traitement médicamenteux , Animaux
10.
Sci Rep ; 14(1): 18418, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39117695

RÉSUMÉ

Bacterial testicular inflammation is one of the important causes of male infertility. Using plant-derived compounds to overcome the side effects of antibiotics is an alternative treatment strategy for many diseases. Schizandrin B (SchB) is a bioactive compound of herbal medicine Schisandra chinensis which has multiple pharmacological effects. However its effect and the mechanism against testicular inflammation are unknown. Here we tackled these questions using models of lipopolysaccharide (LPS)-induced mice and -Sertoli cells (SCs). Histologically, SchB ameliorated the LPS-induced damages of the seminiferous epithelium and blood-testicular barrier, and reduced the production of pro-inflammatory mediators in mouse testes. Furthermore, SchB decreased the levels of pro-inflammatory mediators and inhibited the nuclear factor kB (NF-κB) and MAPK (especially JNK) signaling pathway phosphorylation in LPS-induced mSCs. The bioinformatics analysis based on receptor prediction and the molecular docking was further conducted. We targeted androgen receptor (AR) and illustrated that AR might bind with SchB in its function. Further experiments indicate that the AR expression was upregulated by LPS stimulation, while SchB treatment reversed this phenomenon; similarly, the expression of the JNK-related proteins and apoptotic-related protein were also reversed after AR activator treatment. Together, SchB mitigates LPS-induced inflammation and apoptosis by inhibiting the AR-JNK pathway.


Sujet(s)
Apoptose , Cyclooctanes , Lignanes , Lipopolysaccharides , Composés polycycliques , Cellules de Sertoli , Animaux , Mâle , Cyclooctanes/pharmacologie , Composés polycycliques/pharmacologie , Composés polycycliques/usage thérapeutique , Lignanes/pharmacologie , Lignanes/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Souris , Cellules de Sertoli/effets des médicaments et des substances chimiques , Cellules de Sertoli/métabolisme , Récepteurs aux androgènes/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Simulation de docking moléculaire , Testicule/effets des médicaments et des substances chimiques , Testicule/métabolisme , Testicule/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme
11.
Exp Biol Med (Maywood) ; 249: 10123, 2024.
Article de Anglais | MEDLINE | ID: mdl-39119118

RÉSUMÉ

Antiretroviral drugs have made significant progress in treating HIV-1 and improving the quality of HIV-1-infected individuals. However, due to their limited permeability into the brain HIV-1 replication persists in brain reservoirs such as perivascular macrophages and microglia, which cause HIV-1-associated neurocognitive disorders. Therefore, it is highly desirable to find a novel therapy that can cross the blood-brain barrier (BBB) and target HIV-1 pathogenesis in brain reservoirs. A recently developed 2-amino-3-methylpentanoic acid [2-morpholin-4-yl-ethyl]-amide (LM11A-31), which is a p75 neutrotrophin receptor (p75NTR) modulator, can cross the BBB. In this study, we examined whether LM11A-31 treatment can suppress HIV-1 replication, oxidative stress, cytotoxicity, and inflammatory response in macrophages. Our results showed that LM11A-31 (100 nM) alone and/or in combination with positive control darunavir (5.5 µM) significantly suppresses viral replication and reduces cytotoxicity. Moreover, the HIV-1 suppression by LM11A-31 was comparable to the HIV-1 suppression by darunavir. Although p75NTR was upregulated in HIV-1-infected macrophages compared to uninfected macrophages, LM11A-31 did not significantly reduce the p75NTR expression in macrophages. Furthermore, our study illustrated that LM11A-31 alone and/or in combination with darunavir significantly suppress pro-inflammatory cytokines including IL-1ß, IL-8, IL-18, and TNF-α and chemokines MCP-1 in HIV-induced macrophages. The suppression of these cytokines and chemokines by LM11A-31 was comparable to darunavir. In contrast, LM11A-31 did not significantly alter oxidative stress, expression of antioxidant enzymes, or autophagy marker proteins in U1 macrophages. The results suggest that LM11A-31, which can cross the BBB, has therapeutic potential in suppressing HIV-1 and inflammatory response in brain reservoirs, especially in macrophages.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Macrophages , Morpholines , Réplication virale , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Humains , Réplication virale/effets des médicaments et des substances chimiques , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/virologie , Morpholines/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Darunavir/pharmacologie , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Récepteurs facteur croissance nerf/métabolisme , Cytokines/métabolisme , Isoleucine/analogues et dérivés , Protéines de tissu nerveux
12.
Cells ; 13(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39120289

