Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters











Publication year range
1.
Reproduction ; 135(5): 733-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18304985

ABSTRACT

Two experiments were conducted to determine whether an estradiol challenge could cause a female-type LH surge in castrated male- and female-oriented rams (MORs and FORs). Administration of 17beta-estradiol to castrated MORs and FORs and ovariectomized ewes caused an initial reduction in LH secretion followed for 12-20 h by a surge release of LH in the ewes. No surge release of LH occurred in the MORs and FORs. The pattern of changes in LH secretion within rams and ewes did not differ between the breeding and nonbreeding seasons. Treatment failed to elicit female-typical receptive sexual behaviors in the rams but did stimulate increased sexual receptivity in the ewes as determined by the measures of responsiveness to the teaser ram. Overall, no differences were found in hypothalamic-hypophyseal function in response to exogenous estradiol between MORs and FORs. These data are interpreted to suggest that in contrast to sexual attraction, the neural mechanisms controlling the LH surge and female receptivity are defeminized in MORs.


Subject(s)
Estradiol/pharmacology , Luteinizing Hormone/blood , Sheep/blood , Animals , Castration , Female , Luteinizing Hormone/antagonists & inhibitors , Male , Pregnenediones/pharmacology , Progesterone/blood , Seasons , Secretory Rate/drug effects , Sexual Behavior, Animal
2.
Endocrine ; 29(3): 501-11, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16943590

ABSTRACT

This study tested the hypothesis that aromatization of testosterone to estradiol is necessary for sexual differentiation of the sheep brain. Pregnant ewes (n = 10) were treated with the aromatase inhibitor 1,4,6- androstatriene-3,17-dione (ATD) during the period of gestation when the sheep brain is maximally sensitive to the behavior-modifying effects of exogenous testosterone (embryonic d 50-80; 147 d is term). Control (n = 10) ewes received vehicle injections. Fifteen control lambs (7 males and 8 females) and 17 ATD-exposed lambs (7 males and 10 females) were evaluated for sexually dimorphic behavioral and neuroendocrine traits as adults. Prenatal ATD exposure had no significant effect on serum concentrations of androgen at birth, growth rates, expression of juvenile play behaviors, or the onset of puberty in male and female lambs. Rams exposed to ATD prenatally exhibited a modest, but significant, decrease in mounting behavior at 18 mo of age. However, prenatal ATD exposure did not interfere with defeminization of adult sexual partner preferences, receptive behavior, or the LH surge mechanism. In summary, our results indicate that aromatization is necessary for complete behavioral masculinization in sheep. However, before we can conclude that aromatization does not play a role in defeminization of the sheep brain, it will be necessary to evaluate whether intrauterine exposure of male fetuses to higher doses of ATD for a more extended period of time can disrupt normal neuroendocrine and behavioral development.


Subject(s)
Aromatase Inhibitors/pharmacology , Brain/drug effects , Brain/embryology , Pregnancy, Animal , Sex Differentiation/drug effects , Androstatrienes/pharmacology , Animals , Animals, Newborn/blood , Animals, Newborn/growth & development , Behavior, Animal , Female , Fertility/drug effects , Luteinizing Hormone/metabolism , Male , Maternal-Fetal Exchange/drug effects , Pregnancy , Pregnancy, Animal/blood , Sexual Maturation , Sheep , Steroids/blood , Testosterone/blood
3.
J Comp Neurol ; 439(2): 208-23, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11596049

ABSTRACT

This study mapped the regional locations of cells expressing cytochrome P450 aromatase (P450AROM) and androgen receptor (AR) mRNAs in the adult male macaque hypothalamus and amygdala by in situ hybridization histochemistry using monkey-specific cRNA probes. High densities of P450AROM and AR mRNA-containing neurons were observed in discrete hypothalamic areas involved in the regulation of gonadotropin secretion and reproductive behavior. P450AROM mRNA-containing neurons were most abundant in the medial preoptic nucleus, bed nucleus of the stria terminalis, and anterior hypothalamic area, whereas AR mRNA-containing neurons were most numerous in the ventromedial nucleus, arcuate nucleus, and tuberomamillary nucleus. Moderate to heavily labeled P450AROM mRNA-containing cells were present in the cortical and medial amygdaloid nuclei, which are known to have strong reciprocal inputs with the hypothalamus. Heavily labeled P450AROM mRNA-containing cells were found in the accessory basal amygdala nucleus, which projects to the cingulate cortex and hippocampus, areas that are important in the expression of emotional behaviors and memory processing. In contrast to P450AROM, the highest density of AR mRNA labeling in the temporal lobe was associated with the cortical amygdaloid nucleus and the pyramidal cells of the hippocampus. All areas that contained P450AROM mRNA-expressing cells also contained AR mRNA-expressing cells, but there were areas in which AR mRNA was expressed but not P450AROM mRNA. The apparent relative differences in the expression of P450AROM and AR mRNA-containing neurons within the monkey brain suggests that T acts through different signaling pathways in specific brain areas or within different cells from the same region.


Subject(s)
Amygdala/metabolism , Aromatase/genetics , Hypothalamus/metabolism , Macaca fascicularis/metabolism , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Reproduction/physiology , Amygdala/cytology , Animals , Base Sequence , Biological Assay , DNA, Complementary/chemistry , Hypothalamus/cytology , In Situ Hybridization , Macaca fascicularis/anatomy & histology , Male , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/analysis , Septal Nuclei/cytology , Septal Nuclei/metabolism , Sequence Homology, Nucleic Acid , Sex Factors
4.
J Steroid Biochem Mol Biol ; 79(1-5): 247-53, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11850231

ABSTRACT

In adult male primates, estrogens play a role in both gonadotropin feedback and sexual behavior. Inhibition of aromatization in intact male monkeys acutely elevates serum levels of luteinizing hormone, an effect mediated, at least partially, within the brain. High levels of aromatase (CYP19) are present in the monkey brain and regulated by androgens in regions thought to be involved in the central regulation of reproduction. Androgens regulate aromatase pretranslationally and androgen receptor activation is correlated with the induction of aromatase activity. Aromatase and androgen receptor mRNAs display both unique and overlapping distributions within the hypothalamus and limbic system suggesting that androgens and androgen-derived estrogens regulate complimentary and interacting genes within many neural networks. Long-term castrated monkeys, like men, exhibit an estrogen-dependent neural deficit that could be an underlying cause of the insensitivity to testosterone that develops in states of chronic androgen deficiency. Future studies of in situ estrogen formation in brain in the primate model are important for understanding the importance of aromatase not only for reproduction, but also for neural functions such as memory and cognition that appear to be modulated by estrogens.


Subject(s)
Aromatase/metabolism , Brain/enzymology , Androgens/metabolism , Animals , Aromatase/genetics , Brain/physiology , Drug Resistance , Estrogens/metabolism , Feedback , Gene Expression Regulation, Enzymologic , Humans , Luteinizing Hormone/metabolism , Macaca mulatta , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Sexual Behavior, Animal/physiology , Testosterone/metabolism , Testosterone/pharmacology , Tissue Distribution
5.
Endocrine ; 16(1): 15-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11822822

ABSTRACT

To understand the role of estrogen in testicular and epididymal function of rhesus monkeys, we measured steroids in the spermatic and peripheral venus circulation and aromatase activity and its mRNA in testis and epididymis. Testosterone, estradiol-17beta, and estrone, but not androstenedione, were elevated in the spermatic vein serum compared to the peripheral circulation. Aromatase activity in testis and in caput epididymis (259+/-16 [SEM] vs 274+/-47 fmol of 3H2O/mg of protein/h [n = 10], respectively) was significantly higher (p < 0.01) than in corpus and cauda (124+/-28 and 113+/-33 fmol of 3H2O/mg of protein/h [n = 10], respectively). In the ribonuclease protection assay, two P450arom mRNA transcripts were identified in testis and epididymis. One corresponded with the aromatase full-length transcript and the other was a truncated isoform. The latter was significantly more abundant than the former (p < 0.01). Our results demonstrate that the monkey testis and, to a lesser extent, the epididymis can aromatize androgens. However, in the epididymis, like in some areas of the brain, there was a discrepancy between the aromatase activity and the mRNA. The fact that P450arom mRNA and aromatase activity do not correlate in the epididymis may indicate that aromatase activity is not strictly regulated at the level of RNA expression and that other mechanisms for this regulation should be considered.


Subject(s)
Aromatase/metabolism , Epididymis/enzymology , Macaca mulatta/metabolism , Testis/enzymology , Animals , Aromatase/genetics , Male , RNA, Messenger/metabolism , Tissue Distribution
6.
Biol Reprod ; 63(3): 872-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10952934

ABSTRACT

Testosterone at physiological levels cannot exert negative feedback action on LH secretion in long-term castrated male monkeys. The cellular basis of this refractoriness is unknown. To study it, we compared two groups of male rhesus macaques: one group (group 1, n = 4) was castrated and immediately treated with testosterone for 30 days; the second group (group 2, n = 4) was castrated and treated with testosterone for 9 days beginning 21 days after castration. Feedback control of LH by testosterone in group 1 was normal, whereas insensitivity to its action was found in group 2. Using the endpoints of concentrations of aromatase activity (P450(AROM) messenger RNA [mRNA]) and androgen receptor mRNA in the medial preoptic anterior hypothalamus and in the medial basal hypothalamus, we found that aromatase activity in both of these tissues was significantly lower, P: < 0.01, in group 2 compared with group 1 males. P450(AROM) mRNA and androgen receptor mRNA did not differ, however. Our data suggest that the cellular basis of testosterone insensitivity after long-term castration may reside in the reduced capacity of specific brain areas to aromatize testosterone. Because P450(AROM) mRNA did not change in group 2 males, we hypothesize that an estrogen-dependent neural deficit, not involving the regulation of the P450(AROM) mRNA, occurs in long-term castrated monkeys.


Subject(s)
Luteinizing Hormone/metabolism , Orchiectomy , Testosterone/pharmacology , Androstenedione/blood , Animals , Aromatase/genetics , Aromatase/metabolism , Circadian Rhythm , Dihydrotestosterone/blood , Estradiol/blood , Feedback , Hypothalamus, Anterior/chemistry , Hypothalamus, Middle/chemistry , Luteinizing Hormone/blood , Macaca mulatta , Male , Preoptic Area/chemistry , RNA, Messenger/analysis , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Testosterone/blood , Testosterone/metabolism
7.
J Trauma Stress ; 13(3): 369-80, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10948479

ABSTRACT

Information involving the development of the DSM-IV version of the Children's PTSD Inventory is described. Independent ratings by highly experienced judges denote that the instrument encompassed the universe of definition that it was intended to measure (i.e., the DSM-IV criteria for PTSD). The instrument was administered to 82 traumatized and 22 nontraumatized youths at Bellevue Hospital. Moderate to high Cronbach alphas (.53-.89) were evident at the subtest level. An alpha of .95 was evident at the diagnostic level. In terms of inter-rater reliability, 98.1% agreement was evident at the diagnostic level. Inter-rater intraclass correlation coefficients (ICCs) ranged from .88 to .96 at the subtest level and .98 at the diagnostic level. Good to excellent kappas (.66-1.00) were reported for inter-rater reliability at the subtest level. An inter-rater reliability kappa of .96 was evident at the diagnostic level. In terms of test-retest reliability, 97.6% agreement was evident at the diagnostic level. Good to excellent test-retest kappas (.66-1.00) and ICCs (.66-.94) were observed. A test-retest kappa of .91 and an ICC of .88 was observed at the diagnostic level.


Subject(s)
Psychiatric Status Rating Scales/standards , Stress Disorders, Post-Traumatic/classification , Adolescent , Child , Female , Humans , Male , Observer Variation , Psychology, Adolescent , Psychology, Child , Reproducibility of Results , Sensitivity and Specificity , Urban Population
8.
J Neuroendocrinol ; 12(7): 656-64, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10849210

ABSTRACT

The regional distribution of neurones expressing aromatase mRNA in the ram hypothalamus was examined by in situ hybridization using 33P-labelled cRNA probes. The highest amounts of hybridization signal were observed in the central part of the medial preoptic nucleus and posterior medial part of the bed nucleus of the stria terminalis. Moderate amounts of hybridization signal were observed in the anteroventral periventricular preoptic nucleus, medial preoptic nucleus and a broad band extending between the medial preoptic nucleus and bed nucleus of the stria terminalis. Low levels of hybridization signal were observed in the organum vasculosum of the lamina terminalis, anterior part of the medial preoptic nucleus, and central part of the ventromedial nucleus of the hypothalamus. The presence of aromatase mRNA within neurones of the steroid-sensitive hypothalamic circuit supports a role for aromatization in the mechanism of testosterone action on reproductive function in male sheep. The distribution of aromatase mRNA in the ovine hypothalamus was similar to that described for other vertebrate species, suggesting a high degree of functional conservation across species.


Subject(s)
Aromatase/genetics , Hypothalamus/metabolism , RNA, Messenger/metabolism , Sheep/metabolism , Animals , Base Sequence/genetics , DNA Transposable Elements/genetics , DNA, Complementary/genetics , In Situ Hybridization , Male , Molecular Sequence Data , Tissue Distribution
9.
Biol Reprod ; 62(6): 1818-22, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10819787

ABSTRACT

We demonstrated previously that testosterone regulates aromatase activity in the anterior/dorsolateral hypothalamus of male rhesus macaques. To determine the level of the androgen effect, we developed a ribonuclease protection assay to study the effects of testosterone or dihydrotestosterone (DHT) on aromatase (P450(AROM)) mRNA in selected brain areas. Adult male rhesus monkeys were treated with testosterone or DHT. Steroids in serum were quantified by RIA. Fourteen brain regions were analyzed for P450(AROM) mRNA. Significant elevations of its message over controls (P<0.05) were found in the medial preoptic area/anterior hypothalamus of both androgen treatment groups and the medial basal hypothalamus of the testosterone-treated males. Other brain areas were not affected by androgen treatment. We conclude that testosterone and DHT regulate P450(AROM) mRNA in brain regions that mediate reproductive behaviors and gonadotropin release. The P450(AROM) mRNA of other brain areas is not androgen dependent. Brain-derived estrogens may also be important for maintaining neural circuitry in brain areas not related to reproduction. The control of P450(AROM) mRNA in these areas may differ from what we report here, but it is equally important to understand the function of in situ estrogen formation in these areas.


Subject(s)
Androgens/pharmacology , Aromatase/genetics , Brain/metabolism , Gene Expression Regulation, Enzymologic/drug effects , RNA, Messenger/metabolism , Animals , Dihydrotestosterone/blood , Dihydrotestosterone/pharmacology , Estradiol/blood , Hypothalamus, Anterior/metabolism , Hypothalamus, Middle/metabolism , Macaca mulatta , Male , Preoptic Area/metabolism , Testosterone/blood , Testosterone/pharmacology
11.
Biol Reprod ; 60(5): 1251-6, 1999 May.
Article in English | MEDLINE | ID: mdl-10208992

ABSTRACT

Because the distribution and hormonal regulation of the androgen receptor (AR) mRNA in brains and pituitaries of adult rhesus monkeys have not been studied, we cloned and sequenced a 329-base pair segment of the 5' coding region of the rhesus AR cDNA. Monkey AR cDNA was 99% identical with the human sequence and 96% homologous with the rat sequence. Using a ribonuclease protection assay, we studied the distribution and regulation of AR mRNA in brains and anterior pituitary glands of three groups of male rhesus monkeys: intact (n = 3), castrated (Cx, n = 4), and Cx treated with testosterone (n = 6). Serum testosterone levels of Cx males treated with testosterone differed significantly (p < 0.05) in the morning but not in the evening hours from those in intact controls. Serum LH concentrations were significantly suppressed (p < 0.05) in both morning and evening serum samples of testosterone-treated males compared to intact controls. We found the highest concentrations of AR mRNA in the medial basal hypothalamus, the bed nucleus of the stria terminalis, the medial preoptic area-anterior hypothalamus, and the lateral dorsomedial hypothalamus. Intermediate amounts were found in the septum and amygdala. Low amounts were found in the hippocampus, cingulate cortex, parietal cortex, and cerebellum. The anterior pituitary gland also contained a large amount of AR mRNA. Surprisingly, neither Cx for 3 wk nor Cx plus testosterone replacement for 3 wk significantly affected AR mRNA in any brain area or in the pituitary gland. The present study demonstrates that the effectiveness of testosterone as a regulator of LH secretion in male monkeys is not related to changes of AR mRNA in the brain or pituitary gland. It appears that AR mRNA in the monkey brain and pituitary gland is not regulated at the transcriptional level by androgen.


Subject(s)
Brain Chemistry/genetics , Luteinizing Hormone/metabolism , Pituitary Gland/metabolism , RNA, Messenger/biosynthesis , Receptors, Androgen/biosynthesis , Animals , Base Sequence , Macaca mulatta , Male , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/metabolism
12.
J Reprod Fertil Suppl ; 54: 259-69, 1999.
Article in English | MEDLINE | ID: mdl-10692860

ABSTRACT

The components of heterosexual behaviour in rams are reviewed as a basis for understanding partner preference behaviour. A small percentage of rams will not mate with oestrous females and if given a choice will display courtship behaviour towards another ram in preference to a female. Some of the endocrine profiles of these male-oriented rams differ from those of heterosexual controls. These differences include reduced serum concentrations of testosterone, oestradiol and oestrone, reduced capacity to produce testosterone in vitro, and reduced capacity to aromatize androgens in the preoptic-anterior hypothalamus of the brain. Our observation that aromatase activity is significantly lower in the preoptic-anterior hypothalamic area of male-oriented rams than in female-oriented rams may indicate an important neurochemical link to sexual behaviour that should be investigated. The defect in steroid hormone production by the adult testes of the male-oriented ram may represent a defect that can be traced to the fetal testes. If this contention is correct, partner preference behaviour of rams may also be traceable to fetal development and represent a phenomenon of sexual differentiation.


Subject(s)
Aromatase/metabolism , Gonadal Steroid Hormones/physiology , Hypothalamus/metabolism , Sexual Behavior, Animal/physiology , Sheep/physiology , Testis/metabolism , Animals , Brain/enzymology , Breeding , Hypothalamus/enzymology , Male , Orchiectomy , Sex Differentiation/physiology
13.
Endocrine ; 11(1): 23-30, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10668637

ABSTRACT

Immunocytochemical detection of androgen receptors (ARs) in several compartments of the macaque ovary, including the germinal epithelium, follicle, and corpus luteum, suggests a role for androgens in modulating ovarian function via the classical receptor-mediated pathway. To examine AR mRNA expression in the rhesus monkey ovary, total RNA was isolated from whole ovaries, the germinal epithelium-enriched cortical and medullary compartments of the ovary, and corpora lutea from early (d 3-5), mid (d 6-8), mid-late (d 10-12), and late (d 13-15) stages of the luteal phase of the menstrual cycle. RNA was also obtained from luteinized granulosa cells from monkeys receiving gonadotropin treatment to stimulate the development of multiple ovarian follicles. After reverse transcription of total RNA using oligo-dT as a primer, polymerase chain reaction (PCR) was used to amplify a unique 329 bp segment of the monkey AR hormone-binding region. Reverse transcriptase (RT)-PCR products of the expected size were detected in all ovarian and control tissues. Sequence analysis of the AR cDNA from the macaque ovary revealed 99% nucleotide homology and 100% predicted amino acid homology to the cDNA for the hormone-binding region of human AR. Northern analysis demonstrated the presence of a major AR mRNA species at 9.5 kb in corpus luteum, luteinized granulosa cells, and prostate, with additional bands detected in the corpus luteum and prostate at 7.9 and 3.4 kb, respectively. A sensitive RNase protection assay was used to examine AR mRNA levels in ovarian tissues and showed AR mRNA expression throughout the life-span of the corpus luteum. Thus, detection of AR mRNA in the primate ovary, including the periovulatory follicle and corpus luteum, supports the concept that these tissues are targets for receptor-mediated androgen action during the menstrual cycle.


Subject(s)
Ovary/metabolism , RNA, Messenger/biosynthesis , Receptors, Androgen/biosynthesis , Animals , Autoradiography , Blotting, Northern , Corpus Luteum/metabolism , Female , Luteal Phase/physiology , Macaca mulatta , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/metabolism , Steroids/metabolism
14.
Brain Res ; 811(1-2): 105-10, 1998 Nov 16.
Article in English | MEDLINE | ID: mdl-9804911

ABSTRACT

Conversion of androgen to estrogen within the brain by cytochrome P450 aromatase is a component of the system controlling the display of normal male reproductive behavior and negative feedback inhibition of LH secretion in rams. In the present study, we used the highly sensitive 3H2O assay to measure aromatase activity in microdissected regions of the basal diencephalon and amygdala of intact and castrated rams. We found that aromatase activity was heterogeneously distributed. The highest activity was found in the medial amygdala, cortical amygdala, and bed nucleus of the stria terminalis. Intermediate levels of aromatase activity were found within the medial preoptic area/anterior hypothalamus, periventricular preoptic area, lateral preoptic area/anterior hypothalamus, ventromedial hypothalamus, and lateral hypothalamus. The lowest activity was present in the septum, infundibulum/median eminence, and dorsal medial hypothalamus. After castration, aromatase was significantly reduced in the medial preoptic area/anterior hypothalamus, periventricular preoptic area, lateral preoptic area/anterior hypothalamus, and infundibular nucleus/median eminence. In contrast, levels in castrate rams were unchanged in several other regions, most notably the bed nucleus of the stria terminalis and medial and cortical amygdala. These results provide a quantitative profile of aromatase activity in discrete regions of the ram hypothalamus and limbic system. They also demonstrate that aromatase activity is reduced in some, but not all brain regions after castration which suggests that different regulatory mechanisms control aromatase within different neuronal populations of the ram.


Subject(s)
Amygdala/enzymology , Aromatase/metabolism , Hypothalamus/enzymology , Animals , Dissection , Male , Orchiectomy , Sheep , Testis/metabolism
15.
Endocrine ; 8(1): 51-60, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9666345

ABSTRACT

We localized immunoreactive androgen receptors in the central nervous system (CNS) of fetal and adult male rhesus macaques by immunocytochemisty using an affinity-purified polyclonal antibody to the first 21 amino acids of the human androgen receptor (AR). This antibody caused a shift in the mobility of AR-bound 3H-DHT on a sucrose gradient and recognized a protein of approx 116 kDa on Western blot. Other criteria for specificity are presented. We localized AR in the diencephalon of male rhesus monkey fetuses. Immunoreactive neurons were found in the medial hypothalamic area and the ventromedial nucleus of the hypothalamus on days 47, 61, and 124 of gestation. At 124 d of gestation, AR immunoreactivity was aslo found in the arcuate nucleus. AR immunostaining was not found in other diencephalic structures in fetal life, including the preoptic area. In the adult monkey, neurons in ventromedial, dorsomedial, and arcuate nuclei of the hypothalamus; cortical, medial, and accessory basal nuclei of the amygdala; and regions of the hippocampus and the anterior pituitary gland contained immunoreactive AR. These data indicate that AR is found in specific areas of the CNS early in fetal development, but they also appear in other brain areas as the fetus grows. At 124 d of gestation (term, 167 d), the hypothalamic location of immunoreactive AR is similar to the adult.


Subject(s)
Brain Chemistry , Brain/growth & development , Immunohistochemistry , Receptors, Androgen/analysis , Amygdala/chemistry , Amygdala/embryology , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Arcuate Nucleus of Hypothalamus/embryology , Blotting, Western , Brain/embryology , Diencephalon/chemistry , Diencephalon/embryology , Gestational Age , Humans , Hypothalamus, Middle/chemistry , Hypothalamus, Middle/embryology , Macaca mulatta , Male , Pituitary Gland, Anterior/chemistry , Pituitary Gland, Anterior/embryology
16.
Biol Reprod ; 57(4): 772-7, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9314579

ABSTRACT

The conversion of androgens to estrogens by aromatase cytochrome P450 (P450arom) is an important step in the mechanism of androgen action in the brain. However, the distribution of P450arom mRNA in adult rhesus monkey brains has not been studied because specific probes have not been available. To address this deficit, we cloned and sequenced a 455-basepair segment of the 5' coding region of the rhesus P450arom cDNA. Total RNA was extracted from a rhesus monkey placenta (Day 47 of gestation and subjected to reverse transcriptase (RT) polymerase chain reaction (PCR) using consensus oligonucleotide primers selected from published human and rat P450arom DNA sequences. The RT-PCR product was subcloned into a vector and sequenced. The monkey P450arom cDNA was 97% identical to the human sequence but shared only 86% homology with the rat sequence. We then developed a ribonuclease protection assay using a monkey P450arom cDNA and studied the distribution of P450arom mRNA in adult monkey brains. This assay protected two RNA fragments, one 455 nucleotides (nt) in length and the second approximately 300 nt. The relative distribution of P450arom mRNA (the 455-nt fragment) between brain areas of the adult (n = 3) was high in the bed nucleus of the stria terminalis > medial preoptic/anterior hypothalamus > amygdala; intermediate in the medial basal hypothalamus (infundibular nucleus, median eminence, ventromedial nucleus) > lateral preoptic/anterior hypothalamus; and low in the septum > lateral-dorsal-medial hypothalamus. P450arom mRNA was undetectable in cingulate and parietal cortex, hippocampus, and cerebellum. P450arom activity, as measured by the 3H2O assay, correlated well with the distribution of P450arom mRNA (the 455-nt protected fragment; r = 0.9) in the same tissues. A shorter protected RNA fragment was found in the medial basal hypothalamus, the bed nucleus of the stria terminalis, the amygdala, and the cingulate and parietal cortex but not in the other brain areas investigated. Its presence did not correlate with aromatase activity in brain tissue. This study describes the development of a ribonuclease protection assay using a monkey cDNA produced by RT-PCR and its usefulness for studying the distribution of P450arom mRNA in brains of nonhuman primates.


Subject(s)
Aromatase/metabolism , Brain Chemistry/physiology , RNA, Messenger/metabolism , Animals , Base Sequence , Cloning, Molecular , DNA Probes , Humans , Macaca mulatta , Male , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/isolation & purification , Rats , Ribonucleases/metabolism
17.
J Steroid Biochem Mol Biol ; 61(3-6): 365-74, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9365212

ABSTRACT

The basis of functional gender differences in adult responsiveness to testosterone (T) is not yet understood. Conversion of T to estradiol by cytochrome P450 aromatase in the medial preoptic area is required for the full expression of male sexual behavior in rats. High levels of aromatase are found in the medial preoptic nucleus (MPN) and in an interconnected group of sexually dimorphic nuclei which mediate masculine sexual behavior. Within this neural circuit, aromatase is regulated by T, acting through an androgen receptor (AR)-mediated mechanism. This arrangement constitutes a feedforward system because T is both the regulator and the major substrate of aromatase. Preoptic aromatase is thus more active in adult males than in females because of normal sex differences in circulating androgen levels. However, the mechanism of enzyme induction also appears to be sexually dimorphic because equivalent physiological doses of T stimulate aromatase to a greater extent in males than in females. Dose-response studies indicate that the sex difference is apparent over a range of circulating T concentrations and constitute a gender difference in T efficacy, but not potency. Sex differences in aromatase correlate with sex differences in nuclear AR concentrations in most regions of the sexually dimorphic neural circuit, but not in MPN. These results suggest that males may have larger populations of target cells in which aromatase is regulated by androgen, but the lack of a gender difference in AR levels in the MPN suggests that differences in post-receptor mechanisms could also be involved. Measurements of aromatase mRNA in androgen-treated gonadectomized rats demonstrate that sex difference in regulation is exerted pretranslationally. Taken together these results demonstrate a sexually dimorphic mechanism that could potentially limit the action of T in females, and may relate to the enhanced expression of T-stimulated sexual behaviors in males.


Subject(s)
Androgens/physiology , Aromatase/physiology , Brain/physiology , Gene Expression Regulation, Enzymologic , Animals , Female , Male , Rats , Sex Factors
18.
Brain Res Bull ; 44(4): 351-7, 1997.
Article in English | MEDLINE | ID: mdl-9370199

ABSTRACT

Brain aromatase plays an important role in the regulation of adult reproductive behavior in male rodents. This report focuses on recent experiments from our laboratory that examined the distribution and regulation of aromatase mRNA in the rat brain. Aromatase mRNA was measured by a highly sensitive ribonuclease protection assay using a 32P-labeled antisense RNA probe that was complimentary to the 5' coding region of rat aromatase mRNA. This probe protects two RNA fragments in rat brain tissue: a 430-nt length fragment and a shorter 300-nt fragment. The presence of the 300-nt RNA fragment is not associated with enzyme activity in the rat brain and appears to represent an alternative brain-specific aromatase transcript whose function, if any, is unknown. In contrast, the 430-nt RNA fragment represents mRNA, which is thought to encode functional aromatase enzyme because its levels are correlated with aromatase activity concentrations in preoptic area, hypothalamus, amygdala, and ovary. Aromatase activity and mRNA levels in the preoptic area and hypothalamus decreased by 7 days after castration and were maintained at intact levels by treatment with testosterone and dihyhdrotestosterone, but not with estradiol. In contrast, neither aromatase activity nor mRNA levels in the amygdala are affected by castration or hormone replacement. In addition, sex differences in the regulation of aromatase mRNA were apparent in both the preoptic area and hypothalamus. These results demonstrate that androgens regulate the transcription or stability of aromatase mRNA in specific brain areas. Moreover, they suggest that gender differences in androgen responsiveness play an important role in regulating gene expression in the adult rat brain.


Subject(s)
Aromatase/biosynthesis , Brain/enzymology , Gene Expression Regulation, Enzymologic , Transcription, Genetic , Alternative Splicing , Animals , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Male , Orchiectomy , Organ Specificity , Ovary/enzymology , Preoptic Area/enzymology , RNA Probes , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Testosterone/pharmacology , Transcription, Genetic/drug effects
19.
Cell Mol Neurobiol ; 17(6): 627-48, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9442350

ABSTRACT

1. The central nervous systems (CNS) of males and females differ in the control mechanisms for the release of gonadotropins from the anterior pituitary gland as well as the capacity to display sex specific behaviors. 2. In guinea pigs and monkeys, these differences are organized through the actions of prenatal androgens secreted by the fetal testes. In both males and females androgen receptors have been identified within the brain during the period in development in which organization of the CNS occurs. Sex differences between the ratio of cytosolic and nuclear androgen receptors are due to the amount of endogenous androgen present in the circulation of the developing fetus. Thus, at least part of the biochemical machinery necessary for androgen action resides in the CNS during the period of sexual differentiation. 3. In addition to the physiological differences that have been observed, morphological differences that are androgen dependent have been found in the medial preoptic nucleus and the bed nucleus of the stria terminalis of the guinea pig. The location of these sex differences in brain morphology coincides roughly with the location of steroid binding neurons. 4. In some species the in situ conversion of testosterone to dihydrotestosterone (DHT) by the 5 alpha-reductases or to estradiol-17 beta by cytochrome P450 aromatase mediates testosterone's action. The gonadotropin surge mechanism of adult guinea pigs exposed to a 5a-reductase inhibitor in utero during the critical period for sexual differentiation was unaffected in either males or females even though the development of the external organs of reproduction of males was feminized by the treatment. Likewise, the gonadotropin surge mechanism of subjects exposed to an aromatase inhibitor in utero during the critical period for sexual differentiation was unaffected by this treatment. 5. The mechanism controlling negative feedback, however, was affected in both males and females. Subjects that were exposed to an aromatase inhibitor while developing in utero could not respond to the negative feedback actions of estrogen on gonadotropin release in adulthood. 6. The surge mechanism for the control of gonadotropin secretion in nonhuman primates is not sexually differentiated as it is in rodents. Castrated male monkeys release surge amounts of LH in response to an estrogen challenge. Both infant and adult dimorphic behaviors of rhesus monkeys are organized by the prenatal actions of androgen.


Subject(s)
Brain/embryology , Gonadal Steroid Hormones/physiology , Neurosecretory Systems/embryology , Sex Characteristics , Animals , Embryonic and Fetal Development , Female , Guinea Pigs , Male , Pregnancy , Primates , Reproduction/physiology
20.
J Steroid Biochem Mol Biol ; 59(3-4): 315-22, 1996 Nov.
Article in English | MEDLINE | ID: mdl-9010323

ABSTRACT

To study the tissue-specific control of androgen receptors by circulating androgens, guinea pigs were castrated or castrated and treated with crystalline testosterone propionate. Levels of serum androgens and LH were measured and brain and peripheral tissues were collected for determination of androgen receptor levels under differing androgen states. We found that circulating androgen levels changed rapidly following castration or treatment with exogenous androgen. LH secretion was coupled to circulating androgens; high levels of androgen quickly suppressed LH secretion, whereas LH levels did not rise significantly above intact levels until 7 days following castration. Androgen receptor levels were effected by circulating androgens. Cytosolic androgen receptors (ARc) increased significantly in the preoptic area (POA), septum, anterior pituitary, prostate and seminal vesicle following castration, whereas nuclear androgen receptors (ARn) decreased significantly in the POA, septum, medial basal hypothalamus (MBH), amygdala, parietal cortex, pituitary, prostate and seminal vesicle. Exogenous androgens, which increased serum steroid levels significantly above that in intact animals, decreased ARc below control levels in MBH, amygdala, pituitary and seminal vesicle. High circulating androgens increased ARn above intact levels in the MBH, pituitary and prostate. It thus appears that circulating androgens regulate LH secretion and have profound, but tissue-specific effects on androgen receptors in the guinea pig.


Subject(s)
Brain/drug effects , Luteinizing Hormone/metabolism , Pituitary Gland/drug effects , Receptors, Androgen/metabolism , Testosterone/pharmacology , Androgens/blood , Androstenedione/blood , Animals , Cell Nucleus/metabolism , Cytosol/metabolism , Dihydrotestosterone/blood , Guinea Pigs , Luteinizing Hormone/blood , Male , Orchiectomy , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL