Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 14(8): e0220694, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31461461

RESUMO

A large number of genetic studies in yeast rely on the use of expression vectors. To facilitate the experimental approach of these studies, several collections of expression vectors have been generated (YXplac, pRS series, etc.). Subsequently, these collections have been expanded by adding more diversity to many of the plasmid features, including new selection markers and new promoter sequences. However, the ever growing number of plasmid features makes it unrealistic for research labs to maintain an up-to-date collection of plasmids. Here, we developed the COSPLAY toolbox: a Golden Gate approach based on the scheme of a simple modular plasmid that recapitulates and completes all the properties of the pRS plasmids. The COSPLAY toolbox contains a basal collection of individual functional modules. Moreover, we standardized a simple and rapid, software-assisted protocol which facilitates the addition of new personalized modules. Finally, our toolbox includes the possibility to select a genomic target location and to perform a single copy integration of the expression vector.


Assuntos
Clonagem Molecular/métodos , Vetores Genéticos/genética , Plasmídeos/genética , Saccharomyces cerevisiae/genética , Biblioteca Gênica , Genes Reporter , Engenharia Genética/métodos , Software , Transformação Genética
2.
Cell Rep ; 28(2): 408-422.e4, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31291577

RESUMO

Budding yeast cells undergo a limited number of divisions before they enter senescence and die. Despite recent mechanistic advances, whether and how molecular events are temporally and causally linked during the transition to senescence remain elusive. Here, using real-time observation of the accumulation of extrachromosomal rDNA circles (ERCs) in single cells, we provide evidence that ERCs build up rapidly with exponential kinetics well before any physiological decline. We then show that ERCs fuel a massive increase in ribosomal RNA (rRNA) levels in the nucleolus, which do not mature into functional ribosomes. This breakdown in nucleolar coordination is followed by a loss of nuclear homeostasis, thus defining a chronology of causally related events leading to cell death. A computational analysis supports a model in which a series of age-independent processes lead to an age-dependent increase in cell mortality, hence explaining the emergence of aging in budding yeast.


Assuntos
DNA Ribossômico/genética , Saccharomycetales/genética , Transcrição Gênica/genética , Senescência Celular , Homeostase
3.
Methods Cell Biol ; 147: 29-40, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30165960

RESUMO

The generation of complex temporal stress patterns may be instrumental to investigate the adaptive properties of individual cells submitted to environmental stress on physiological timescale. However, it is difficult to accurately control stress concentration over time in bulk experiments. Here, we describe a microfluidics-based protocol to induce tightly controllable H2O2 stress in budding yeast while constantly monitoring cell growth with single cell resolution over multi-generation timescale. Moreover, we describe a simple methodology to produce ramping H2O2 stress to investigate the homeostatic properties of the H2O2 scavenging system.


Assuntos
Técnicas Analíticas Microfluídicas/métodos , Estresse Mecânico , Dimetilpolisiloxanos/química , Peróxido de Hidrogênio/química , Fatores de Tempo
4.
Elife ; 72018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29972352

RESUMO

Coordination of cell growth with division is essential for proper cell function. In budding yeast, although some molecular mechanisms responsible for cell size control during G1 have been elucidated, the mechanism by which cell size homeostasis is established remains to be discovered. Here, we developed a new technique based on quantification of histone levels to monitor cell cycle progression in individual cells with unprecedented accuracy. Our analysis establishes the existence of a mechanism controlling bud size in G2/M that prevents premature onset of anaphase, and controls the overall size variability. While most G1 mutants do not display impaired size homeostasis, mutants in which cyclin B-Cdk regulation is altered display large size variability. Our study thus demonstrates that size homeostasis is not controlled by a G1-specific mechanism alone but is likely to be an emergent property resulting from the integration of several mechanisms that coordinate cell and bud growth with division.


Assuntos
Ciclo Celular , Homeostase , Saccharomyces cerevisiae/citologia , Anáfase , Ciclo Celular/genética , Ciclina B/metabolismo , Fluorescência , Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Histonas/biossíntese , Hidroxiureia/farmacologia , Metáfase , Viabilidade Microbiana , Microfluídica , Modelos Biológicos , Mutação/genética , Saccharomyces cerevisiae/genética , Imagem com Lapso de Tempo
5.
Elife ; 62017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28418333

RESUMO

Homeostatic systems that rely on genetic regulatory networks are intrinsically limited by the transcriptional response time, which may restrict a cell's ability to adapt to unanticipated environmental challenges. To bypass this limitation, cells have evolved mechanisms whereby exposure to mild stress increases their resistance to subsequent threats. However, the mechanisms responsible for such adaptive homeostasis remain largely unknown. Here, we used live-cell imaging and microfluidics to investigate the adaptive response of budding yeast to temporally controlled H2O2 stress patterns. We demonstrate that acquisition of tolerance is a systems-level property resulting from nonlinearity of H2O2 scavenging by peroxiredoxins and our study reveals that this regulatory scheme induces a striking hormetic effect of extracellular H2O2 stress on replicative longevity. Our study thus provides a novel quantitative framework bridging the molecular architecture of a cellular homeostatic system to the emergence of nonintuitive adaptive properties.


Assuntos
Retroalimentação , Peróxido de Hidrogênio/toxicidade , Oxidantes/toxicidade , Estresse Oxidativo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/fisiologia , Estresse Fisiológico , Microscopia Intravital , Microfluídica , Imagem Óptica
6.
Neurosci Lett ; 643: 16-21, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28192197

RESUMO

Oxymorphone, one of oxycodone's metabolic products, is a potent opioid receptor agonist which is thought to contribute to the analgesic effect of its parent compound and may have high potential abuse liability. Nonetheless, the in vivo pharmacological binding profile of this drug is still unclear. This study uses mice lacking mu (MOP), kappa (KOP) or delta (DOP) opioid receptors as well as mice lacking all three opioid receptors to provide full characterisation of oxymorphone binding sites in the brain. Saturation binding studies using [3H]oxymorphone revealed high affinity binding sites in mouse brain displaying Kd of 1.7nM and Bmax of 147fmol/mg. Furthermore, we performed quantitative autoradiography binding studies using [3H]oxymorphone in mouse brain. The distribution of [3H]oxymorphone binding sites was found to be similar to the selective MOP agonist [3H]DAMGO in the mouse brain. [3H]Oxymorphone binding was completely abolished across the majority of the brain regions in mice lacking MOP as well as in mice lacking all three opioid receptors. DOP and KOP knockout mice retained [3H]oxymorphone binding sites suggesting oxymorphone may not target DOP or KOP. These results confirm that the MOP, and not the DOP or the KOP is the main high affinity binding target for oxymorphone.


Assuntos
Encéfalo/metabolismo , Oximorfona/farmacologia , Receptores Opioides/metabolismo , Animais , Autorradiografia/métodos , Sítios de Ligação , Camundongos Knockout , Receptores Opioides/deficiência , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
7.
Biol Psychiatry ; 81(9): 778-788, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28185645

RESUMO

BACKGROUND: Mu opioid receptors (MORs) are central to pain control, drug reward, and addictive behaviors, but underlying circuit mechanisms have been poorly explored by genetic approaches. Here we investigate the contribution of MORs expressed in gamma-aminobutyric acidergic forebrain neurons to major biological effects of opiates, and also challenge the canonical disinhibition model of opiate reward. METHODS: We used Dlx5/6-mediated recombination to create conditional Oprm1 mice in gamma-aminobutyric acidergic forebrain neurons. We characterized the genetic deletion by histology, electrophysiology, and microdialysis; probed neuronal activation by c-Fos immunohistochemistry and resting-state functional magnetic resonance imaging; and investigated main behavioral responses to opiates, including motivation to obtain heroin and palatable food. RESULTS: Mutant mice showed MOR transcript deletion mainly in the striatum. In the ventral tegmental area, local MOR activity was intact, and reduced activity was only observed at the level of striatonigral afferents. Heroin-induced neuronal activation was modified at both sites, and whole-brain functional networks were altered in live animals. Morphine analgesia was not altered, and neither was physical dependence to chronic morphine. In contrast, locomotor effects of heroin were abolished, and heroin-induced catalepsy was increased. Place preference to heroin was not modified, but remarkably, motivation to obtain heroin and palatable food was enhanced in operant self-administration procedures. CONCLUSIONS: Our study reveals dissociable MOR functions across mesocorticolimbic networks. Thus, beyond a well-established role in reward processing, operating at the level of local ventral tegmental area neurons, MORs also moderate motivation for appetitive stimuli within forebrain circuits that drive motivated behaviors.


Assuntos
Comportamento Alimentar/fisiologia , Neurônios GABAérgicos/fisiologia , Heroína/administração & dosagem , Motivação/fisiologia , Entorpecentes/administração & dosagem , Prosencéfalo/fisiologia , Receptores Opioides mu/fisiologia , Animais , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/fisiologia , Feminino , Neurônios GABAérgicos/metabolismo , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Morfina/administração & dosagem , Motivação/efeitos dos fármacos , Vias Neurais/fisiologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Receptores Opioides mu/genética , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiologia
8.
Biophys J ; 110(7): 1605-1614, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27074685

RESUMO

Budding yeast cells have a finite replicative life span; that is, a mother cell produces only a limited number of daughter cells before it slows division and dies. Despite the gradual aging of the mother cell, all daughters are born rejuvenated and enjoy a full replicative lifespan. It has been proposed that entry of mother cells into senescence is driven by the progressive accumulation and retention of damaged material, including protein aggregates. This additionally allows the daughter cells to be born damage free. However, the mechanism underlying such asymmetrical segregation of protein aggregates by mother and daughter cells remains controversial, in part because of the difficulties inherent in tracking the dynamics and fate of protein aggregates in vivo. To overcome such limitations, we have developed single-cell real-time imaging methodology to track the formation of heat-induced protein aggregates in otherwise unperturbed dividing cells. By combining the imaging data with a simple computational model of protein aggregation, we show that the establishment of asymmetrical partitioning of protein aggregates upon division is driven by the large bud-specific dilution rate associated with polarized growth and the absence of significant mother/bud exchange of protein aggregates during the budded phase of the cell cycle. To our knowledge, this study sheds new light on the mechanism of establishment of a segregation bias, which can be accounted for by simple physical arguments.


Assuntos
Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Agregados Proteicos , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Divisão Celular , Cinética , Transporte Proteico , Saccharomyces cerevisiae/citologia , Temperatura
9.
Biol Psychiatry ; 77(4): 404-15, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25444168

RESUMO

BACKGROUND: The delta opioid receptor (DOR) is broadly expressed throughout the nervous system; it regulates chronic pain, emotional responses, motivation, and memory. Neural circuits underlying DOR activities have been poorly explored by genetic approaches. We used conditional mouse mutagenesis to elucidate receptor function in GABAergic neurons of the forebrain. METHODS: We characterized DOR distribution in the brain of Dlx5/6-CreXOprd1(fl/fl) (Dlx-DOR) mice and tested main central DOR functions through behavioral testing. RESULTS: The DOR proteins were strongly deleted in olfactory bulb and striatum and remained intact in cortex and basolateral amygdala. Olfactory perception, circadian activity, and despair-like behaviors were unchanged. In contrast, locomotor stimulant effects of SNC80 (DOR agonist) and SKF81297 (D1 agonist) were abolished and increased, respectively. The Dlx-DOR mice showed lower levels of anxiety in the elevated plus maze, opposing the known high anxiety in constitutive DOR knockout animals. Also, Dlx-DOR mice reached the food more rapidly in a novelty suppressed feeding task, despite their lower motivation for food reward observed in an operant paradigm. Finally, c-fos protein staining after novelty suppressed feeding was strongly reduced in amygdala, concordant with the low anxiety phenotype of Dlx-DOR mice. CONCLUSIONS: We demonstrate that DORs expressed in the forebrain mediate the described locomotor effect of SNC80 and inhibit D1-stimulated hyperactivity. Our data also reveal an unanticipated anxiogenic role for this particular DOR subpopulation, with a potential novel adaptive role. In emotional responses, DORs exert dual anxiolytic and anxiogenic roles, both of which may have implications in the area of anxiety disorders.


Assuntos
Ansiedade/fisiopatologia , Neurônios GABAérgicos/metabolismo , Prosencéfalo/metabolismo , Receptores Opioides delta/metabolismo , Animais , Comportamento Animal/fisiologia , Benzamidas/farmacologia , Benzazepinas/farmacologia , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Agonistas de Dopamina/farmacologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Motivação/fisiologia , Atividade Motora/efeitos dos fármacos , Bulbo Olfatório/metabolismo , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo , Receptores de Dopamina D1/agonistas , Receptores Opioides delta/agonistas , Receptores Opioides delta/análise , Receptores Opioides delta/genética
10.
Behav Brain Res ; 278: 429-34, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25447299

RESUMO

The delta opioid receptor (DOR) has raised much interest for the development of new therapeutic drugs, particularly to treat patients suffering from mood disorders and chronic pain. Unfortunately, the prototypal DOR agonist SNC80 induces mild epileptic seizures in rodents. Although recently developed agonists do not seem to show convulsant properties, mechanisms and neuronal circuits that support DOR-mediated epileptic seizures remain to be clarified. DORs are expressed throughout the nervous system. In this study we tested the hypothesis that SNC80-evoked seizures stem from DOR activity at the level of forebrain GABAergic transmission, whose inhibition is known to facilitate the development of epileptic seizures. We generated a conditional DOR knockout mouse line, targeting the receptor gene specifically in GABAergic neurons of the forebrain (Dlx-DOR). We measured effects of SNC80 (4.5, 9, 13.5 and 32 mg/kg), ARM390 (10, 30 and 60 mg/kg) or ADL5859 (30, 100 and 300 mg/kg) administration on electroencephalograms (EEGs) recorded in Dlx-DOR mice and their control littermates (Ctrl mice). SNC80 produced dose-dependent seizure events in Ctrl mice, but these effects were not detected in Dlx-DOR mice. As expected, ARM390 and ADL5859 did not trigger any detectable change in mice from both genotypes. These results demonstrate for the first time that SNC80-induced DOR activation induces epileptic seizures via direct inhibition of GABAergic forebrain neurons, and supports the notion of differential activities between first and second-generation DOR agonists.


Assuntos
Analgésicos Opioides/toxicidade , Benzamidas/toxicidade , Neurônios GABAérgicos/metabolismo , Piperazinas/toxicidade , Prosencéfalo/patologia , Receptores Opioides delta/metabolismo , Convulsões , Animais , Benzamidas/farmacologia , Benzopiranos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Eletroencefalografia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Knockout , Prosencéfalo/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Receptores Opioides delta/genética , Convulsões/induzido quimicamente , Convulsões/genética , Convulsões/patologia
11.
Brain Struct Funct ; 220(2): 677-702, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24623156

RESUMO

Opioid receptors are G protein-coupled receptors (GPCRs) that modulate brain function at all levels of neural integration, including autonomic, sensory, emotional and cognitive processing. Mu (MOR) and delta (DOR) opioid receptors functionally interact in vivo, but whether interactions occur at circuitry, cellular or molecular levels remains unsolved. To challenge the hypothesis of MOR/DOR heteromerization in the brain, we generated redMOR/greenDOR double knock-in mice and report dual receptor mapping throughout the nervous system. Data are organized as an interactive database offering an opioid receptor atlas with concomitant MOR/DOR visualization at subcellular resolution, accessible online. We also provide co-immunoprecipitation-based evidence for receptor heteromerization in these mice. In the forebrain, MOR and DOR are mainly detected in separate neurons, suggesting system-level interactions in high-order processing. In contrast, neuronal co-localization is detected in subcortical networks essential for survival involved in eating and sexual behaviors or perception and response to aversive stimuli. In addition, potential MOR/DOR intracellular interactions within the nociceptive pathway offer novel therapeutic perspectives.


Assuntos
Encéfalo/metabolismo , Rede Nervosa/metabolismo , Neurônios/metabolismo , Receptores Opioides delta/análise , Receptores Opioides mu/análise , Animais , Feminino , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Neuropsychopharmacology ; 39(11): 2694-705, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24874714

RESUMO

Addiction is a chronic disorder involving recurring intoxication, withdrawal, and craving episodes. Escaping this vicious cycle requires maintenance of abstinence for extended periods of time and is a true challenge for addicted individuals. The emergence of depressive symptoms, including social withdrawal, is considered a main cause for relapse, but underlying mechanisms are poorly understood. Here we establish a mouse model of protracted abstinence to heroin, a major abused opiate, where both emotional and working memory deficits unfold. We show that delta and kappa opioid receptor (DOR and KOR, respectively) knockout mice develop either stronger or reduced emotional disruption during heroin abstinence, establishing DOR and KOR activities as protective and vulnerability factors, respectively, that regulate the severity of abstinence. Further, we found that chronic treatment with the antidepressant drug fluoxetine prevents emergence of low sociability, with no impact on the working memory deficit, implicating serotonergic mechanisms predominantly in emotional aspects of abstinence symptoms. Finally, targeting the main serotonergic brain structure, we show that gene knockout of mu opioid receptors (MORs) in the dorsal raphe nucleus (DRN) before heroin exposure abolishes the development of social withdrawal. This is the first result demonstrating that intermittent chronic MOR activation at the level of DRN represents an essential mechanism contributing to low sociability during protracted heroin abstinence. Altogether, our findings reveal crucial and distinct roles for all three opioid receptors in the development of emotional alterations that follow a history of heroin exposure and open the way towards understanding opioid system-mediated serotonin homeostasis in heroin abuse.


Assuntos
Dependência de Heroína/fisiopatologia , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Comportamento Social , Síndrome de Abstinência a Substâncias/fisiopatologia , Animais , Antidepressivos de Segunda Geração/farmacologia , Depressão/metabolismo , Modelos Animais de Doenças , Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/metabolismo , Fluoxetina/farmacologia , Heroína/farmacologia , Dependência de Heroína/psicologia , Masculino , Transtornos da Memória/fisiopatologia , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Entorpecentes/farmacologia , Receptores Opioides kappa/genética , Receptores Opioides mu/genética , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/psicologia
13.
Eur J Pharmacol ; 731: 1-7, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24657279

RESUMO

Several methodological approaches suggest that receptor heteromers exist in cell systems, but their presence in physiological tissue is widely contentious. We describe a novel method to determine if heterodimers exist in brain tissue sections using autoradiographic binding comparisons from single and double gene knockout mice, where tissues either have a full receptor complement and can form heterodimers, or are incapable of making heterodimers. We have tested this model, which we have named Knockout Subtraction Autoradiography, to determine if heterodimerisation of the kappa (KOP) and delta opioid (DOP) receptors occurs, as evidence from binding studies in cell systems suggest they are present in the brain. Using labeling of putative KOP receptor/DOP receptor heterodimers with either [(3)H]bremazocine or with [(3)H]naltrindole, two ligands which were used to provide evidence suggesting that these opioid receptor subtypes heterodimerize, we have applied a subtraction equation model based on the principle that receptor gene double knockout of either MOP receptor/KOP receptor (DOP receptor expression only) or MOP receptor/DOP receptor (KOP receptor expression only) produces tissue incapable of making the KOP receptor/DOP receptor heterodimer. We have shown in most brain regions that the labeling fits a simple additive model of monomer labeling, but that in a few brain regions opioid receptor heterodimerization does occur. The data does not support the conclusion that KOP receptor/DOP receptor heterodimerisation is widespread in the central nervous system, but does indicate that this novel methodology can detect heterodimerisation, when ligands with distinct binding affinities for monomer and heterodimer forms exist.


Assuntos
Autorradiografia/métodos , Encéfalo/metabolismo , Técnicas de Inativação de Genes , Multimerização Proteica , Receptores Opioides delta/química , Receptores Opioides kappa/química , Técnica de Subtração , Animais , Benzomorfanos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Naltrexona/análogos & derivados , Naltrexona/metabolismo , Estrutura Quaternária de Proteína , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética
14.
PLoS One ; 8(9): e74706, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24069332

RESUMO

Opiates are powerful drugs to treat severe pain, and act via mu opioid receptors distributed throughout the nervous system. Their clinical use is hampered by centrally-mediated adverse effects, including nausea or respiratory depression. Here we used a genetic approach to investigate the potential of peripheral mu opioid receptors as targets for pain treatment. We generated conditional knockout (cKO) mice in which mu opioid receptors are deleted specifically in primary afferent Nav1.8-positive neurons. Mutant animals were compared to controls for acute nociception, inflammatory pain, opiate-induced analgesia and constipation. There was a 76% decrease of mu receptor-positive neurons and a 60% reduction of mu-receptor mRNA in dorsal root ganglia of cKO mice. Mutant mice showed normal responses to heat, mechanical, visceral and chemical stimuli, as well as unchanged morphine antinociception and tolerance to antinociception in models of acute pain. Inflammatory pain developed similarly in cKO and controls mice after Complete Freund's Adjuvant. In the inflammation model, however, opiate-induced (morphine, fentanyl and loperamide) analgesia was reduced in mutant mice as compared to controls, and abolished at low doses. Morphine-induced constipation remained intact in cKO mice. We therefore genetically demonstrate for the first time that mu opioid receptors partly mediate opiate analgesia at the level of Nav1.8-positive sensory neurons. In our study, this mechanism operates under conditions of inflammatory pain, but not nociception. Previous pharmacology suggests that peripheral opiates may be clinically useful, and our data further demonstrate that Nav1.8 neuron-associated mu opioid receptors are feasible targets to alleviate some forms of persistent pain.


Assuntos
Analgesia , Analgésicos Opioides/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Receptores Opioides mu/genética , Animais , Constipação Intestinal/induzido quimicamente , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Deleção de Genes , Expressão Gênica , Técnicas de Inativação de Genes , Ordem dos Genes , Marcação de Genes , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Camundongos , Camundongos Knockout , Morfina/efeitos adversos , Morfina/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Nociceptividade/efeitos dos fármacos , Dor/tratamento farmacológico , Dor/genética , Medição da Dor , Ligação Proteica , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Limiar Sensorial/efeitos dos fármacos
15.
J Biotechnol ; 166(4): 182-6, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-23732834

RESUMO

In vivo conditional knock-out of a protein is a method of choice to decipher its biological function. It can be achieved by encoding the cre-recombinase on a recombinant virus to exert spatio-temporal control of its expression and enzymatic activity and, subsequently, of the target gene deletion. Recombinant baculoviruses have been successfully used to express a wide range of proteins in insect cells. More recently, their potential to infect mammalian cells has been addressed but, so far, their ability to yield a conditional knock-out as a result of efficient in vivo cre-recombinase gene delivery has not been examined. Cre-recombinase fused to the green fluorescent protein was cloned under the control of the CAG promoter in a recombinant Autographa californica baculovirus expressing the vesicular stomatitis virus envelope G protein for increased mammalian cell infection. Gene delivery was evaluated in vitro in mammalian cells, neuroblastoma and mouse primary neuronal cultures as well as in vivo in the mouse brain. Infection with adeno-associated viruses encoding the cre-recombinase fused to the green fluorescent protein was performed as a positive control. Our results indicate that baculovirus infection leads to functional cre-recombinase expression in non-neuronal and neuroblastoma cell lines but not in mouse primary neuronal cultures or brain.


Assuntos
Baculoviridae , Técnicas de Transferência de Genes , Integrases/genética , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Dependovirus/genética , Dependovirus/metabolismo , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Camundongos , Infecções por Parvoviridae/genética
16.
J Neurosci ; 32(21): 7301-10, 2012 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-22623675

RESUMO

G-protein-coupled receptors (GPCRs) mediate numerous physiological functions and represent prime therapeutic targets. Receptor trafficking upon agonist stimulation is critical for GPCR function, but examining this process in vivo remains a true challenge. Using knock-in mice expressing functional fluorescent delta opioid receptors under the control of the endogenous promoter, we visualized in vivo internalization of this native GPCR upon physiological stimulation. We developed a paradigm in which animals were made dependent on morphine in a drug-paired context. When re-exposed to this context in a drug-free state, mice showed context-dependent withdrawal signs and activation of the hippocampus. Receptor internalization was transiently detected in a subset of CA1 neurons, uncovering regionally restricted opioid peptide release. Importantly, a pool of surface receptors always remained, which contrasts with the in vivo profile previously established for exogenous drug-induced internalization. Therefore, a distinct response is observed at the receptor level upon a physiological or pharmacological stimulation. Altogether, direct in vivo GPCR visualization enables mapping receptor stimulation promoted by a behavioral challenge and represents a powerful approach to study endogenous GPCR physiology.


Assuntos
Hipocampo/metabolismo , Transporte Proteico , Receptores Opioides delta/metabolismo , Animais , Encefalina Metionina/metabolismo , Feminino , Técnicas de Introdução de Genes , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Imagem Molecular , Morfina/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Síndrome de Abstinência a Substâncias/metabolismo
17.
Psychopharmacology (Berl) ; 223(1): 99-106, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22526530

RESUMO

RATIONALE: The exact role of delta opioid receptors in drug-induced conditioned place preference (CPP) remains debated. Under classical experimental conditions, morphine-induced CPP is decreased in mice lacking delta opioid receptors (Oprd1 (-/-)). Morphine self-administration, however, is maintained, suggesting that drug-context association rather than drug reward is deficient in these animals. OBJECTIVES: This study further examined the role of delta opioid receptors in mediating drug-cue associations, which are necessary for the expression of morphine-induced CPP. METHODS: We first identified experimental conditions under which Oprd1 (-/-) mice are able to express CPP to morphine (5, 10 or 20 mg/kg) in a drug-free state and observed that, in this paradigm, CPP was dependent on circadian time conditions. We then took advantage of this particularity to assess the ability of various cues (internal or discrete), predicting either drug or food reward, to restore CPP induced by morphine (10 mg/kg) in Oprd1 (-/-) mice in conditions under which they normally fail to express CPP. RESULTS: We found that presentation of circadian, drug or auditory cues, predicting morphine or food reward, restored morphine CPP in Oprd1 (-/-) mice, which then performed as well as control mice. CONCLUSIONS: This study reveals that, in contrast to spatial cues, internal or discrete morphine-predicting stimuli permit full expression of morphine CPP in Oprd1 (-/-) mice. Delta receptors, therefore, appear to play a crucial role in modulating spatial contextual cue-related responses. This activity may be critical when context gains control over behavior, as is the case for context-induced relapse in drug abuse.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Morfina/farmacologia , Receptores Opioides delta/genética , Recompensa , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Ritmo Circadiano , Sinais (Psicologia) , Relação Dose-Resposta a Droga , Comportamento Alimentar , Feminino , Masculino , Camundongos , Camundongos Knockout , Morfina/administração & dosagem
18.
J Neuroimmune Pharmacol ; 6(4): 608-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21938479

RESUMO

Here, we evaluated the influence of endogenous opioid activation on immune responses by examining consequences of all three opioid receptor gene (mu, delta and kappa) inactivation. In triple-opioid receptor knockout mice, splenocytes and thymocytes numbers, lymphocyte subsets as well as proliferation and cytokines induced by in vitro stimulation of T lymphocytes were measured. Compared with wild-type mice, similar lymphocyte distribution in thymus and spleen as well as comparable T lymphocyte proliferation were observed, while lower levels of IL-2 and IFNγ as well as higher levels of IL-4 and IL-10 were found in triple-opioid receptor knockout mice. Together, our results indicate a shift from TH1 to TH2 cytokines in triple-opioid receptor knockout animals, suggesting that global endogenous opioid tone drives T lymphocytes toward a TH1 profile under non-pathological conditions.


Assuntos
Peptídeos Opioides/imunologia , Receptores Opioides delta/imunologia , Receptores Opioides kappa/imunologia , Receptores Opioides mu/imunologia , Células Th1/imunologia , Animais , Citocinas/biossíntese , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Opioides delta/deficiência , Receptores Opioides delta/genética , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Timo/citologia , Timo/imunologia
19.
Pain ; 152(6): 1238-1248, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21295407

RESUMO

Opioid receptors are major actors in pain control and are broadly distributed throughout the nervous system. A major challenge in pain research is the identification of key opioid receptor populations within nociceptive pathways, which control physiological and pathological pain. In particular, the respective contribution of peripheral vs. central receptors remains unclear, and it has not been addressed by genetic approaches. To investigate the contribution of peripheral delta opioid receptors in pain control, we created conditional knockout mice where delta receptors are deleted specifically in peripheral Na(V)1.8-positive primary nociceptive neurons. Mutant mice showed normal pain responses to acute heat and to mechanical and formalin stimuli. In contrast, mutant animals showed a remarkable increase of mechanical allodynia under both inflammatory pain induced by complete Freund adjuvant and neuropathic pain induced by partial sciatic nerve ligation. In these 2 models, heat hyperalgesia was virtually unchanged. SNC80, a delta agonist administered either systemically (complete Freund adjuvant and sciatic nerve ligation) or into a paw (sciatic nerve ligation), reduced thermal hyperalgesia and mechanical allodynia in control mice. However, these analgesic effects were absent in conditional mutant mice. In conclusion, this study reveals the existence of delta opioid receptor-mediated mechanisms, which operate at the level of Na(V)1.8-positive nociceptive neurons. Delta receptors in these neurons tonically inhibit mechanical hypersensitivity in both inflammatory and neuropathic pain, and they are essential to mediate delta opioid analgesia under conditions of persistent pain. This delta receptor population represents a feasible therapeutic target to alleviate chronic pain while avoiding adverse central effects. The conditional knockout of delta-opioid receptor in primary afferent Na(V)1.8 neurons augmented mechanical allodynia in persistent pain models and abolished delta opioid analgesia in these models.


Assuntos
Gânglios Espinais/patologia , Nociceptores/fisiologia , Dor/genética , Dor/patologia , Receptores Opioides delta/deficiência , Analgésicos Opioides/uso terapêutico , Análise de Variância , Animais , Benzamidas/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Adjuvante de Freund/efeitos adversos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Inflamação/induzido quimicamente , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Canal de Sódio Disparado por Voltagem NAV1.8 , Nociceptores/efeitos dos fármacos , Dor/etiologia , Medição da Dor/métodos , Piperazinas/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Canais de Sódio/genética , Canais de Sódio/metabolismo , Isótopos de Enxofre/farmacocinética
20.
Biol Psychiatry ; 69(7): 700-3, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21168121

RESUMO

BACKGROUND: Converging experimental data indicate that δ opioid receptors contribute to mediate drug reinforcement processes. Whether their contribution reflects a role in the modulation of drug reward or an implication in conditioned learning, however, has not been explored. In the present study, we investigated the impact of δ receptor gene knockout on reinforced conditioned learning under several experimental paradigms. METHODS: We assessed the ability of δ receptor knockout mice to form drug-context associations with either morphine (appetitive)- or lithium (aversive)-induced Pavlovian place conditioning. We also examined the efficiency of morphine to serve as a positive reinforcer in these mice and their motivation to gain drug injections, with operant intravenous self-administration under fixed and progressive ratio schedules and at two different doses. RESULTS: Mutant mice showed impaired place conditioning in both appetitive and aversive conditions, indicating disrupted context-drug association. In contrast, mutant animals displayed intact acquisition of morphine self-administration and reached breaking-points comparable to control subjects. Thus, reinforcing effects of morphine and motivation to obtain the drug were maintained. CONCLUSION: Collectively, the data suggest that δ receptor activity is not involved in morphine reinforcement but facilitates place conditioning. This study reveals a novel aspect of δ opioid receptor function in addiction-related behaviors.


Assuntos
Analgésicos Opioides/administração & dosagem , Condicionamento Operante/fisiologia , Deficiências da Aprendizagem/genética , Morfina/administração & dosagem , Receptores Opioides delta/deficiência , Reforço Psicológico , Animais , Comportamento Apetitivo/efeitos dos fármacos , Comportamento Animal , Condicionamento Operante/efeitos dos fármacos , Discriminação Psicológica/efeitos dos fármacos , Relação Dose-Resposta a Droga , Reação de Fuga/efeitos dos fármacos , Deficiências da Aprendizagem/fisiopatologia , Camundongos , Camundongos Knockout , Motivação/efeitos dos fármacos , Autoadministração , Percepção Espacial/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...