Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
1.
BMC Musculoskelet Disord ; 24(1): 538, 2023 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-37391737

RÉSUMÉ

BACKGROUND: Development of valid and feasible quality indicators (QIs) is needed to track quality initiatives for osteoarthritis pain management in primary care settings. METHODS: Literature search identified published guidelines that were reviewed for QI extraction. A panel of 14 experts was assembled, including primary care physicians, rheumatologists, orthopedic surgeons, pain specialists, and outcomes research pharmacists. A screening survey excluded QIs that cannot be reliably extracted from the electronic health record or that are irrelevant for osteoarthritis in primary care settings. A validity screening survey used a 9-point Likert scale to rate the validity of each QI based on predefined criteria. During expert panel discussions, stakeholders revised QI wording, added new QIs, and voted to include or exclude each QI. A priority survey used a 9-point Likert scale to prioritize the included QIs. RESULTS: Literature search identified 520 references published from January 2015 to March 2021 and 4 additional guidelines from professional/governmental websites. The study included 41 guidelines. Extraction of 741 recommendations yielded 115 candidate QIs. Feasibility screening excluded 28 QIs. Validity screening and expert panel discussion excluded 73 QIs and added 1 QI. The final set of 15 prioritized QIs focused on pain management safety, education, weight-management, psychological wellbeing, optimizing first-line medications, referral, and imaging. CONCLUSION: This multi-disciplinary expert panel established consensus on QIs for osteoarthritis pain management in primary care settings by combining scientific evidence with expert opinion. The resulting list of 15 prioritized, valid, and feasible QIs can be used to track quality initiatives for osteoarthritis pain management.


Sujet(s)
Arthrose , Gestion de la douleur , Humains , Indicateurs qualité santé , Douleur , Arthrose/complications , Arthrose/diagnostic , Arthrose/thérapie , Soins de santé primaires
2.
J Infus Nurs ; 46(1): 28-35, 2023.
Article de Anglais | MEDLINE | ID: mdl-36571825

RÉSUMÉ

The use of midline catheters has increased to reduce excessive use of central venous access devices, and additional data on midline catheter complications are needed. This study aimed to describe midline catheter complications among hospitalized patients. This retrospective study included a random sample of 300 hospitalized patients with a midline catheter insertion in 2019. The primary outcome was a composite end point of 8 complications: occlusion, bleeding at insertion site, infiltration/extravasation, catheter-related thrombosis, accidental removal, phlebitis, hematoma, and catheter-related infection. Midline catheter failure was defined as removal prior to the end of therapy due to complications. Among 300 midline catheters, the incidence of the composite end point of 1 or more midline complications was 38% (95% confidence interval, 33%-44%). Complications included occlusion (17.0%), bleeding at insertion site (12.0%), infiltration/extravasation (10.0%), catheter-related thrombosis (4.0%), accidental removal (3.0%), phlebitis (0.3%), hematoma (0.3%), and catheter-related infection (0.3%). Midline catheter failure occurred in 16% of midline catheters (n = 48) due to infiltration/extravasation (n = 27), accidental removal (n = 10), catheter-related thrombosis (n = 9), occlusion (n = 4), and catheter-related infection (n = 1). Three catheters had 2 types of failure. The most common complications of occlusion and bleeding rarely resulted in midline catheter failure. The most common causes of midline catheter failure were infiltration/extravasation, accidental removal, and catheter-related thrombosis.


Sujet(s)
Infections sur cathéters , Cathétérisme veineux central , Cathétérisme périphérique , Phlébite , Thrombose , Humains , Infections sur cathéters/étiologie , Infections sur cathéters/complications , Incidence , Études rétrospectives , Cathéters/effets indésirables , Phlébite/épidémiologie , Phlébite/étiologie , Thrombose/étiologie , Thrombose/complications , Cathétérisme périphérique/méthodes , Hématome/étiologie , Hématome/complications , Cathéters à demeure/effets indésirables
3.
Genes Brain Behav ; 21(7): e12827, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35878875

RÉSUMÉ

ProSAAS is a neuroendocrine protein that is cleaved by neuropeptide-processing enzymes into more than a dozen products including the bigLEN and PEN peptides, which bind and activate the receptors GPR171 and GPR83, respectively. Previous studies have suggested that proSAAS-derived peptides are involved in physiological functions that include body weight regulation, circadian rhythms and anxiety-like behavior. In the present study, we find that proSAAS knockout mice display robust anxiety-like behaviors in the open field, light-dark emergence and elevated zero maze tests. These mutant mice also show a reduction in cued fear and an impairment in fear-potentiated startle, indicating an important role for proSAAS-derived peptides in emotional behaviors. ProSAAS knockout mice exhibit reduced water consumption and urine production relative to wild-type controls. No differences in food consumption and overall energy expenditure were observed between the genotypes. However, the respiratory exchange ratio was elevated in the mutants during the light portion of the light-dark cycle, indicating decreased fat metabolism during this period. While proSAAS knockout mice show normal circadian patterns of activity, even upon long-term exposure to constant darkness, they were unable to shift their circadian clock upon exposure to a light pulse. Taken together, these results show that proSAAS-derived peptides modulate a wide range of behaviors including emotion, metabolism and the regulation of the circadian clock.


Sujet(s)
Neuropeptides/métabolisme , Animaux , Anxiété/génétique , Rythme circadien/génétique , Acte consommatoire , Souris , Souris de lignée C57BL , Souris knockout , Peptides , Récepteurs couplés aux protéines G
4.
PLoS One ; 17(6): e0270329, 2022.
Article de Anglais | MEDLINE | ID: mdl-35749484

RÉSUMÉ

OBJECTIVE: Immunoglobulin-like Domain-Containing Receptor 1 (ILDR1) is expressed on nutrient sensing cholecystokinin-positive enteroendocrine cells of the gastrointestinal tract and it has the unique ability to induce fat-mediated CCK secretion. However, the role of ILDR1 in CCK-mediated regulation of satiety is unknown. In this study, we examined the effects of ILDR1 on food intake and metabolic activity using mice with genetically-deleted Ildr1. METHODS: The expression of ILDR1 in murine tissues and the measurement of adipocyte cell size were evaluated by light and fluorescence confocal microscopy. The effects of Ildr1 deletion on mouse metabolism were quantitated using CLAMS chambers and by targeted metabolomics assays of multiple tissues. Hormone levels were measured by ELISA. The effects of Ildr1 gene deletion on glucose and insulin levels were determined using in vivo oral glucose tolerance, meal tolerance, and insulin tolerance tests, as well as ex vivo islet perifusion. RESULTS: ILDR1 is expressed in a wide range of tissues. Analysis of metabolic data revealed that although Ildr1-/- mice consumed more food than wild-type littermates, they gained less weight on a high fat diet and exhibited increased metabolic activity. Adipocytes in Ildr1-/- mice were significantly smaller than in wild-type mice fed either low or high fat diets. ILDR1 was expressed in both alpha and beta cells of pancreatic islets. Based on oral glucose and mixed meal tolerance tests, Ildr1-/- mice were more effective at lowering post-prandial glucose levels, had improved insulin sensitivity, and glucose-regulated insulin secretion was enhanced in mice lacking ILDR1. CONCLUSION: Ildr1 loss significantly modified metabolic activity in these mutant mice. While Ildr1 gene deletion increased high fat food intake, it reduced weight gain and improved glucose tolerance. These findings indicate that ILDR1 modulates metabolic responses to feeding in mice.


Sujet(s)
Hyperglycémie , Insulinorésistance , Récepteurs de surface cellulaire/métabolisme , Animaux , Cholécystokinine , Alimentation riche en graisse , Délétion de gène , Glucose/métabolisme , Hyperglycémie/génétique , Insuline/métabolisme , Insulinorésistance/génétique , Souris , Souris de lignée C57BL , Obésité/génétique , Obésité/métabolisme
5.
Neurosci Lett ; 661: 11-17, 2017 Nov 20.
Article de Anglais | MEDLINE | ID: mdl-28916300

RÉSUMÉ

Fear-conditioned analgesia (FCA) is modulated by brain areas involved in the descending inhibitory pain pathway such as the basolateral (BLA) and central amygdala (CEA). The BLA contains Ca2+/calmodulin-dependent protein kinase II (CaMKII) and parvalbumin (PV) neurons. CEA neurons are primarily inhibitory (GABAergic) that comprise enkephalin (ENK) interneurons and corticotropin-releasing factor (CRF) - neurons that project to the periaqueductal grey. The purpose of our experiment was to determine the pattern of activation of CaMKII/PV and ENK/CRF neurons following the expression of acute pain, conditioned fear, and FCA. A significant reduction was observed in nociceptive behaviors in mice re-exposed to a contextually-aversive environment. Using NeuN and cFos as markers for activated neurons, CaMKII, PV, ENK, or CRF were used to identify neuronal subtypes. We find that mice expressing conditioned fear displayed an increase in c-Fos/CaMKII co-localization in the lateral amygdala and BLA compared to controls. Additionally a significant increase in cFos/CRF co-localization was observed in mice expressing FCA. These results show that amygdala processing of conditioned contextual aversive, nociceptive, and FCA behaviors involve different neuronal phenotypes and neural circuits between, within, and from various amygdala nuclei. This information will be important in developing novel therapies for treating pain and emotive disorders in humans.


Sujet(s)
Douleur aigüe/physiopathologie , Amygdale (système limbique)/physiopathologie , Noyau central de l'amygdale/métabolisme , Peur/physiologie , Neurones/métabolisme , Analgésie/méthodes , Animaux , Conditionnement psychologique/physiologie , Mâle , Souris de lignée C57BL , Gestion de la douleur , Substance grise centrale du mésencéphale/métabolisme , Substance grise centrale du mésencéphale/physiopathologie
6.
J Biol Chem ; 292(14): 5943-5956, 2017 04 07.
Article de Anglais | MEDLINE | ID: mdl-28213524

RÉSUMÉ

G protein-coupled receptors (GPCRs) regulate many animal behaviors. GPCR signaling is mediated by agonist-promoted interactions of GPCRs with heterotrimeric G proteins, GPCR kinases (GRKs), and arrestins. To further elucidate the role of GRKs in regulating GPCR-mediated behaviors, we utilized the genetic model system Caenorhabditis elegans Our studies demonstrate that grk-2 loss-of-function strains are egg laying-defective and contain low levels of serotonin (5-HT) and high levels of the 5-HT metabolite 5-hydroxyindole acetic acid (5-HIAA). The egg laying defect could be rescued by the expression of wild type but not by catalytically inactive grk-2 or by the selective expression of grk-2 in hermaphrodite-specific neurons. The addition of 5-HT or inhibition of 5-HT metabolism also rescued the egg laying defect. Furthermore, we demonstrate that AMX-2 is the primary monoamine oxidase that metabolizes 5-HT in C. elegans, and we also found that grk-2 loss-of-function strains have abnormally high levels of AMX-2 compared with wild-type nematodes. Interestingly, GRK-2 was also found to interact with and promote the phosphorylation of AMX-2. Additional studies reveal that 5-HIAA functions to inhibit egg laying in a manner dependent on the 5-HT receptor SER-1 and the G protein GOA-1. These results demonstrate that GRK-2 modulates 5-HT metabolism by regulating AMX-2 function and that 5-HIAA may function in the SER-1 signaling pathway.


Sujet(s)
Protéines de Caenorhabditis elegans/biosynthèse , Caenorhabditis elegans/métabolisme , Kinases associées à des récepteurs couplés à une protéine G/biosynthèse , Régulation de l'expression des gènes codant pour des enzymes/physiologie , Monoamine oxidase/métabolisme , Sérotonine/métabolisme , Transduction du signal/physiologie , Animaux , Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/métabolisme , Kinases associées à des récepteurs couplés à une protéine G/génétique , Sous-unités alpha Gi-Go des protéines G/génétique , Sous-unités alpha Gi-Go des protéines G/métabolisme , Acide 5-hydroxy-indole-3-acétique/métabolisme , Monoamine oxidase/génétique , Récepteurs de la sérotonine de type 5-HT2/génétique , Récepteurs de la sérotonine de type 5-HT2/métabolisme , Sérotonine/génétique
7.
Nature ; 537(7619): 185-190, 2016 09 08.
Article de Anglais | MEDLINE | ID: mdl-27533032

RÉSUMÉ

Morphine is an alkaloid from the opium poppy used to treat pain. The potentially lethal side effects of morphine and related opioids-which include fatal respiratory depression-are thought to be mediated by µ-opioid-receptor (µOR) signalling through the ß-arrestin pathway or by actions at other receptors. Conversely, G-protein µOR signalling is thought to confer analgesia. Here we computationally dock over 3 million molecules against the µOR structure and identify new scaffolds unrelated to known opioids. Structure-based optimization yields PZM21-a potent Gi activator with exceptional selectivity for µOR and minimal ß-arrestin-2 recruitment. Unlike morphine, PZM21 is more efficacious for the affective component of analgesia versus the reflexive component and is devoid of both respiratory depression and morphine-like reinforcing activity in mice at equi-analgesic doses. PZM21 thus serves as both a probe to disentangle µOR signalling and a therapeutic lead that is devoid of many of the side effects of current opioids.


Sujet(s)
Analgésiques morphiniques/effets indésirables , Analgésiques morphiniques/composition chimique , Découverte de médicament , Récepteur mu/agonistes , Thiophènes/composition chimique , Thiophènes/pharmacologie , Urée/analogues et dérivés , Analgésie/méthodes , Analgésiques morphiniques/pharmacologie , Animaux , Sous-unités alpha Gi-Go des protéines G/métabolisme , Cellules HEK293 , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Simulation de docking moléculaire , Douleur/traitement médicamenteux , Récepteur mu/déficit , Récepteur mu/génétique , Récepteur mu/métabolisme , Spiranes/pharmacologie , Relation structure-activité , Thiophènes/effets indésirables , Urée/effets indésirables , Urée/composition chimique , Urée/pharmacologie
8.
Genesis ; 54(8): 439-46, 2016 08.
Article de Anglais | MEDLINE | ID: mdl-27194399

RÉSUMÉ

DREADDs, designer receptors exclusively activated by designer drugs, are engineered G protein-coupled receptors (GPCR) which can precisely control GPCR signaling pathways (for example, Gq, Gs, and Gi). This chemogenetic technology for control of GPCR signaling has been successfully applied in a variety of in vivo studies, including in mice, to remotely control GPCR signaling, for example, in neurons, glia cells, pancreatic ß-cells, or cancer cells. In order to fully explore the in vivo applications of the DREADD technology, we generated hM3Dq and hM4Di strains of mice which allow for Cre recombinase-mediated restricted expression of these pathway-selective DREADDs. With the many Cre driver lines now available, these DREADD lines will be applicable to studying a wide array of research and preclinical questions. genesis 54:439-446, 2016. © 2016 Wiley Periodicals, Inc.


Sujet(s)
Ciblage de gène/méthodes , Récepteurs couplés aux protéines G/métabolisme , Animaux , Cellules cultivées , Integrases/génétique , Ligands , Souris , Souris de lignée C57BL , Liaison aux protéines
9.
Proc Natl Acad Sci U S A ; 113(13): 3428-35, 2016 Mar 29.
Article de Anglais | MEDLINE | ID: mdl-26929359

RÉSUMÉ

Disrupted brain iron homeostasis is a common feature of neurodegenerative disease. To begin to understand how neuronal iron handling might be involved, we focused on dopaminergic neurons and asked how inactivation of transport proteins affected iron homeostasis in vivo in mice. Loss of the cellular iron exporter, ferroportin, had no apparent consequences. However, loss of transferrin receptor 1, involved in iron uptake, caused neuronal iron deficiency, age-progressive degeneration of a subset of dopaminergic neurons, and motor deficits. There was gradual depletion of dopaminergic projections in the striatum followed by death of dopaminergic neurons in the substantia nigra. Damaged mitochondria accumulated, and gene expression signatures indicated attempted axonal regeneration, a metabolic switch to glycolysis, oxidative stress, and the unfolded protein response. We demonstrate that loss of transferrin receptor 1, but not loss of ferroportin, can cause neurodegeneration in a subset of dopaminergic neurons in mice.


Sujet(s)
Neurones dopaminergiques/métabolisme , Fer/métabolisme , Dégénérescence nerveuse/étiologie , Dégénérescence nerveuse/métabolisme , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Transporteurs de cations/déficit , Transporteurs de cations/génétique , Transporteurs de cations/métabolisme , Neurones dopaminergiques/anatomopathologie , Femelle , Homéostasie , Mâle , Souris , Souris de souche-129 , Souris de lignée C57BL , Souris knockout , Dégénérescence nerveuse/anatomopathologie , Récepteurs à la transferrine/déficit , Récepteurs à la transferrine/génétique , Récepteurs à la transferrine/métabolisme
10.
Invest New Drugs ; 34(2): 149-58, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26728879

RÉSUMÉ

D2C7-(scdsFv)-PE38KDEL (D2C7-IT) is a novel immunotoxin that reacts with wild-type epidermal growth factor receptor (EGFRwt) and mutant EGFRvIII proteins overexpressed in glioblastomas. This study assessed the toxicity of intracerebral administration of D2C7-IT to support an initial Food and Drug Administration Investigational New Drug application. After the optimization of the formulation and administration, two cohorts (an acute and chronic cohort necropsied on study days 5 and 34) of Sprague-Dawley (SD) rats (four groups of 5 males and 5 females) were infused with the D2C7-IT formulation at total doses of 0, 0.05, 0.1, 0.4 µg (the acute cohort) and 0, 0.05, 0.1, 0.35 µg (the chronic cohort) for approximately 72 h by intracerebral convection-enhanced delivery using osmotic pumps. Mortality was observed in the 0.40 µg (5/10 rats) and 0.35 µg (4/10 rats) high-dose groups of each cohort. Body weight loss and abnormal behavior were only revealed in the rats treated with high doses of D2C7-IT. No dose-related effects were observed in clinical laboratory tests in either cohort. A gross pathologic examination of systemic tissues from the high-dose and control groups in both cohorts exhibited no dose-related or drug-related pathologic findings. Brain histopathology revealed the frequent occurrence of dose-related encephalomalacia, edema, and demyelination in the high-dose groups of both cohorts. In this study, the maximum tolerated dose of D2C7-IT was determined to be between 0.10 and 0.35 µg, and the no-observed-adverse-effect-level was 0.05 µg in SD rats. Both parameters were utilized to design the Phase I/II D2C7-IT clinical trial.


Sujet(s)
Convection , Systèmes de délivrance de médicaments , Évaluation préclinique de médicament , Immunoconjugués/administration et posologie , Immunoconjugués/toxicité , Immunotoxines/administration et posologie , Immunotoxines/toxicité , Anticorps à chaîne unique/administration et posologie , Anticorps à chaîne unique/toxicité , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Femelle , Concentration inhibitrice 50 , Injections ventriculaires , Mâle , Rat Sprague-Dawley
11.
Neuropsychopharmacology ; 41(5): 1404-15, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26383016

RÉSUMÉ

Elucidating how the brain's serotonergic network mediates diverse behavioral actions over both relatively short (minutes-hours) and long period of time (days-weeks) remains a major challenge for neuroscience. Our relative ignorance is largely due to the lack of technologies with robustness, reversibility, and spatio-temporal control. Recently, we have demonstrated that our chemogenetic approach (eg, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)) provides a reliable and robust tool for controlling genetically defined neural populations. Here we show how short- and long-term activation of dorsal raphe nucleus (DRN) serotonergic neurons induces robust behavioral responses. We found that both short- and long-term activation of DRN serotonergic neurons induce antidepressant-like behavioral responses. However, only short-term activation induces anxiogenic-like behaviors. In parallel, these behavioral phenotypes were associated with a metabolic map of whole brain network activity via a recently developed non-invasive imaging technology DREAMM (DREADD Associated Metabolic Mapping). Our findings reveal a previously unappreciated brain network elicited by selective activation of DRN serotonin neurons and illuminate potential therapeutic and adverse effects of drugs targeting DRN neurons.


Sujet(s)
Anxiété/physiopathologie , Dépression/physiopathologie , Noyau dorsal du raphé/physiologie , Neurones sérotonergiques/physiologie , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Comportement animal/physiologie , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/physiologie , Rythme circadien , Drogues fabriquées clandestinement/administration et posologie , Noyau dorsal du raphé/effets des médicaments et des substances chimiques , Noyau dorsal du raphé/métabolisme , Mâle , Souris , Souris transgéniques , Neurones sérotonergiques/effets des médicaments et des substances chimiques , Neurones sérotonergiques/métabolisme , Facteurs temps
12.
Nat Neurosci ; 18(6): 883-91, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25938885

RÉSUMÉ

Psychiatric and neurodevelopmental disorders may arise from anomalies in long-range neuronal connectivity downstream of pathologies in dendritic spines. However, the mechanisms that may link spine pathology to circuit abnormalities relevant to atypical behavior remain unknown. Using a mouse model to conditionally disrupt a critical regulator of the dendritic spine cytoskeleton, the actin-related protein 2/3 complex (Arp2/3), we report here a molecular mechanism that unexpectedly reveals the inter-relationship of progressive spine pruning, elevated frontal cortical excitation of pyramidal neurons and striatal hyperdopaminergia in a cortical-to-midbrain circuit abnormality. The main symptomatic manifestations of this circuit abnormality are psychomotor agitation and stereotypical behaviors, which are relieved by antipsychotics. Moreover, this antipsychotic-responsive locomotion can be mimicked in wild-type mice by optogenetic activation of this circuit. Collectively these results reveal molecular and neural-circuit mechanisms, illustrating how diverse pathologies may converge to drive behaviors relevant to psychiatric disorders.


Sujet(s)
Neuroleptiques/pharmacologie , Épines dendritiques/physiologie , Dopamine/métabolisme , Locomotion/effets des médicaments et des substances chimiques , Néostriatum/métabolisme , Réseau nerveux/métabolisme , Protéine-3 apparentée à l'actine/génétique , Protéine-3 apparentée à l'actine/métabolisme , Protéine-2 de type angiopoïétine , Protéines semblables à l'angiopoïétine , Angiopoïétines/génétique , Angiopoïétines/métabolisme , Animaux , Femelle , Mâle , Mésencéphale/effets des médicaments et des substances chimiques , Souris , Souris knockout , Mutation , Néostriatum/effets des médicaments et des substances chimiques , Réseau nerveux/effets des médicaments et des substances chimiques , Optogénétique , Techniques de patch-clamp , Cortex préfrontal/effets des médicaments et des substances chimiques , Agitation psychomotrice/psychologie , Cellules pyramidales/effets des médicaments et des substances chimiques , Comportement stéréotypé/effets des médicaments et des substances chimiques
13.
Neuron ; 80(2): 429-41, 2013 Oct 16.
Article de Anglais | MEDLINE | ID: mdl-24139043

RÉSUMÉ

We analyzed four families that presented with a similar condition characterized by congenital microcephaly, intellectual disability, progressive cerebral atrophy, and intractable seizures. We show that recessive mutations in the ASNS gene are responsible for this syndrome. Two of the identified missense mutations dramatically reduce ASNS protein abundance, suggesting that the mutations cause loss of function. Hypomorphic Asns mutant mice have structural brain abnormalities, including enlarged ventricles and reduced cortical thickness, and show deficits in learning and memory mimicking aspects of the patient phenotype. ASNS encodes asparagine synthetase, which catalyzes the synthesis of asparagine from glutamine and aspartate. The neurological impairment resulting from ASNS deficiency may be explained by asparagine depletion in the brain or by accumulation of aspartate/glutamate leading to enhanced excitability and neuronal damage. Our study thus indicates that asparagine synthesis is essential for the development and function of the brain but not for that of other organs.


Sujet(s)
Aspartate-ammonia ligase/déficit , Aspartate-ammonia ligase/génétique , Encéphale/enzymologie , Encéphale/anatomopathologie , Prédisposition génétique à une maladie/génétique , Microcéphalie/enzymologie , Microcéphalie/génétique , Adolescent , Animaux , Atrophie/complications , Atrophie/enzymologie , Atrophie/génétique , Enfant , Femelle , Humains , Nourrisson , Nouveau-né , Déficience intellectuelle/complications , Déficience intellectuelle/enzymologie , Déficience intellectuelle/génétique , Déficience intellectuelle/anatomopathologie , Mâle , Souris , Souris transgéniques , Microcéphalie/complications , Microcéphalie/anatomopathologie , Mutation faux-sens/génétique , Pedigree , Syndrome
14.
Proc Natl Acad Sci U S A ; 110(40): 16211-6, 2013 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-24043826

RÉSUMÉ

Multiple peptide systems, including neuropeptide Y, leptin, ghrelin, and others, are involved with the control of food intake and body weight. The peptide LENSSPQAPARRLLPP (BigLEN) has been proposed to act through an unknown receptor to regulate body weight. In the present study, we used a combination of ligand-binding and receptor-activity assays to characterize a Gαi/o protein-coupled receptor activated by BigLEN in the mouse hypothalamus and Neuro2A cells. We then selected orphan G protein-coupled receptors expressed in the hypothalamus and Neuro2A cells and tested each for activation by BigLEN. G protein-coupled receptor 171 (GPR171) is activated by BigLEN, but not by the C terminally truncated peptide LittleLEN. The four C-terminal amino acids of BigLEN are sufficient to bind and activate GPR171. Overexpression of GPR171 leads to an increase, and knockdown leads to a decrease, in binding and signaling by BigLEN and the C-terminal peptide. In the hypothalamus GPR171 expression complements the expression of BigLEN, and its level and activity are elevated in mice lacking BigLEN. In mice, shRNA-mediated knockdown of hypothalamic GPR171 leads to a decrease in BigLEN signaling and results in changes in food intake and metabolism. The combination of GPR171 shRNA together with neutralization of BigLEN peptide by antibody absorption nearly eliminates acute feeding in food-deprived mice. Taken together, these results demonstrate that GPR171 is the BigLEN receptor and that the BigLEN-GPR171 system plays an important role in regulating responses associated with feeding and metabolism in mice.


Sujet(s)
Poids/physiologie , Comportement alimentaire/physiologie , Neuropeptides/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Analyse de variance , Animaux , Technique de Western , Cellules CHO , Cricetinae , Cricetulus , AMP cyclique/métabolisme , Immunohistochimie , Système de signalisation des MAP kinases/physiologie , Souris , Souris de lignée C57BL , Phosphorylation , Réaction de polymérisation en chaine en temps réel
15.
Neuropsychopharmacology ; 37(2): 321-37, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-21956448

RÉSUMÉ

Systemic administration of amphetamine (AMPH) induces phosphorylation of MeCP2 at Ser421 (pMeCP2) in select populations of neurons in the mesolimbocortical brain regions. Because AMPH simultaneously activates multiple monoamine neurotransmitter systems, here we examined the ability of dopamine (DA), serotonin (5-HT), and norepinephrine (NE) to induce pMeCP2. Selective blockade of the DA transporter (DAT) or the 5-HT transporter (SERT), but not the NE transporter (NET), was sufficient to induce pMeCP2 in the CNS. DAT blockade induced pMeCP2 in the prelimbic cortex (PLC) and nucleus accumbens (NAc), whereas SERT blockade induced pMeCP2 only in the NAc. Administration of selective DA and 5-HT receptor agonists was also sufficient to induce pMeCP2; however, the specific combination of DA and 5-HT receptors activated determined the regional- and cell-type specificity of pMeCP2 induction. The D(1)-class DA receptor agonist SKF81297 induced pMeCP2 widely; however, coadministration of the D(2)-class agonist quinpirole restricted the induction of pMeCP2 to GABAergic interneurons of the NAc. Intra-striatal injection of the adenylate cyclase activator forskolin was sufficient to induce pMeCP2 in medium-spiny neurons, suggesting that the combinatorial regulation of cAMP by different classes of DA and 5-HT receptors may contribute to the cell-type specificity of pMeCP2 induction. Consistent with the regulation of pMeCP2 by multiple monoamine neurotransmitters, genetic disruption of any single monoamine transporter in DAT-, SERT-, and NET-knockout mice failed to eliminate AMPH-induced pMeCP2 in the NAc. Together, these studies indicate that combinatorial signaling through DA and 5-HT receptors can regulate the brain region- and cell-type specific pMeCP2 in the CNS.


Sujet(s)
Encéphale/métabolisme , Dopamine/physiologie , Régulation de l'expression des gènes/physiologie , Protéine-2 de liaison au CpG méthylé/métabolisme , Sérotonine/physiologie , Animaux , Encéphale/effets des médicaments et des substances chimiques , Cellules cultivées , Citalopram/pharmacologie , Colforsine/pharmacologie , Agonistes de la dopamine/pharmacologie , Interactions médicamenteuses , Neurones GABAergiques/effets des médicaments et des substances chimiques , Neurones GABAergiques/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Souris , Souris knockout , Microinjections , Imagerie moléculaire/méthodes , Morpholines/pharmacologie , Activité motrice/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Phosphorylation/physiologie , Pipérazines/pharmacologie , Transporteurs plasmiques de neurotransmetteurs/antagonistes et inhibiteurs , Transporteurs plasmiques de neurotransmetteurs/génétique , Quipazine/pharmacologie , Réboxétine
16.
J Pharm Sci ; 98(4): 1587-601, 2009 Apr.
Article de Anglais | MEDLINE | ID: mdl-18780336

RÉSUMÉ

Pharmacokinetics of amitriptyline and nortriptyline were evaluated after intravenous (2.5-10 mg/kg) and oral (10-100 mg/kg) administration of amitriptyline to rats. The hepatic, gastric, and intestinal first-pass effects of amitriptyline were also measured at a dose of 10 mg/kg. The areas under the plasma concentration-time curve (AUCs) of amitriptyline were dose-proportional following both intravenous and oral administration. After oral administration of amitriptyline, approximately 1.50% of the dose was not absorbed, the extent of absolute oral bioavalability (F) was approximately 6.30%, and the hepatic and intestinal first-pass effects of amitriptyline were approximately 9% and 87% of the oral dose, respectively. Although the hepatic first-pass effect was 78.9% after absorption into the portal vein, the value was only 9% of the oral dose due to considerable intestinal first-pass effect in rats. The low F of amitriptyline in rats was primarily attributable to considerable intestinal first-pass effect. This study proves the little contribution of considerable hepatic first-pass effect to low F of amitriptyline due to great intestinal first-pass effect in rats. The lower F value of amitriptyline in rats than that in humans (46 +/- 48%) was due to grater metabolism of amitriptyline in rats' liver and/or small intestine.


Sujet(s)
Amitriptyline/pharmacocinétique , Intestin grêle/métabolisme , Foie/métabolisme , Nortriptyline/pharmacocinétique , Administration par voie orale , Amitriptyline/administration et posologie , Amitriptyline/sang , Animaux , Biodisponibilité , Relation dose-effet des médicaments , Muqueuse gastrique/métabolisme , Injections veineuses , Mâle , Nortriptyline/administration et posologie , Nortriptyline/sang , Rats , Rat Sprague-Dawley , Distribution tissulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...