Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 658
Filtrer
1.
Nutrients ; 16(12)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38931158

RÉSUMÉ

Previous studies have reported that TT genotype carriers of the adenosine A2a receptor (ADORA2A) gene rs5751876 polymorphism have better ergogenic and anti-inflammatory responses to caffeine intake compared to C allele carriers. The aim of the present study was twofold: (1) to investigate the association of the ADORA2A rs5751876 polymorphism with acute caffeine supplementation on hormonal (growth hormone and testosterone) response to resistance exercise (RE); (2) to examine the relationship between the rs5751876 polymorphism and the resting levels of growth hormone and testosterone in athletes who are light caffeine consumers. A double-blind, crossover, placebo-controlled study involving 30 resistance-trained men (age 21.7 ± 4.1) was conducted to assess the impact of caffeine supplementation on serum growth hormone (GH) and testosterone (TS) levels before, immediately after, and 15 min post-RE. One hour before engaging in resistance exercise, subjects were randomly administered 6 mg of caffeine per kg of body mass or a placebo (maltodextrin). After a 7-day washout period, the same protocol was repeated. Resting testosterone and growth hormone levels were examined in the sera of 94 elite athletes (31 females, age 21.4 ± 2.8; 63 males, age 22.9 ± 3.8). Caffeine consumption led to significantly greater increases in GH and TS in men with the TT genotype compared to C allele carriers. Furthermore, in the group of athletes, carriers of the TT genotype had significantly higher testosterone (p = 0.0125) and growth hormone (p = 0.0365) levels compared to C allele carriers. In conclusion, the ADORA2A gene rs5751876 polymorphism may modify the effect of caffeine intake on the hormonal response to exercise.


Sujet(s)
Caféine , Études croisées , Compléments alimentaires , Récepteur A2A à l'adénosine , Entraînement en résistance , Testostérone , Humains , Caféine/administration et posologie , Mâle , Méthode en double aveugle , Récepteur A2A à l'adénosine/génétique , Jeune adulte , Testostérone/sang , Adulte , Femelle , Athlètes , Polymorphisme de nucléotide simple , Génotype , Hormone de croissance humaine/sang , Polymorphisme génétique , Exercice physique
2.
Sci Rep ; 14(1): 12724, 2024 06 03.
Article de Anglais | MEDLINE | ID: mdl-38830861

RÉSUMÉ

Evidence has shown that both sleep loss and daily caffeine intake can induce changes in grey matter (GM). Caffeine is frequently used to combat sleepiness and impaired performance caused by insufficient sleep. It is unclear (1) whether daily use of caffeine could prevent or exacerbate the GM alterations induced by 5-day sleep restriction (i.e. chronic sleep restriction, CSR), and (2) whether the potential impact on GM plasticity depends on individual differences in the availability of adenosine receptors, which are involved in mediating effects of caffeine on sleep and waking function. Thirty-six healthy adults participated in this double-blind, randomized, controlled study (age = 28.9 ± 5.2 y/; F:M = 15:21; habitual level of caffeine intake < 450 mg; 29 homozygous C/C allele carriers of rs5751876 of ADORA2A, an A2A adenosine receptor gene variant). Each participant underwent a 9-day laboratory visit consisting of one adaptation day, 2 baseline days (BL), 5-day sleep restriction (5 h time-in-bed), and a recovery day (REC) after an 8-h sleep opportunity. Nineteen participants received 300 mg caffeine in coffee through the 5 days of CSR (CAFF group), while 17 matched participants received decaffeinated coffee (DECAF group). We examined GM changes on the 2nd BL Day, 5th CSR Day, and REC Day using magnetic resonance imaging and voxel-based morphometry. Moreover, we used positron emission tomography with [18F]-CPFPX to quantify the baseline availability of A1 adenosine receptors (A1R) and its relation to the GM plasticity. The results from the voxel-wise multimodal whole-brain analysis on the Jacobian-modulated T1-weighted images controlled for variances of cerebral blood flow indicated a significant interaction effect between caffeine and CSR in four brain regions: (a) right temporal-occipital region, (b) right dorsomedial prefrontal cortex (DmPFC), (c) left dorsolateral prefrontal cortex (DLPFC), and (d) right thalamus. The post-hoc analyses on the signal intensity of these GM clusters indicated that, compared to BL, GM on the CSR day was increased in the DECAF group in all clusters  but decreased in the thalamus, DmPFC, and DLPFC in the CAFF group. Furthermore, lower baseline subcortical A1R availability predicted a larger GM reduction in the CAFF group after CSR of all brain regions except for the thalamus. In conclusion, our data suggest an adaptive GM upregulation after 5-day CSR, while concomitant use of caffeine instead leads to a GM reduction. The lack of consistent association with individual A1R availability may suggest that CSR and caffeine affect thalamic GM plasticity predominantly by a different mechanism. Future studies on the role of adenosine A2A receptors in CSR-induced GM plasticity are warranted.


Sujet(s)
Caféine , Substance grise , Imagerie par résonance magnétique , Tomographie par émission de positons , Récepteur A1 à l'adénosine , Privation de sommeil , Humains , Caféine/administration et posologie , Caféine/pharmacologie , Mâle , Adulte , Substance grise/imagerie diagnostique , Substance grise/métabolisme , Substance grise/effets des médicaments et des substances chimiques , Substance grise/anatomopathologie , Récepteur A1 à l'adénosine/métabolisme , Récepteur A1 à l'adénosine/génétique , Tomographie par émission de positons/méthodes , Femelle , Imagerie par résonance magnétique/méthodes , Méthode en double aveugle , Privation de sommeil/métabolisme , Privation de sommeil/imagerie diagnostique , Jeune adulte , Récepteur A2A à l'adénosine/métabolisme , Récepteur A2A à l'adénosine/génétique
3.
Int J Mol Sci ; 25(11)2024 Jun 02.
Article de Anglais | MEDLINE | ID: mdl-38892324

RÉSUMÉ

SARS-CoV-2 infection ranges from mild to severe presentations, according to the intensity of the aberrant inflammatory response. Purinergic receptors dually control the inflammatory response: while adenosine A2A receptors (A2ARs) are anti-inflammatory, ATP P2X7 receptors (P2X7Rs) exert pro-inflammatory effects. The aim of this study was to assess if there were differences in allelic and genotypic frequencies of a loss-of-function SNP of ADORA2A (rs2298383) and a gain-of-function single nucleotide polymorphism (SNP) of P2RX7 (rs208294) in the severity of SARS-CoV-2-associated infection. Fifty-five individuals were enrolled and categorized according to the severity of the infection. Endpoint genotyping was performed in blood cells to screen for both SNPs. The TT genotype (vs. CT + CC) and the T allele (vs. C allele) of P2RX7 SNP were found to be associated with more severe forms of COVID-19, whereas the association between ADORA2A SNP and the severity of infection was not significantly different. The T allele of P2RX7 SNP was more frequent in people with more than one comorbidity and with cardiovascular conditions and was associated with colorectal cancer. Our findings suggest a more prominent role of P2X7R rather than of A2AR polymorphisms in SARS-CoV-2 infection, although larger population-based studies should be performed to validate our conclusions.


Sujet(s)
COVID-19 , Polymorphisme de nucléotide simple , Récepteurs purinergiques P2X7 , Humains , Mâle , Adulte d'âge moyen , Sujet âgé , Sujet âgé de 80 ans ou plus , Récepteurs purinergiques P2X7/génétique , Récepteurs purinergiques P2X7/métabolisme , Récepteur A2A à l'adénosine/génétique , Acuité des besoins du patient , COVID-19/complications , COVID-19/génétique , COVID-19/anatomopathologie , Génotype , Fréquence d'allèle , Maladies cardiovasculaires/complications , Maladies cardiovasculaires/génétique , Tumeurs du côlon/complications , Tumeurs du côlon/génétique
4.
Nat Commun ; 15(1): 5129, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879678

RÉSUMÉ

Glucagon, a hormone released from pancreatic α-cells, is critical for maintaining euglycemia and plays a key role in the pathophysiology of diabetes. To stimulate the development of new classes of therapeutic agents targeting glucagon release, key α-cell signaling pathways that regulate glucagon secretion need to be identified. Here, we focused on the potential importance of α-cell Gs signaling on modulating α-cell function. Studies with α-cell-specific mouse models showed that activation of α-cell Gs signaling causes a marked increase in glucagon secretion. We also found that intra-islet adenosine plays an unexpected autocrine/paracrine role in promoting glucagon release via activation of α-cell Gs-coupled A2A adenosine receptors. Studies with α-cell-specific Gαs knockout mice showed that α-cell Gs also plays an essential role in stimulating the activity of the Gcg gene, thus ensuring proper islet glucagon content. Our data suggest that α-cell enriched Gs-coupled receptors represent potential targets for modulating α-cell function for therapeutic purposes.


Sujet(s)
Sous-unités alpha Gs des protéines G , Cellules à glucagon , Glucagon , Souris knockout , Transduction du signal , Glucagon/métabolisme , Animaux , Cellules à glucagon/métabolisme , Souris , Sous-unités alpha Gs des protéines G/métabolisme , Adénosine/métabolisme , Récepteur A2A à l'adénosine/métabolisme , Récepteur A2A à l'adénosine/génétique , Mâle , Souris de lignée C57BL , Ilots pancréatiques/métabolisme
5.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38731962

RÉSUMÉ

ADORA2A (adenosine A2a receptor) and ADORA2B propagate immunoregulatory signals, including restricting both innate and adaptive immunity, though recent data also suggest a tumor suppressor effect in certain settings. We evaluated the RNA expression from 514 tumors in a clinical-grade laboratory; 489 patients with advanced/metastatic disease had clinical outcome correlates. Transcript expression was standardized to internal housekeeping genes and ranked (0-100 scale) relative to 735 specimens from 35 different cancer types. Transcript abundance rank values were defined as "low/moderate" (0-74) or "high" (75-100) percentile RNA expression ranks. Overall, 20.8% of tumors had high ADORA2A (≥75 percentile RNA rank). The greatest proportion of high ADORA2A expressors was found in neuroendocrine and breast cancers and sarcomas, whereas the lowest was found in colorectal and ovarian cancers, albeit with patient-to-patient variability. In multivariable logistic regression analysis, there was a significant positive correlation between high ADORA2A RNA expression and a high expression of the immune checkpoint-related molecules PD-1 (p = 0.015), VISTA (p ≤ 0.001), CD38 (p = 0.031), and CD39 (p ≤ 0.001). In 217 immunotherapy-treated patients, high ADORA2A did not correlate significantly with progression-free (p = 0.51) or overall survival (OS) (p = 0.09) from the initiation of the checkpoint blockade. However, high versus not-high ADORA2A transcript expression correlated with longer OS from the time of advanced/metastatic disease (N = 489 patients; (HR 0.69 (95% CI 0.51-0.95) (p = 0.02)). Therefore, high ADORA2A transcript levels may be a favorable prognostic factor, unrelated to immunotherapy. Importantly, ascertaining co-expression patterns of ADORA2A with PD-1 and VISTA in individual tumors as a basis for the precision co-targeting of ADORA2A and these other checkpoint-related molecules warrants investigation in clinical trials.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Tumeurs , Récepteur A2A à l'adénosine , Transcriptome , Femelle , Humains , Mâle , Marqueurs biologiques tumoraux/génétique , Tumeurs/génétique , Tumeurs/anatomopathologie , Pronostic , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme
6.
Nat Commun ; 15(1): 3661, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38688901

RÉSUMÉ

Optochemistry, an emerging pharmacologic approach in which light is used to selectively activate or deactivate molecules, has the potential to alleviate symptoms, cure diseases, and improve quality of life while preventing uncontrolled drug effects. The development of in-vivo applications for optochemistry to render brain cells photoresponsive without relying on genetic engineering has been progressing slowly. The nucleus accumbens (NAc) is a region for the regulation of slow-wave sleep (SWS) through the integration of motivational stimuli. Adenosine emerges as a promising candidate molecule for activating indirect pathway neurons of the NAc expressing adenosine A2A receptors (A2ARs) to induce SWS. Here, we developed a brain-permeable positive allosteric modulator of A2ARs (A2AR PAM) that can be rapidly photoactivated with visible light (λ > 400 nm) and used it optoallosterically to induce SWS in the NAc of freely behaving male mice by increasing the activity of extracellular adenosine derived from astrocytic and neuronal activity.


Sujet(s)
Adénosine , Noyau accumbens , Récepteur A2A à l'adénosine , Sommeil à ondes lentes , Animaux , Noyau accumbens/métabolisme , Noyau accumbens/effets des médicaments et des substances chimiques , Noyau accumbens/physiologie , Mâle , Récepteur A2A à l'adénosine/métabolisme , Récepteur A2A à l'adénosine/génétique , Souris , Adénosine/métabolisme , Adénosine/pharmacologie , Régulation allostérique , Sommeil à ondes lentes/physiologie , Sommeil à ondes lentes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Lumière , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Humains , Agonistes des récepteurs A2 à l'adénosine/pharmacologie
7.
J Clin Invest ; 134(8)2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38441967

RÉSUMÉ

Antitumor responses of CD8+ T cells are tightly regulated by distinct metabolic fitness. High levels of glutathione (GSH) are observed in the majority of tumors, contributing to cancer progression and treatment resistance in part by preventing glutathione peroxidase 4-dependent (GPX4-dependent) ferroptosis. Here, we show the necessity of adenosine A2A receptor (A2AR) signaling and the GSH/GPX4 axis in orchestrating metabolic fitness and survival of functionally competent CD8+ T cells. Activated CD8+ T cells treated ex vivo with simultaneous inhibition of A2AR and lipid peroxidation acquire a superior capacity to proliferate and persist in vivo, demonstrating a translatable means to prevent ferroptosis in adoptive cell therapy. Additionally, we identify a particular cluster of intratumoral CD8+ T cells expressing a putative gene signature of GSH metabolism (GMGS) in association with clinical response and survival across several human cancers. Our study addresses a key role of GSH/GPX4 and adenosinergic pathways in fine-tuning the metabolic fitness of antitumor CD8+ T cells.


Sujet(s)
Tumeurs , Récepteur A2A à l'adénosine , Humains , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme , Tumeurs/génétique , Tumeurs/thérapie , Tumeurs/métabolisme , Lymphocytes T CD8+/métabolisme , Glutathion/métabolisme
8.
Pharmacol Res ; 203: 107156, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38522762

RÉSUMÉ

Cardiovascular diseases (CVDs) are the leading cause of death worldwide, and morbidity and mortality rates continue to rise. Atherosclerosis constitutes the principal etiology of CVDs. Endothelial injury, inflammation, and dysfunction are the initiating factors of atherosclerosis. Recently, we reported that endothelial adenosine receptor 2 A (ADORA2A), a G protein-coupled receptor (GPCR), plays critical roles in neovascularization disease and cerebrovascular disease. However, the precise role of endothelial ADORA2A in atherosclerosis is still not fully understood. Here, we showed that ADORA2A expression was markedly increased in the aortic endothelium of humans with atherosclerosis or Apoe-/- mice fed a high-cholesterol diet. In vivo studies unraveled that endothelial-specific Adora2a deficiency alleviated endothelial-to-mesenchymal transition (EndMT) and prevented the formation and instability of atherosclerotic plaque in Apoe-/- mice. Moreover, pharmacologic inhibition of ADORA2A with KW6002 recapitulated the anti-atherogenic phenotypes observed in genetically Adora2a-deficient mice. In cultured human aortic endothelial cells (HAECs), siRNA knockdown of ADORA2A or KW6002 inhibition of ADORA2A decreased EndMT, whereas adenoviral overexpression of ADORA2A induced EndMT. Mechanistically, ADORA2A upregulated ALK5 expression via a cAMP/PKA/CREB axis, leading to TGFß-Smad2/3 signaling activation, thereby promoting EndMT. In conclusion, these findings, for the first time, demonstrate that blockade of ADORA2A attenuated atherosclerosis via inhibition of EndMT induced by the CREB1-ALK5 axis. This study discloses a new link between endothelial ADORA2A and EndMT and indicates that inhibiting endothelial ADORA2A could be an effective novel strategy for the prevention and treatment of atherosclerotic CVDs.


Sujet(s)
Athérosclérose , Protéine de liaison à l'élément de réponse à l'AMP cyclique , Transition épithélio-mésenchymateuse , Souris de lignée C57BL , Récepteur A2A à l'adénosine , Récepteur de type I du facteur de croissance transformant bêta , Animaux , Humains , Mâle , Souris , Antagonistes des récepteurs A2 à l'adénosine/pharmacologie , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Athérosclérose/génétique , Cellules cultivées , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/anatomopathologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Souris knockout , Récepteur A2A à l'adénosine/métabolisme , Récepteur A2A à l'adénosine/génétique , Récepteur de type I du facteur de croissance transformant bêta/génétique , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Transduction du signal
9.
Int J Biol Sci ; 20(5): 1855-1870, 2024.
Article de Anglais | MEDLINE | ID: mdl-38481815

RÉSUMÉ

Hepatic fibrosis exacerbates mortality and complications in progressive metabolic dysfunction-associated steatohepatitis (MASH). The role of the adenosine 2A receptor (A2aAR) in hepatic fibrosis within the context of MASH remains uncertain. This study aims to elucidate the involvement of the A2aAR signaling pathway and the efficacy of a novel potent A2aAR antagonist in treating hepatic fibrosis in MASH-induced mice fed a chlorine-deficient, L-amino acid-defined, high fat diet (CDAHFD). A2aAR overexpression in LX-2 cells increased fibrosis markers, whereas the known A2aAR antagonist, ZM241385, decreased these markers. A novel A2aAR antagonist, RAD11, not only attenuated fibrosis progression but also exhibited greater inhibition of the A2aAR signaling pathway compared to ZM241385 in mice with MASH, activated primary hepatocytes, and LX-2 cells. RAD11 exhibited a dual antifibrotic mechanism by targeting both activated HSCs and hepatocytes. Its superior antifibrotic efficacy over ZM241385 in the MASH condition stems from its ability to suppress A2aAR-mediated signaling, inhibit HSC activation, reduce hepatic lipogenesis in hepatocytes, and mitigate lipid accumulation-induced oxidative stress-mediated liver damage. This study has shed light on the relationship between A2aAR signaling and hepatic fibrosis, presenting RAD11 as a potent therapeutic agent for managing MASH and hepatic fibrosis.


Sujet(s)
Stéatose hépatique , Cirrhose du foie , Souris , Animaux , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/métabolisme , Transduction du signal , Modèles animaux de maladie humaine , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme , Souris de lignée C57BL
10.
Sci Rep ; 14(1): 4896, 2024 02 28.
Article de Anglais | MEDLINE | ID: mdl-38418830

RÉSUMÉ

This work prepared and investigated the impact of carboxymethyl chitosan nanoparticles (MC-NPs) on the proliferative capability of keloid fibroblasts (KFBs) while analyzing the mechanistic roles of miR-214 and adenosine A2A receptor (A2AR) in fibroblasts within hypertrophic scars. MC-NPs were synthesized through ion cross-linking, were characterized using transmission electron microscopy (TEM) and laser particle size scattering. The influence of MC-NPs on the proliferation capacity of KFBs was assessed using the MTT method. Changes in the expression levels of miR-214 and A2AR in KFBs, normal skin fibroblasts (NFBs), hypertrophic scar tissue, and normal skin tissue were analyzed. KFBs were categorized into anti-miR-214, anti-miR-NC, miR-214 mimics, miR-NC, si-A2AR, si-con, anti-miR-214+ si-con, and anti-miR-214+ si-A2AR groups. Bioinformatics target prediction was conducted to explore the interaction between miR-214 and A2AR. Real-time quantitative PCR and immunoblotting (WB) were employed to detect the expression levels of miR-214, A2AR, apoptotic protein Bax, and TGF-ß in different cells. Cell counting kit-8 (CCK8) and flow cytometry were employed to assess cell proliferation activity and apoptosis. The results indicated that MC-NPs exhibited spherical particles with an average diameter of 236.47 ± 4.98 nm. The cell OD value in the MC-NPs group was lower than that in KFBs (P < 0.05). The mRNA levels of miR-214 in KFBs and hypertrophic scar tissue were lower than those in NFBs and normal tissue (P < 0.001), while the mRNA and protein levels of A2AR were significantly elevated (P < 0.05). Compared to the control group and anti-miR-NC, the anti-miR-214 group showed significantly increased cell OD values and Bcl-2 protein expression (P < 0.001), decreased levels of apoptotic gene Bax protein, TGF-ß gene mRNA, and protein expression (P < 0.001). Continuous complementary binding sites were identified between miR-214 and A2AR. Compared to the control group, the si-A2AR group exhibited a significant decrease in A2AR gene mRNA and protein expression levels (P < 0.001), reduced cell viability (P < 0.001), increased apoptosis rate (P < 0.001), and a significant elevation in TGF-ß protein expression (P < 0.001). miR-214 targetedly regulated the expression of A2AR, inducing changes in TGF-ß content, promoting the proliferation of keloid fibroblasts, and inhibiting cell apoptosis.


Sujet(s)
Chitosane , Cicatrice hypertrophique , Chéloïde , microARN , Humains , Chéloïde/anatomopathologie , Cicatrice hypertrophique/métabolisme , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme , Antagomirs/métabolisme , Chitosane/pharmacologie , Chitosane/métabolisme , Prolifération cellulaire , Facteur de croissance transformant bêta/métabolisme , Apoptose , microARN/métabolisme , Fibroblastes/métabolisme , ARN messager/métabolisme
11.
Neurosci Lett ; 826: 137610, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38157926

RÉSUMÉ

BACKGROUND: Protein misfolding and inclusion body aggregation caused by α-Syn mutations in the brain often cause neurodegeneration and cognitive impairment, among which the A53T point mutation is more common. Inhibition of adenosine A2A receptor (A2AR) can alleviate the pathological symptoms of brain dysfunction caused by A53T-α-Syn protofibrils, but the mechanism of action is still unclear. AIM: This studies aimed to investigate the potential therapeutic role of the A2AR inhibitor KW6002 in a mouse model of brain synucleinopathy. METHODS: A53T-α-Syn fibre precursor cell nuclear protein was injected into the bilateral prefrontal cortex of mice to establish a synucleinopathy animal model, and the A2AR inhibitor KW6002 (5 mg/kg) was injected intraperitoneally to intervene. RESULT: The intracerebral injection of A53T-α-Syn protofibrils triggers the formation of inclusion bodies in the brain, leading to astrocyte activation, an increased number of apoptotic cells, and suppression of autophagic flux. The administration of KW6002 significantly reversed these phenomena. In vitro experiments revealed that A53T-α-Syn protofibrils inhibited HT-22 autophagy in mouse hippocampal neuronal cells, whereas KW6002 increased cellular autophagic flux, upregulated the expression of LAMP2A and Hsc70 proteins and inhibited the expression of SQSTM1 protein. The present study suggests that KW6002 reduces the level of α-Syn phosphorylation by inhibiting A2AR protein, at the same time, enhances the autophagic flux of neuronal cells, resulting in the degradation of A53T-α-Syn protofibrils and thus reducing the neuronal toxicity and apoptosis induced by A53T-α-Syn protofibrils. CONCLUSION: KW6002 has a significant protective effect on neuronal injury induced by A53T-α-Syn.


Sujet(s)
Lésions encéphaliques , Maladie de Parkinson , Purines , Souris , Animaux , Maladie de Parkinson/génétique , alpha-Synucléine/génétique , alpha-Synucléine/métabolisme , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme , Encéphale/métabolisme , Apoptose , Autophagie
12.
Commun Biol ; 6(1): 1218, 2023 11 30.
Article de Anglais | MEDLINE | ID: mdl-38036689

RÉSUMÉ

Mutations that constitutively activate G protein-coupled receptors (GPCRs), known as constitutively activating mutations (CAMs), modify cell signaling and interfere with drugs, resulting in diseases with limited treatment options. We utilize fluorescence imaging at the single-molecule level to visualize the dynamic process of CAM-mediated activation of the human A2A adenosine receptor (A2AAR) in real time. We observe an active-state population for all CAMs without agonist stimulation. Importantly, activating mutations significantly increase the population of an intermediate state crucial for receptor activation, notably distinct from the addition of a partner G protein. Activation kinetics show that while CAMs increase the frequency of transitions to the intermediate state, mutations altering sodium sensitivity increase transitions away from it. These findings indicate changes in GPCR function caused by mutations may be predicted based on whether they favor or disfavor formation of an intermediate state, providing a framework for designing receptors with altered functions or therapies that target intermediate states.


Sujet(s)
Adénosine , Récepteur A2A à l'adénosine , Humains , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme , Protéines G/métabolisme , Transduction du signal , Mutation
13.
Cell Rep Med ; 4(9): 101188, 2023 09 19.
Article de Anglais | MEDLINE | ID: mdl-37729873

RÉSUMÉ

Inhibition of adenosine A2A receptor (A2AR) is a promising approach for cancer immunotherapy currently evaluated in several clinical trials. We here report that anti-obesogenic and anti-inflammatory functions of A2AR, however, significantly restrain hepatocellular carcinoma (HCC) development. Adora2a deletion in mice triggers obesity, non-alcoholic steatohepatitis (NASH), and systemic inflammation, leading to spontaneous HCC and promoting dimethylbenzyl-anthracene (DMBA)- or diethylnitrosamine (DEN)-induced HCC. Conditional Adora2a deletion reveals critical roles of myeloid and hepatocyte-derived A2AR signaling in restraining HCC by limiting hepatic inflammation and steatosis. Remarkably, the impact of A2AR pharmacological blockade on HCC development is dependent on pre-existing NASH. In support of our animal studies, low ADORA2A gene expression in human HCC is associated with cirrhosis, hepatic inflammation, and poor survival. Together, our study uncovers a previously unappreciated tumor-suppressive function for A2AR in the liver and suggests caution in the use of A2AR antagonists in patients with NASH and NASH-associated HCC.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Stéatose hépatique non alcoolique , Humains , Animaux , Souris , Carcinome hépatocellulaire/génétique , Stéatose hépatique non alcoolique/génétique , Récepteur A2A à l'adénosine/génétique , Tumeurs du foie/génétique , Inflammation
14.
Neuroimage ; 280: 120345, 2023 10 15.
Article de Anglais | MEDLINE | ID: mdl-37625500

RÉSUMÉ

The EEG alpha rhythm (∼ 8-13 Hz) is one of the most salient human brain activity rhythms, modulated by the level of attention and vigilance and related to cerebral energy metabolism. Spectral power in the alpha range in wakefulness and sleep strongly varies among individuals based on genetic predisposition. Knowledge about the underlying genes is scarce, yet small studies indicated that the variant rs5751876 of the gene encoding A2A adenosine receptors (ADORA2A) may contribute to the inter-individual variation. The neuromodulator adenosine is directly linked to energy metabolism as product of adenosine tri-phosphate breakdown and acts as a sleep promoting molecule by activating A1 and A2A adenosine receptors. We performed sleep and positron emission tomography studies in 59 healthy carriers of different rs5751876 alleles, and quantified EEG oscillatory alpha power in wakefulness and sleep, as well as A1 adenosine receptor availability with 18F-CPFPX. Oscillatory alpha power was higher in homozygous C-allele carriers (n = 27, 11 females) compared to heterozygous and homozygous carriers of the T-allele (n(C/T) = 23, n(T/T) = 5, 13 females) (F(18,37) = 2.35, p = 0.014, Wilk's Λ = 0.487). Furthermore, a modulatory effect of ADORA2A genotype on A1 adenosine receptor binding potential was found across all considered brain regions (F(18,40) = 2.62, p = 0.006, Wilk's Λ = 0.459), which remained significant for circumscribed occipital region of calcarine fissures after correction for multiple comparisons. In female participants, a correlation between individual differences in oscillatory alpha power and A1 receptor availability was observed. In conclusion, we confirmed that a genetic variant of ADORA2A affects individual alpha power, while a direct modulatory effect via A1 adenosine receptors in females is suggested.


Sujet(s)
Encéphale , Récepteur A2A à l'adénosine , Femelle , Humains , Adénosine , Encéphale/imagerie diagnostique , Électroencéphalographie , Variation génétique , Récepteur A2A à l'adénosine/génétique , Mâle
15.
J Am Heart Assoc ; 12(14): e028421, 2023 07 18.
Article de Anglais | MEDLINE | ID: mdl-37421280

RÉSUMÉ

Background The identification of large-artery stiffness as a major, independent risk factor for cardiovascular disease-associated morbidity and death has focused attention on identifying therapeutic strategies to combat this disorder. Genetic manipulations that delete or inactivate the translin/trax microRNA-degrading enzyme confer protection against aortic stiffness induced by chronic ingestion of high-salt water (4%NaCl in drinking water for 3 weeks) or associated with aging. Therefore, there is heightened interest in identifying interventions capable of inhibiting translin/trax RNase activity, as these may have therapeutic efficacy in large-artery stiffness. Methods and Results Activation of neuronal adenosine A2A receptors (A2ARs) triggers dissociation of trax from its C-terminus. As A2ARs are expressed by vascular smooth muscle cells (VSMCs), we investigated whether stimulation of A2AR on vascular smooth muscle cells promotes the association of translin with trax and, thereby increases translin/trax complex activity. We found that treatment of A7r5 cells with the A2AR agonist CGS21680 leads to increased association of trax with translin. Furthermore, this treatment decreases levels of pre-microRNA-181b, a target of translin/trax, and those of its downstream product, mature microRNA-181b. To check whether A2AR activation might contribute to high-salt water-induced aortic stiffening, we assessed the impact of daily treatment with the selective A2AR antagonist SCH58261 in this paradigm. We found that this treatment blocked aortic stiffening induced by high-salt water. Further, we confirmed that the age-associated decline in aortic pre-microRNA-181b/microRNA-181b levels observed in mice also occurs in humans. Conclusions These findings suggest that further studies are warranted to evaluate whether blockade of A2ARs may have therapeutic potential in treating large-artery stiffness.


Sujet(s)
microARN , Récepteur A2A à l'adénosine , Humains , Souris , Animaux , Récepteur A2A à l'adénosine/génétique , Protéines de liaison à l'ADN/génétique , Protéines de transport/génétique , microARN/génétique , microARN/métabolisme , Aorte/métabolisme , Adénosine , Eau/métabolisme
16.
Gene ; 876: 147503, 2023 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-37220831

RÉSUMÉ

Attention deficit hyperactivity disorder (ADHD) is a common and heritable neurodevelopmental disorder. Particularly, ADHD is known to be related to the dopaminergic system. ADHD symptoms can appear when the dopamine binding affinity diminishes due to dopamine receptor abnormalities, such as the dopamine D2 receptor (D2R). This receptor interacts with the adenosine A2A receptor (A2AR). The A2AR acts as an antagonist of D2R, that is, the increased binding of adenosine with A2AR inhibits the D2R activity. Furthermore, it is found that the single nucleotide polymorphisms of the adenosine A2A receptor gene (ADORA2A) revealed a significant relationship with ADHD in various populations. Therefore, we examined the genetic relationship between ADORA2A polymorphisms (rs2297838, rs5751876, and rs4822492) and Korean ADHD children. A case-control study was performed for 150 cases and 322 controls. Genotyping of ADORA2A polymorphisms was conducted by PCR-RFLP. The results demonstrated that the rs5751876 TC genotype was associated with children with ADHD (p = 0.018). The rs2298383 CC genotype was significantly associated with children with ADHD/HI (p = 0.026). However, when Bonferroni correction was used, the significance vanished (padjusted = 0.054 and padjusted = 0.078, respectively). Haplotype analysis showed that TTC, TCC, and CTG demonstrated a significant difference between ADHD/C children and control groups (padjusted = 0.006, padjusted = 0.011, and padjusted = 0.028, respectively). In conclusion, we propose a possible association between ADORA2A polymorphisms with Korean children having ADHD.


Sujet(s)
Trouble déficitaire de l'attention avec hyperactivité , Récepteur A2A à l'adénosine , Enfant , Humains , Adénosine , Trouble déficitaire de l'attention avec hyperactivité/génétique , Études cas-témoins , Dopamine/métabolisme , Génotype , Polymorphisme de nucléotide simple , Récepteur A2A à l'adénosine/génétique , République de Corée
17.
Structure ; 31(7): 836-847.e6, 2023 07 06.
Article de Anglais | MEDLINE | ID: mdl-37236187

RÉSUMÉ

Cholesterol is a critical component of mammalian cell membranes and an allosteric modulator of G protein-coupled receptors (GPCRs), but divergent views exist on the mechanisms by which cholesterol influences receptor functions. Leveraging the benefits of lipid nanodiscs, i.e., quantitative control of lipid composition, we observe distinct impacts of cholesterol in the presence and absence of anionic phospholipids on the function-related conformational dynamics of the human A2A adenosine receptor (A2AAR). Direct receptor-cholesterol interactions drive activation of agonist-bound A2AAR in membranes containing zwitterionic phospholipids. Intriguingly, the presence of anionic lipids attenuates cholesterol's impact through direct interactions with the receptor, highlighting a more complex role for cholesterol that depends on membrane phospholipid composition. Targeted amino acid replacements at two frequently predicted cholesterol interaction sites showed distinct impacts of cholesterol at different receptor locations, demonstrating the ability to delineate different roles of cholesterol in modulating receptor signaling and maintaining receptor structural integrity.


Sujet(s)
Phospholipides , Récepteurs couplés aux protéines G , Animaux , Humains , Phospholipides/métabolisme , Membrane cellulaire/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Conformation moléculaire , Cholestérol/métabolisme , Simulation de dynamique moléculaire , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/composition chimique , Mammifères/métabolisme
18.
Nutrients ; 15(7)2023 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-37049474

RÉSUMÉ

Caffeine is an adenosine A2A receptor (ADORA2A) antagonist with ergogenic and anti-inflammatory effects. Previous studies have reported that the ADORA2A gene regulates glutamate metabolism and immune responses, with the ADORA2A rs5751876 TT genotype (with high sensitivity to caffeine) showing larger ergogenic effect following caffeine ingestion. We therefore hypothesized that the TT genotype would be associated with greater anti-inflammatory effects of caffeine in response to exercise, and with higher coffee intake in physically active individuals. The aim of the present study was twofold: (1) to investigate the association of the ADORA2A variant with the anti-inflammatory effects of caffeine in response to intense resistance exercise (RE), and (2) to analyze the association of the rs5751876 with coffee intake in physically active individuals (n = 134). Fifteen resistance-trained athletes participated in a randomized, double-blind, placebo-controlled cross-over study, where they consumed 6 mg/kg of caffeine or placebo one hour prior to performing an RE protocol. Blood samples were taken immediately from the arterial vein before, immediately after, and 15 min after RE for the analysis of inflammatory markers myeloperoxidase (MPO) and acetylcholinesterase (AChE). We found that the ADORA2A TT genotype carriers experienced lower exercise-induced inflammatory responses (p < 0.05 for AchE) when compared to the C allele carriers (i.e., CC/CT) one hour following the ingestion of caffeine. Furthermore, the ADORA2A TT genotype was positively associated with coffee intake (p = 0.0143; irrespective of CYP1A2 rs762551 polymorphism). In conclusion, we found that the ADORA2A gene polymorphism is associated with anti-inflammatory effects of caffeine in response to resistance exercise, as well as with habitual coffee intake in physically active individuals.


Sujet(s)
Caféine , Entraînement en résistance , Humains , Récepteur A2A à l'adénosine/génétique , Café , Études croisées , Acetylcholinesterase , Hétérozygote , Anti-inflammatoires/pharmacologie , Cytochrome P-450 CYP1A2/génétique , Génotype
19.
FASEB J ; 37(4): e22838, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36884388

RÉSUMÉ

Osteoarthritis (OA) pathogenesis is associated with reduced chondrocyte homeostasis and increased levels of cartilage cellular senescence. Chondrosenescence is the development of cartilage senescence that increases with aging joints and disrupts chondrocyte homeostasis and is associated with OA. Adenosine A2A receptor (A2AR) activation in cartilage via intra-articular injection of liposomal A2AR agonist, liposomal-CGS21680, leads to cartilage regeneration in vivo and chondrocyte homeostasis. A2AR knockout mice develop early OA isolated chondrocytes demonstrate upregulated expression of cellular senescence and aging-associated genes. Based on these observations, we hypothesized that A2AR activation would ameliorate cartilage senescence. We found that A2AR stimulation of chondrocytes reduced beta-galactosidase staining and regulated levels and cell localization of common senescence mediators p21 and p16 in vitro in the human TC28a2 chondrocyte cell line. In vivo analysis similarly showed A2AR activation reduced nuclear p21 and p16 in obesity-induced OA mice injected with liposomal-CGS21680 and increased nuclear p21 and p16 in A2AR knockout mouse chondrocytes compared to wild-type mice. A2AR agonism also increased activity of the chondrocyte Sirt1/AMPK energy-sensing pathway by enhancing nuclear Sirt1 localization and upregulating T172-phosphorylated (active) AMPK protein levels. Lastly, A2AR activation in TC28a2 and primary human chondrocytes reduced wild-type p53 and concomitantly increased p53 alternative splicing leading to increase in an anti-senescent p53 variant, Δ133p53α. The results reported here indicate that A2AR signaling promotes chondrocyte homeostasis in vitro and reduces OA cartilage development in vivo by reducing chondrocyte senescence.


Sujet(s)
Cartilage articulaire , Arthrose , Souris , Humains , Animaux , Chondrocytes/métabolisme , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme , Sirtuine-1/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , AMP-Activated Protein Kinases/métabolisme , Vieillissement de la cellule/physiologie , Arthrose/métabolisme , Cartilage articulaire/métabolisme
20.
Exp Neurol ; 364: 114378, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36907351

RÉSUMÉ

The formation of fear memory is crucial in emotional disorders such as PTSD and anxiety. Traumatic brain injury (TBI) can cause emotional disorders with dysregulated fear memory formation; however, their cross-interaction remains unclear and hurdled the treatment against TBI-related emotional disorders. While adenosine A2A receptor(A2AR) contributes to the physiological regulation of fear memory, this study aimed to evaluate the A2AR role and possible mechanisms in post-TBI fear memory formation using a craniocerebral trauma model, genetically modified A2AR mutant mice, and pharmacological A2AR agonist CGS21680 and antagonist ZM241385. Our finding showed (i) TBI enhanced mice freezing levels (fear memory) at seven days post-TBI; (ii) The A2AR agonist CGS21680 enhanced the post-TBI freezing levels; conversely, the A2AR antagonist ZM241385 reduced mice freezing level; further (iii) Genetic knockdown of neuronal A2AR in the hippocampal CA1, CA3, and DG regions reduced post-TBI freezing levels, while A2AR knockout in DG region yielded the most reduction in fear memory; finally, (iv) AAV-CaMKII-Cre virus-mediated DG deletion of A2AR on excitatory neurons led to a significant decreased freezing levels post-TBI. These findings indicate that brain trauma increases fear memory retrieval post-TBI, and A2AR on DG excitatory neurons plays a crucial role in this process. Importantly, inhibition of A2AR attenuates fear memory enhancement, which provides a new strategy to prevent fear memory formation/enhancement after TBI.


Sujet(s)
Lésions traumatiques de l'encéphale , Récepteur A2A à l'adénosine , Souris , Animaux , Récepteur A2A à l'adénosine/génétique , Récepteur A2A à l'adénosine/métabolisme , Lésions traumatiques de l'encéphale/traitement médicamenteux , Hippocampe/métabolisme , Peur , Neurones/métabolisme , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...