RÉSUMÉ

This review discusses the potential of targeting the kynurenine pathway (KP) in the treatment of inflammatory diseases. The KP, responsible for the catabolism of the amino acid tryptophan (TRP), produces metabolites that regulate various physiological processes, including inflammation, cell cycle, and neurotransmission. These metabolites, although necessary to maintain immune balance, may accumulate excessively during inflammation, leading to systemic disorders. Key KP enzymes such as indoleamine 2,3-dioxygenase 1 (IDO1), indoleamine 2,3-dioxygenase 2 (IDO2), tryptophan 2,3-dioxygenase (TDO), and kynurenine 3-monooxygenase (KMO) have been considered promising therapeutic targets. It was highlighted that both inhibition and activation of these enzymes may be beneficial, depending on the specific inflammatory disorder. Several inflammatory conditions, including autoimmune diseases, for which modulation of KP activity holds therapeutic promise, have been described in detail. Preclinical studies suggest that this modulation may be an effective treatment strategy for diseases for which treatment options are currently limited. Taken together, this review highlights the importance of further research on the clinical application of KP enzyme modulation in the development of new therapeutic strategies for inflammatory diseases.


Sujet(s)
Inflammation , Cynurénine , Humains , Cynurénine/métabolisme , Inflammation/traitement médicamenteux , Animaux , Thérapie moléculaire ciblée , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs
14.
J Biomed Mater Res B Appl Biomater ; 112(8): e35458, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39122663

RÉSUMÉ

Bacterial infections already pose a significant threat to skin wounds, especially in diabetic patients who have difficulty healing wounds. However, wound or bacterial infections are known to produce excess reactive oxygen species (ROS), and hypoxia may further hinder wound healing and the development of chronic wounds. In this study, a multifunctional hydrogel for ROS scavenging and bacterial inhibition was developed by cross-linking polyvinyl alcohol (PVA) and sodium alginate (SA) with graphene oxide (GO) loaded with silver-platinum hybrid nanoparticles (GO@Ag-Pt). The PVA/SA hydrogel loaded with GO@Ag-Pt exhibited the ability to scavenge different types of ROS, generate O2, and kill a broad spectrum of bacteria in vitro. The silver-platinum hybrid nanoparticles significantly increased the antibacterial ability against Escherichia coli and Staphylococcus aureus compared with silver nanoparticles (AgNps). GO@Ag-Pt loaded hydrogel was effective in treating infections caused by S.aureus, thereby significantly promoting wound healing during the inflammatory phase. Hydrogel therapy significantly reduced the level of ROS and alleviated inflammation levels. Notably, our ROS-scavenging, antibacterial hydrogels can be used to effectively treat various types of wounds, including difficult-to-heal diabetic wounds with bacterial infections. Thus, this study proposes an effective strategy for various chronic wound healing based on ROS clearance and bacteriostatic hydrogels.


Sujet(s)
Antibactériens , Escherichia coli , Hydrogels , Nanoparticules métalliques , Espèces réactives de l'oxygène , Argent , Staphylococcus aureus , Cicatrisation de plaie , Espèces réactives de l'oxygène/métabolisme , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Hydrogels/pharmacologie , Antibactériens/pharmacologie , Antibactériens/composition chimique , Staphylococcus aureus/effets des médicaments et des substances chimiques , Animaux , Nanoparticules métalliques/composition chimique , Argent/composition chimique , Argent/pharmacologie , Escherichia coli/effets des médicaments et des substances chimiques , Souris , Graphite/composition chimique , Graphite/pharmacologie , Inflammation/traitement médicamenteux , Poly(alcool vinylique)/composition chimique , Poly(alcool vinylique)/pharmacologie , Humains , Alginates/composition chimique , Alginates/pharmacologie , Infection de plaie/traitement médicamenteux , Infections à staphylocoques/traitement médicamenteux , Mâle , Oxygène/composition chimique , Piégeurs de radicaux libres/pharmacologie , Piégeurs de radicaux libres/composition chimique
15.
Nutrients ; 16(15)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39125360

RÉSUMÉ

Withania somnifera, commonly known as Ashwagandha, has been popular for many years. Numerous studies have shown that the extract of this plant, due to its wealth of active substances, can induce anti-inflammatory, neuroprotective, immunomodulatory, hepatoprotective, cardioprotective, anti-diabetic, adaptogenic, anti-arthritic, anti-stress, and antimicrobial effects. This review examines the impact of Ashwagandha extract on the vascular endothelium, inflammation, lipid metabolism, and cardiovascular outcomes. Studies have shown that Ashwagandha extracts exhibit an anti-angiogenic effect by inhibiting vascular endothelial growth factor (VEGF)-induced capillary sprouting and formation by lowering the mean density of microvessels. Furthermore, the results of numerous studies highlight the anti-inflammatory role of Ashwagandha extract, as the action of this plant causes a decrease in the expression of pro-inflammatory cytokines. Interestingly, withanolides, present in Ashwagandha root, have shown the ability to inhibit the differentiation of preadipocytes into adipocytes. Research results have also proved that W. somnifera demonstrates cardioprotective effects due to its antioxidant properties and reduces ischemia/reperfusion-induced apoptosis. It seems that this plant can be successfully used as a potential treatment for several conditions, mainly those with increased inflammation. More research is needed to elucidate the exact mechanisms by which the substances contained in W. somnifera extracts can act in the human body.


Sujet(s)
Anti-inflammatoires , Maladies cardiovasculaires , Endothélium vasculaire , Inflammation , Métabolisme lipidique , Extraits de plantes , Withania , Humains , Extraits de plantes/pharmacologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Anti-inflammatoires/pharmacologie , Withania/composition chimique , Maladies cardiovasculaires/traitement médicamenteux , Animaux , Antioxydants/pharmacologie
16.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39125585

RÉSUMÉ

Acute respiratory distress syndrome (ARDS) occurs as an acute onset condition, and patients present with diffuse alveolar damage, refractory hypoxemia, and non-cardiac pulmonary edema. ARDS progresses through an initial exudative phase, an inflammatory phase, and a final fibrotic phase. Pirfenidone, a powerful anti-fibrotic agent, is known as an agent that inhibits the progression of fibrosis in idiopathic pulmonary fibrosis. In this study, we studied the treatment efficiency of pirfenidone on lipopolysaccharide (LPS) and bleomycin-induced ARDS using rats. The ARDS rat model was created by the intratracheal administration of 3 mg/kg LPS of and 3 mg/kg of bleomycin dissolved in 0.2 mL of normal saline. The pirfenidone treatment group was administered 100 or 200 mg/kg of pirfenidone dissolved in 0.5 mL distilled water orally 10 times every 2 days for 20 days. The administration of LPS and bleomycin intratracheally increased lung injury scores and significantly produced pro-inflammatory cytokines. ARDS induction increased the expressions of transforming growth factor (TGF)-ß1/Smad-2 signaling factors. Additionally, matrix metalloproteinase (MMP)-9/tissue inhibitor of metalloproteinase (TIMP)-1 imbalance occurred, resulting in enhanced fibrosis-related factors. Treatment with pirfenidone strongly suppressed the expressions of TGF-ß1/Smad-2 signaling factors and improved the imbalance of MMP-9/TIMP-1 compared to the untreated group. These effects led to a decrease in fibrosis factors and pro-inflammatory cytokines, promoting the recovery of damaged lung tissue. These results of this study showed that pirfenidone administration suppressed inflammation and fibrosis in the ARDS animal model. Therefore, pirfenidone can be considered a new early treatment for ARDS.


Sujet(s)
Bléomycine , Lipopolysaccharides , Pyridones , , Transduction du signal , Animaux , Pyridones/pharmacologie , Pyridones/usage thérapeutique , /traitement médicamenteux , /métabolisme , /anatomopathologie , /induit chimiquement , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Mâle , Bléomycine/effets indésirables , Inhibiteur tissulaire de métalloprotéinase-1/métabolisme , Protéine Smad2/métabolisme , Rat Sprague-Dawley , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Modèles animaux de maladie humaine , Matrix metalloproteinase 9/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta/métabolisme , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Protéines Smad/métabolisme
17.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125655

RÉSUMÉ

Pancreatic cancer is a very aggressive disease with a dismal prognosis. The tumor microenvironment exerts immunosuppressive activities through the secretion of several cytokines, including interleukin (IL)-1. The IL-1/IL-1 receptor (IL-1R) axis is a key regulator in tumor-promoting T helper (Th)2- and Th17-type inflammation. Th2 cells are differentiated by dendritic cells endowed with Th2-polarizing capability by the thymic stromal lymphopoietin (TSLP) that is secreted by IL-1-activated cancer-associated fibroblasts (CAFs). Th17 cells are differentiated in the presence of IL-1 and other IL-1-regulated cytokines. In pancreatic cancer, the use of a recombinant IL-1R antagonist (IL1RA, anakinra, ANK) in in vitro and in vivo models has shown efficacy in targeting the IL-1/IL-1R pathway. In this study, we have developed sphingomyelin nanosystems (SNs) loaded with ANK (ANK-SNs) to compare their ability to inhibit Th2- and Th17-type inflammation with that of the free drug in vitro. We found that ANK-SNs inhibited TSLP and other pro-tumor cytokines released by CAFs at levels similar to ANK. Importantly, inhibition of IL-17 secretion by Th17 cells, but not of interferon-γ, was significantly higher, and at lower concentrations, with ANK-SNs compared to ANK. Collectively, the use of ANK-SNs might be beneficial in reducing the effective dose of the drug and its toxic effects.


Sujet(s)
Antagoniste du récepteur à l'interleukine-1 , Interleukine-1 , Tumeurs du pancréas , Sphingomyéline , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Humains , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Interleukine-1/métabolisme , Sphingomyéline/métabolisme , Cytokines/métabolisme , Lignée cellulaire tumorale , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Lymphocytes auxiliaires Th2/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th2/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques
18.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39125657

RÉSUMÉ

Well-controlled type 1 diabetes (T1DM) is characterized by inflammation and endothelial dysfunction, thus constituting a suitable model of subclinical cardiovascular disease (CVD). miR-199b-5p overexpression in murine CVD has shown proatherosclerotic effects. We hypothesized that miR-199b-5p would be overexpressed in subclinical CVD yet downregulated following metformin therapy. Inflammatory and vascular markers were measured in 29 individuals with T1DM and 20 matched healthy controls (HCs). miR-199b-5p expression in CFU-Hill's colonies was analyzed from each study group, and correlations with inflammatory/vascular health indices were evaluated. Significant upregulation of miR-199b-5p was observed in T1DM, which was significantly downregulated by metformin. miR-199b-5p correlated positively with vascular endothelial growth factor-D and c-reactive protein (CRP: nonsignificant). ROC analysis determined miR-199b-5p to define subclinical CVD by discriminating between HCs and T1DM individuals. ROC analyses of HbA1c and CRP showed that the upregulation of miR-199b-5p in T1DM individuals defined subclinical CVD at HbA1c > 44.25 mmol and CRP > 4.35 × 106 pg/mL. Ingenuity pathway analysis predicted miR-199b-5p to inhibit the target genes SIRT1, ETS1, and JAG1. Metformin was predicted to downregulate miR-199b-5p via NFATC2 and STAT3 and reverse its downstream effects. This study validated the antiangiogenic properties of miR-199b-5p and substantiated miR-199b-5p overexpression as a biomarker of subclinical CVD. The downregulation of miR-199b-5p by metformin confirmed its cardio-protective effect.


Sujet(s)
Maladies cardiovasculaires , Metformine , microARN , microARN/génétique , microARN/métabolisme , Metformine/pharmacologie , Metformine/usage thérapeutique , Humains , Mâle , Femelle , Maladies cardiovasculaires/traitement médicamenteux , Maladies cardiovasculaires/génétique , Maladies cardiovasculaires/métabolisme , Adulte , Diabète de type 1/génétique , Diabète de type 1/traitement médicamenteux , Diabète de type 1/métabolisme , Inflammation/génétique , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Protéine C-réactive/métabolisme , Protéine C-réactive/génétique , Adulte d'âge moyen , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique , Protéine jagged-1/métabolisme , Protéine jagged-1/génétique , Marqueurs biologiques , Études cas-témoins
19.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39126007

RÉSUMÉ

Diabetic retinopathy (DR) is one of the most prevalent secondary complications associated with diabetes. Specifically, Type 1 Diabetes Mellitus (T1D) has an immune component that may determine the evolution of DR by compromising the immune response of the retina, which is mediated by microglia. In the early stages of DR, the permeabilization of the blood-retinal barrier allows immune cells from the peripheral system to interact with the retinal immune system. The use of new bioactive molecules, such as 3-(2,4-dihydroxyphenyl)phthalide (M9), with powerful anti-inflammatory activity, might represent an advance in the treatment of diseases like DR by targeting the immune systems responsible for its onset and progression. Our research aimed to investigate the molecular mechanisms involved in the interaction of specific cells of the innate immune system during the progression of DR and the reduction in inflammatory processes contributing to the pathology. In vitro studies were conducted exposing Bv.2 microglial and Raw264.7 macrophage cells to proinflammatory stimuli for 24 h, in the presence or absence of M9. Ex vivo and in vivo approaches were performed in BB rats, an animal model for T1D. Retinal explants from BB rats were cultured with M9. Retinas from BB rats treated for 15 days with M9 via intraperitoneal injection were analyzed to determine survival, cellular signaling, and inflammatory markers using qPCR, Western blot, or immunofluorescence approaches. Retinal structure images were acquired via Spectral-Domain-Optical Coherence Tomography (SD-OCT). Our results show that the treatment with M9 significantly reduces inflammatory processes in in vitro, ex vivo, and in vivo models of DR. M9 works by inhibiting the proinflammatory responses during DR progression mainly affecting immune cell responses. It also induces an anti-inflammatory response, primarily mediated by microglial cells, leading to the synthesis of Arginase-1 and Hemeoxygenase-1(HO-1). Ultimately, in vivo administration of M9 preserves the retinal integrity from the degeneration associated with DR progression. Our findings demonstrate a specific interaction between both retinal and systemic immune cells in the progression of DR, with a differential response to treatment, mainly driven by microglia in the anti-inflammatory action. In vivo treatment with M9 induces a switch in immune cell phenotypes and functions that contributes to delaying the DR progression, positioning microglial cells as a new and specific therapeutic target in DR.


Sujet(s)
Diabète de type 1 , Rétinopathie diabétique , Modèles animaux de maladie humaine , Microglie , Animaux , Rétinopathie diabétique/traitement médicamenteux , Rétinopathie diabétique/anatomopathologie , Rétinopathie diabétique/immunologie , Rats , Diabète de type 1/traitement médicamenteux , Diabète de type 1/immunologie , Diabète de type 1/métabolisme , Diabète de type 1/complications , Souris , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Rétine/effets des médicaments et des substances chimiques , Rétine/anatomopathologie , Rétine/métabolisme , Cellules RAW 264.7 , Mâle , Benzofuranes/pharmacologie , Benzofuranes/usage thérapeutique , Immunomodulation/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications , Rats de lignée BB
20.
J Cell Mol Med ; 28(15): e18589, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39135202

RÉSUMÉ

Sepsis causes systemic inflammatory responses and acute lung injury (ALI). Despite modern treatments, sepsis-related ALI mortality remains high. Aqueous extract of Descuraniae Semen (AEDS) exerts anti-endoplasmic reticulum (ER) stress, antioxidant and anti-inflammatory effects. AEDS alleviates inflammation and oedema in ALI. Sodium-potassium-chloride co-transporter isoform 1 (NKCC1) is essential for regulating alveolar fluid and is important in ALI. The NKCC1 activity is regulated by upstream with-no-lysine kinase-4 (WNK4) and STE20/SPS1-related proline/alanine-rich kinase (SPAK). This study aimed to investigate the effects of AEDS on lipopolysaccharide (LPS)-induced ALI model in A549 cells, considering the regulation of ER stress, WNK4-SPAK-NKCC1 cascades, inflammation and apoptosis. Cell viability was investigated by the CCK-8 assay. The expressions of the proteins were assessed by immunoblotting analysis assays. The levels of pro-inflammatory cytokines were determined by ELISA. The expression of cytoplasmic Ca2+ in A549 cells was determined using Fluo-4 AM. AEDS attenuates LPS-induced inflammation, which is associated with increased pro-inflammatory cytokine expression and activation of the WNK4-SPAK-NKCC1 pathway. AEDS inhibits the WNK4-SPAK-NKCC1 pathway by regulating of Bcl-2, IP3R and intracellular Ca2+. WNK4 expression levels are significantly higher in the WNK4-overexpressed transfected A549 cells and significantly decrease after AEDS treatment. AEDS attenuates LPS-induced inflammation by inhibiting the WNK4-SPAK-NKCC1 cascade. Therefore, AEDS is regarded as a potential therapeutic agent for ALI.


Sujet(s)
Stress du réticulum endoplasmique , Inflammation , Lipopolysaccharides , Protein-Serine-Threonine Kinases , Transduction du signal , Membre-2 de la famille-12 des transporteurs de solutés , Humains , Protein-Serine-Threonine Kinases/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Cellules A549 , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , Membre-2 de la famille-12 des transporteurs de solutés/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Extraits de plantes/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Anti-inflammatoires/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE