Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Neuroendocrinology ; 113(11): 1177-1188, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37348474

RESUMO

INTRODUCTION: Women are more vulnerable to cocaine's reinforcing effects and have a more rapid course to addiction after initial cocaine use as compared to men. Studies in rodents similarly indicate an enhanced sensitivity to the reinforcing effects of cocaine in females versus males. Levels of estradiol (E2) are correlated with vulnerability to the rewarding actions of cocaine. Here, we asked if sex chromosome complement (SCC) influences vulnerability to cocaine use. METHODS: We used the four-core genotype mouse that produces gonadal males and females with either XX or XY SCC. Mice were gonadectomized and implanted with either an estradiol (E2) or cholesterol-filled pellet. This allowed us to determine the effects of SCC in the absence (cholesterol-treated) and presence of tonic high physiological hormone levels (estradiol). Acquisition of cocaine self-administration was determined over a 12-day period using an escalated dose procedure (0.3 mg/kg/infusion, sessions 1-6; 0.6 mg/kg/infusion, sessions 6-12). RESULTS: Without estradiol treatment, a greater percentage of castrated XY mice acquired cocaine self-administration and did so at a faster rate than XX castrates and ovariectomized XY females. These same XY males acquired sooner, infused more cocaine, and directed more nose pokes to the rewarded nose-poke hole than XX castrates and XY males receiving E2. CONCLUSION: Our results suggest that in gonadal male mice, SCC and estradiol can modulate the reinforcing effects of cocaine which may influence the likelihood of cocaine use.


Assuntos
Cocaína , Humanos , Camundongos , Animais , Masculino , Feminino , Estradiol/farmacologia , Cromossomos Sexuais , Genótipo , Colesterol
4.
Pharmacol Rev ; 75(2): 217-249, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36781217

RESUMO

Sex/gender effects have been demonstrated for multiple aspects of addiction, with one of the most commonly cited examples being the "telescoping effect" where women meet criteria and/or seek treatment of substance use disorder (SUD) after fewer years of drug use as compared with men. This phenomenon has been reported for multiple drug classes including opioids, psychostimulants, alcohol, and cannabis, as well as nonpharmacological addictions, such as gambling. However, there are some inconsistent reports that show either no difference between men and women or opposite effects and a faster course to addiction in men than women. Thus, the goals of this review are to evaluate evidence for and against the telescoping effect in women and to determine the conditions/populations for which the telescoping effect is most relevant. We also discuss evidence from preclinical studies, which strongly support the validity of the telescoping effect and show that female animals develop addiction-like features (e.g., compulsive drug use, an enhanced motivation for the drug, and enhanced drug-craving/vulnerability to relapse) more readily than male animals. We also discuss biologic factors that may contribute to the telescoping effect, such as ovarian hormones, and its neurobiological basis focusing on the mesolimbic dopamine reward pathway and the corticomesolimbic glutamatergic pathway considering the critical roles these pathways play in the rewarding/reinforcing effects of addictive drugs and SUD. We conclude with future research directions, including intervention strategies to prevent the development of SUD in women. SIGNIFICANCE STATEMENT: One of the most widely cited gender/sex differences in substance use disorder (SUD) is the "telescoping effect," which reflects an accelerated course in women versus men for the development and/or seeking treatment for SUD. This review evaluates evidence for and against a telescoping effect drawing upon data from both clinical and preclinical studies. We also discuss the contribution of biological factors and underlying neurobiological mechanisms and highlight potential targets to prevent the development of SUD in women.


Assuntos
Comportamento Aditivo , Jogo de Azar , Transtornos Relacionados ao Uso de Substâncias , Animais , Masculino , Feminino , Fatores Sexuais , Recompensa
5.
Physiol Behav ; 243: 113630, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34710466

RESUMO

An increase in opioid-overdose deaths was evident before the COVID-19 pandemic, and has escalated since its onset. Fentanyl, a highly potent synthetic opioid, is the primary driver of these recent trends. The current study used two inbred mouse strains, C57BL/6 J and A/J, to investigate the genetics of behavioral responses to fentanyl. Mice were tested for conditioned place preference and fentanyl-induced locomotor activity. C57BL/6J mice formed a conditioned place preference to fentanyl injections and fentanyl increased their activity. Neither effect was noted in A/J mice. We conducted RNA-sequencing on the nucleus accumbens of mice used for fentanyl-induced locomotor activity. Surprisingly, we noted few differentially expressed genes using treatment as the main factor. However many genes differed between strains. We validated differences in two genes: suppressor APC domain containing 1 (Sapcd1) and Glyoxalase 1 (Glo1), with quantitative PCR on RNA from the nucleus accumbens and prefrontal cortex (). In both regions A/J mice had significantly higher expression of both genes than did C57BL/6 J. In prefrontal cortex, fentanyl treatment decreased Glo1 mRNA. Glyoxalase 1 catalyzes the detoxification of reactive alpha-oxoaldehydes such as glyoxal and methylglyoxal, is associated with anxiety and activity levels, and its inhibition reduces alcohol intake. We suggest that future studies assess the ability of Glo1 and related metabolites to modify opioid intake.


Assuntos
COVID-19 , Fentanila , Animais , Fentanila/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pandemias , SARS-CoV-2
6.
J Autism Dev Disord ; 52(10): 4608-4624, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34596807

RESUMO

Investigations into the etiology of autism spectrum disorders have been largely confined to two realms: variations in DNA sequence and somatic developmental exposures. Here we suggest a third route-disruption of the germline epigenome induced by exogenous toxicants during a parent's gamete development. Similar to cases of germline mutation, these molecular perturbations may produce dysregulated transcription of brain-related genes during fetal and early development, resulting in abnormal neurobehavioral phenotypes in offspring. Many types of exposures may have these impacts, and here we discuss examples of anesthetic gases, tobacco components, synthetic steroids, and valproic acid. Alterations in parental germline could help explain some unsolved phenomena of autism, including increased prevalence, missing heritability, skewed sex ratio, and heterogeneity of neurobiology and behavior.


Assuntos
Anestésicos Inalatórios , Transtorno do Espectro Autista , Transtorno Autístico , Transtorno do Espectro Autista/genética , Transtorno Autístico/genética , Células Germinativas , Humanos , Ácido Valproico
7.
Endocrinology ; 161(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32761086

RESUMO

The organizational/activational hypothesis suggests that gonadal steroid hormones like testosterone (T) and estradiol (E2) are important at 2 different times during the lifespan when they perform 2 different functions. First steroids "organize" brain structures early in life and during puberty, and in adults these same hormones "activate" sexually dimorphic behaviors. This hypothesis has been tested and proven valid for a large number of behaviors (learning, memory, social, and sexual behaviors). Sex differences in drug addiction are well established both for humans and animal models. Previous research in this field has focused primarily on cocaine self-administration by rats. Traditionally, observed sex differences have been explained by the sex-specific concentrations of gonadal hormones present at the time of the drug-related behavior. Studies with gonadectomized rodents establishes an activational role for E2 that facilitates vulnerability in females, and when E2 is combined with progesterone, addiction is attenuated. Literature on organizational actions of steroids is sparse but predicts that T, after it is aromatized to E2, changes aspects of the neural reward system. Here we summarize these data and propose that sex chromosome complement also plays a role in determining sex-specific drug-taking behavior. Future research is needed to disentangle the effects of hormones and sex chromosome complement, and we propose the four core genotype mouse model as an effective tool for answering these questions.


Assuntos
Diferenciação Sexual/fisiologia , Transtornos Relacionados ao Uso de Substâncias/etiologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Hormônios Esteroides Gonadais/farmacologia , Humanos , Recompensa , Fatores de Risco , Caracteres Sexuais , Fatores Sexuais , Transtornos Relacionados ao Uso de Substâncias/epidemiologia
8.
Horm Behav ; 125: 104821, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32721403

RESUMO

Women acquire cocaine habits faster and are more motivated to obtain drug than men. In general, female rodents acquire intravenous cocaine self-administration (SA) faster and show greater locomotor responses to cocaine than males. Sex differences are attributed to differences in circulating estradiol. We used the four core genotype (FCG) mouse to ask whether sex chromosome complement influences vulnerability to cocaine's reinforcing and/or locomotor-activating effects. The FCG cross produces ovary-bearing mice with XX or XY genotypes (XXF, XYF) and testes-bearing mice with XX or XY genotypes (XXM, XYM). A greater percentage of gonadal females acquired cocaine SA via infusions into jugular catheters as compared with XYM mice, but XXM mice were not significantly different than any other group. Discrimination of the active versus inactive nose poke holes and cocaine intake were in general greater in gonadal females than in gonadal males. Progressive ratio tests for motivation revealed an interaction between sex chromosomes and gonads: XYM mice were more motivated to self-administer cocaine taking more infusions than mice in any other group. Locomotor responses to cocaine exposure revealed effects of sex chromosomes. After acute exposure, activity was greater in XX than in XY mice and the reverse was true for behavioral sensitization. Mice with XY genotypes displayed more activity than XX mice when given cocaine after a 10-day drug-free period. Our data demonstrate that sex chromosome complement alone and/or interacting with gonadal status can modify cocaine's reinforcing and locomotor-activating effects. These data should inform current studies of sex differences in drug use.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/genética , Predisposição Genética para Doença , Cromossomos Sexuais/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Cocaína/farmacologia , Estradiol/fisiologia , Feminino , Genótipo , Gônadas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Caracteres Sexuais
9.
Int J Mol Sci ; 21(9)2020 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-32365465

RESUMO

Bisphenol A (BPA) is an endocrine-disrupting compound detected in the urine of more than 92% of humans, easily crosses the placental barrier, and has been shown to influence gene expression during fetal brain development. The purpose of this study was to investigate the effect of in utero BPA exposure on gene expression in the anterior hypothalamus, the basal nucleus of the stria terminalis (BNST), and hippocampus in C57BL/6 mice. Mice were exposed in utero to human-relevant doses of BPA, and then RNA sequencing was performed on male PND 28 tissue from whole hypothalamus (n = 3/group) that included the medial preoptic area (mPOA) and BNST to determine whether any genes were differentially expressed between BPA-exposed and control mice. A subset of genes was selected for further study using RT-qPCR on adult tissue from hippocampus to determine whether any differentially expressed genes (DEGs) persisted into adulthood. Two different RNA-Seq workflows indicated a total of 259 genes that were differentially expressed between BPA-exposed and control mice. Gene ontology analysis indicated that those DEGs were overrepresented in categories relating to mating, cell-cell signaling, behavior, neurodevelopment, neurogenesis, synapse formation, cognition, learning behaviors, hormone activity, and signaling receptor activity, among others. Ingenuity Pathway Analysis was used to interrogate novel gene networks and upstream regulators, indicating the top five upstream regulators as huntingtin, beta-estradiol, alpha-synuclein, Creb1, and estrogen receptor (ER)-alpha. In addition, 15 DE genes were identified that are suspected in autism spectrum disorders.


Assuntos
Poluentes Ocupacionais do Ar/efeitos adversos , Transtorno do Espectro Autista/etiologia , Transtorno do Espectro Autista/metabolismo , Compostos Benzidrílicos/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Exposição Materna/efeitos adversos , Fenóis/efeitos adversos , Animais , Biologia Computacional/métodos , Bases de Dados Genéticas , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Perfilação da Expressão Gênica , Hormônios/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transdução de Sinais , Transcriptoma
10.
Horm Behav ; 119: 104677, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31927019

RESUMO

It is our hope this mini-review will stimulate discussion and new research. Here we briefly examine the literature on transgenerational actions of endocrine disrupting chemicals (EDCs) on brain and behavior and their underlying epigenetic mechanisms including: DNA methylation, histone modifications, and non-coding RNAs. We stress that epigenetic modifications need to be examined in a synergistic manner, as they act together in situ on chromatin to change transcription. Next we highlight recent work from one of our laboratories (VGC). The data provide new evidence that the sperm genome is poised for transcription. In developing sperm, gene enhancers and promoters are accessible for transcription and these activating motifs are also found in preimplantation embryos. Thus, DNA modifications associated with transcription factors during fertilization, in primordial germ cells (PGCs), and/or during germ cell maturation may be passed to offspring. We discuss the implications of this model to EDC exposures and speculate on whether natural variation in hormone levels during fertilization and PGC migration may impart transgenerational effects on brain and behavior. Lastly we discuss how this mechanism could apply to neural sexual differentiation.


Assuntos
Comportamento/efeitos dos fármacos , Disruptores Endócrinos/farmacologia , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/fisiologia , Efeitos Tardios da Exposição Pré-Natal/genética , Animais , Pesquisa Comportamental/métodos , Pesquisa Comportamental/tendências , Efeito de Coortes , Metilação de DNA/efeitos dos fármacos , Exposição Ambiental/análise , Exposição Ambiental/estatística & dados numéricos , Feminino , Humanos , Masculino , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/embriologia , Sistema Nervoso/crescimento & desenvolvimento , Neuroendocrinologia/métodos , Neuroendocrinologia/tendências , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Reprodução/efeitos dos fármacos , Diferenciação Sexual/efeitos dos fármacos
11.
Endocrinology ; 160(8): 1854-1867, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31188430

RESUMO

Bisphenol A (BPA) is a ubiquitous endocrine-disrupting chemical. Developmental exposure produces changes in behavior and gene expression in the brain. Here, we examined social recognition behaviors in mice from the third familial generation (F3) after exposure to gestational BPA. Second-generation mice were bred in one of four mating combinations to reveal whether characteristics in F3 were acquired via maternal or paternal exposures. After repeated habituation to the same mouse, offspring of dams from the BPA lineage failed to display increased investigation of a novel mouse. Genes involved in excitatory postsynaptic densities (PSDs) were examined in F3 brains using quantitative PCR. Differential expression of genes important for function and stability of PSDs were assessed at three developmental ages. Several related PSD genes-SH3 and multiple ankyrin repeat domains 1 (Shank1), Homer scaffolding protein 1c (Homer1c), DLG associated protein 1 (Gkap), and discs large MAGUK scaffold protein 4 (PSD95)-were differentially expressed in control- vs BPA-lineage brains. Using a second strain of F3 inbred mice exposed to BPA, we noted the same differences in Shank1 and PSD95 expression in C57BL/6J mice. In sum, transgenerational BPA exposure disrupted social interactions in mice and dysregulated normal expression of PSD genes during neural development. The fact that the same genetic effects were found in two different mouse strains and in several brain regions increased potential for translation. The genetic and functional relationship between PSD and abnormal neurobehavioral disorders is well established, and our data suggest that BPA may contribute in a transgenerational manner to neurodevelopmental diseases.


Assuntos
Compostos Benzidrílicos/toxicidade , Feto/efeitos dos fármacos , Fenóis/toxicidade , Densidade Pós-Sináptica/efeitos dos fármacos , Comportamento Social , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/fisiologia
12.
Toxicology ; 420: 66-72, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30959087

RESUMO

Bisphenol S (2,2-bisulfone, BPS) and Bisphenol F (2,2-bis [4-hydroxyphenol]methane, BPF) are analogs of Bisphenol A (2,2-bis[4-hydroxyphenyl]propane, BPA), a widely used endocrine disrupting compound present in polycarbonate plastics, thermal receipts and epoxy resins that line food cans. Here we examined effects of BPA, BPS, and BPF in low concentrations on differentiation in murine 3T3-L1 preadipocytes. We also fed adult male mice chow with one of three doses of BPF (0, 0.5, 5, 50 mg/kg chow, or approximately 0.044, 0.44 and 4.4 mg/kg body weight per day) for 12 weeks, collected body weights, food intake, and tested for glucose tolerance. The doses of BPF used produced mean concentrations of 0, 6.2, 43.6, and 561 ng/mL in plasma. In 3T3-L1 cells BPS had the greatest effects, along with BPA, both increased expression of several genes required for preadipocyte differentiation over 12 days in culture. In contrast, BPF decreased expression of several genes late in differentiation. This dichotomy was also reflected in lipid accumulation as BPA and BPS treated cells had elevated lipid concentrations compared to controls or cells treated with BPF. Male mice fed either the highest or lowest concentrations of BPF gained less weight than controls with no effects on glucose levels or glucose tolerance. Plasma levels of BPF reflected doses in food with no overlap between doses. In summary, our results suggest that BPS has a strong potential to be obesogenic while effects of BPF are subtler and potentially in the opposite direction.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Fenóis/toxicidade , Sulfonas/toxicidade , Aumento de Peso/efeitos dos fármacos , Células 3T3-L1 , Adipócitos/metabolismo , Adipogenia/genética , Animais , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fatores de Tempo
13.
Horm Behav ; 101: 68-76, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28964733

RESUMO

Bisphenol A (BPA) is an endocrine disrupting chemical used in the production of polycarbonate plastics and resins. Exposure to BPA during gestation has been proposed as a risk factor for the development of neurobehavioral disorders, such as autism spectrum disorder. To address the behavioral impact of developmental exposure to BPA, we tested offspring of mice exposed to a daily low dose of BPA during pregnancy. We also asked if preconception exposure of the sire affected behaviors in offspring. Sires that consumed BPA for 50days prior to mating weighed less than controls, but no effects on any reproductive measures were noted. Juvenile offspring exposed to BPA maternally, but not paternally, spent less time in the open arms of the elevated plus maze than controls, indicating increased anxiety-like behavior. However, neither parental exposure group differed significantly from controls in the social recognition task. We also assessed the behaviors of maternally exposed offspring in two novel tasks: ultrasonic vocalizations (USVs) in pups and operant reversal learning in adults. Maternal BPA exposure increased the duration and median frequency of USVs emitted by pups during maternal separation. In the reversal learning task, females responded more accurately and earned more rewards than males. Additionally, control females received more rewards than BPA females during the acquisition phase of the task. These are among the first studies conducted to ask if BPA exposure via the sire affects offspring behavior and the first study to report effects of gestational BPA exposure on pup USVs and adult operant responding.


Assuntos
Comportamento Animal/efeitos dos fármacos , Compostos Benzidrílicos/farmacologia , Exposição Materna/efeitos adversos , Exposição Paterna/efeitos adversos , Fenóis/farmacologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Ansiedade/induzido quimicamente , Ansiedade/fisiopatologia , Disruptores Endócrinos/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Comportamento Sexual Animal/efeitos dos fármacos
14.
Endocrinology ; 159(1): 132-144, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29165653

RESUMO

Bisphenol A (BPA) is a ubiquitous man-made endocrine disrupting compound (EDC). Developmental exposure to BPA changes behavioral and reproductive phenotypes, and these effects can last for generations. We exposed embryos to BPA, producing two lineages: controls and BPA exposed. In the third filial generation (F3), brain tissues containing the preoptic area, the bed nucleus of the stria terminalis, and the anterior hypothalamus were collected. RNA sequencing (RNA-seq) and subsequent data analyses revealed 50 differentially regulated genes in the brains of F3 juveniles from BPA vs control lineages. BPA exposure can lead to loss of imprinting, and one of the two imprinted genes in our data set, maternally expressed gene 3 (Meg3), has been associated with EDCs and neurobehavioral phenotypes. We used quantitative polymerase chain reaction to examine the two imprinted genes in our data set, Meg3 and microRNA-containing gene Mirg (residing in the same loci). Confirming the RNA-seq, Meg3 messenger RNA was higher in F3 brains from the BPA lineage than in control brains. This was true in brains from mice produced with two different BPA paradigms. Next, we used pyrosequencing to probe differentially methylated regions of Meg3. We found transgenerational effects of BPA on imprinted genes in brain. Given these results, and data on Meg3 methylation in humans, we suggest this gene may be a biomarker indicative of early life environmental perturbation.


Assuntos
Compostos Benzidrílicos/toxicidade , Encéfalo/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Exposição Materna/efeitos adversos , Fenóis/toxicidade , Animais , Encéfalo/metabolismo , Cruzamentos Genéticos , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Hipotálamo Anterior/efeitos dos fármacos , Hipotálamo Anterior/metabolismo , Lactação , Masculino , Camundongos Endogâmicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Gravidez , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , RNA Longo não Codificante/agonistas , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Distribuição Aleatória , Núcleos Septais/efeitos dos fármacos , Núcleos Septais/metabolismo , Caracteres Sexuais
15.
PLoS One ; 12(2): e0171977, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28199414

RESUMO

Di-(2-ethylhexyl) phthalate (DEHP) is an endocrine disrupting chemical commonly used as a plasticizer in medical equipment, food packaging, flooring, and children's toys. DEHP exposure during early development has been associated with adverse neurobehavioral outcomes in children. In animal models, early exposure to DEHP results in abnormal development of the reproductive system as well as altered behavior and neurodevelopment. Based on these data, we hypothesized that developmental exposure to DEHP would decrease social interactions and increase anxiety-like behaviors in mice in a dose-dependent manner, and that the effects would persist over generations. C57BL/6J mice consumed one of three DEHP doses (0, 5, 40, and 400 µg/kg body weight) throughout pregnancy and during the first ten days of lactation. The two higher doses yielded detectable levels of DEHP metabolites in serum. Pairs of mice from control, low, and high DEHP doses were bred to create three dose lineages in the third generation (F3). Average anogenital index (AGI: anogenital distance/body weight) was decreased in F1 males exposed to the low dose of DEHP and in F1 females exposed to the highest dose. In F1 mice, juvenile pairs from the two highest DEHP dose groups displayed fewer socially investigative behaviors and more exploratory behaviors as compared with control mice. The effect of DEHP on these behaviors was reversed in F3 mice as compared with F1 mice. F1 mice exposed to low and medium DEHP doses spent more time in the closed arms of the elevated plus maze than controls, indicating increased anxiety-like behavior. The generation-dependent effects on behavior and AGI suggest complex mechanisms by which DEHP directly impacts reproductive and neurobehavioral development and influences germline-inherited traits.


Assuntos
Comportamento Animal/efeitos dos fármacos , Dietilexilftalato/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Dietilexilftalato/sangue , Dietilexilftalato/metabolismo , Feminino , Masculino , Exposição Materna , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Comportamento Social
16.
Endocrinology ; 158(1): 21-30, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27841950

RESUMO

Bisphenol A (BPA) is an endocrine-disrupting compound used to manufacture plastics; it is present in linings of food cans, bottles, thermal receipts, and many other everyday items and is detectable in human urine and blood. Exposure to BPA during development can disrupt sexual differentiation of some brain regions. Moreover, BPA can have transgenerational effects on gene expression and behaviors. Here, we used a diet and breeding regimen that produces transgenerational effects on behaviors. C57BL/6J mice consumed control or BPA-containing diets during pregnancy. We examined vasopressin (AVP) and estrogen receptor α (ERα) immunoreactivity (ir) in sexually dimorphic brain regions from first-generation (F1) offspring and transgenerational effects of BPA in third-generation offspring. In all but one brain region examined, the expected sex differences were noted in both generations of control mice. In F1 mice, a diet by sex interaction was present for AVP-ir in the lateral septum and posterodorsal medial amygdala. In both regions, BPA exposure reduced immunoreactivity in male brains. An interaction between diet and sex for ERα-ir in the ventromedial hypothalamus was caused by reduced immunoreactivity in BPA-exposed females. Of interest, BPA had transgenerational effects on ERα-ir in the anteroventral periventricular nucleus and bed nucleus of the stria terminalis. Our data show that BPA produces immunoreactive differences in ERα-ir generations after exposure to BPA. We speculate that actions of BPA in utero on ERα-ir in brain have long-term consequences for reproduction and social behavior.


Assuntos
Compostos Benzidrílicos/toxicidade , Encéfalo/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Fenóis/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Caracteres Sexuais , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Distribuição Aleatória , Núcleos Septais/metabolismo , Vasopressinas/metabolismo
17.
Endocrinology ; 157(8): 2969-71, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27477860

RESUMO

Nowadays, we are bombarded with information on a large number of endocrine-disrupting chemicals. We hear and read about endocrine-disrupting chemicals on blogs, the web, news stories, television specials, advertisements, and of course scientific articles. Reports claim these ubiquitous compounds are responsible for increased rates of cancer, autism, obesity, hypospadias, and infertility, just to name a few. But it was not always this way. In fact, the scientific study of endocrine-disrupting chemicals is relatively new: a recent PubMed search found a total of 6184 hits for the term, 739 articles in 2015 as compared with 4, 20 years ago in 1995.


Assuntos
Disruptores Endócrinos/isolamento & purificação , Endocrinologia/organização & administração , Endocrinologia/tendências , Exposição Ambiental , Estrogênios/isolamento & purificação , Sociedades Médicas , Animais , Disruptores Endócrinos/farmacologia , Exposição Ambiental/análise , Poluentes Ambientais/isolamento & purificação , Poluentes Ambientais/farmacologia , Estrogênios/farmacologia , História do Século XX , História do Século XXI , Humanos , Células MCF-7 , Sociedades Médicas/história , Sociedades Médicas/organização & administração , Sociedades Médicas/tendências
18.
Horm Behav ; 80: 132-138, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26940434

RESUMO

The importance of gonadal steroids in modulating male sexual behavior is well established. Individual differences in male sexual behavior, independent of gonadal steroids, are prevalent across a wide range of species, including man. However, the genetic mechanisms underlying steroid-independent male sexual behavior are poorly understood. A high proportion of B6D2F1 hybrid male mice demonstrates steroid-independent male sexual behavior (identified as "maters"), providing a mouse model that opens up avenues of investigation into the mechanisms regulating male sexual behavior in the absence of gonadal hormones. Recent studies have revealed several proteins that play a significant factor in regulating steroid-independent male sexual behavior in B6D2F1 male mice, including amyloid precursor protein (APP), tau, and synaptophysin. The specific goals of our study were to determine whether steroid-independent male sexual behavior was a heritable trait by determining if it was dependent upon the behavioral phenotype of the B6D2F1 sire, and whether the differential expression of APP, tau, and synaptophysin in the medial preoptic area found in the B6D2F1 sires that did and did not mate after gonadectomy was similar to those found in their male offspring. After adult B6D2F1 male mice were bred with C57BL/6J female mice, they and their male offspring (BXB1) were orchidectomized and identified as either maters or "non-maters". A significant proportion of the BXB1 maters was sired only from B6D2F1 maters, indicating that the steroid-independent male sexual behavior behavioral phenotype of the B6D2F1 hybrid males, when crossed with C57BL/6J female mice, is inherited by their male offspring. Additionally, APP, tau, and synaptophysin were elevated in in the medial preoptic area in both the B6D2F1 and BXB1 maters relative to the B6D2F1 and BXB1 non-maters, respectively, suggesting a potential genetic mechanism for the inheritance of steroid-independent male sexual behavior.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Expressão Gênica/genética , Hormônios Esteroides Gonadais/fisiologia , Área Pré-Óptica/fisiologia , Comportamento Sexual Animal/fisiologia , Sinaptofisina/genética , Proteínas tau/genética , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Fenótipo , Comportamento Sexual , Esteroides
19.
Endocrinology ; 157(5): 1967-79, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27010449

RESUMO

Calbindin-D(28K) (Calb1), a high-affinity calcium buffer/sensor, shows abundant expression in neurons and has been associated with a number of neurobehavioral diseases, many of which are sexually dimorphic in incidence. Behavioral and physiological end points are affected by experimental manipulations of calbindin levels, including disruption of spatial learning, hippocampal long-term potentiation, and circadian rhythms. In this study, we investigated novel aspects of calbindin function on social behavior, anxiety-like behavior, and fear conditioning in adult mice of both sexes by comparing wild-type to littermate Calb1 KO mice. Because Calb1 mRNA and protein are sexually dimorphic in some areas of the brain, we hypothesized that sex differences in behavioral responses of these behaviors would be eliminated or revealed in Calb1 KO mice. We also examined gene expression in the amygdala and prefrontal cortex, two areas of the brain intimately connected with limbic system control of the behaviors tested, in response to sex and genotype. Our results demonstrate that fear memory and social behavior are altered in male knockout mice, and Calb1 KO mice of both sexes show less anxiety. Moreover, gene expression studies of the amygdala and prefrontal cortex revealed several significant genotype and sex effects in genes related to brain-derived neurotrophic factor signaling, hormone receptors, histone deacetylases, and γ-aminobutyric acid signaling. Our findings are the first to directly link calbindin with affective and social behaviors in rodents; moreover, the results suggest that sex differences in calbindin protein influence behavior.


Assuntos
Tonsila do Cerebelo/metabolismo , Comportamento Animal/fisiologia , Calbindinas/genética , Medo/fisiologia , Córtex Pré-Frontal/metabolismo , Comportamento Social , Animais , Ansiedade/genética , Ansiedade/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Calbindinas/metabolismo , Condicionamento Psicológico/fisiologia , Feminino , Expressão Gênica , Histona Desacetilases/metabolismo , Masculino , Memória/fisiologia , Camundongos , Camundongos Knockout , Transdução de Sinais/fisiologia , Ácido gama-Aminobutírico/metabolismo
20.
Endocrinology ; 156(9): 3077-83, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26168342

RESUMO

Exposure to di-(2-ethylhexyl) phthalate (DEHP) has been linked to male reproductive abnormalities. Here, we assessed transgenerational actions of DEHP on several behaviors and stress responses. We used 2 doses of DEHP (150- and 200-mg/kg body weight) and a treatment regimen previously shown to produce transgenerational effects on male reproduction. Mice, 3 generations removed from DEHP exposure (F3), were tested for social behavior and anxiety on the elevated plus maze. We collected blood and pituitaries from undisturbed and restrained mice. Body weights, anogenital distances, and reproductive organ weights were collected at killing. In social interaction tests juvenile males from the DEHP lineage (200 mg/kg) displayed more digging and less self-grooming than did controls. Interestingly, 150-mg/kg lineage males, killed in early puberty, had smaller seminal vesicle weights than their controls. However, the 200-mg/kg males (killed on average 10 d later) did not show this effect. Females from a DEHP lineage had lower corticosterone concentrations than controls after restraint stress. We also found sex- and DEHP-specific mRNA expression changes in the pituitary in 2 of the 6 stress-related genes we measured. In particular, Gnas mRNA was elevated by the combination of DEHP lineage and stress. Thus, transgenerational effects of DEHP are noted in male behavior, and in females, DEHP had transgenerational effects on levels of corticosterone. Both of these results may be related to transgenerational modifications in the expression of several pituitary hormones involved in the hypothalamic-pituitary-adrenal axis.


Assuntos
Comportamento Animal/efeitos dos fármacos , Corticosterona/sangue , Dietilexilftalato/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico/sangue , Animais , Feminino , Genitália/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Gravidez , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Estresse Psicológico/induzido quimicamente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...