Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 92
Filtrer
1.
J Neurochem ; 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38770633

RÉSUMÉ

Here, we describe the characterization of a radioligand selective for GluN2B-containing NMDA receptors, 3-[3H] 1-(azetidin-1-yl)-2-(6-(4-fluoro-3-methyl-phenyl)pyrrolo[3,2-b]pyridin-1-yl)ethanone ([3H]-JNJ- GluN2B-5). In rat cortical membranes, the compound bound to a single site, and the following kinetic parameters were measured; association rate constant Kon = 0.0066 ± 0.0006 min-1 nM-1, dissociation rate constant Koff = 0.0210 ± 0.0001 min-1 indicating calculated KD = Koff/Kon = 3.3 ± 0.4 nM, (mean ± SEM, n = 3). The equilibrium dissociation constant determined from saturation binding experiments in rat cortex was KD of 2.6 ± 0.3 nM (mean ± SEM, n = 3). In contrast to the widely used GluN2B radioligand [3H]-Ro 25-6981, whose affinity Ki for sigma 1 and sigma 2 receptors are 2 and 189 nM, respectively, [3H]-JNJ-GluN2B-5 exhibits no measurable affinity for sigma 1 and sigma 2 receptors (Ki > 10 µM for both) providing distinct selectivity advantages. Anatomical distribution of [3H]-JNJ-GluN2B-5 binding sites in rat, mouse, dog, monkey, and human brain tissue was studied using in vitro autoradiography, which showed high specific binding in the hippocampus and cortex and negligible binding in the cerebellum. Enhanced selectivity for GluN2B-containing receptors translated to a good signal-to-noise ratio in both in vitro radioligand binding and in vitro autoradiography assays. In conclusion, [3H]-JNJ-GluN2B-5 is a high-affinity GluN2B radioligand with excellent signal-to-noise ratio and unprecedented selectivity.

2.
Brain Behav Immun ; 115: 356-373, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37914101

RÉSUMÉ

Chronic stress is linked to increased anxiety. Repeated social defeat (RSD) in mice causes anxiety that is dependent on activated neurons, reactive microglia, and accumulation of monocytes in the brain. This response requires interactions between the immune system and central nervous system (CNS). Neuronal activation within threat appraisal regions is a key response to RSD, however, it is unclear how microglia become activated. One potential explanation is that microglia express a purinergic non-selective ligand gated adenosine-triphosphate (ATP) receptor 7 (P2X7). Activation of P2X7 promotes the release of chemokines and cytokines, and recruitment of monocytes to the brain. Thus, the purpose of this study was to determine if a novel P2X7 antagonist blocked neuronal and microglia interactions and the corresponding anxiety following RSD. Male mice were administered (i.p.) a P2X7 antagonist, JNJ-54471300, prior to each cycle of RSD. Fourteen hours after RSD, behavioral deficits including social avoidance and anxiety-like were determined. Moreover, several immune parameters were assessed. RSD caused neuronal activation in stress-responsive regions, monocyte production and release, splenomegaly, and social avoidance. These parameters were unaffected by P2X7 antagonism. RSD-associated proportional area of Iba-1+ microglia, monocyte accumulation in the brain, IL-1ß mRNA expression in enriched myeloid cells, plasma IL-6, and anxiety-like behavior were ameliorated by P2X7 antagonism. Gene expression analysis in the hippocampus and amygdala showed regional specific responses to RSD and some were reversed with P2X7 antagonism. Overall, blocking P2X7 activation attenuated RSD-induced microglia reactivity with corresponding reduction in neuroinflammation, monocyte accumulation, and anxiety-like behavior in male mice.


Sujet(s)
Microglie , Monocytes , Souris , Mâle , Animaux , Monocytes/métabolisme , Microglie/métabolisme , Défaite sociale , Anxiété , Encéphale/métabolisme , Canaux ioniques/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Adénosine triphosphate
3.
J Pharmacol Exp Ther ; 387(2): 226-234, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37679045

RÉSUMÉ

The type-5 muscarinic acetylcholine receptor (mAChR, M5) is almost exclusively expressed in dopamine (DA) neurons of the ventral tegmental area and substantia nigra pars compacta; therefore, they are ideally located to modulate DA signaling and underlying behaviors. However, the role of M5 in shaping DA release is still poorly characterized. In this study, we first quantitatively mapped the expression of M5 in different neurons of the mouse midbrain, then used voltammetry in mouse striatum to evaluate the effect of M5-selective modulators on DA release. The M5 negative allosteric modulator ML375 significantly decreased electrically evoked DA release and blocked the effect of Oxotremorine-M (Oxo-M; nonselective mAChR agonist) on DA release in the presence of an acetylcholine nicotinic receptor blocker. Conversely, the M5 positive allosteric modulator VU 0365114 significantly increased electrically evoked DA release and the Oxo-M effect on DA release. We then assessed M5's impact on mesolimbic circuit function in vivo. Although psychostimulant-induced locomotor activity models in knockout mice have previously been used to characterize the role of M5 in DA transmission, the results of these studies conflict, leading us to select a different in vivo model, namely a cocaine self-administration paradigm. In contrast to a previous study that also used this model, in the current study, administration of ML375 did not decrease cocaine self-administration in rats (using fixed and progressive ratio). These conflicting results illustrate the complexity of M5 modulation and the need to further characterize its involvement in the regulation of dopamine signaling, central to multiple neuropsychiatric diseases. SIGNIFICANCE STATEMENT: This work describes the type-5 muscarinic receptor (M5) pattern of expression within the midbrain as well as its physiological modulation by selective compounds at the axon terminal level in the striatum, where M5 directly shapes dopamine transmission. It offers the first direct readout of mesolimbic dopamine release modulation by M5, highlighting its role in regulating neurocircuits implicated in the pathophysiology of neuropsychiatric disorders such as substance use disorders, major depressive disorder, and schizophrenia.

4.
Br J Pharmacol ; 180(13): 1710-1729, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-36637008

RÉSUMÉ

BACKGROUND AND PURPOSE: Neonatal seizures represent a clinical emergency. However, current anti-seizure medications fail to resolve seizures in ~50% of infants. The P2X7 receptor (P2X7R) is an important driver of inflammation, and evidence suggests that P2X7R contributes to seizures and epilepsy in adults. However, no genetic proof has yet been provided to determine what contribution P2X7R makes to neonatal seizures, its effects on inflammatory signalling during neonatal seizures, and the therapeutic potential of P2X7R-based treatments on long-lasting brain excitability. EXPERIMENTAL APPROACH: Neonatal seizures were induced by global hypoxia in 7-day-old mouse pups (P7). The role of P2X7Rs during seizures was analysed in P2X7R-overexpressing and knockout mice. Treatment of wild-type mice after hypoxia with the P2X7R antagonist JNJ-47965567 was used to determine the effects of the P2X7R on long-lasting brain hyperexcitability. Cell type-specific P2X7R expression was analysed in P2X7R-EGFP reporter mice. RNA sequencing was used to monitor P2X7R-dependent hippocampal downstream signalling. KEY RESULTS: P2X7R deletion reduced seizure severity, whereas P2X7R overexpression exacerbated seizure severity and reduced responsiveness to anti-seizure medication. P2X7R deficiency led to an anti-inflammatory phenotype in microglia, and treatment of mice with a P2X7R antagonist reduced long-lasting brain hyperexcitability. RNA sequencing identified several pathways altered in P2X7R knockout mice after neonatal hypoxia, including a down-regulation of genes implicated in inflammation and glutamatergic signalling. CONCLUSION AND IMPLICATIONS: Treatments based on targeting the P2X7R may represent a novel therapeutic strategy for neonatal seizures with P2X7Rs contributing to the generation of neonatal seizures, driving inflammatory processes and long-term hyperexcitability states.


Sujet(s)
Récepteurs purinergiques P2X7 , Crises épileptiques , Animaux , Souris , Animaux nouveau-nés , Encéphale/métabolisme , Épilepsie/induit chimiquement , Épilepsie/métabolisme , Hypoxie/complications , Inflammation/traitement médicamenteux , Souris knockout , Récepteurs purinergiques P2X7/génétique , Récepteurs purinergiques P2X7/métabolisme , Crises épileptiques/métabolisme
5.
Epilepsia ; 64(2): 511-523, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36507708

RÉSUMÉ

OBJECTIVE: The P2X7 receptor (P2X7R) is an important contributor to neuroinflammation, responding to extracellularly released adenosine triphosphate. Expression of the P2X7R is increased in the brain in experimental and human epilepsy, and genetic or pharmacologic targeting of the receptor can reduce seizure frequency and severity in preclinical models. Experimentally induced seizures also increase levels of the P2X7R in blood. Here, we tested 18 F-JNJ-64413739, a positron emission tomography (PET) P2X7R antagonist, as a potential noninvasive biomarker of seizure-damage and epileptogenesis. METHODS: Status epilepticus was induced via an intra-amygdala microinjection of kainic acid. Static PET studies (30 min duration, initiated 30 min after tracer administration) were conducted 48 h after status epilepticus via an intravenous injection of 18 F-JNJ-64413739. PET images were coregistered with a brain magnetic resonance imaging atlas, tracer uptake was determined in the different brain regions and peripheral organs, and values were correlated to seizure severity during status epilepticus. 18 F-JNJ-64413739 was also applied to ex vivo human brain slices obtained following surgical resection for intractable temporal lobe epilepsy. RESULTS: P2X7R radiotracer uptake correlated strongly with seizure severity during status epilepticus in brain structures including the cerebellum and ipsi- and contralateral cortex, hippocampus, striatum, and thalamus. In addition, a correlation between radiotracer uptake and seizure severity was also evident in peripheral organs such as the heart and the liver. Finally, P2X7R radiotracer uptake was found elevated in brain sections from patients with temporal lobe epilepsy when compared to control. SIGNIFICANCE: Taken together, our data suggest that P2X7R-based PET imaging may help to identify seizure-induced neuropathology and temporal lobe epilepsy patients with increased P2X7R levels possibly benefitting from P2X7R-based treatments.


Sujet(s)
Épilepsie temporale , État de mal épileptique , Souris , Humains , Mâle , Animaux , Épilepsie temporale/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Récepteurs purinergiques P2X7/usage thérapeutique , Encéphale/imagerie diagnostique , Encéphale/métabolisme , État de mal épileptique/induit chimiquement , État de mal épileptique/imagerie diagnostique , État de mal épileptique/métabolisme , Crises épileptiques/traitement médicamenteux
6.
Front Pharmacol ; 14: 1308478, 2023.
Article de Anglais | MEDLINE | ID: mdl-38259288

RÉSUMÉ

There remains a need for new drug targets for treatment-resistant temporal lobe epilepsy. The ATP-gated P2X7 receptor coordinates neuroinflammatory responses to tissue injury. Previous studies in mice reported that the P2X7 receptor antagonist JNJ-47965567 suppressed spontaneous seizures in the intraamygdala kainic acid model of epilepsy and reduced attendant gliosis in the hippocampus. The drug-resistance profile of this model is not fully characterised, however, and newer P2X7 receptor antagonists with superior pharmacokinetic profiles have recently entered clinical trials. Using telemetry-based continuous EEG recordings in mice, we demonstrate that spontaneous recurrent seizures in the intraamygdala kainic acid model are refractory to the common anti-seizure medicine levetiracetam. In contrast, once-daily dosing of JNJ-54175446 (30 mg/kg, intraperitoneal) resulted in a significant reduction in spontaneous recurrent seizures which lasted several days after the end of drug administration. Using a combination of immunohistochemistry and ex vivo radiotracer assay, we find that JNJ-54175446-treated mice at the end of recordings display a reduction in astrogliosis and altered microglia process morphology within the ipsilateral CA3 subfield of the hippocampus, but no difference in P2X7 receptor surface expression. The present study extends the characterisation of the drug-resistance profile of the intraamygdala kainic acid model in mice and provides further evidence that targeting the P2X7 receptor may have therapeutic applications in the treatment of temporal lobe epilepsy.

7.
Sci Rep ; 11(1): 10447, 2021 05 17.
Article de Anglais | MEDLINE | ID: mdl-34001933

RÉSUMÉ

Microglia, CNS resident innate immune cells, respond strongly to activation of TLR3 and TLR4, which recognize viral dsRNA poly(I:C) and bacterial endotoxin LPS, respectively. However, few studies have thoroughly and parallelly compared functional phenotypes and downstream mechanisms between LPS- and poly(I:C)-exposed primary microglia. Here, we investigated the responses of mouse primary microglia upon LPS and poly(I:C) stimulation by detecting various phenotypes ranging from morphology, proliferation, secretion, chemotaxis, to phagocytosis. Furthermore, we explored their sequential gene expression and the downstream signal cascades. Interestingly, we found that the microglial activation pattern induced by LPS was distinguished from that induced by poly(I:C). Regarding microglial morphology, LPS caused an ameboid-like shape while poly(I:C) induced a bushy shape. Microglial proliferation was also facilitated by LPS but not by poly(I:C). In addition, LPS and poly(I:C) modulated microglial chemotaxis and phagocytosis differently. Furthermore, genome-wide analysis provided gene-level support to these functional differences, which may be associated with NF-κb and type I interferon pathways. Last, LPS- and poly(I:C)-activated microglia mediated neurotoxicity in a co-culture system. This study extends our understanding of TLR roles in microglia and provides insights into selecting proper inflammatory microglial models, which may facilitate identification of new targets for therapeutic application.


Sujet(s)
Lipopolysaccharides/pharmacologie , Microglie/effets des médicaments et des substances chimiques , Poly I-C/pharmacologie , Animaux , Cellules cultivées , Chimiotaxie/effets des médicaments et des substances chimiques , Chimiotaxie/immunologie , Techniques de coculture , Femelle , Interféron de type I/métabolisme , Souris , Microglie/immunologie , Facteur de transcription NF-kappa B/métabolisme , Neurones , Phagocytose/effets des médicaments et des substances chimiques , Phagocytose/immunologie , Culture de cellules primaires , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie , Récepteur de type Toll-3/agonistes , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-4/agonistes , Récepteur de type Toll-4/métabolisme
8.
PLoS One ; 16(4): e0243683, 2021.
Article de Anglais | MEDLINE | ID: mdl-33909614

RÉSUMÉ

Identification of genomic mutations by molecular testing plays an important role in diagnosis, prognosis, and treatment of myeloid neoplasms. Next-generation sequencing (NGS) is an efficient method for simultaneous detection of clinically significant genomic mutations with high sensitivity. Various NGS based in-house developed and commercial myeloid neoplasm panels have been integrated into routine clinical practice. However, some genes frequently mutated in myeloid malignancies are particularly difficult to sequence with NGS panels (e.g., CEBPA, CARL, and FLT3). We report development and validation of a 48-gene NGS panel that includes genes that are technically challenging for molecular profiling of myeloid neoplasms including acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN). Target regions were captured by hybridization with complementary biotinylated DNA baits, and NGS was performed on an Illumina NextSeq500 instrument. A bioinformatics pipeline that was developed in-house was used to detect single nucleotide variations (SNVs), insertions/deletions (indels), and FLT3 internal tandem duplications (FLT3-ITD). An analytical validation study was performed on 184 unique specimens for variants with allele frequencies ≥5%. Variants identified by the 48-gene panel were compared to those identified by a 35-gene hematologic neoplasms panel using an additional 137 unique specimens. The developed assay was applied to a large cohort (n = 2,053) of patients with suspected myeloid neoplasms. Analytical validation yielded 99.6% sensitivity (95% CI: 98.9-99.9%) and 100% specificity (95% CI: 100%). Concordance of variants detected by the 2 tested panels was 100%. Among patients with suspected myeloid neoplasms (n = 2,053), 54.5% patients harbored at least one clinically significant mutation: 77% in AML patients, 48% in MDS, and 45% in MPN. Together, these findings demonstrate that the assay can identify mutations associated with diagnosis, prognosis, and treatment options of myeloid neoplasms even in technically challenging genes.


Sujet(s)
Tumeurs hématologiques , Séquençage nucléotidique à haut débit , Leucémie aigüe myéloïde , Mutation , Syndromes myélodysplasiques , Protéines tumorales , Tumeurs hématologiques/génétique , Tumeurs hématologiques/métabolisme , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/métabolisme , Protéines tumorales/génétique , Protéines tumorales/métabolisme
9.
Dis Model Mech ; 13(10)2020 10 30.
Article de Anglais | MEDLINE | ID: mdl-33174532

RÉSUMÉ

Neuroinflammation is one of the main physiopathological mechanisms of amyotrophic lateral sclerosis (ALS), produced by the chronic activation of microglia in the CNS. This process is triggered by the persistent activation of the ATP-gated P2X7 receptor (P2RX7, hereafter referred to as P2X7R). The present study aimed to evaluate the effects of the chronic treatment with the P2X7R antagonist JNJ-47965567 in the development and progression of ALS in the SOD1G93A murine model. SOD1G93A mice were intraperitoneally (i.p.) injected with either 30 mg/kg of JNJ-47965567 or vehicle 4 times per week, from pre-onset age (here, postnatal day 60; P60) until study endpoint. Body weight, motor coordination, phenotypic score, disease onset and survival were measured throughout the study, and compared between vehicle- and drug-injected groups. Treatment with the P2X7R antagonist JNJ-47965567 delayed disease onset, reduced body weight loss and improved motor coordination and phenotypic score in female SOD1G93A mice, although it did not increase lifespan. Interestingly, neither beneficial nor detrimental effects were observed in males in any of the analyzed parameters. Treatment did not affect motor neuron survival or ChAT, Iba-1 and P2X7R protein expression in endpoint individuals of mixed sexes. Overall, chronic administration of JNJ-47965567 for 4 times per week to SOD1G93A mice from pre-onset stage altered disease progression in female individuals while it did not have any effect in males. Our results suggest a partial, yet important, effect of P2X7R in the development and progression of ALS.


Sujet(s)
Sclérose latérale amyotrophique/traitement médicamenteux , Sclérose latérale amyotrophique/physiopathologie , Évolution de la maladie , Activité motrice , Nicotinamide/analogues et dérivés , Pipérazines/administration et posologie , Pipérazines/usage thérapeutique , Antagonistes des récepteurs purinergiques P2X/administration et posologie , Antagonistes des récepteurs purinergiques P2X/usage thérapeutique , Récepteurs purinergiques P2X7/métabolisme , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Détermination du point final , Femelle , Humains , Souris de lignée C57BL , Souris transgéniques , Activité motrice/effets des médicaments et des substances chimiques , Motoneurones/effets des médicaments et des substances chimiques , Motoneurones/anatomopathologie , Nicotinamide/administration et posologie , Nicotinamide/composition chimique , Nicotinamide/pharmacologie , Nicotinamide/usage thérapeutique , Pipérazines/composition chimique , Pipérazines/pharmacologie , Antagonistes des récepteurs purinergiques P2X/composition chimique , Antagonistes des récepteurs purinergiques P2X/pharmacologie , Test du rotarod , Analyse de survie , Perte de poids/effets des médicaments et des substances chimiques
10.
J Clin Invest ; 130(11): 6158-6170, 2020 11 02.
Article de Anglais | MEDLINE | ID: mdl-33074244

RÉSUMÉ

The α6ß4 nicotinic acetylcholine receptor (nAChR) is enriched in dorsal root ganglia neurons and is an attractive non-opioid therapeutic target for pain. However, difficulty expressing human α6ß4 receptors in recombinant systems has precluded drug discovery. Here, genome-wide screening identified accessory proteins that enable reconstitution of human α6ß4 nAChRs. BARP, an auxiliary subunit of voltage-dependent calcium channels, promoted α6ß4 surface expression while IRE1α, an unfolded protein response sensor, enhanced α6ß4 receptor assembly. Effects on α6ß4 involve BARP's N-terminal region and IRE1α's splicing of XBP1 mRNA. Furthermore, clinical efficacy of nicotinic agents in relieving neuropathic pain best correlated with their activity on α6ß4. Finally, BARP-knockout, but not NACHO-knockout mice lacked nicotine-induced antiallodynia, highlighting the functional importance of α6ß4 in pain. These results identify roles for IRE1α and BARP in neurotransmitter receptor assembly and unlock drug discovery for the previously elusive α6ß4 receptor.


Sujet(s)
Agonistes cholinergiques/pharmacologie , Endoribonucleases/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Récepteurs cholinergiques/biosynthèse , Animaux , Endoribonucleases/génétique , Cellules HEK293 , Humains , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Souris , Souris knockout , Protein-Serine-Threonine Kinases/génétique , Épissage des ARN/effets des médicaments et des substances chimiques , Rats , Récepteurs cholinergiques/génétique , Protéine-1 liant la boite X/génétique
11.
Adv Pharmacol ; 89: 357-386, 2020.
Article de Anglais | MEDLINE | ID: mdl-32616213

RÉSUMÉ

Scopolamine is a nonselective muscarinic antagonist that has shown relatively rapid antidepressant effects, although to date the results are from limited clinical studies. Scopolamine reportedly has downstream signaling effects thought to be linked to neuroplasticity within glutamatergic synapses and consequent antidepressant action. In psychiatry, clinically validated pathways are unusual and thus merit further research in an effort develop more effect medicines for patients with mood disorders. Thus, we are faced with a unique opportunity to build on the clinical observation associated with scopolamine through reverse translation to identify of targets that retain the clinical efficacy while reducing the side effect profile. This chapter reviews the clinical antidepressant findings with scopolamine, including discussion of differential response across patient subgroups, as well as a review of biomarkers that predict clinical outcome. The preclinical data associated with scopolamine also are reviewed and convey a vision for narrowing in on the therapeutic muscarinic receptor subtype(s) that support the antidepressant effects to guide the development of next generation antimuscarinic drug targets for depression.


Sujet(s)
Antidépresseurs/usage thérapeutique , Antagonistes muscariniques/usage thérapeutique , Scopolamine/usage thérapeutique , Animaux , Choline/antagonistes et inhibiteurs , Dépression/traitement médicamenteux , Humains , Scopolamine/effets indésirables , Résultat thérapeutique
12.
Prog Med Chem ; 59: 63-99, 2020.
Article de Anglais | MEDLINE | ID: mdl-32362329

RÉSUMÉ

P2X7 has continued to be a target of immense interest since it is implicated in several peripheral and central nervous system disorders that result from inflammation. This review primarily describes new P2X7 receptor antagonists that have been investigated and disclosed in patent applications or primary literature since 2015. While a crystal structure of the receptor to aid in the design of novel chemical structures remains elusive, many of the chemotypes that have been disclosed contain similarities, with an amide motif present in all series that have been explored to date. Several of the recent antagonists described are brain penetrant, and two compounds are currently in clinical trials for CNS indications. Additionally, brain penetrant PET ligands have been developed that aid in measuring target engagement and these ligands can potentially be used as biomarkers.


Sujet(s)
Troubles de l'humeur/traitement médicamenteux , Stéatose hépatique non alcoolique/traitement médicamenteux , Antagonistes des récepteurs purinergiques P2X/pharmacologie , Récepteurs purinergiques P2X7/métabolisme , Humains , Ligands , Structure moléculaire , Troubles de l'humeur/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Antagonistes des récepteurs purinergiques P2X/composition chimique
13.
Epigenetics ; 15(1-2): 183-198, 2020.
Article de Anglais | MEDLINE | ID: mdl-31282290

RÉSUMÉ

DNA methylation (DNAm) and microRNAs (miRNAs) have been implicated in a wide-range of human diseases. While often studied in isolation, DNAm and miRNAs are not independent. We analyzed associations of expression of 283 miRNAs with DNAm at >400K CpG sites in whole blood obtained from 3565 individuals and identified 227 CpGs at which differential methylation was associated with the expression of 40 nearby miRNAs (cis-miR-eQTMs) at FDR<0.01, including 91 independent CpG sites at r2 < 0.2. cis-miR-eQTMs were enriched for CpGs in promoter and polycomb-repressed state regions, and 60% were inversely associated with miRNA expression. Bidirectional Mendelian randomization (MR) analysis further identified 58 cis-miR-eQTMCpG-miRNA pairs where DNAm changes appeared to drive miRNA expression changes and opposite directional effects were unlikely. Integration of genetic variants in joint analyses revealed an average partial between cis-miR-eQTM CpGs and miRNAs of 2% after conditioning on site-specific genetic variation, suggesting that DNAm is an important epigenetic regulator of miRNA expression. Finally, two-step MR analysis was performed to identify putatively causal CpGs driving miRNA expression in relation to human complex traits. We found that an imprinted region on 14q32 that was previously identified in relation to age at menarche is enriched with cis-miR-eQTMs. Nine CpGs and three miRNAs at this locus tested causal for age at menarche, reflecting novel epigenetic-driven molecular pathways underlying this complex trait. Our study sheds light on the joint genetic and epigenetic regulation of miRNA expression and provides insights into the relations of miRNAs to their targets and to complex phenotypes.


Sujet(s)
Méthylation de l'ADN , Épigénome , microARN/génétique , Hérédité multifactorielle , Chromosomes humains de la paire 14/génétique , Ilots CpG , Épigénomique/méthodes , Étude d'association pangénomique/méthodes , Empreinte génomique , Humains , Ménarche/génétique , Analyse de randomisation mendélienne/méthodes , microARN/métabolisme , Locus de caractère quantitatif , Transcriptome
15.
Front Pharmacol ; 10: 840, 2019.
Article de Anglais | MEDLINE | ID: mdl-31507408

RÉSUMÉ

Alzheimer's disease (AD) is a large and increasing unmet medical need with no disease-modifying treatment currently available. Genetic evidence from genome-wide association studies (GWASs) and gene network analysis has clearly revealed a key role of the innate immune system in the brain, of which microglia are the most important element. Single-nucleotide polymorphisms (SNPs) in genes predominantly expressed in microglia have been associated with altered risk of developing AD. Furthermore, microglia-specific pathways are affected on the messenger RNA (mRNA) expression level in post-mortem AD tissue and in mouse models of AD. Together these findings have increased the interest in microglia biology, and numerous scientific reports have proposed microglial molecules and pathways as drug targets for AD. Target identification and validation are generally the first steps in drug discovery. Both target validation and drug lead identification for central nervous system (CNS) targets and diseases entail additional significant obstacles compared to peripheral targets and diseases. This makes CNS drug discovery, even with well-validated targets, challenging. In this article, we will illustrate the special challenges of AD drug discovery by discussing the viability/practicality of possible microglia drug targets including cluster of differentiation 33 (CD33), KCa3.1, kynurenines, ionotropic P2 receptor 7 (P2X7), programmed death-1 (PD-1), Toll-like receptors (TLRs), and triggering receptor expressed in myeloid cells 2 (TREM2).

16.
Methods Mol Biol ; 1970: 101-120, 2019.
Article de Anglais | MEDLINE | ID: mdl-30963491

RÉSUMÉ

MicroRNAs are small noncoding RNA molecules with great importance in regulating a large number of diverse biological processes in health and disease. MicroRNAs can bind to both coding and noncoding RNAs and regulate their stability and expression. Genetic variants and somatic mutations may alter microRNA sequences and their target sites and therefore impact microRNA-target recognition. Aberrant microRNA-target interactions have been associated with many diseases. In recent years, computational resources have been developed for retrieving, annotating, and analyzing the impact of mutations on microRNA-target recognition. In this chapter, we provide an overview on the computational analysis of mutations impacting microRNA target recognition, followed by a detailed tutorial on how to use three major Web-based bioinformatics resources: PolymiRTS ( http://compbio.uthsc.edu/miRSNP ), a database of genetic variants impacting microRNA target recognition; SomamiR ( http://compbio.uthsc.edu/SomamiR ), a database of somatic mutations affecting the interactions between microRNAs and their targets in mRNAs and noncoding RNAs; and miR2GO ( http://compbio.uthsc.edu/miR2GO ), a computational tool for knowledge-based functional analysis of genetic variants and somatic mutations in microRNA seed regions.


Sujet(s)
Marqueurs biologiques/analyse , Biologie informatique/méthodes , Maladie/génétique , microARN/génétique , Mutation , Polymorphisme de nucléotide simple , ARN messager/génétique , Logiciel , Régulation de l'expression des gènes , Humains , microARN/métabolisme , ARN messager/métabolisme
17.
Eur J Pharmacol ; 853: 299-307, 2019 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-30965058

RÉSUMÉ

Transient receptor potential melastatin type 2 (TRPM2) is a cation channel activated by free intracellular ADP-ribose and reactive oxygen species. TRPM2 signaling has been linked to the pathophysiology of CNS disorders such as neuropathic pain, bipolar disorder and Alzheimer's disease. In this manuscript, we describe the discovery of JNJ-28583113, a potent brain penetrant TRPM2 antagonist. Ca2+ flux assays in cells overexpressing TRPM2 and electrophysiological recordings were used to test the pharmacology of JNJ-28583113. JNJ-28583113 was assayed in vitro on GSK-3 phosphorylation levels, cell death, cytokine release in microglia and unbiased morphological phenotypic analysis. Finally, we dosed animals to evaluate its pharmacokinetic properties. Our results showed that JNJ-28583113 is a potent (126 ±â€¯0.5 nM) TRPM2 antagonist. Blocking TRPM2 caused phosphorylation of GSK3α and ß subunits. JNJ-28583113 also protected cells from oxidative stress induced cell death as well as morphological changes induced by non-cytotoxic concentrations of H2O2. In addition, inhibiting TRPM2 blunted cytokine release in response to pro-inflammatory stimuli in microglia. Lastly, we showed that JNJ-28583113 was brain penetrant but not suitable for systemic dosing as it was rapidly metabolized in vivo. While the in-vitro pharmacology of JNJ-28583113 is the best in class, its in-vivo properties would need optimization to assist in further probing key roles of TRPM2 in CNS pathophysiology.


Sujet(s)
Découverte de médicament , Pyrazoles/pharmacologie , Canaux cationiques TRPM/antagonistes et inhibiteurs , Animaux , Cellules HEK293 , Cellules HeLa , Humains , Mâle , Souris , Rats
18.
J Nucl Med ; 60(8): 1154-1159, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-30733317

RÉSUMÉ

The P2X7 receptor is an adenosine triphosphate-gated ion channel, which is abundantly expressed in glial cells within the central nervous system and in the periphery. P2X7 receptor activation leads to the release of the proinflammatory cytokine IL-1ß in the brain, and antagonism of the P2X7 receptor is a novel therapeutic strategy to dampen adenosine triphosphate-dependent IL-1ß signaling. PET ligands for the P2X7 receptor will not only be valuable to assess central target engagement of drug candidates but also hold promise as surrogate markers of central neuroinflammation. Herein we describe the in vitro and in vivo evaluation of 18F-JNJ-64413739, an 18F-labeled PET ligand for imaging the P2X7 receptor in the brain. Methods: P2X7 receptor affinity and specificity, pharmacokinetics, metabolic stability, blood-brain barrier permeability, and off-target binding of JNJ-64413739 were evaluated in a series of in vitro, ex vivo, and in vivo assays. 18F-JNJ-64413739 was radiolabeled via a one-step nucleophilic aromatic substitution. The tracer was also studied in rhesus macaques, and PET images were analyzed with an arterial plasma input function-based Logan graphical analysis. Results: The potency (half-maximal inhibitory concentration) of the P2X7 receptor antagonist JNJ-64413739 is 1.0 ± 0.2 nM and 2.0 ± 0.6 nM at the recombinant human and rat P2X7 receptor, respectively, and the binding affinity is 2.7 nM (rat cortex binding assay) and 15.9 nM (human P2X7 receptor). In nonhuman primate PET imaging studies, dose-dependent receptor occupancy of JNJ-54175446 was observed in 2 rhesus monkeys. At a 0.1 mg/kg dose (intravenous) of JNJ-54175446, the receptor occupancy was calculated to be 17% by Logan graphical analysis, whereas a dose of 2.5 mg/kg yielded a receptor occupancy of 60%. Conclusion: The preclinical evaluation of 18F-JNJ-64413739 demonstrates that the tracer engages the P2X7 receptor. Reproducible and dose-dependent receptor occupancy studies with the P2X7 receptor antagonist JNJ-54175446 were obtained in rhesus monkeys. This novel PET tracer exhibits in vitro and in vivo characteristics suitable for imaging the P2X7 receptor in the brain and warrants further studies in humans.


Sujet(s)
Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Pyrimidines/pharmacologie , Radiopharmaceutiques/pharmacologie , Récepteurs purinergiques P2X7/métabolisme , Animaux , Encéphale/effets des médicaments et des substances chimiques , Femelle , Humains , Techniques in vitro , Inflammation/imagerie diagnostique , Cinétique , Ligands , Macaca mulatta , Mâle , Souris knockout , Perméabilité/effets des médicaments et des substances chimiques , Tomographie par émission de positons , Liaison aux protéines , Radiochimie , Rats , Distribution tissulaire , Résultat thérapeutique
19.
Mol Imaging Biol ; 21(5): 871-878, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-30632003

RÉSUMÉ

PURPOSE: The P2X7 receptor, an adenosine triphosphate (ATP)-gated purinoreceptor, has emerged as one of the key players in neuroinflammatory processes. Therefore, developing a positron emission tomography (PET) tracer for imaging of P2X7 receptors in vivo presents a promising approach to diagnose, monitor, and study neuroinflammation in a variety of brain disorders. To fulfill the goal of developing a P2X7 PET ligand as a biomarker of neuroinflammation, [18F]JNJ-64413739 has been recently disclosed. PROCEDURES: We evaluated [18F]JNJ-64413739 in a rat model of neuroinflammation induced by an intracerebral injection of lipopolysaccharide (LPS). In vivo brain uptake was determined by PET imaging. Upregulation of neuroinflammatory biomarkers was determined by quantitative polymerase chain reaction (qPCR). Distribution of the tracer in the brain was determined by ex vivo autoradiography (ARG). The specificity of [18F]JNJ-64413739 was confirmed by performing blocking experiments with the P2X7 antagonist JNJ-54175446. RESULTS: Brain regions of rats injected with LPS had a significantly increased uptake (34 % ± 3 % s.e.m., p = 0.036, t test, standardized uptake value measured over the entire scanning period) of [18F]JNJ-64413739 relative to the corresponding brain regions of control animals injected with phosphate-buffered saline (PBS). The uptake in the contralateral regions and cerebellum was not significantly different between the groups of animals. The increase in uptake of [18F]JNJ-64413739 at the LPS-injected site observed by PET imaging was concordant with ex vivo ARG, upregulation of neuroinflammatory biomarkers, and elevated P2X7 expression levels. CONCLUSIONS: While further work is needed to study [18F]JNJ-64413739 in other types of neuroinflammation, the current results favorably characterize [18F]JNJ-64413739 as a potential PET tracer of central neuroinflammation.


Sujet(s)
Encéphale/imagerie diagnostique , Inflammation/imagerie diagnostique , Inflammation/anatomopathologie , Peptides/composition chimique , Tomographie par émission de positons , Radiopharmaceutiques/composition chimique , Récepteurs purinergiques P2X7/métabolisme , Animaux , Marqueurs biologiques/métabolisme , Encéphale/anatomopathologie , Modèles animaux de maladie humaine , Lipopolysaccharides , Peptides/pharmacocinétique , Projets pilotes , Tomographie par émission de positons couplée à la tomodensitométrie , Pyridines/composition chimique , Rats , Triazoles/composition chimique
20.
Methods Mol Biol ; 1938: 37-47, 2019.
Article de Anglais | MEDLINE | ID: mdl-30617971

RÉSUMÉ

Astrocytes play fundamental roles in development and plasticity of the central nervous system. Dysfunction of astrocytes contributes to most of neurological diseases ranging from neurodegenerative diseases to psychological disorders. To better understand the involvement of astrocytes in both physiological and pathological conditions, and further elucidate their underlying mechanisms, it is critical to establish a reliable isolation and culture method for the cells. In this chapter, we describe the isolation and culture protocols of astrocytes from postnatal and adult mouse brains.


Sujet(s)
Astrocytes/cytologie , Encéphale/cytologie , Techniques de culture cellulaire , Séparation cellulaire , Facteurs âges , Animaux , Astrocytes/métabolisme , Marqueurs biologiques , Séparation cellulaire/méthodes , Immunohistochimie , Souris , Phénotype
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE