Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Psychopharmacology (Berl) ; 233(4): 549-69, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26758282

RESUMO

RATIONALE: For several decades, elite athletes and a growing number of recreational consumers have used anabolic androgenic steroids (AAS) as performance enhancing drugs. Despite mounting evidence that illicit use of these synthetic steroids has detrimental effects on affective states, information available on sex-specific actions of these drugs is lacking. OBJECTIVES: The focus of this review is to assess information to date on the importance of sex and its interaction with other environmental factors on affective behaviors, with an emphasis on data derived from non-human studies. METHODS: The PubMed database was searched for relevant studies in both sexes. RESULTS: Studies examining AAS use in females are limited, reflecting the lower prevalence of use in this sex. Data, however, indicate significant sex-specific differences in AAS effects on anxiety-like and aggressive behaviors, interactions with other drugs of abuse, and the interplay of AAS with other environmental factors such as diet and exercise. CONCLUSIONS: Current methods for assessing AAS use have limitations that suggest biases of both under- and over-reporting, which may be amplified for females who are poorly represented in self-report studies of human subjects and are rarely used in animal studies. Data from animal literature suggest that there are significant sex-specific differences in the impact of AAS on aggression, anxiety, and concomitant use of other abused substances. These results have relevance for human females who take these drugs as performance-enhancing substances and for transgender XX individuals who may illicitly self-administer AAS as they transition to a male gender identity.


Assuntos
Anabolizantes/efeitos adversos , Androgênios/efeitos adversos , Transtornos do Humor/induzido quimicamente , Transtornos do Humor/psicologia , Caracteres Sexuais , Agressão/efeitos dos fármacos , Agressão/psicologia , Anabolizantes/administração & dosagem , Androgênios/administração & dosagem , Animais , Ansiedade/induzido quimicamente , Ansiedade/psicologia , Feminino , Humanos , Masculino , Autoadministração , Esteroides/administração & dosagem , Esteroides/efeitos adversos , Resultado do Tratamento
2.
Horm Behav ; 66(2): 283-97, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24768711

RESUMO

Anabolic androgenic steroids (AAS) are taken by both sexes to enhance athletic performance and body image, nearly always in conjunction with an exercise regime. Although taken to improve physical attributes, chronic AAS use can promote negative behavior, including anxiety. Few studies have directly compared the impact of AAS use in males versus females or assessed the interaction of exercise and AAS. We show that AAS increase anxiety-like behaviors in female but not male mice and that voluntary exercise accentuates these sex-specific differences. We also show that levels of the anxiogenic peptide corticotrophin releasing factor (CRF) are significantly greater in males, but that AAS selectively increase CRF levels in females, thus abrogating this sex-specific difference. Exercise did not ameliorate AAS-induced anxiety or alter CRF levels in females. Exercise was anxiolytic in males, but this behavioral outcome did not correlate with CRF levels. Brain-derived neurotrophic factor (BDNF) has also been implicated in the expression of anxiety. As with CRF, levels of hippocampal BDNF mRNA were significantly greater in males than females. AAS and exercise were without effect on BDNF mRNA in females. In males, anxiolytic effects of exercise correlated with increased BDNF mRNA, however AAS-induced changes in BDNF mRNA and anxiety did not. In sum, we find that AAS elicit sex-specific differences in anxiety and that voluntary exercise accentuates these differences. In addition, our data suggest that these behavioral outcomes may reflect convergent actions of AAS and exercise on a sexually differentiated CRF signaling system within the extended amygdala.


Assuntos
Anabolizantes/toxicidade , Androgênios/toxicidade , Ansiedade/induzido quimicamente , Ansiedade/psicologia , Condicionamento Físico Animal/fisiologia , Comportamento Sexual Animal/fisiologia , Esteroides/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Comportamento de Escolha , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reflexo de Sobressalto/efeitos dos fármacos , Caracteres Sexuais , Comportamento Social
3.
Neuroendocrinology ; 96(2): 141-51, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22576754

RESUMO

Anabolic androgenic steroids (AAS) comprise a large and growing class of synthetic androgens used clinically to promote tissue-building in individuals suffering from genetic disorders, injuries, and diseases. Despite these beneficial therapeutic applications, the predominant use of AAS is illicit: these steroids are self-administered to promote athletic performance and body image. Hand in hand with the desired anabolic actions of the AAS are untoward effects on the brain and behavior. While the signaling routes by which the AAS impose both beneficial and harmful actions may be quite diverse, key endpoints are likely to include ligand-gated and voltage-dependent ion channels that govern the activity of electrically excitable tissues. Here, we review the known effects of AAS on molecular targets that play critical roles in controlling electrical activity, with a specific focus on the effects of AAS on neurotransmission mediated by GABA(A) receptors in the central nervous system.


Assuntos
Anabolizantes/farmacologia , Neurônios/metabolismo , Transmissão Sináptica/fisiologia , Congêneres da Testosterona/farmacologia , Animais , Encéfalo/citologia , Humanos , Canais Iônicos/metabolismo , Neurônios/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Esteroides/metabolismo , Transmissão Sináptica/efeitos dos fármacos
4.
Trends Neurosci ; 35(6): 382-92, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22516619

RESUMO

Anabolic androgenic steroids (AAS) are illicitly administered to enhance athletic performance and body image. Although conferring positive actions on performance, steroid abuse is associated with changes in anxiety and aggression. AAS users are often keenly invested in understanding the biological actions of these drugs. Thus, mechanistic information on AAS actions is important not only for the biomedical community, but also for steroid users. Here we review findings from animal studies on the impact of AAS exposure on neural systems that are crucial for the production of anxiety and aggression, and compare the effects of the different classes of AAS and their potential signaling mechanisms, as well as context-, age- and sex-dependent aspects of their actions.


Assuntos
Agressão/efeitos dos fármacos , Anabolizantes/efeitos adversos , Ansiedade/fisiopatologia , Encéfalo/fisiopatologia , Medo/efeitos dos fármacos , Substâncias para Melhoria do Desempenho/efeitos adversos , Esteroides/efeitos adversos , Ansiedade/induzido quimicamente , Humanos , Modelos Neurológicos
5.
Neuropsychopharmacology ; 37(6): 1483-99, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22298120

RESUMO

Increased anxiety is commonly observed in individuals who illicitly administer anabolic androgenic steroids (AAS). Behavioral effects of steroid abuse have become an increasing concern in adults and adolescents of both sexes. The dorsolateral bed nucleus of the stria terminalis (dlBnST) has a critical role in the expression of diffuse anxiety and is a key site of action for the anxiogenic neuromodulator, corticotropin releasing factor (CRF). Here we demonstrate that chronic, but not acute, exposure of female mice during adolescence to AAS augments anxiety-like behaviors; effects that were blocked by central infusion of the CRF receptor type 1 antagonist, antalarmin. AAS treatment selectively increased action potential (AP) firing in neurons of the central amygdala (CeA) that project to the dlBnST, increased the frequency of GABA(A) receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in dlBnST target neurons, and decreased both c-FOS immunoreactivity (IR) and AP frequency in these postsynaptic cells. Acute application of antalarmin abrogated the enhancement of GABAergic inhibition induced by chronic AAS exposure whereas application of CRF to brain slices of naïve mice mimicked the actions of this treatment. These results, in concert with previous data demonstrating that chronic AAS treatment results in enhanced levels of CRF mRNA in the CeA and increased CRF-IR in the dlBnST neuropil, are consistent with a mechanism in which the enhanced anxiety elicited by chronic AAS exposure involves augmented inhibitory activity of CeA afferents to the dlBnST and CRF-dependent enhancement of GABAergic inhibition in this brain region.


Assuntos
Androgênios/toxicidade , Ansiedade/patologia , Hormônio Liberador da Corticotropina/farmacologia , Prosencéfalo/efeitos dos fármacos , Estimulação Acústica , Potenciais de Ação/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Análise de Variância , Animais , Ansiedade/induzido quimicamente , Ansiedade/tratamento farmacológico , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Pirimidinas/farmacologia , Pirróis/farmacologia , RNA Mensageiro/metabolismo , Filtro Sensorial/efeitos dos fármacos , Fatores de Tempo
6.
Hum Mol Genet ; 21(4): 730-50, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22048958

RESUMO

We have identified a point mutation in Npc1 that creates a novel mouse model (Npc1(nmf164)) of Niemann-Pick type C1 (NPC) disease: a single nucleotide change (A to G at cDNA bp 3163) that results in an aspartate to glycine change at position 1005 (D1005G). This change is in the cysteine-rich luminal loop of the NPC1 protein and is highly similar to commonly occurring human mutations. Genetic and molecular biological analyses, including sequencing the Npc1(spm) allele and identifying a truncating mutation, confirm that the mutation in Npc1(nmf164) mice is distinct from those in other existing mouse models of NPC disease (Npc1(nih), Npc1(spm)). Analyses of lifespan, body and spleen weight, gait and other motor activities, as well as acoustic startle responses all reveal a more slowly developing phenotype in Npc1(nmf164) mutant mice than in mice with the null mutations (Npc1(nih), Npc1(spm)). Although Npc1 mRNA levels appear relatively normal, Npc1(nmf164) brain and liver display dramatic reductions in Npc1 protein, as well as abnormal cholesterol metabolism and altered glycolipid expression. Furthermore, histological analyses of liver, spleen, hippocampus, cortex and cerebellum reveal abnormal cholesterol accumulation, glial activation and Purkinje cell loss at a slower rate than in the Npc1(nih) mouse model. Magnetic resonance imaging studies also reveal significantly less demyelination/dysmyelination than in the null alleles. Thus, although prior mouse models may correspond to the severe infantile onset forms of NPC disease, Npc1(nmf164) mice offer many advantages as a model for the late-onset, more slowly progressing forms of NPC disease that comprise the large majority of human cases.


Assuntos
Proteínas de Transporte/genética , Modelos Animais de Doenças , Glicoproteínas de Membrana/genética , Doença de Niemann-Pick Tipo C/genética , Mutação Puntual/genética , Idade de Início , Alelos , Animais , Astrócitos/patologia , Encéfalo/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Colesterol/metabolismo , Análise Mutacional de DNA , Progressão da Doença , Estresse do Retículo Endoplasmático , Gangliosídeos/metabolismo , Homozigoto , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Metabolismo dos Lipídeos , Pulmão/citologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Microglia/patologia , Bainha de Mielina , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Doença de Niemann-Pick Tipo C/fisiopatologia , Fenótipo , Deficiências na Proteostase , Células de Purkinje/patologia , RNA Mensageiro/análise , RNA Mensageiro/genética , Reflexo de Sobressalto , Taxa de Sobrevida
7.
Neuropharmacology ; 61(4): 653-64, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21645530

RESUMO

Disruption of reproductive function is a hallmark of abuse of anabolic androgenic steroids (AAS) in female subjects. To understand the central actions of AAS, patch clamp recordings were made in estrous, diestrous and AAS-treated mice from gonadotropin releasing hormone (GnRH) neurons, neurons in the medial preoptic area (mPOA) and neurons in the anteroventroperiventricular nucleus (AVPV); regions known to provide GABAergic and kisspeptin inputs to the GnRH cells. Action potential (AP) frequency was significantly higher in GnRH neurons of estrous mice than in AAS-treated or diestrous animals. No significant differences in AAS-treated, estrous or diestrous mice were evident in the amplitude or kinetics of spontaneous postsynaptic currents (sPSCs), miniature PSCs or tonic currents mediated by GABA(A) receptors or in GABA(A) receptor subunit expression in GnRH neurons. In contrast, the frequency of GABA(A) receptor-mediated sPSCs in GnRH neurons showed an inverse correlation with AP frequency across the three hormonal states. Surprisingly, AP activity in the medial preoptic area (mPOA), a likely source of GABAergic afferents to GnRH cells, did not vary in concert with the sPSCs in the GnRH neurons. Furthermore, pharmacological blockade of GABA(A) receptors did not alter the pattern in which there was lower AP frequency in GnRH neurons of AAS-treated and diestrous versus estrous mice. These data suggest that AAS do not impose their effects either directly on GnRH neurons or on putative GABAergic afferents in the mPOA. AP activity recorded from neurons in kisspeptin-rich regions of the AVPV and the expression of kisspeptin mRNA and peptide did vary coordinately with AP activity in GnRH neurons. Our data demonstrate that AAS treatment imposes a "diestrous-like" pattern of activity in GnRH neurons and suggest that this effect may arise from suppression of presynaptic kisspeptin-mediated excitatory drive arising from the AVPV. The actions of AAS on neuroendocrine regulatory circuits may contribute the disruption of reproductive function observed in steroid abuse.


Assuntos
Anabolizantes/administração & dosagem , Metiltestosterona/administração & dosagem , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Potenciais Sinápticos/efeitos dos fármacos , Animais , Feminino , Hormônio Liberador de Gonadotropina/biossíntese , Hormônio Liberador de Gonadotropina/fisiologia , Camundongos , Camundongos Transgênicos , Núcleos da Linha Média do Tálamo/fisiologia , Neurônios/metabolismo , Área Pré-Óptica/fisiologia , Sinapses/fisiologia , Potenciais Sinápticos/fisiologia
8.
Psychoneuroendocrinology ; 35(10): 1473-85, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20537804

RESUMO

In the past several decades, the therapeutic use of anabolic androgenic steroids (AAS) has been overshadowed by illicit use of these drugs by elite athletes and a growing number of adolescents to enhance performance and body image. As with adults, AAS use by adolescents is associated with a range of behavioral effects, including increased anxiety and altered responses to stress. It has been suggested that adolescents, especially adolescent females, may be particularly susceptible to the effects of these steroids, but few experiments in animal models have been performed to test this assertion. Here we show that chronic exposure of adolescent female mice to a mixture of three commonly abused AAS (testosterone cypionate, nandrolone decanoate and methandrostenolone; 7.5 mg/kg/day for 5 days) significantly enhanced anxiety-like behavior as assessed by the acoustic startle response (ASR), but did not augment the fear-potentiated startle response (FPS) or alter sensorimotor gating as assessed by prepulse inhibition of the acoustic startle response (PPI). AAS treatment also significantly increased the levels of corticotropin releasing factor (CRF) mRNA and somal-associated CRF immunoreactivity in the central nucleus of the amygdala (CeA), as well as neuropil-associated immunoreactivity in the dorsal aspect of the anterolateral division of the bed nucleus of the stria terminalis (dBnST). AAS treatment did not alter CRF receptor 1 or 2 mRNA in either the CeA or the dBnST; CRF immunoreactivity in the ventral BnST, the paraventricular nucleus (PVN) or the median eminence (ME); or peripheral levels of corticosterone. These results suggest that chronic AAS treatment of adolescent female mice may enhance generalized anxiety, but not sensorimotor gating or learned fear, via a mechanism that involves increased CRF-mediated signaling from CeA neurons projecting to the dBnST.


Assuntos
Anabolizantes/farmacologia , Androgênios/farmacologia , Ansiedade/psicologia , Hormônio Liberador da Corticotropina/biossíntese , Esteroides/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Corticosterona/sangue , Medo/psicologia , Feminino , Habituação Psicofisiológica/efeitos dos fármacos , Imuno-Histoquímica , Metandrostenolona/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Nandrolona/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reflexo de Sobressalto/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Núcleos Septais/efeitos dos fármacos
9.
J Neurosci ; 30(19): 6497-506, 2010 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-20463213

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons are the central regulators of reproduction. GABAergic transmission plays a critical role in pubertal activation of pulsatile GnRH secretion. Self-administration of excessive doses of anabolic androgenic steroids (AAS) disrupts reproductive function and may have critical repercussions for pubertal onset in adolescent users. Here, we demonstrate that chronic treatment of adolescent male mice with the AAS 17alpha-methyltestosterone significantly decreased action potential frequency in GnRH neurons, reduced the serum gonadotropin levels, and decreased testes mass. AAS treatment did not induce significant changes in GABAA receptor subunit mRNA levels or alter the amplitude or decay kinetics of GABAA receptor-mediated spontaneous postsynaptic currents (sPSCs) or tonic currents in GnRH neurons. However, AAS treatment significantly increased action potential frequency in neighboring medial preoptic area (mPOA) neurons and GABAA receptor-mediated sPSC frequency in GnRH neurons. In addition, physical isolation of the more lateral aspects of the mPOA from the medially localized GnRH neurons abrogated the AAS-induced increase in GABAA receptor-mediated sPSC frequency and the decrease in action potential firing in the GnRH cells. Our results indicate that AAS act predominantly on steroid-sensitive presynaptic neurons within the mPOA to impart significant increases in GABAA receptor-mediated inhibitory tone onto downstream GnRH neurons, resulting in diminished activity of these pivotal mediators of reproductive function. These AAS-induced changes in central GABAergic circuits of the forebrain may significantly contribute to the disruptive actions of these drugs on pubertal maturation and the development of reproductive competence in male steroid abusers.


Assuntos
Anabolizantes/toxicidade , Hormônio Liberador de Gonadotropina/metabolismo , Metiltestosterona/toxicidade , Neurônios/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Androgênios/toxicidade , Animais , Gonadotropinas/sangue , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/fisiopatologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , RNA Mensageiro/metabolismo , Transtornos Relacionados ao Uso de Substâncias/sangue , Transtornos Relacionados ao Uso de Substâncias/patologia , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/fisiologia , Transmissão Sináptica/fisiologia , Testículo/efeitos dos fármacos , Testículo/patologia
10.
J Neurosci ; 30(4): 1463-70, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20107073

RESUMO

AMPA receptors (AMPARs) are tetrameric ligand-gated ion channels that couple the energy of glutamate binding to the opening of a transmembrane channel. Crystallographic and electrophysiological analysis of AMPARs has suggested a coupling between (1) cleft closure in the bilobate ligand-binding domain (LBD), (2) the resulting separation of transmembrane helix attachment points across subunit dimers, and (3) agonist efficacy. In general, more efficacious agonists induce greater degrees of cleft closure and transmembrane separation than partial agonists. Several apparent violations of the cleft-closure/efficacy paradigm have emerged, although in all cases, intradimer separation remains as the driving force for channel opening. Here, we examine the structural basis of partial agonism in GluA4 AMPARs. We find that the L651V substitution enhances the relative efficacy of kainate without increasing either LBD cleft closure or transmembrane separation. Instead, the conformational change relative to the wild-type:kainate complex involves a twisting motion with the efficacy contribution opposite from that expected based on previous analyses. As a result, channel opening may involve transmembrane rearrangements with a significant rotational component. Furthermore, a two-dimensional analysis of agonist-induced GluA2 LBD motions suggests that efficacy is not a linearly varying function of lobe 2 displacement vectors, but is rather determined by specific conformational requirements of the transmembrane domains.


Assuntos
Agonistas de Aminoácidos Excitatórios/química , Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/metabolismo , Receptores de AMPA/agonistas , Receptores de AMPA/química , Transmissão Sináptica/fisiologia , Sítios de Ligação , Linhagem Celular , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cristalografia por Raios X , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Canais Iônicos/agonistas , Canais Iônicos/química , Ácido Caínico/farmacologia , Ligantes , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Proteômica , Receptores de AMPA/metabolismo
11.
J Neurosci ; 29(40): 12484-96, 2009 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-19812324

RESUMO

Anabolic androgenic steroids (AAS) can promote detrimental effects on social behaviors for which GABA type A (GABA(A)) receptor-mediated circuits in the forebrain play a critical role. While all AAS bind to androgen receptors (AR), they may also be aromatized to estrogens and thus potentially impart effects via estrogen receptors (ER). Chronic exposure of wild-type male mice to a combination of chemically distinct AAS increased action potential (AP) frequency, selective GABA(A) receptor subunit mRNAs, and GABAergic synaptic current decay in the medial preoptic area (mPOA). Experiments performed with pharmacological agents and in AR-deficient Tfm mutant mice suggest that the AAS-dependent enhancement of GABAergic transmission in wild-type mice is AR-mediated. In AR-deficient mice, the AAS elicited dramatically different effects, decreasing AP frequency, spontaneous IPSC amplitude and frequency and the expression of selective GABA(A) receptor subunit mRNAs. Surprisingly, in the absence of AR signaling, the data indicate that the AAS do not act as ER agonists, but rather suggest a novel in vivo action in which the AAS inhibit aromatase and impair endogenous ER signaling. These results show that the AAS have the capacity to alter neuronal function in the forebrain via multiple steroid signaling mechanisms and suggest that effects of these steroids in the brain will depend not only on the balance of AR- versus ER-mediated regulation for different target genes, but also on the ability of these drugs to alter steroid metabolism and thus the endogenous steroid milieu.


Assuntos
Metiltestosterona/administração & dosagem , Nandrolona/análogos & derivados , Neurônios/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Receptores de GABA/efeitos dos fármacos , Testosterona/análogos & derivados , Potenciais de Ação/efeitos dos fármacos , Anabolizantes/administração & dosagem , Antagonistas de Receptores de Andrógenos , Androgênios/administração & dosagem , Animais , Inibidores da Aromatase/farmacologia , Combinação de Medicamentos , Estradiol/metabolismo , Estrogênios/metabolismo , Estrogênios/farmacologia , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Transgênicos , Nandrolona/administração & dosagem , Decanoato de Nandrolona , Neurônios/metabolismo , Área Pré-Óptica/citologia , Receptores Androgênicos/administração & dosagem , Receptores Androgênicos/deficiência , Transmissão Sináptica/efeitos dos fármacos , Testosterona/administração & dosagem , Testosterona/metabolismo
12.
Biochemistry ; 47(52): 13831-41, 2008 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-19102704

RESUMO

AMPA receptors are glutamate-gated ion channels that are essential mediators of synaptic signals in the central nervous system. They form tetramers that are assembled as combinations of subunits GluR1-4, each of which contains a ligand-binding domain (LBD). Crystal structures of the GluR2 LBD have revealed an agonist-binding cleft, which is located between two lobes and which acts like a Venus flytrap. In general, agonist efficacy is correlated with the extent of cleft closure. However, recent observations show that cleft closure is not the sole determinant of the relative efficacy for glutamate receptors. In addition, these studies have focused on the GluR2 subunit, which is the specific target of a physiologically important RNA-editing modification in vivo. We therefore sought to test the generality of the cleft closure-efficacy correlation for other AMPA-R subunits. Here, we present crystal structures of the GluR4(flip) LBD in complex with both full and partial agonists. As for GluR2, both agonists stabilize a closed-cleft conformation, and the partial agonist induces a smaller cleft closure than the full agonist. However, a detailed analysis of LBD-kainate interactions reveals the importance of subtle backbone conformational changes in the ligand-binding pocket in determining the magnitude of agonist-associated conformational changes. Furthermore, the GluR4 subunit exhibits a different correlation between receptor activation and LBD cleft closure than does GluR2.


Assuntos
Receptores de AMPA/agonistas , Receptores de AMPA/química , Animais , Sítios de Ligação , Cristalografia por Raios X , Ligantes , Ligação Proteica , Conformação Proteica , Ratos , Receptores de AMPA/metabolismo
13.
J Neurochem ; 103(3): 1258-71, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17714454

RESUMO

Incorporation of the epsilon subunit into the GABAA receptor has been suggested to confer unusual, but variable, biophysical and pharmacological characteristics to both recombinant and native receptors. Due to their structural similarity with the gamma subunits, epsilon subunits have been assumed to substitute at the single position of the gamma subunit in assembled receptors. However, prior work suggests that functional variability in epsilon-containing receptors may reflect alternative sites of incorporation and of not just one, but possibly multiple epsilon subunits in the pentameric receptor complex. Here we present data indicating that increased expression of epsilon, in conjunction with alpha2 and beta3 subunits, results in expression of GABAA receptors with correspondingly altered rectification, deactivation and levels of spontaneous openings, but not increased total current density. We also provide data that the epsilon subunit, like the beta3 subunit, can self-export and data from chimeric receptors suggesting that similarities between the assembly domains of the beta3 and the epsilon subunits may allow the epsilon subunit to replace the beta, as well as the gamma, subunit. The substitution of an epsilon for a beta, as well as the gamma subunit and formation of receptors with alternative patterns of assembly with respect to epsilon incorporation may underlie the observed variability in both biophysical and pharmacological properties noted not only in recombinant, but also in native receptors.


Assuntos
Membrana Celular/metabolismo , Receptores de GABA-A/biossíntese , Linhagem Celular , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Modelos Moleculares , Inibição Neural/genética , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transporte Proteico/fisiologia , Receptores de GABA-A/química , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transmissão Sináptica/genética , Transfecção , Zinco/metabolismo , Zinco/farmacologia , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia
14.
Neuropharmacology ; 52(7): 1439-53, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17433821

RESUMO

The hypothalamus, the seat of neuroendocrine control, is exquisitely sensitive to gonadal steroids. For decades it has been known that androgens, estrogens and progestins, acting through nuclear hormone receptors, elicit both organizational and activational effects in the hypothalamus and basal forebrain that are essential for reproductive function. While changes in gene expression mediated by these classical hormone pathways are paramount in governing both sexual differentiation and the neural control of reproduction, it is also clear that steroids impart critical control of neuroendocrine functions through non-genomic mechanisms. Specifically, endogenous neurosteroid derivatives of deoxycorticosterone, progesterone and testosterone, as well and synthetic anabolic androgenic steroids that are self-administered as drugs of abuse, elicit acute effects via allosteric modulation of gamma-aminobutyric acid type A receptors. GABAergic transmission within the hypothalamus and basal forebrain is a key regulator of pubertal onset, the expression of sexual behaviors, pregnancy and parturition. Summarized here are the known actions of steroid modulators on GABAergic transmission within the hypothalamus/basal forebrain, with a focus on the medial preoptic area and the supraoptic/paraventricular nuclei that are known to be central players in the control of reproduction.


Assuntos
Hipotálamo/fisiologia , Receptores de GABA-B/fisiologia , Reprodução/fisiologia , Esteroides/metabolismo , Transmissão Sináptica/fisiologia , Animais , Humanos , Hipotálamo/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Esteroides/farmacologia , Transmissão Sináptica/efeitos dos fármacos
15.
Brain Res ; 1126(1): 122-38, 2006 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-17010954

RESUMO

Illicit use of anabolic androgenic steroids (AAS) has become a prevalent health concern not only among male professional athletes, but, disturbingly, among a growing number of women and adolescent girls. Despite the increasing use of AAS among women and adolescents, few studies have focused on the effects of these steroids in females, and female adolescent subjects are particularly underrepresented. Among the hallmarks of AAS abuse are changes in reproductive behaviors. Here, we discuss work from our laboratories on the actions of AAS on the onset of puberty and sexual behaviors in female rodents, AAS interactions and sex- and age-specific effects of these steroids on neural transmission mediated by gamma-aminobutyric acid receptors within forebrain neuroendocrine control regions that may underlie AAS-induced changes in these behaviors.


Assuntos
Sistemas Neurossecretores/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Congêneres da Testosterona/efeitos adversos , Ácido gama-Aminobutírico/metabolismo , Fatores Etários , Animais , Feminino , Humanos , Sistemas Neurossecretores/crescimento & desenvolvimento , Sistemas Neurossecretores/metabolismo , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Puberdade/efeitos dos fármacos , Puberdade/metabolismo , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Reprodução/fisiologia , Diferenciação Sexual/efeitos dos fármacos , Diferenciação Sexual/fisiologia , Comportamento Sexual Animal/fisiologia
16.
Endocrinology ; 147(9): 4192-204, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16777973

RESUMO

Endocrine-disrupting compounds (EDCs) may interfere with neuronal development due to high levels of accumulation in biological tissue and potentially aberrant steroid signaling. Treatment of dissociated embryonic Xenopus spinal cord neurons with the EDC, nonylphenol (NP), did not alter cell survival or neurite outgrowth but inhibited neurotrophin-induced neurite outgrowth, effects that were recapitulated by treatment with comparable concentrations of 17 beta-estradiol (E2) and beta-estradiol 6-(O-carboxy-methyl)oxime: BSA (E2-BSA), but not a synthetic androgen. Effects of NP were not inhibited by the nuclear estrogen receptor antagonist, ICI 182,780, but were inhibited by the G protein antagonist, pertussis toxin. Nerve growth factor (NGF)-induced neurite outgrowth in Xenopus neurons was shown to require MAPK signaling. NP did not affect TrkA expression, MAPK signaling, or phosphatidylinositol 3' kinase-Akt-glycogen synthase kinase 3 beta (PI3K-Akt-GSK3 beta) signaling in Xenopus. The ability of NP to inhibit NGF-induced neurite outgrowth without altering survival was recapitulated in the rat pheochromocytoma (PC12) cell line. As with Xenopus neurons, the inhibitory actions of NP in PC12 cells were not antagonized by ICI 182,780 and did not involve alterations in signaling along either the MAPK or PI3K-Akt-GSK3 beta pathways. NP did significantly inhibit the ability of NGF to increase protein kinase A activity in this cell line. These data have important implications with respect to potentially deleterious effects of NP exposure during early neural development and highlight the fact that bioaccumulation of EDCs, such as NP, may elicit very disparate effects along divergent signaling pathways than those that arise from the actions of physiological levels of endogenous estrogens.


Assuntos
Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Fenóis/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estradiol/farmacologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Quinases da Glicogênio Sintase/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuritos/fisiologia , Neuritos/ultraestrutura , Células PC12 , Toxina Pertussis/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/fisiologia , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/citologia , Medula Espinal/embriologia , Xenopus laevis/embriologia
17.
J Physiol ; 573(Pt 3): 571-93, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16543268

RESUMO

GABAergic transmission regulates the activity of gonadotrophin-releasing hormone (GnRH) neurons in the preoptic area/hypothalamus that control the onset of puberty and the expression of reproductive behaviours. One of the hallmarks of illicit use of anabolic androgenic steroids (AAS) is disruption of behaviours under neuroendocrine control. GnRH neurons are among a limited population of cells that express high levels of the epsilon-subunit of the GABAA receptor. To better understand the actions of AAS on neuroendocrine mechanisms, we have characterized modulation of GABAA receptor-mediated currents in mouse native GnRH neurons and in heterologous cells expressing recombinant alpha2beta3epsilon-receptors. GnRH neurons exhibited robust currents in response to millimolar concentrations of GABA and a picrotoxin (PTX)-sensitive, bicuculline-insensitive current that probably arises from spontaneous openings of GABAA receptors. The AAS 17alpha-methyltestosterone (17alpha-MeT) inhibited spontaneous and GABA-evoked currents in GnRH neurons. For recombinant alpha2beta3epsilon-receptors, 17alpha-MeT inhibited phasic and tonic GABA-elicited responses, accelerated desensitization and slowed paired pulse response recovery. Single channel analysis indicated that GABA-evoked events could be described by three open dwell components and that 17alpha-MeT enhanced residence in the intermediate dwell state. This AAS also inhibited a PTX-sensitive, spontaneous current (open probability, approximately 0.15-0.2) in a concentration-dependent fashion (IC50 approximately 9 microm). Kinetic modelling indicated that the inhibition induced by 17alpha-MeT occurs by an allosteric block in which the AAS interacts preferentially with a closed state and promotes accumulation in that state. Finally, studies with a G302S mutant epsilon-subunit suggest that this residue within the transmembrane domain TM2 plays a role in mediating AAS binding and modulation. In sum, our results indicate that inclusion of the epsilon-subunit significantly alters the profile of AAS modulation and that this allosteric inhibition of native GnRH neurons should be considered with regard to AAS disruption of neuroendocrine control.


Assuntos
Anabolizantes/toxicidade , Encéfalo/efeitos dos fármacos , Antagonistas GABAérgicos/toxicidade , Metiltestosterona/toxicidade , Receptores de GABA-A/efeitos dos fármacos , Potenciais de Ação , Regulação Alostérica , Sequência de Aminoácidos , Anabolizantes/metabolismo , Animais , Sítios de Ligação , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Antagonistas GABAérgicos/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Ativação do Canal Iônico , Canais Iônicos/efeitos dos fármacos , Cinética , Metiltestosterona/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estrutura Terciária de Proteína , Receptores de GABA-A/química , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Transfecção , Ácido gama-Aminobutírico/farmacologia
18.
Neuropharmacology ; 49(3): 300-16, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15993433

RESUMO

Neural transmission mediated by circuits expressing alpha2 subunit-containing gamma-aminobutyric acid type A (GABA(A)) receptors is critical for the expression of behaviors known to be altered by anabolic androgenic steroids (AAS). Here we show that micromolar concentrations of AAS, which reflect levels found in steroid abusers, induce positive modulation of currents from alpha2beta3 gamma2L recombinant receptors elicited by pulses of GABA that mimic synaptic conditions in a manner that is mechanistically distinct from modulation induced at alpha1beta3 gamma2L receptors. Specifically, at alpha2-containing receptors, the AAS, 17alpha-methyltestosterone (17alpha-MeT) enhanced peak current, slowed deactivation, diminished desensitization, and promoted entry of receptors into more distal states along the activation pathway. Analysis of GABA(A) receptor-mediated synaptic currents in primary cortical neurons followed by single cell real-time RT-PCR demonstrated that 17alpha-MeT enhancement of synaptic currents is proportional to the ratio of alpha2 to alpha1 subunit mRNA. Finally, we show that the modulation elicited by AAS is not comparable to that produced by micromolar concentrations of other positive allosteric modulators at alpha2-containing receptors. In sum, these data indicate that AAS elicit effects on GABA(A) receptor function that depend significantly on alpha subunit composition and that the mechanism of AAS modulation of GABA(A) receptors is distinct from that of other positive allosteric modulators.


Assuntos
Anabolizantes/farmacologia , Subunidades Proteicas/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Congêneres da Testosterona/farmacologia , Animais , Ansiolíticos/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Feminino , Agonistas GABAérgicos/farmacologia , Humanos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Estatísticos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Gravidez , Subunidades Proteicas/genética , Pirazóis/farmacologia , Piridinas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de GABA-A/genética , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinapses/fisiologia , Transfecção , Zolpidem , Ácido gama-Aminobutírico/farmacologia
19.
Neurosci Biobehav Rev ; 27(5): 413-36, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-14505684

RESUMO

Anabolic-androgenic steroids (AAS) are synthetic derivatives of testosterone originally designed for therapeutic uses to provide enhanced anabolic potency with negligible androgenic effects. Although AAS continue to be used clinically today, the medical benefits of low therapeutic doses of AAS stand in sharp contrast to the potential health risks associated with the excessive doses self-administered not only by elite athletes and body builders, but by a growing number of recreational users, including adolescent boys and girls. The deleterious effects of AAS on peripheral organs and the incidence of altered behaviors in AAS abusers have been well documented in a number of excellent current reviews for clinical populations. However, a comparable synthesis of nonclinical studies has not been made. Our purpose in this review is to summarize the literature for animal models of the effects of supraphysiological doses of AAS (e.g. those that mimic human abuse regimes) on behaviors and on the neural circuitry for these behaviors. In particular, we have focused on studies in rodents that have examined how AAS alter aggression, sexual behaviors, anxiety, reward, learning, and locomotion and how AAS alter the expression and function of neurotransmitter systems and other signaling molecules that underlie these behaviors.


Assuntos
Anabolizantes/efeitos adversos , Comportamento/efeitos dos fármacos , Receptores Androgênicos/efeitos dos fármacos , Esteroides/efeitos adversos , Agressão/efeitos dos fármacos , Animais , Ansiedade/induzido quimicamente , Modelos Animais de Doenças , Dopamina/metabolismo , Hormônios/metabolismo , Humanos , Aprendizagem/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Entorpecentes/metabolismo , Fatores de Crescimento Neural/metabolismo , Neuropeptídeos/metabolismo , Neurotransmissores/metabolismo , Receptores Androgênicos/metabolismo , Receptores Dopaminérgicos/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Receptores Opioides/efeitos dos fármacos , Receptores de Serotonina/efeitos dos fármacos , Serotonina/metabolismo , Comportamento Sexual/efeitos dos fármacos , Transtornos Relacionados ao Uso de Substâncias
20.
Horm Behav ; 43(4): 454-64, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12788291

RESUMO

ICI 182,780 (ICI) is a pure antiestrogen that when administered systemically does not cross the blood-brain barrier, thus its actions are limited to the periphery. Four experiments were conducted to test the effects of ICI on the display of sexual behaviors in ovariectomized rats. Experiment 1 examined the effects of three doses of ICI (250, 500, and 750 micro g/rat) on sexual receptivity and paced mating behavior in rats primed with estradiol benzoate (EB) in combination with progesterone (P). Experiments 2 and 3 compared the display of sexual behaviors in rats primed with EB+P or EB alone and administered either 250 micro g ICI (Experiment 2) or 500 micro g ICI (Experiment 3). Experiment 4 tested the effects of ICI (250 and 500 micro g) on the expression of estrogen-induced progestin receptors in the uterus. ICI did not affect the display of sexual receptivity in any experiment. In rats primed with EB+P, paced mating behavior was altered by the 500 and 750 micro g, but not the 250 micro g, doses of ICI. The lowest (250 micro g) dose of ICI did alter paced mating behavior in rats primed with EB alone. The effects of ICI on paced mating behavior were manifested by a substantial lengthening of contact-return latencies following intromissions and ejaculations. The percentage of exits were not affected by ICI. Estrogen stimulation of uterine weight and induction of uterine progestin receptors was suppressed by ICI (250 and 500 micro g). ICI effects on paced mating behavior in hormone-primed female rats are likely to reflect antiestrogenic actions in the periphery, including interference with the estrogen induction of progestin receptors.


Assuntos
Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Fulvestranto , Masculino , Tamanho do Órgão , Ovariectomia , Progesterona/farmacologia , Ratos , Ratos Long-Evans , Tempo de Reação/efeitos dos fármacos , Útero/anatomia & histologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...