Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
J Neuroendocrinol ; 25(10): 939-55, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23927378

RESUMO

Expression of the primary female sex behaviour, lordosis, in laboratory animals depends on oestrogen-induced expression of progesterone receptor (PgR) within a defined cell group in the ventrolateral portion of the ventromedial nucleus of the hypothalamus (VMH). The minimal latency from oestradiol administration to lordosis is 18 h. During that time, ligand-bound oestrogen receptors (ER), members of a nuclear receptor superfamily, recruit transcriptional coregulators, which induce covalent modifications of histone proteins, thus leading to transcriptional activation or repression of target genes. The present study aimed to investigate the early molecular epigenetic events underlying oestrogen-regulated transcriptional activation of the Pgr gene in the VMH of female mice. Oestradiol (E2) administration induced rapid and transient global histone modifications in the VMH of ovariectomised female mice. Histone H3 N-terminus phosphorylation (H3S10phK14Ac), acetylation (H3Ac) and methylation (H3K4me3) exhibited distinct temporal patterns facilitative to the induction of transcription. A transcriptional repressive (H3K9me3) modification showed a different temporal pattern. Collectively, this should create a permissive environment for the transcriptional activity necessary for lordosis, within 3-6 h after E2 treatment. In the VMH, changes in the H3Ac and H3K4me3 levels of histone H3 were also detected at the promoter region of the Pgr gene within the same time window, although they were delayed in the preoptic area. Moreover, examination of histone modifications associated with the promoter of another ER-target gene, oxytocin receptor (Oxtr), revealed gene- and brain-region specific effects of E2 treatment. In the VMH of female mice, E2 treatment resulted in the recruitment of ERα to the oestrogen-response-elements-containing putative enhancer site of Pgr gene, approximately 200 kb upstream of the transcription start site, although it failed to increase ERα association with the more proximal promoter region. Finally, E2 administration led to significant changes in the mRNA expression of several ER coregulators in a brain-region dependent manner. Taken together, these data indicate that, in the hypothalamus and preoptic area of female mice, early responses to E2 treatment involve highly specific changes in chromatin structure, dependent on cell group, gene, histone modification studied, promoter/enhancer site and time following E2.


Assuntos
Estradiol/administração & dosagem , Histonas/metabolismo , Hipotálamo/metabolismo , Área Pré-Óptica/metabolismo , Acetilação , Animais , Sequência de Bases , Western Blotting , Imunoprecipitação da Cromatina , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Feminino , Camundongos , Reação em Cadeia da Polimerase
2.
Behav Brain Res ; 229(2): 349-58, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22285420

RESUMO

Deep brain stimulation (DBS) has shown promise in the treatment of many neurological and psychiatric disorders as well as a disorder of consciousness, the minimally conscious state (MCS). In the clinic, DBS is always monotonic standard pulses; however, we have hypothesized that temporally patterned pulses might be more efficient in achieving desired behavioral responses. Here we present two experiments on DBS of the central thalamus to increase arousal, as measured by motor activity, and to affect the electroencephalogram (EEG). In the first, we optimized amplitude and frequency in standard stimulation of the central thalamus in intact mice. In the second, the optimized fixed frequency was compared to two alternative temporal patterns, chaotic and random, which were physically identical to each other and fixed frequency in all ways except temporal pattern. In both experiments and with all types of stimulation, DBS of the central thalamus increased arousal as measured by motor activity. These data also revealed that temporal patterning of pulses can modulate response to stimulation. That temporal patterns in DBS of the central thalamus were found to alter motor activity response implies possible usefulness of temporal patterns in DBS of other contexts. More investigation into exactly how temporally patterned stimulation may affect neuronal circuit dynamics is necessary.


Assuntos
Nível de Alerta/fisiologia , Ondas Encefálicas/fisiologia , Estimulação Encefálica Profunda/métodos , Modelos Logísticos , Tálamo/fisiologia , Animais , Estimulação Encefálica Profunda/estatística & dados numéricos , Eletroencefalografia/métodos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Atividade Motora/fisiologia , Fatores de Tempo
3.
J Neuroendocrinol ; 21(4): 406-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19226347

RESUMO

The sagittalis nucleus (SGN) of the hypothalamus is a newly-identified nucleus that is located in the interstitial area between the arcuate and ventromedial nuclei of the rat hypothalamus and for which the long axis of the nucleus is oriented sagittally. Interestingly, the SGN exhibits structural and physiological sex differences, as defined by Nissl staining and oestrogen receptor (ER)alpha immunoreactivity (-ir), being larger in males than females. The structural sex difference is established by sex steroid action in neonates because the treatment of female pups with testosterone propionate masculinised the SGN. The phenotypical sex difference in ERalpha-ir is mediated hormonally in adulthood. Ovariectomy of female rats caused a significant increase in ERalpha-ir in the SGN, and eliminated the physiological sex difference, but with recovery to the level of gonad-intact females when given oestradiol replacement. Adult females have oestrous cycle-related variations in ERalpha-ir in the SGN, with levels at a nadir during the evening of pro-oestrous. The discovery of the SGN, a target of sex steroid action, provides a new opportunity for explaining hormonal regulation of sexually-differentiated behavioural and endocrine functions.


Assuntos
Hipotálamo/anatomia & histologia , Hipotálamo/metabolismo , Caracteres Sexuais , Animais , Contagem de Células , Metabolismo Energético/fisiologia , Receptor alfa de Estrogênio/metabolismo , Ciclo Estral/fisiologia , Feminino , Hormônios Esteroides Gonadais/metabolismo , Hipotálamo/crescimento & desenvolvimento , Masculino , Neurônios/fisiologia , Tamanho do Órgão
4.
Horm Behav ; 54(3): 347-54, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18539279

RESUMO

Reverse engineering takes the facts we know about a device or a process and reasons backwards to infer the principles underlying the structure-function relations. The goal of this review is to apply this approach to a well-studied hormone-controlled behavior, namely the reproductive stance of female rodents, lordosis. We first provide a brief overview on the considerable amount of progress in the analysis of female reproductive behavior. Then, we propose an analysis of the mechanisms of this behavior from a reverse-engineering perspective with the goal of generating novel hypotheses about the properties of the circuitry elements. In particular, the previously proposed neuronal circuit modules, feedback signals, and genomic mechanisms are considered to make predictions in this manner. The lordosis behavior itself appears to proceed ballistically once initiated, but negative and positive hormonal feedback relations are evident in its endocrine controls. Both rapid membrane-initiated and slow genomic hormone effects contribute to the behavior's control. We propose that the value of the reverse-engineering approach is based on its ability to provide testable, mechanistic hypotheses that do not emerge from either traditional evolutionary or simple reductionistic perspectives, and several are proposed in this review. These novel hypotheses may generalize to brain functions beyond female reproductive behavior. In this way, the reverse-engineering perspective can further develop our conceptual frameworks for behavioral and systems neuroscience.


Assuntos
Engenharia Biomédica , Estrogênios/fisiologia , Postura/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Cricetinae , Retroalimentação Fisiológica/fisiologia , Feminino , Hipotálamo/fisiologia , Cinética , Mesencéfalo/fisiologia , Rede Nervosa/fisiologia , Redes Neurais de Computação , Neurônios/fisiologia , Propriocepção/fisiologia , Ratos , Receptores de Estrogênio/fisiologia , Transfecção
5.
J Neuroendocrinol ; 18(9): 692-702, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16879168

RESUMO

Rapidly emerging evidence suggests that glial cells in the central nervous system are sensitive to oestrogen actions. However, the functional consequences of the cellular mechanisms of these cells have proven difficult to study in vivo because of the intimate relationships between neurones and glia. Microarray technology offers the potential to uncover steroid hormone regulation of glial-specific genes that may play a role in hormone-dependent neuronal-glial interactions. Analysis of transcriptomes from the medial basal hypothalamus (MBH) of oestradiol and vehicle-treated adult ovariectomised mice revealed an up-regulation of several glial specific genes by oestradiol, including glutamine synthetase (GS), which facilitates the conversion of glutamate to glutamine and plays an integral role in amino acid neurotransmission. In situ hybridisation confirmed that oestradiol treatment resulted in an up-regulation of GS gene expression in the arcuate and ventromedial nuclei of the MBH, as well as the medial amygdala and hippocampus. Moreover, oestradiol increased protein expression of GS in both the MBH and hippocampus. Neurones are incapable of de novo net synthesis of glutamate from glucose and are dependent on glial-provided precursors such as glutamine to renew their amino acid transmitter pools. Thus, oestradiol induced expression of GS suggests a significant role for glial cells in hormonal modulation of glutamatergic neurotransmission important to female reproductive behaviours, neuroendocrine physiology and cognitive functions.


Assuntos
Estradiol/fisiologia , Glutamato-Amônia Ligase/metabolismo , Hipocampo/enzimologia , Hipotálamo/enzimologia , Neuroglia/metabolismo , Animais , Comunicação Celular/fisiologia , Feminino , Glutamato-Amônia Ligase/genética , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise , Distribuição Aleatória , Transmissão Sináptica/fisiologia , Regulação para Cima
6.
J Neuroendocrinol ; 14(3): 175-83, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11999716

RESUMO

Progesterone receptors play a central role in neuroendocrine and behavioural regulation. To gain insight into the sex- and tissue-specific regulation of progesterone receptors, protein binding on a progesterone receptor-oestrogen response element and mRNA levels for progesterone receptor (PR)-A and PR-B were compared between female and male rats following oestradiol benzoate replacement treatment in hypothalamic and pituitary tissue. Both male and female pituitary protein extracts demonstrated an increase in nuclear protein binding activity to a progesterone receptor-oestrogen response element following oestradiol benzoate treatment. However, there was a greater difference in total binding activity seen in the female pituitary extracts compared to male pituitary protein extracts. In both cases, reflecting the binding data, oestradiol benzoate pretreatment led to an increase in pituitary PR-B messenger RNA, although this increase was significantly larger in females than in males. Oestradiol benzoate treatment also led to a significant increase in specific binding of hypothalamic nuclear proteins to the progesterone receptor oestrogen response element from both females and male hypothalamic extracts. In addition, PR-B messenger RNA was induced by oestradiol benzoate treatment in the female rat hypothalamus, under circumstances where no PR-A could be detected. The male also demonstrated an increase in PR-B messenger RNA following oestradiol benzoate treatment, with undetectable levels of PR-A, although to a lesser degree than that seen in the female. The predominance of PR-B over PR-A messenger RNA in rat hypothalamus and pituitary, and the quantitative differences between female and male rats, could both contribute to the greater responsiveness of female rats to progesterone with respect to control over luteinizing hormone release from the pituitary, and lordosis behaviour regulated by hypothalamic neurones.


Assuntos
Estradiol/farmacologia , Hipotálamo/metabolismo , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Receptores de Progesterona/genética , Caracteres Sexuais , Animais , Feminino , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Útero/metabolismo
7.
J Comp Neurol ; 437(3): 286-95, 2001 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-11494256

RESUMO

Estrogen receptors (ER) and thyroid hormone receptors (TR) are members of the nuclear receptor family of transcription factors that induce or repress the expression of target genes. Previous behavioral studies in female rodents have demonstrated that thyroid hormones can antagonize the effects of estrogen in the central nervous system (CNS), particularly by attenuating estrogen's ability to facilitate reproductive behaviors. Additional molecular studies have suggested a mechanism for this antagonism by showing that ligand-activated ER alpha and TRs have the potential to interact in their transcriptional controls. Although the expression patterns of ER alpha and TRs in the rodent brain appear to overlap in behaviorally relevant areas, it remained to be determined whether these two classes of proteins coexist in vivo at the level of single neurons. To address this possibility, we employed a highly sensitive double-label in situ hybridization technique using digoxigenin and (35)S-labeled cRNA probes to analyze, in detail, the expression of ER alpha mRNA with TR alpha 1 and TR alpha 2 mRNAs in the same neurons of the ovariectomized (OVX) adult mouse brain. Our results demonstrate that a large majority of the ER alpha-positive neurons also expresses TR alpha 1 and TR alpha 2 mRNAs. Quantitative examination of the cellular expression in the ventromedial and arcuate nuclei of the hypothalamus (VMH and Arc) showed that 81.5% and 80.5% of the neurons endowed with ER alpha mRNA also contain TR alpha 1 and TR alpha 2 mRNAs, respectively. In the amygdala, more than 60.5% and 67% of ER alpha-positive cells also contain TR alpha 1 and TR alpha 2 mRNAs, respectively. These findings provide the first anatomical evidence that ER and TR can be found in the same neurons, including hypothalamic neurons. This coexpression of ER alpha and TR provides the cellular basis for a new level of neuronal integration in a brain region where estrogens control female reproductive behaviors.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hipotálamo/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Receptores dos Hormônios Tireóideos/genética , Tonsila do Cerebelo/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Receptor alfa de Estrogênio , Feminino , Hipotálamo/citologia , Hibridização In Situ , Camundongos , Neurônios/citologia , Isoformas de Proteínas/genética , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/metabolismo
8.
Brain Res Mol Brain Res ; 88(1-2): 144-54, 2001 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-11295240

RESUMO

Rodent female reproductive behavior is facilitated by the genomic targets of estrogen (E) and progesterone (P) in neuroendocrine regions of the brain. Using the differential display-PCR technique to identify these targets we discovered a novel hormone-sensitive mRNA in the female rat brain that is substantially reduced in the ventromedial hypothalamus (VMH) after 3 h of P treatment, following 24 h of E priming. Northern blots show that it is a single transcript of approximately 1.7 kb. The sequence of the corresponding full-length cDNA indicates that this gene is the rat homolog of mouse SCAMP-4, the fourth member identified in a family of proteins known as secretory carrier membrane proteins (SCAMPs). In situ hybridization studies show that SCAMP-4 mRNA is relatively low throughout the rat forebrain, with the highest levels observed in the VMH, habenula and hippocampus. The SCAMP-4 message is also less abundant in the habenula and VMH during proestrus, when circulating levels of E and P are at their peak, than during diestrus-1 when circulating hormone levels are low. Amino acid sequence analysis indicates that SCAMP-4 lacks the putative calcium binding and leucine zipper structures, as well as protein-protein interacting NPF domains common among most SCAMP family members, but is the only member identified to date to contain a putative protein kinase C (PKC) phosphorylation site. Fluorescent microscopy of cells transfected with a SCAMP-4/GFP fusion construct reveals distinct fluorescence in subcellular aggregates that may contain secretory vesicles. In addition to our results in the VMH, the finding of high levels of SCAMP-4 message in the habenula, a brain area rich in mast cells, together with previous reports linking mast cell secretion with courtship behavior also suggest a possible role for SCAMP-4 in reproductive behaviors associated with mast cell activity in the central nervous system (CNS).


Assuntos
Química Encefálica/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Progesterona/farmacologia , Comportamento Sexual Animal/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Química Encefálica/efeitos dos fármacos , Proteínas de Transporte/genética , DNA Complementar/análise , Diestro/fisiologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Perfilação da Expressão Gênica , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Dados de Sequência Molecular , Neuroblastoma , Ovariectomia , Hipófise/química , Hipófise/fisiologia , Postura/fisiologia , Proestro/fisiologia , Ratos , Ratos Sprague-Dawley , Vesículas Secretórias/química , Vesículas Secretórias/fisiologia , Células Tumorais Cultivadas
9.
Proc Natl Acad Sci U S A ; 97(23): 12816-21, 2000 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-11070092

RESUMO

The ventromedial hypothalamus (VMH) plays a central role in the regulation of the female reproductive behavior lordosis, a behavior dependent upon the sequential activation of receptors for the ovarian steroid hormones estradiol (E) and progesterone (P). These receptors function as transcription factors to alter the expression of target genes. To discover behaviorally relevant genes targeted by E and P in the VMH, we used the differential display PCR to identify messenger RNAs that are differentially expressed in the hypothalamus of ovariectomized (ovx) rats treated with E alone compared with ovariectomized rats treated with E and P. We show here that one interesting mRNA within the hypothalamus that is repressed by P after E priming encodes the protein 25-Dx, the rat homolog of the human membrane-associated P-binding protein Hpr6.6. Neurons in the brain containing the highest levels of 25-Dx are located in several nuclei of the basal forebrain, including the VMH. 25-Dx expression is also higher in the hypothalamus of female P receptor "knockout" mice than in their wild-type littermates. These findings suggest a mechanism in which the activation of nuclear P receptor represses expression of a membrane P receptor, 25-Dx, during lordosis facilitation.


Assuntos
Hipotálamo/metabolismo , Proteínas de Membrana/metabolismo , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Comportamento Sexual Animal , Animais , Sequência de Bases , Encéfalo/metabolismo , Encéfalo/patologia , Membrana Celular/metabolismo , DNA Complementar , Estradiol/administração & dosagem , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Expressão Gênica , Hipotálamo/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/metabolismo , Sistemas Neurossecretores/metabolismo , Postura/fisiologia , Progesterona/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/genética , Reprodução/fisiologia , Caracteres Sexuais
10.
Neurosci Res ; 38(1): 85-92, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10997581

RESUMO

We examined two molecular responses to estrogen, reduction in estrogen receptor alpha (ER alpha) mRNA and increase in progesterone receptor (PR) mRNA, in the hypothalamus of 3- (young) and 10-month-old (middle-aged) cycling, and 15-month-old (old) acyclic, Fischer 344 female rats. The rats were ovariectomized and then given silastic capsules containing 5% 17beta-estradiol. or empty implants, and killed 4 days after implantation. By means of in situ hybridization, we found that, in young rats, estrogen reduced ER alpha mRNA in both the ventromedial hypothalamus (VMH) and arcuate nucleus (ARC) but not in the preoptic area (POA). In contrast, the effect of estrogen on ER alpha mRNA in the VMH and ARC of middle-aged and old rats was not statistically significant. On the other hand in all regions the induction of PR mRNA by estrogen was at least as strong in middle-aged and old as in young rats. The present study revealed that the induction of PR mRNA by estrogen in the hypothalamus was not impaired with age but ER alpha mRNA in the VMH and ARC was significantly impaired with age, but not in the POA.


Assuntos
Envelhecimento/metabolismo , Hipotálamo/metabolismo , RNA Mensageiro/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Biomarcadores , Receptor alfa de Estrogênio , Estrogênios/metabolismo , Estrogênios/farmacologia , Feminino , Hipotálamo/efeitos dos fármacos , Hibridização In Situ , Ovariectomia , Área Pré-Óptica/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores de Estrogênio/genética , Receptores de Progesterona/genética
11.
J Neuroendocrinol ; 11(1): 1-10, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9918223

RESUMO

The oxytocin (OT) gene promoter has a composite hormone response element, such that several members of the steroid/thyroid hormone superfamily of nuclear receptors can interact at this response element in vitro. To investigate this in brain tissue, parallel to foregoing behavioural experiments, we used in situ hybridization histochemistry to seek interactions between estrogen and thyroid hormones on OT mRNA in the hypothalamus. In ovariectomized (OVX) rats, high doses of triiodothyronine (T3) elevated OT mRNA levels in the paraventricular (PVN) nucleus, while treatment with estradiol benzoate (EB) alone had no significant effect. In contrast, animals that were thyroidectomized (TX) in addition to OVX had dramatically elevated levels of OT gene expression in the PVN following EB treatment. That is, endogenous thyroid hormones interfered with EB-induction of gene expression. Moreover, in both OVX and TX/OVX animals, OT gene expression was reduced to values equivalent to controls when T3 was given together with EB. Particular subdivisions of the PVN responded differentially to T3 and EB treatment, demonstrating marked heterogeneity of OT-containing neurons in this nucleus. Thus, parallel to and perhaps related to the manner in which thyroid hormones reduced estrogen-stimulated behaviour, endogenous or exogenous thyroid hormones interfered with estrogen stimulation of OT mRNA. These data demonstrate competition between nuclear proteins, transcription factors, in hypothalamic neurons.


Assuntos
Estradiol/análogos & derivados , Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Neurônios/metabolismo , Ocitocina/genética , Hormônios Tireóideos/farmacologia , Animais , Estradiol/farmacologia , Feminino , Hibridização In Situ , Ovariectomia , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Tireoidectomia , Tri-Iodotironina/farmacologia
12.
Brain Res Mol Brain Res ; 55(1): 115-25, 1998 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-9645966

RESUMO

Ap-1 proteins such as Fos and Jun are nuclear transcription factors that have been postulated to function as third messengers in signal transduction pathways to regulate target gene expression. Using electrophoretic mobility shift assays (EMSA), we have studied estrogen (E) effects on regulation of AP-1 DNA binding activity in the rat hypothalamus and pituitary. AP-1 binding is defined herein as the specific association with a consensus AP-1 site during EMSA. Specific AP-1 binding activity was observed in nuclear extracts from the hypothalamus and pituitary of ovariectomized (OVX) female and castrated (CAS) male rats. Treatment with E increased the levels of AP-1 binding activity in the pituitary and uterus, whereas E decreased the levels of AP-1 binding in the hypothalamus, of OVX females. These effects were observed within 60 min and maintained for at least 72 h after a single dose of estrogen. Estrogen-induced changes in AP-1 binding were much more prominent in OVX females than in CAS males. Treatment with progesterone in OVX females had no significant effects on AP-1 binding activity in either pituitary or hypothalamus. Analysis of AP-1 binding activity in both hypothalamus and pituitary by supershift, immunodepletion and shift-Western blot indicated that part of the AP-1 binding was due to the presence of Fos and Jun proteins. However, Western blot analysis shows that the levels of Fos and Jun proteins in the hypothalamic nuclear extracts were not altered by E treatment. We conclude that E produced tissue and sex-differentiated alterations in AP-1 DNA binding activity in the hypothalamus and pituitary of female rats, which may be related to differential estrogenic actions on gene regulation.


Assuntos
DNA/metabolismo , Estradiol/análogos & derivados , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Hipófise/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Animais , Western Blotting , Núcleo Celular/metabolismo , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Hipotálamo/metabolismo , Masculino , Proteínas do Tecido Nervoso/química , Orquiectomia , Especificidade de Órgãos , Ovariectomia , Hipófise/metabolismo , Progesterona/farmacologia , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/análise , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-jun/análise , Proteínas Proto-Oncogênicas c-jun/genética , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Tamoxifeno/farmacologia , Testosterona/farmacologia , Fator de Transcrição AP-1/química , Útero/efeitos dos fármacos , Útero/metabolismo
13.
J Neuroendocrinol ; 9(4): 317-26, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9147295

RESUMO

Gonadal steroids and physiological stressors affect the regulation of proenkephalin (PPE) gene expression in the paraventricular (PVN) and ventromedial (VMH) hypothalamic nuclei. To examine the effects of these modulators at the cellular level, the current study utilized a transgenic mouse line that expresses a human proenkephalin promoter/bacterial beta-galactosidase fusion gene (ENK-1). Previous studies have demonstrated that the regulatory sequences included in this transgene are sufficient to support appropriate transcriptional regulation of the reported gene in the PVN of male ENK-1 mice in response to stress. The present experimental paradigm was designed to examine possible interactions of sex and circulating estrogen levels with the opioid responses to acute systemic stressors, an intraperitoneal injection of hypertonic (1.5 M) or isotonic (0.15 M) saline. Adult ENK-1 mice were gonadally intact, gonadectomized, or 21 days postpartum. Forty-eight hours before perfusion, castrated males and ovariectomized females received either 10 micrograms estradiol benzoate or oil vehicle and 4 animals per group received no further treatment. Six h before perfusion, remaining animals received a single intraperitoneal injection of either hypertonic or isotonic saline. Tissues were sectioned through the hypothalamus and processed for X-gal histochemistry. In the VMH of ovariectomized females that received isotonic saline, estrogen significantly elevated transgene expression. This effect was not seen in females that only received estrogen or in those that received the severe systemic stressor of a injection of hypertonic saline. Estrogen and stress did not interact to elevate transgene expression in the VMH of males. A different pattern of expression was observed in the PVN; injection of hypertonic saline induced transgene expression only in gonadally intact males and in castrated males given estrogen. These findings demonstrate that stress and estrogen have sex-specific and site-specific regulatory effects on the expression of a PPE promoter transgene in hypothalamic neurons.


Assuntos
Encefalinas/genética , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Precursores de Proteínas/genética , Estresse Fisiológico/metabolismo , beta-Galactosidase/genética , Animais , Feminino , Humanos , Hipotálamo Médio/metabolismo , Masculino , Camundongos , Ovariectomia , Núcleo Hipotalâmico Paraventricular/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão , Caracteres Sexuais
14.
Brain Res ; 740(1-2): 291-306, 1996 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-8973827

RESUMO

The distribution of the enzymes NADPH diaphorase and nitric oxide synthase in the ventromedial nucleus of the hypothalamus of cycling and ovariectomized/estrogen-treated and control female rats was demonstrated using histochemical and immunocytochemical methods. Serial section analysis of vibratome sections through the entire ventromedial nucleus showed that NADPH diaphorase cellular staining was localized primarily in the ventrolateral subdivision. NADPH diaphorase staining was visible in both neuronal perikarya and processes. Light microscopic immunocytochemistry using affinity-purified polyclonal antibodies to brain nitric oxide synthase revealed a similar pattern of labelling within the ventromedial nucleus and within neurons of the ventrolateral subdivision of the ventromedial nucleus. Control experiments involved omitting the primary antibodies; no labelling was visible under these conditions. Some, but not all, neurons in the ventrolateral subdivision of the ventromedial nucleus contained both NADPH diaphorase and brain nitric oxide synthase as demonstrated by co-localization of these two enzymes in individual cells of this area. That NADPH diaphorase and brain nitric oxide synthase were found in estrogen-binding cells was shown by co-localization of NADPH diaphorase and estrogen receptor and brain nitric oxide synthase and estrogen receptor at the light and ultrastructural levels, respectively. Our studies suggest that brain nitric oxide synthase is present and may be subject to estrogenic influences in lordosis-relevant neurons in the ventrolateral subdivision of the ventromedial nucleus. The hypothalamus is a primary subcortical regulatory center controlling sympathetic function. Therefore, not only is nitric oxide likely to be important for reproductive behavior, but also for the regulation of responses to emotional stress and other autonomic functions.


Assuntos
Hipotálamo/enzimologia , NADPH Desidrogenase/metabolismo , Óxido Nítrico Sintase/metabolismo , Animais , Feminino , Imuno-Histoquímica , Microscopia Eletrônica , NADPH Desidrogenase/ultraestrutura , Ovariectomia , Ratos , Ratos Sprague-Dawley
15.
Neuroendocrinology ; 64(5): 357-63, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8930936

RESUMO

Expression and estrogen regulation of the genes for nitric-oxide (NO)-synthesizing enzymes (NO synthase, NOS) were investigated by in situ hybridization. This study focused on regions of the hypothalamus that contain estrogen receptors and regulate specific neuroendocrine functions related to female sexual behavior and food intake, among others. Ovariectomized (OVX) rats were treated with vehicle or 3 micrograms/100 g estradiol benzoate (EB) for 7 days. Brains were sectioned and hybridized with antisense riboprobes for neuronal NOS, macrophage NOS and endothelial NOS. In the hypothalamus, mRNA was clearly detectable only for the neuronal NOS with the probes used. A strong hybridization signal was observed in the supraoptic paraventricular and ventromedial nuclei (SON, PVN and VMN, respectively). Quantitative analysis showed an increase in neuronal NOS mRNA in the VMN of the OVX rats treated with EB. The increase was mainly in the ventrolateral aspect of the VMN. No significant changes were observed in the hypothalamic SON and PVN. The data suggest that the expression of neuronal NOS mRNA in VMN can be regulated by estrogen.


Assuntos
Estradiol/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipotálamo/enzimologia , Óxido Nítrico Sintase/genética , RNA Mensageiro/metabolismo , Animais , Feminino , Humanos , Hipotálamo Médio/enzimologia , Imuno-Histoquímica , Hibridização In Situ , Ovariectomia , Ratos , Ratos Sprague-Dawley
16.
Proc Natl Acad Sci U S A ; 93(22): 12587-92, 1996 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-8901626

RESUMO

Estrogen receptor (ER) and thyroid hormone receptors (TRs) are ligand-dependent nuclear transcription factors that can bind to an identical half-site, AGGTCA, of their cognate hormone response elements. By in vitro transfection analysis in CV-1 cells, we show that estrogen induction of chloramphenicol acetyltransferase (CAT) activity in a construct containing a CAT reporter gene under the control of a minimal thymidine kinase (tk) promoter and a copy of the consensus ER response element was attenuated by cotransfection of TR alpha 1 plus triiodothyronine treatment. This inhibitory effect of TR was ligand-dependent and isoform-specific. Neither TR beta 1 nor TR beta 2 cotransfection inhibited estrogen-induced CAT activity, although both TR alpha and TR beta can bind to a consensus ER response element. Furthermore, cotransfection of a mutated TR alpha 1 that lacks binding to the AGGTCA sequence also inhibited the estrogen effect. Thus, the repression of estrogen action by liganded TR alpha 1 may involve protein-protein interactions although competition of ER and TR at the DNA level cannot be excluded. A similar inhibitory effect of liganded TR alpha 1 on estrogen induction of CAT activity was observed in a construct containing the preproenkephalin (PPE) promoter. A study in hypophysectomized female rats demonstrated that the estrogen-induced increase in PPE mRNA levels in the ventromedial hypothalamus was diminished by coadministration of triiodothyronine. These results suggest that ER and TR may interact to modulate estrogen-sensitive gene expression, such as for PPE, in the hypothalamus.


Assuntos
Estrogênios/fisiologia , Regulação da Expressão Gênica , Receptores de Estrogênio/genética , Receptores dos Hormônios Tireóideos/genética , Animais , Cloranfenicol O-Acetiltransferase/genética , Encefalinas/genética , Feminino , Genes Reporter , Hipofisectomia , Hipotálamo/metabolismo , Regiões Promotoras Genéticas , Precursores de Proteínas/genética , RNA Mensageiro/metabolismo , Ratos , Receptores de Estrogênio/fisiologia , Receptores dos Hormônios Tireóideos/fisiologia , Timidina Quinase/genética , Transcrição Gênica , Tri-Iodotironina/farmacologia
17.
J Neuroendocrinol ; 8(5): 325-36, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8736432

RESUMO

In the absence of universal equations expressing neurobiological findings, the safest theoretical approach for the neuroendocrinologist is to start from axiomatic requirements for biologically adaptive neural mechanisms, in our case for reproduction. From this emerge two themes: the likely importance of interactions between internal (hormonal) and external signals in controlling gene expression relevant to reproductive functions; and, second, the vision of molecular interactions on DNA subserving environmental impacts on reproduction. The first theoretical notion has so far yielded data showing a role for synaptic inputs during the onset of estradiol actions for the hormone's induction of enkephalin mRNA, a finding which parallels earlier behavioral results. As well, noxious somatosensory inputs interact with estrogens and progesterone in their influence on enkephalin gene expression. The second theme led to novel investigations of thyroid influences on reproductive molecular biology and behavior, including the ability of exogenous or endogenous thyroid hormones to reduce female mating responses. Since elevated thyroid hormone levels could signal environmental cold, our experiments offer the possibility of explaining ethological facts at a molecular level. More generally, nuclear hormone receptor interactions on the surface of DNA may offer a new level of neural integration revealed first by hormone effects in neuroendocrine cells.


Assuntos
Hipotálamo/fisiologia , Reprodução/fisiologia , Animais , Poluentes Ambientais/farmacologia , Expressão Gênica/fisiologia , Hormônios/genética , Humanos , Hipotálamo/citologia , Biologia Molecular , Sistemas Neurossecretores/fisiologia , Reprodução/genética
18.
Neuroendocrinology ; 62(5): 454-66, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8559277

RESUMO

Preproenkephalin (PPE) gene expression is specifically induced by estrogen in hypothalamus of ovariectomized (OVX) females, better than in male rats. To study estrogen actions on gene regulation, we have presently characterized protein-DNA interactions by use of a consensus estrogen response element (ERE) and a putative ERE from PPE gene, with nuclear extracts from hypothalamus. By use of the electrophoretic mobility shift assay (EMSA), ERE binding activity was detected in nuclear extracts from neuronal tissues including hypothalamus, hippocampus, striatum, cerebellum and frontal cortex, and non-neuronal tissues such as pituitary and uterus, but not lung of OVX female rats with a consensus ERE, as well as a 129-bp PCR fragment from PPE promoter and a hairpin oligonucleotide that contains a putative ERE of the rat PPE gene. The ERE binding was eliminated by the addition of specific ERE-containing oligonucleotide, but not control oligonucleotides. Protein and DNA associated and dissociated very rapidly. By use of supershift assay, interactions of estrogen receptor with ERE were demonstrated in hypothalamic nuclear extracts. The initial levels of specific ERE binding in the hypothalamic nuclear extracts were comparable between castrated male and OVX female rats. However, estrogen treatment, either estradiol or estradiol benzoate, produced a rapid and tissue-specific induction of a slow mobility complex of ERE binding in hypothalamic nuclear extracts from females, better than in male rats, presumably from other associated factors, or a conformational change or other posttranslational modifications. This estrogen-induced slow mobility complex of ERE binding in hypothalamus was not observed after treatment with progesterone or tamoxifen. These results suggest that specific ERE binding is present in rat hypothalamic nuclear proteins, which may contribute to the upregulation of PPE gene expression by estrogen, and that the sexually differentiated action of estrogen may be related to an estrogen-induced conformational change, but not to the initial level of ERE-binding activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Encefalinas/genética , Estrogênios/farmacologia , Hipotálamo/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Precursores de Proteínas/genética , Animais , Ligação Competitiva , Feminino , Expressão Gênica/genética , Cinética , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
19.
J Comp Neurol ; 360(4): 685-97, 1995 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-8801259

RESUMO

Two separate forms of glutamic acid decarboxylase, now termed GAD65 and GAD67, are the rate limiting enzymes for synthesis of gamma-aminobutyric acid (GABA). Because of the significance of GABA to neuroendocrine processes, numerous attempts have been made to determine the impact of gonadal steroids on enzyme functioning with inconclusive results. Therefore, we attempted to determine the impact of estradiol on mRNA levels for each form of GAD by quantitative in situ hybridization histochemistry in various brain regions. Ovariectomized rats were treated with estradiol benzoate or oil vehicle on 2 consecutive days and the brains collected on the third day. DNA probes specific for GAD65 and GAD67 were radiolabeled with CTP32 using asymmetric polymerase chain reaction. Results of in situ hybridizations for each probe on alternate sections from the same animals were analyzed for magnocellular preoptic area (McPOA), dorsal medial nucleus of the hypothalamus (DMN), zona incerta (ZI), and midbrain central gray (MCG). In the McPOA, estradiol exerted opposite effects on the frequency distribution of pixels per cell for two GAD mRNA probes, significantly increasing GAD65 (P < .05) and decreasing GAD67 (P < .01; Kolmogorov-Smirnov). In the DMN, estradiol treatment significantly increased GAD67 by 60% (P < .05; two-way ANOVA) but decreased GAD65 mRNA by 73% (P < .01). Note the direction of effects are opposite between McPOA and DMN. In MCG, analysis showed no estradiol effect on GAD mRNA levels/cells, but the proportion of cells expressing detectable levels of GAD65 or GAD67 increased by 33-40% in estradiol-treated rats (chi 2, P < .001).


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Estrogênios/farmacologia , Glutamato Descarboxilase/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/efeitos dos fármacos , Animais , Sondas de DNA , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hibridização In Situ , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/metabolismo
20.
Brain Res Mol Brain Res ; 28(1): 129-34, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7707866

RESUMO

To gain a better understanding of the relationship between the female rat reproductive system and preproenkephalin (PPE) expressing neurons under physiological conditions, we examined changes in PPE mRNA levels in the mediobasal hypothalamus during the rat estrous cycle by means of northern blotting and in situ hybridization histochemistry (ISHH). In the Northern blot studies, we found that PPE mRNA levels in the mediobasal hypothalamus were significantly increased by noon of proestrus compared to those in the morning and stayed high until diestrus day 1, and returned toward low levels on diestrous day 2. In contrast, measured as controls, glyceraldehyde-3-phosphate-dehydrogenase mRNA levels were significantly higher on proestrus regardless of time of day compared to diestrus day 2, and levels of calcineurin mRNA on proestrous and estrous were significantly lower than diestrous day 1 and day 2. ISHH studies revealed that these changes in PPE mRNA levels were specific in the ventromedial hypothalamic nucleus pars ventrolateralis (VMHVL), since we could not see any significant changes in signal in other parts including ventromedial hypothalamic nucleus pars dorsomedialis and arcuate hypothalamic nucleus. In the VMHVL, PPE mRNA levels in the afternoon of proestrous were significantly higher than those in the afternoon of diestrous day 2 whereas no significant change in PPE mRNA was observed in the caudate-putamen. The present study provides additional information relevant to possible implications of PPE gene expression in female reproductive systems, since changes in PPE mRNA levels may be associated with estrogen as well as progesterone or other hormonal concentrations during the estrous cycle.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Núcleo Caudado/metabolismo , Encefalinas/biossíntese , Estro/fisiologia , Hipotálamo/metabolismo , Precursores de Proteínas/biossíntese , Animais , Autorradiografia , Encefalinas/metabolismo , Feminino , Hibridização In Situ , Precursores de Proteínas/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA