Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Behav Brain Res ; 395: 112842, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32745660

RESUMO

Current estimates indicate that millions of people in the United States abuse opioid drugs, which may also affect their offspring. To determine whether parental exposure to morphine alters reward and affective behaviors in subsequent generations we exposed male and female C57BL/6NTac mice to morphine (75 mg) or placebo pellets for 4 weeks. Naïve mice were used as mating partners to create subsequent generations (F1 and F2). Adult male and female F1 and F2 mice were tested in the morphine conditioned place preference paradigm (CPP), marble burying (MB), acoustic startle response (ASR), and open field tests (OFT). Paternal morphine exposure resulted in significantly attenuated preference scores amongst F1 male offspring, but significantly higher preference scores amongst F1 female offspring at the lowest CPP dose tested (5 mg/kg). In contrast, maternal exposure to morphine did not affect morphine reward in the F1 generation; however, the F2 male offspring of morphine-exposed F0 females displayed significantly higher CPP preference scores. Preference scores in F2 females were not affected by F0 male or female morphine exposure. Sex-specific alterations in affective behaviors were observed only in the offspring of F0 males exposed to morphine with F1 males spending less time in the center of the open field and F1 females spending more time in the center of the open field. One generation later, affective behaviors were no longer altered in F2 males but F2 females from the F0 male morphine exposure buried more marbles in the MB test. In summary, early exposure to morphine in males and females causes lineage-specific inheritance of reward and affective behaviors.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Afeto/efeitos dos fármacos , Morfina/efeitos adversos , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Feminino , Masculino , Exposição Materna/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Comportamento Obsessivo , Exposição Paterna/efeitos adversos , Reflexo de Sobressalto/efeitos dos fármacos , Recompensa , Fatores Sexuais
2.
Stress ; 22(1): 142-150, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30457440

RESUMO

Chronic administration of nicotine or exposure to stress can produce long-lasting behavioral and physiological changes in humans and animals alike. Further, the impact of nicotine and stress exposure can be inherited by offspring to produce persistent changes in physiology and behavior. To determine if nicotine and stress interact across generations to influence offspring behavior we exposed F0 male mice to nicotine and F1 male and female mice to chronic unpredictable stress during adolescence. We then measured locomotor sensitization to repeated nicotine injections in the subsequent F2 and F3 generations. Stress exposure alone (F1) did not influence locomotor sensitization in any lineage. However, in the F1 male lineage, F0 nicotine exposure abrogated locomotor sensitization in F2 male and transiently enhanced locomotor sensitization in F2 female offspring. These effects were not passed down to the F3 generations or observed in the F1 female lineage. F1 stress exposure modulated the effects of prior F0 nicotine exposure in a sex-dependent manner. Specifically, stress blunted the nicotine-induced enhancement in locomotor sensitization observed in F2 female offspring of F1 males. The effect of F0 nicotine and F1 stress exposure in females appears to have skipped a generation and enhanced nicotine sensitization only in the F3 generation, and only in females. This novel multigenerational exposure paradigm examining the inheritance of two different environmental exposures demonstrates that nicotine responses can be modified by nicotine and stress exposure from previous generations, and these effects are strongly influenced by sex.


Assuntos
Nicotina/farmacologia , Estresse Psicológico/fisiopatologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Distribuição Aleatória
3.
PLoS One ; 13(5): e0196743, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29746501

RESUMO

BACKGROUND AND AIMS: Associated with numerous metabolic and behavioral abnormalities, obesity is classified by metrics reliant on body weight (such as body mass index). However, overnutrition is the common cause of obesity, and may independently contribute to these obesity-related abnormalities. Here, we use dietary challenges to parse apart the relative influence of diet and/or energy balance from body weight on various metabolic and behavioral outcomes. MATERIALS AND METHODS: Seventy male mice (mus musculus) were subjected to the diet switch feeding paradigm, generating groups with various body weights and energetic imbalances. Spontaneous activity patterns, blood metabolite levels, and unbiased gene expression of the nutrient-sensing ventral hypothalamus (using RNA-sequencing) were measured, and these metrics were compared using standardized multivariate linear regression models. RESULTS: Spontaneous activity patterns were negatively related to body weight (p<0.0001) but not diet/energy balance (p = 0.63). Both body weight and diet/energy balance predicted circulating glucose and insulin levels, while body weight alone predicted plasma leptin levels. Regarding gene expression within the ventral hypothalamus, only two genes responded to diet/energy balance (neuropeptide y [npy] and agouti-related peptide [agrp]), while others were related only to body weight. CONCLUSIONS: Collectively, these results demonstrate that individual components of obesity-specifically obesogenic diets/energy imbalance and elevated body mass-can have independent effects on metabolic and behavioral outcomes. This work highlights the shortcomings of using body mass-based indices to assess metabolic health, and identifies novel associations between blood biomarkers, neural gene expression, and animal behavior following dietary challenges.


Assuntos
Comportamento Animal/fisiologia , Peso Corporal/fisiologia , Ritmo Circadiano/fisiologia , Metabolismo Energético/fisiologia , Sono/fisiologia , Vigília/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Biomarcadores/sangue , Dieta/métodos , Ingestão de Energia/fisiologia , Expressão Gênica/fisiologia , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/metabolismo , Obesidade/sangue , Obesidade/metabolismo , Obesidade/fisiopatologia
4.
Neuropsychopharmacology ; 42(8): 1670-1678, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28094283

RESUMO

Adolescence is a time period in development when the brain undergoes substantial remodeling in response to the environment. To determine whether a stressful experience during adolescence affects adult behavior, we exposed adolescent male and female C57BL/6J mice to chronic unpredictable stress (CUS) for 12 days starting at postnatal day 28 (PND28). We also exposed adult male and female mice to CUS for 12 days beginning at PND70 to determine whether adolescence is a sensitive time period when stress can have long-lasting effects on behavior. Regardless of when mice were exposed to stress, they were all tested exactly 30 days later in the marble burying task, elevated zero maze, acoustic startle response, and forced swim test. Adolescent stress exposure increased anxiety-like behaviors in adult male and female mice and decreased acoustic startle response in a sex-dependent manner. However, adult stress exposure did not change anxiety or response to an acoustic tone in adult male or female mice as compared with nonstressed animals. Of interest, increased depression-like behavior in the forced swim test was observed in all mice, regardless of when the stress occurred. Gene expression analysis showed significant upregulation of corticotropin releasing factor receptor 2 (CrfR2) in the amygdala of males subjected to CUS during adolescence, but not in males that experienced CUS during adulthood. In contrast, females, regardless of when they were exposed to CUS, were not affected. These data support clinical evidence suggesting that early-life stress may predispose individuals to increased anxiety and depression later in life.


Assuntos
Envelhecimento/psicologia , Comportamento Animal , Período Crítico Psicológico , Estresse Psicológico/psicologia , Tonsila do Cerebelo/metabolismo , Animais , Feminino , Masculino , Camundongos , Receptores de Hormônio Liberador da Corticotropina/biossíntese , Reflexo de Sobressalto , Caracteres Sexuais
5.
Psychopharmacology (Berl) ; 233(23-24): 3991-4002, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27624598

RESUMO

RATIONALE: Prescription opioid abuse and transition to heroin use are growing problems in the USA. However, the long-term consequences of adolescent prescription opioid abuse on subsequent drug use and affective-like behavior are unknown. OBJECTIVES: This study aims to determine if adolescent exposure to oxycodone alters the rewarding effects of morphine, anxiety-like behavior, and reward-related gene expression later in adulthood. METHODS: Adolescent male C57Bl/6 mice were exposed to oxycodone (3 mg/kg/day) via osmotic minipumps for 28 days. Following a 28-day withdrawal period, mice were tested in morphine-conditioned place preference paradigm (CPP), morphine sensitization, open field, marble burying, and forced swim (FST) tests. To determine if effects were specific to adolescent exposure, adult mice were exposed to oxycodone for 28 days and underwent 28 days of withdrawal prior to the same behavioral testing schedule. Expression of reward-related genes including dopamine receptor 1 (D1) and dopamine transporter (DAT) in the nucleus accumbens (NAc) and ventral tegmental area (VTA) was examined. RESULTS: Adolescent oxycodone exposure significantly increased (300 %) response to morphine CPP during adulthood and significantly reduced D1 expression (30 %) in the NAc and DAT expression (75 %) in the VTA. Adult oxycodone exposure did not affect subsequent responses to morphine CPP. Oxycodone exposure did not affect the development of morphine sensitization or affective-like behaviors. Corticosterone response to a stressor (FST) was significantly reduced (65 %) in mice exposed to oxycodone during adolescence but not adulthood. CONCLUSIONS: Adolescent oxycodone exposure enhances rewarding effects of morphine in adulthood with no effect on other affective-like behaviors.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Morfina/farmacologia , Entorpecentes/farmacologia , Oxicodona/farmacologia , Recompensa , Análise de Variância , Animais , Ansiedade/etiologia , Comportamento Animal/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores Dopaminérgicos/metabolismo , Área Tegmentar Ventral/metabolismo
6.
Prog Biophys Mol Biol ; 118(1-2): 21-33, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25839742

RESUMO

Familial inheritance of drug abuse is composed of both genetic and environmental factors. Additionally, epigenetic transgenerational inheritance may provide a means by which parental drug use can influence several generations of offspring. Recent evidence suggests that parental drug exposure produces behavioral, biochemical, and neuroanatomical changes in future generations. The focus of this review is to discuss these multigenerational and transgenerational phenotypes in the offspring of animals exposed to drugs of abuse. Specifically, changes found following the administration of alcohol, opioids, cocaine, marijuana, and nicotine will be discussed. In addition, epigenetic modifications to the genome following administration of these drugs will be detailed as well as their potential for transmission to the next generation.


Assuntos
Álcoois/toxicidade , Analgésicos Opioides/toxicidade , Cannabis/toxicidade , Cocaína/toxicidade , Exposição Ambiental/efeitos adversos , Padrões de Herança , Nicotina/toxicidade , Transtornos Relacionados ao Uso de Substâncias/genética , Animais , Humanos
7.
J Pharmacol Exp Ther ; 349(2): 348-54, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24627467

RESUMO

Although nicotine mediates its effects through several nicotinic acetylcholine receptor (nAChR) subtypes, it remains to be determined which nAChR subtypes directly mediate heightened anxiety during withdrawal. Relative success in abstinence has been found with the nAChR partial agonist varenicline (Chantix; Pfizer, Groton, CT); however, treatment with this drug fails to alleviate anxiety in individuals during nicotine withdrawal. Therefore, it is hypothesized that success can be found by the repurposing of other nAChR partial agonists for cessation therapies that target anxiety. It is noteworthy that the selective partial agonists for α4ß2, ABT-089 [2-methyl-3-[2(S)-pyrrolidinylmethoxy]pyridine], and α7, ABT-107 [5-(6-[(3R)-1-azabicyclo[2.2.2]oct-3-yloxy] pyridazin-3-yl)-1H-indole] (AbbVie, North Chicago, IL), have not been evaluated as possible therapeutics for nicotine cessation. Therefore, we examined the effect of ABT-089 and ABT-107 on anxiety during withdrawal from nicotine in the novelty-induced hypophagia (NIH) paradigm. We found that short-term administration of ABT-089 and ABT-107 alleviate anxiety-like behavior during withdrawal from nicotine while long-term administration of ABT-089 but not ABT-107 reduces anxiety-like behavior during withdrawal. After behavioral testing, brains were harvested and ß2-containing nAChRs were measured using [(3)H]epibaditine. ABT-089 and ABT-107 do not upregulate nAChRs, which is in contrast to the upregulation of nAChRs observed after nicotine. Furthermore, ABT-089 is anxiogenic in nicotine naive animals, suggesting that the effects on anxiety are specifically related to the nicotine-dependent state. Together, these studies identify additional nAChR partial agonists that may aid in the rational development of smoking cessation aids.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Indóis/farmacologia , Nicotina/efeitos adversos , Agonistas Nicotínicos/farmacologia , Piridinas/farmacologia , Pirrolidinas/farmacologia , Quinuclidinas/farmacologia , Receptores Nicotínicos/metabolismo , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Animais , Ansiolíticos/uso terapêutico , Ansiedade/metabolismo , Ansiedade/psicologia , Encéfalo/metabolismo , Agonismo Parcial de Drogas , Indóis/uso terapêutico , Masculino , Camundongos , Agonistas Nicotínicos/uso terapêutico , Piridinas/uso terapêutico , Pirrolidinas/uso terapêutico , Quinuclidinas/uso terapêutico , Ensaio Radioligante , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/psicologia , Fatores de Tempo , Regulação para Cima
8.
BMC Urol ; 11: 19, 2011 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-21864408

RESUMO

BACKGROUND: The PTEN/Phosphatidylinositol 3'-kinase (PI3-kinase) growth factor signaling pathway plays a critical role in epithelial tumor development in a multitude of tissue types. Deletion of the Pten tumor suppressor gene in murine urothelial cells in vivo results in upregulation of cyclin-dependent kinase inhibitor p21. We have previously shown in mice that p21 expression blocks an increase in urothelial cell proliferation due to Pten deletion. In this study, we utilized human urothelial carcinoma cells UMUC-3 and UMUC-14 to identify the signaling pathways downstream of PI3-kinase that regulate p21. METHODS: Cells were treated with a combination of PI3-kinase stimulating growth factors and kinase inhibitors, or transfected with exogenous genes in order to identify the signaling events that are necessary for p21 induction. Mice with conditional deletion of Pten in bladder urothelium were also examined for evidence of PI3-kinase pathway signaling events that affect p21 expression. RESULTS: When cells were treated with PI3-kinase activating growth factors EGF or PDGF, we found that p21 levels increased, in a manner similar to that observed in mice. We used the inhibitors LY294002, Akti-1/2, and rapamycin, to show that p21 induction is dependent upon PI3-kinase and AKT activity, and partially dependent on mTOR. We treated the cells with proteasome inhibitor MG-132 and found that p21 may be degraded in the proteasome to regulate protein levels. Importantly, our findings show that GSK-3ß plays a role in diminishing p21 levels in cells. Treatment of cells with the GSK-3ß inhibitor SB-216763 increased p21 levels, while exogenous expression of GSK-3ß caused a decrease in p21, indicating that GSK-3ß actively reduces p21 levels. We found that a combined treatment of LY294002 and SB-216763 improved the cytotoxic effect against UMUC-3 and UMUC-14 carcinoma cells over LY294002 alone, suggesting potential therapeutic uses for GSK-3ß inhibitors. Immunohistochemical staining in bladders from wild-type and Pten-deleted mice indicated that GSK-3ß inhibitory phosphorylation increases when Pten is deleted. CONCLUSION: PI3-kinase and AKT cause an upregulation of p21 by suppressing GSK-3ß activity and activating mTOR in both cultured human urothelial carcinoma cells and mouse urothelial cells in vivo.


Assuntos
Carcinoma de Células de Transição/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Quinase 3 da Glicogênio Sintase/biossíntese , Fosfatidilinositol 3-Quinases/biossíntese , Serina-Treonina Quinases TOR/biossíntese , Neoplasias da Bexiga Urinária/metabolismo , Animais , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...