Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 298(10): 102458, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36063995

RESUMO

Glutamate acts at eight metabotropic glutamate (mGlu) receptor subtypes expressed in a partially overlapping fashion in distinct brain circuits. Recent evidence indicates that specific mGlu receptor protomers can heterodimerize and that these heterodimers can exhibit different pharmacology when compared to their homodimeric counterparts. Group III mGlu agonist-induced suppression of evoked excitatory potentials and induction of long-term potentiation at Schaffer collateral-CA1 (SC-CA1) synapses in the rodent hippocampus can be blocked by the selective mGlu7 negative allosteric modulator (NAM), ADX71743. Curiously, a different mGlu7 NAM, 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one, failed to block these responses in brain slices despite its robust activity at mGlu7 homodimers in vitro. We hypothesized that this might result from heterodimerization of mGlu7 with another mGlu receptor protomer and focused on mGlu8 as a candidate given the reported effects of mGlu8-targeted compounds in the hippocampus. Here, we used complemented donor acceptor-resonance energy transfer to study mGlu7/8 heterodimer activation in vitro and observed that ADX71743 blocked responses of both mGlu7/7 homodimers and mGlu7/8 heterodimers, whereas 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazonolo[4,5-c]pyridin-4(5H)-one only antagonized responses of mGlu7/7 homodimers. Taken together with our electrophysiology observations, these results suggest that a receptor with pharmacology consistent with an mGlu7/8 heterodimer modulates the activity of SC-CA1 synapses. Building on this hypothesis, we identified two additional structurally related mGlu7 NAMs that also differ in their activity at mGlu7/8 heterodimers, in a manner consistent with their ability to inhibit synaptic transmission and plasticity at SC-CA1. Thus, we propose that mGlu7/8 heterodimers are a key molecular target for modulating the activity of hippocampal SC-CA1 synapses.


Assuntos
Ácido Glutâmico , Receptores de Glutamato Metabotrópico , Sinapses , Hipocampo/metabolismo , Potenciação de Longa Duração , Receptores de Glutamato Metabotrópico/metabolismo , Sinapses/metabolismo , Animais , Roedores , Saccharomyces cerevisiae , Eletrofisiologia
2.
Cereb Cortex ; 28(12): 4291-4304, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29136107

RESUMO

Dysregulated fear memory can lead to a broad spectrum of anxiety disorders. The brain systems underlying fear memory are manifold, with the hippocampus being prominently involved by housing fear-related spatial memories as engrams, which are created and stored through neural changes such as synaptic plasticity. Although metabotropic glutamate (mGlu) receptors contribute significantly to both fear behavior and hippocampal synaptic plasticity, the relationship between these two phenomena has not been fully elucidated. Here, we report that contextual fear extinction induces a novel form of metaplasticity mediated by mGlu5 at the hippocampal SC-CA1 synapse. Further, blockade of mGlu5 prevents both contextual fear extinction and expression of this metaplasticity. This form of metaplasticity was absent in a mouse model of MECP2-duplication syndrome, corresponding to a complete deficit in extinction learning. These findings suggest that mGlu5-dependent metaplasticity within the hippocampus may play a critical role in extinction of contextual fear.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Hipocampo/fisiologia , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Receptores de Ácido Caínico/fisiologia , Animais , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor CB1 de Canabinoide/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia
3.
J Pharmacol Exp Ther ; 365(2): 291-300, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29523700

RESUMO

Mutations in the MeCP2 gene are responsible for the neurodevelopmental disorder Rett syndrome (RTT). MeCP2 is a DNA-binding protein whose abundance and ability to complex with histone deacetylase 3 is linked to the regulation of chromatin structure. Consequently, loss-of-function mutations in MeCP2 are predicted to have broad effects on gene expression. However, to date, studies in mouse models of RTT have identified a limited number of gene or pathway-level disruptions, and even fewer genes have been identified that could be considered amenable to classic drug discovery approaches. Here, we performed RNA sequencing (RNA-seq) on nine motor cortex and six cerebellar autopsy samples from RTT patients and controls. This approach identified 1887 significantly affected genes in the motor cortex and 2110 genes in the cerebellum, with a global trend toward increased expression. Pathway-level analysis identified enrichment in genes associated with mitogen-activated protein kinase signaling, long-term potentiation, and axon guidance. A survey of our RNA-seq results also identified a significant decrease in expression of the CHRM4 gene, which encodes a receptor [muscarinic acetylcholine receptor 4 (M4)] that is the subject of multiple large drug discovery efforts for schizophrenia and Alzheimer's disease. We confirmed that CHRM4 expression was decreased in RTT patients, and, excitingly, we demonstrated that M4 potentiation normalizes social and cognitive phenotypes in Mecp2+/- mice. This work provides an experimental paradigm in which translationally relevant targets can be identified using transcriptomics in RTT autopsy samples, back-modeled in Mecp2+/- mice, and assessed for preclinical efficacy using existing pharmacological tool compounds.


Assuntos
Terapia de Alvo Molecular , Receptor Muscarínico M4/metabolismo , Síndrome de Rett/tratamento farmacológico , Síndrome de Rett/genética , Análise de Sequência de RNA , Animais , Autopsia , Cerebelo/metabolismo , Humanos , Camundongos , Córtex Motor/metabolismo , Receptor Muscarínico M4/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patologia
4.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1596-1604, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28213125

RESUMO

Huntington's disease (HD) is caused by a mutation in the huntingtin gene (HTT), resulting in profound striatal neurodegeneration through an unknown mechanism. Perturbations in the urea cycle have been reported in HD models and in HD patient blood and brain. In neurons, arginase is a central urea cycle enzyme, and the metal manganese (Mn) is an essential cofactor. Deficient biological responses to Mn, and reduced Mn accumulation have been observed in HD striatal mouse and cell models. Here we report in vivo and ex vivo evidence of a urea cycle metabolic phenotype in a prodromal HD mouse model. Further, either in vivo or in vitro Mn supplementation reverses the urea-cycle pathology by restoring arginase activity. We show that Arginase 2 (ARG2) is the arginase enzyme present in these mouse brain models, with ARG2 protein levels directly increased by Mn exposure. ARG2 protein is not reduced in the prodromal stage, though enzyme activity is reduced, indicating that altered Mn bioavailability as a cofactor leads to the deficient enzymatic activity. These data support a hypothesis that mutant HTT leads to a selective deficiency of neuronal Mn at an early disease stage, contributing to HD striatal urea-cycle pathophysiology through an effect on arginase activity.


Assuntos
Corpo Estriado/metabolismo , Doença de Huntington/metabolismo , Manganês/metabolismo , Neurônios/metabolismo , Ureia/metabolismo , Animais , Arginase/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Doença de Huntington/patologia , Masculino , Camundongos , Neurônios/patologia
5.
Zootaxa ; 3790: 534-42, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24869885

RESUMO

Two new species of gall-inducing wasps, Selitrichodes casuarinae Fisher & La Salle sp. n. and Selitrichodes utilis Fisher & La Salle sp. n., are described from Micronesia (Guam, Rota and Palau Islands) and Australia respectively. These species induce galls on Casuarina and can cause extensive damage to the trees. Their status as pest or beneficial species is discussed.


Assuntos
Biodiversidade , Tumores de Planta/parasitologia , Árvores/parasitologia , Vespas/anatomia & histologia , Animais , Austrália , Feminino , Masculino , Vespas/classificação , Vespas/fisiologia
6.
Plant J ; 71(1): 147-60, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22381039

RESUMO

α-Kleisins are core components of meiotic and mitotic cohesin complexes. Arabidopsis contains four genes that encode α-kleisin proteins: SYN1, SYN2, SYN3 and SYN4. SYN1, a REC8 ortholog, is essential for meiosis, while SYN2 and SYN4 appear to be SCC1 orthologs and function in mitosis. SYN3 is essential for megagametogenesis and is enriched in the nucleolus of meiotic and mitotic cells. In this study the role of SYN3 during meiosis was investigated by characterization of plants that express SYN3-RNAi constructs from either meiotic DMC1, native SYN3, or inducible PX7 promoters. Reduction of SYN3 caused defects in homologous chromosome synapsis and synaptonemal complex (SC) formation during male and female meiosis. Consistent with this observation, relatively little signal for the SC component ZYP1 was detected on the chromosomes of SYN3-RNAi plants. ZYP1 transcript levels were relatively normal, but several transcripts for genes that encode proteins involved in meiotic recombination were altered, which suggested that a reduction in SYN3 may inhibit meiotic progression by alteration of meiotic gene expression.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Proteínas de Ciclo Celular/metabolismo , Pareamento Cromossômico , Meiose , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação da Expressão Gênica de Plantas , Células Germinativas Vegetais/crescimento & desenvolvimento , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA
7.
J Exp Anal Behav ; 118(1): 132-147, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35607847

RESUMO

The present study determined whether behavioral economic demand analysis could characterize mothers' decision to exclusively breastfeed in the workplace. Females, aged between 18 and 50 who have given birth in the past three years, completed a novel demand task with hypothetical scenarios, in which they returned to work with a 2-month-old baby. Participants rated their likelihood of breastfeeding their baby at a workplace lactation room versus formula-feeding their baby at their desk. The distance to the lactation room ranged from 10 s to 60 min. This assessment was conducted with and without hypothetical financial incentives for 6-month exclusive breastfeeding. Primary dependent measures were demand intensity and change in demand elasticity, which could conceptually represent initiation and continuation of breastfeeding, respectively. Demand for breastfeeding was more intense and less elastic (i.e., more likely to initiate and continue breastfeeding) among mothers with an experience of 6-month exclusive breastfeeding and under the condition with the financial incentives. The novel demand task can potentially provide a useful behavioral marker for quantifying mothers' decision to initiate and continue exclusive breastfeeding in the workplace, informing workplace policy regarding lactation rooms, identifying risk for early cessation, and developing and individualizing an intervention to assist mothers to exclusively breastfeed in the workplace.


Assuntos
Aleitamento Materno , Mães , Adolescente , Adulto , Economia Comportamental , Feminino , Humanos , Lactente , Pessoa de Meia-Idade , Motivação , Local de Trabalho , Adulto Jovem
8.
Neurotherapeutics ; 19(4): 1340-1352, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35670902

RESUMO

Rett syndrome (RTT) is a neurodevelopmental disorder that is characterized by developmental regression, loss of communicative ability, stereotyped hand wringing, cognitive impairment, and central apneas, among many other symptoms. RTT is caused by loss-of-function mutations in a methyl-reader known as methyl-CpG-binding protein 2 (MeCP2), a protein that links epigenetic changes on DNA to larger chromatin structure. Historically, target identification for RTT has relied heavily on Mecp2 knockout mice; however, we recently adopted the alternative approach of performing transcriptional profiling in autopsy samples from RTT patients. Through this mechanism, we identified muscarinic acetylcholine receptors (mAChRs) as potential therapeutic targets. Here, we characterized a cohort of 40 temporal cortex samples from individuals with RTT and quantified significantly decreased levels of the M1, M2, M3, and M5 mAChRs subtypes relative to neurotypical controls. Of these four subtypes, M1 expression demonstrated a linear relationship with MeCP2 expression, such that M1 levels were only diminished in contexts where MeCP2 was also significantly decreased. Further, we show that M1 potentiation with the positive allosteric modulator (PAM) VU0453595 (VU595) rescued social preference, spatial memory, and associative memory deficits, as well as decreased apneas in Mecp2+/- mice. VU595's efficacy on apneas in Mecp2+/- mice was mediated by the facilitation of the transition from inspiration to expiration. Molecular analysis correlated rescue with normalized global gene expression patterns in the brainstem and hippocampus, as well as increased Gsk3ß inhibition and NMDA receptor trafficking. Together, these data suggest that M1 PAMs could represent a new class of RTT therapeutics.


Assuntos
Síndrome de Rett , Camundongos , Animais , Síndrome de Rett/tratamento farmacológico , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Apneia , Receptores de N-Metil-D-Aspartato , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Camundongos Knockout , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Cromatina
9.
Neuropsychopharmacology ; 46(12): 2148-2157, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34035469

RESUMO

Clinical and translational studies suggest that prefrontal cortex (PFC) dysregulation is a hallmark feature of several affective disorders. Thus, investigating the mechanisms involved in the regulation of PFC function and synaptic plasticity could aid in developing new medications. In recent years, the mGlu2 and mGlu3 subtypes of metabotropic glutamate (mGlu) receptors have emerged as exciting potential targets for the treatment of affective disorders, as mGlu2/3 antagonists exert antidepressant-like effects across many rodent models. Several recent studies suggest that presynaptic mGlu2 receptors may contribute to these effects by regulating excitatory transmission at synapses from the thalamus to the PFC. Interestingly, we found that mGlu3 receptors also inhibit excitatory drive to the PFC but act by inducing long-term depression (LTD) at amygdala-PFC synapses. It remains unclear, however, whether blockade of presynaptic, postsynaptic, or glial mGlu3 receptors contribute to long-term effects on PFC circuit function and antidepressant-like effects of mGlu2/3 antagonists. To address these outstanding questions, we leveraged transgenic Grm3fl/fl mice and viral-mediated gene transfer to genetically ablate mGlu3 receptors from pyramidal cells in the frontal cortex of adult mice of all sexes. Consistent with a role for mGlu3 in PFC pyramidal cells, mGlu3-dependent amygdala-cortical LTD was eliminated following mGlu3 receptor knockdown. Furthermore, knockdown mice displayed a modest, task-specific anxiolytic phenotype and decreased passive coping behaviors. These studies reveal that postsynaptic mGlu3 receptors are critical for mGlu3-dependent LTD and provide convergent genetic evidence suggesting that modulating cortical mGlu3 receptors may provide a promising new approach for the treatment of mood disorders.


Assuntos
Ácido Glutâmico , Receptores de Glutamato Metabotrópico/genética , Animais , Camundongos , Plasticidade Neuronal , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo
10.
JCI Insight ; 6(4)2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33476302

RESUMO

The metabotropic glutamate receptor 7 (mGlu7) is a G protein-coupled receptor that has been recently linked to neurodevelopmental disorders. This association is supported by the identification of GRM7 variants in patients with autism spectrum disorder, attention deficit hyperactivity disorder, and severe developmental delay. One GRM7 mutation previously reported in 2 patients results in a single amino acid change, I154T, within the mGlu7 ligand-binding domain. Here, we report 2 new patients with this mutation who present with severe developmental delay and epilepsy. Functional studies of the mGlu7-I154T mutant reveal that this substitution resulted in significant loss of mGlu7 protein expression in HEK293A cells and in mice. We show that this occurred posttranscriptionally at the level of protein expression and trafficking. Similar to mGlu7-global KO mice, mGlu7-I154T animals exhibited reduced motor coordination, deficits in contextual fear learning, and seizures. This provides functional evidence that a disease-associated mutation affecting the mGlu7 receptor was sufficient to cause neurological dysfunction in mice and further validates GRM7 as a disease-causing gene in the human population.


Assuntos
Transtorno do Espectro Autista/genética , Predisposição Genética para Doença/genética , Fenótipo , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Criança , Pré-Escolar , Epilepsia , Medo , Feminino , Proteínas de Ligação ao GTP , Humanos , Lactente , Aprendizagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Transtornos do Neurodesenvolvimento/genética , Linhagem , Convulsões
11.
Insects ; 11(6)2020 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-32586012

RESUMO

The Queensland fruit fly (Bactrocera tryoni; Q-fly) is an Australian endemic horticultural pest species, which has caused enormous economic losses. It has the potential to expand its range to currently Q-fly-free areas and poses a serious threat to the Australian horticultural industry. A large number of studies have investigated the correlation between environmental factors and Q-fly development, reproduction, and expansion. However, it is still not clear how Q-fly morphological traits vary with the environment. Our study focused on three morphological traits (body size, wing shape, and fluctuating asymmetry) in Q-fly samples collected from 1955 to 1965. We assessed how these traits vary by sex, and in response to latitude, environmental variables, and geographic distance. First, we found sexual dimorphism in body size and wing shape, but not in fluctuating asymmetry. Females had a larger body size but shorter and wider wings than males, which may be due to reproductive and/or locomotion differences between females and males. Secondly, the body size of Q-flies varied with latitude, which conforms to Bergmann's rule. Finally, we found Q-fly wing shape was more closely related to temperature rather than aridity, and low temperature and high aridity may lead to high asymmetry in Q-fly populations.

12.
Genes Brain Behav ; 19(7): e12654, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32248644

RESUMO

Neurodevelopmental disorders are characterized by deficits in communication, cognition, attention, social behavior and/or motor control. Previous studies have pointed to the involvement of genes that regulate synaptic structure and function in the pathogenesis of these disorders. One such gene, GRM7, encodes the metabotropic glutamate receptor 7 (mGlu7 ), a G protein-coupled receptor that regulates presynaptic neurotransmitter release. Mutations and polymorphisms in GRM7 have been associated with neurodevelopmental disorders in clinical populations; however, limited preclinical studies have evaluated mGlu7 in the context of this specific disease class. Here, we show that the absence of mGlu7 in mice is sufficient to alter phenotypes within the domains of social behavior, associative learning, motor function, epilepsy and sleep. Moreover, Grm7 knockout mice exhibit an attenuated response to amphetamine. These findings provide rationale for further investigation of mGlu7 as a potential therapeutic target for neurodevelopmental disorders such as idiopathic autism, attention deficit hyperactivity disorder and Rett syndrome.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/genética , Epilepsia/genética , Transtornos do Neurodesenvolvimento/genética , Receptores de Glutamato Metabotrópico/genética , Animais , Feminino , Aprendizagem , Masculino , Camundongos , Transtornos do Neurodesenvolvimento/fisiopatologia , Fenótipo , Receptores de Glutamato Metabotrópico/deficiência , Sono , Comportamento Social
13.
Sci Signal ; 12(610)2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31796631

RESUMO

Highly selective, positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have emerged as an exciting new approach to potentially improve cognitive function in patients suffering from Alzheimer's disease and schizophrenia. Discovery programs have produced a structurally diverse range of M1 receptor PAMs with distinct pharmacological properties, including different extents of agonist activity and differences in signal bias. This includes biased M1 receptor PAMs that can potentiate coupling of the receptor to activation of phospholipase C (PLC) but not phospholipase D (PLD). However, little is known about the role of PLD in M1 receptor signaling in native systems, and it is not clear whether biased M1 PAMs display differences in modulating M1-mediated responses in native tissue. Using PLD inhibitors and PLD knockout mice, we showed that PLD was necessary for the induction of M1-dependent long-term depression (LTD) in the prefrontal cortex (PFC). Furthermore, biased M1 PAMs that did not couple to PLD not only failed to potentiate orthosteric agonist-induced LTD but also blocked M1-dependent LTD in the PFC. In contrast, biased and nonbiased M1 PAMs acted similarly in potentiating M1-dependent electrophysiological responses that were PLD independent. These findings demonstrate that PLD plays a critical role in the ability of M1 PAMs to modulate certain central nervous system (CNS) functions and that biased M1 PAMs function differently in brain regions implicated in cognition.


Assuntos
Córtex Cerebral/enzimologia , Plasticidade Neuronal , Fosfolipase D/genética , Fosfolipase D/metabolismo , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo , Sítio Alostérico , Animais , Células CHO , Cálcio/química , Cognição , Cricetinae , Cricetulus , Eletrofisiologia , Feminino , Humanos , Depressão Sináptica de Longo Prazo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Córtex Pré-Frontal/enzimologia , Transdução de Sinais , Fosfolipases Tipo C/metabolismo
14.
Front Mol Neurosci ; 11: 387, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30405350

RESUMO

Neurodevelopmental disorders (NDDs) are characterized by a wide range of symptoms including delayed speech, intellectual disability, motor dysfunction, social deficits, breathing problems, structural abnormalities, and epilepsy. Unfortunately, current treatment strategies are limited and innovative new approaches are sorely needed to address these complex diseases. The metabotropic glutamate receptors are a class of G protein-coupled receptors that act to modulate neurotransmission across many brain structures. They have shown great promise as drug targets for numerous neurological and psychiatric diseases. Moreover, the development of subtype-selective allosteric modulators has allowed detailed studies of each receptor subtype. Here, we focus on the metabotropic glutamate receptor 7 (mGlu7) as a potential therapeutic target for NDDs. mGlu7 is expressed widely throughout the brain in regions that correspond to the symptom domains listed above and has established roles in synaptic physiology and behavior. Single nucleotide polymorphisms and mutations in the GRM7 gene have been associated with idiopathic autism and other NDDs in patients. In rodent models, existing literature suggests that decreased mGlu7 expression and/or function may lead to symptoms that overlap with those of NDDs. Furthermore, potentiation of mGlu7 activity has shown efficacy in a mouse model of Rett syndrome. In this review, we summarize current findings that provide rationale for the continued development of mGlu7 modulators as potential therapeutics.

15.
Front Mol Neurosci ; 11: 444, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30588231

RESUMO

[This corrects the article DOI: 10.3389/fnmol.2018.00387.].

16.
ACS Chem Neurosci ; 9(9): 2210-2217, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-29227625

RESUMO

Rett syndrome and MECP2 Duplication syndrome are neurodevelopmental disorders attributed to loss-of-function mutations in, or duplication of, the gene encoding methyl-CpG-binding protein 2 (MeCP2), respectively. We recently reported decreased expression and function of the metabotropic glutamate receptor 7 (mGlu7) in a mouse model of Rett syndrome. Positive allosteric modulation of mGlu7 activity was sufficient to improve several disease phenotypes including cognition. Here, we tested the hypothesis that mGlu7 expression would be reciprocally regulated in a mouse model of MECP2 Duplication syndrome, such that negative modulation of mGlu7 activity would exert therapeutic benefit. To the contrary, we report that mGlu7 is not functionally increased in mice overexpressing MeCP2 and that neither genetic nor pharmacological reduction of mGlu7 activity impacts phenotypes that are antiparallel to those observed in Rett syndrome model mice. These data expand our understanding of how mGlu7 expression and function is affected by changes in MeCP2 dosage and have important implications for the therapeutic development of mGlu7 modulators.


Assuntos
Ansiedade/metabolismo , Medo/fisiologia , Aprendizagem/fisiologia , Deficiência Intelectual Ligada ao Cromossomo X/genética , Receptores de Glutamato Metabotrópico/genética , Regulação Alostérica , Animais , Ansiedade/fisiopatologia , Ansiedade/psicologia , Benzoxazóis/farmacologia , Modelos Animais de Doenças , Agonistas de Aminoácidos Excitatórios/farmacologia , Medo/efeitos dos fármacos , Medo/psicologia , Aprendizagem/efeitos dos fármacos , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/fisiopatologia , Deficiência Intelectual Ligada ao Cromossomo X/psicologia , Camundongos , Fenótipo , Ácidos Fosfínicos/farmacologia , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/metabolismo
17.
Surgery ; 161(2): 308-311, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28096037

RESUMO

Nicole Fisher, MPP, PhD, is the founder and chief executive officer at HHR Strategies, a health care and human rights focused advising firm. Additionally, she is a senior policy advisor and expert on health economics, technology, and reform, specifically as they affect vulnerable populations. Nicole runs a health innovation and policy page at Forbes, highlighting and advising companies, ideas, and people that are changing the health landscape. She also is currently pursuing her PhD at the University of North Carolina in the Health Policy and Management Department. Her writing has appeared in many publications and her talks can be found on the United Nations website. Before pursuing her PhD in health policy, Nicole earned her Master's degree in Public Policy from the University of Chicago and her undergraduate degree from the University of Missouri.


Assuntos
Atenção à Saúde/organização & administração , Planejamento em Saúde/organização & administração , Política de Saúde/tendências , Avaliação de Resultados em Cuidados de Saúde , Medo , Previsões , Humanos , Inovação Organizacional , Formulação de Políticas , Estados Unidos
18.
Sci Transl Med ; 9(403)2017 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-28814546

RESUMO

Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the methyl-CpG binding protein 2 (MECP2) gene. The cognitive impairments seen in mouse models of RTT correlate with deficits in long-term potentiation (LTP) at Schaffer collateral (SC)-CA1 synapses in the hippocampus. Metabotropic glutamate receptor 7 (mGlu7) is the predominant mGlu receptor expressed presynaptically at SC-CA1 synapses in adult mice, and its activation on GABAergic interneurons is necessary for induction of LTP. We demonstrate that pathogenic mutations in MECP2 reduce mGlu7 protein expression in brain tissue from RTT patients and in MECP2-deficient mouse models. In rodents, this reduction impairs mGlu7-mediated control of synaptic transmission. We show that positive allosteric modulation of mGlu7 activity restores LTP and improves contextual fear learning, novel object recognition, and social memory. Furthermore, mGlu7 positive allosteric modulation decreases apneas in Mecp2+/- mice, suggesting that mGlu7 may be a potential therapeutic target for multiple aspects of the RTT phenotype.


Assuntos
Cognição , Receptores de Glutamato Metabotrópico/metabolismo , Respiração , Síndrome de Rett/metabolismo , Síndrome de Rett/fisiopatologia , Comportamento Social , Animais , Apneia/tratamento farmacológico , Apneia/fisiopatologia , Autopsia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Córtex Motor/efeitos dos fármacos , Córtex Motor/metabolismo , Córtex Motor/patologia , Plasticidade Neuronal/efeitos dos fármacos , Fenótipo , Ácidos Picolínicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Respiração/efeitos dos fármacos , Síndrome de Rett/patologia , Transcrição Gênica/efeitos dos fármacos
19.
J Autism Dev Disord ; 46(10): 3369-76, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27465244

RESUMO

Neurofibromatosis type 1 (NF1) is an inherited neurocutaneous disorder associated with neurodevelopmental disorders including autism spectrum disorder (ASD). The frequency of ASD/NF1 co-occurrence has been subject to debate since the 1980s. This relationship was investigated in a large population-based sample of 8-year-old children identified with ASD (N = 12,271) by the Centers for Disease Control and Prevention's Autism and Developmental Disabilities Monitoring (ADDM) Network. Twenty-two (1-in-558) children with ASD had diagnosed NF1, exceeding NF1 general population estimates by four to five fold. Children with ASD/NF1 versus ASD without NF1 were significantly less likely to receive a community-based ASD diagnosis (p = 0.04) and understand non-verbal communication (p = 0.001). These findings underscore the importance of including social-communication ability among relevant developmental concerns in children with NF1.


Assuntos
Transtorno do Espectro Autista/diagnóstico , Transtorno do Espectro Autista/epidemiologia , Redes Comunitárias , Manual Diagnóstico e Estatístico de Transtornos Mentais , Neurofibromatose 1/diagnóstico , Neurofibromatose 1/epidemiologia , Centers for Disease Control and Prevention, U.S./normas , Criança , Redes Comunitárias/normas , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/epidemiologia , Feminino , Humanos , Masculino , Transtornos do Neurodesenvolvimento/diagnóstico , Transtornos do Neurodesenvolvimento/epidemiologia , Comunicação não Verbal , Prevalência , Estados Unidos/epidemiologia
20.
Public Health Genomics ; 18(1): 26-39, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25359560

RESUMO

BACKGROUND: Michigan's BioTrust for Health, a public health research biobank comprised of residual dried bloodspot (DBS) cards from newborn screening contains over 4 million samples collected without written consent. Participant-centric initiatives are IT tools that hold great promise to address the consent challenges in biobank research. METHODS: Working with Private Access Inc., a pioneer in patient-centric web solutions, we created and pilot tested a dynamic informed consent simulation, paired with an educational website, focusing on consent for research utilizing DBSs in Michigan's BioTrust for Health. RESULTS: Out of 187 pilot testers recruited in 2 groups, 137 completed the consent simulation and exit survey. Over 50% indicated their willingness to set up an account if the simulation went live and to recommend it to others. Participants raised concerns about the process of identity verification and appeared to have little experience with sharing health information online. CONCLUSIONS: Applying online, dynamic approaches to address the consent challenges raised by biobanks with legacy sample collections should be explored, given the positive reaction to our pilot test and the strong preference for active consent. Balancing security and privacy with accessibility and ease of use will continue to be a challenge.


Assuntos
Acesso à Informação/ética , Bancos de Espécimes Biológicos , Simulação por Computador , Confidencialidade/ética , Consentimento Livre e Esclarecido , Adulto , Bancos de Espécimes Biológicos/ética , Bancos de Espécimes Biológicos/organização & administração , Pesquisa Biomédica/ética , Pesquisa Biomédica/organização & administração , Feminino , Humanos , Recém-Nascido , Disseminação de Informação , Masculino , Michigan , Pessoa de Meia-Idade , Triagem Neonatal , Projetos Piloto , Manejo de Espécimes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA