Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Geroscience ; 42(6): 1733-1749, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32876855

RESUMO

As human lifespan increases and the population ages, diseases of aging such as Alzheimer's disease (AD) are a major cause for concern. Although calorie restriction (CR) as an intervention has been shown to increase healthspan in many species, few studies have examined the effects of CR on brain aging in primates. Using postmortem tissue from a cohort of extremely aged rhesus monkeys (22-44 years old, average age 31.8 years) from a longitudinal CR study, we measured immunohistochemically labeled amyloid beta plaques in Brodmann areas 32 and 46 of the prefrontal cortex, areas that play key roles in cognitive processing, are sensitive to aging and, in humans, are also susceptible to AD pathogenesis. We also evaluated these areas for cortical neuron loss, which has not been observed in younger cohorts of aged monkeys. We found a significant increase in plaque density with age, but this was unaffected by diet. Moreover, there was no change in neuron density with age or treatment. These data suggest that even in the oldest-old rhesus macaques, amyloid beta plaques do not lead to overt neuron loss. Hence, the rhesus macaque serves as a pragmatic animal model for normative human aging but is not a complete model of the neurodegeneration of AD. This model of aging may instead prove most useful for determining how even the oldest monkeys are protected from AD, and this information may therefore yield valuable information for clinical AD treatments.


Assuntos
Peptídeos beta-Amiloides , Amiloidose , Peptídeos beta-Amiloides/metabolismo , Animais , Restrição Calórica , Macaca mulatta/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo
2.
Genes Brain Behav ; 16(3): 361-368, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27736018

RESUMO

Estradiol supplementation has been shown to enhance cognitive performance in old ovariectomized rhesus macaques (Macaca mulatta). To determine if similar benefits could be achieved in perimenopausal animals using alternative hormonal supplements, we administered dehydroepiandrosterone (DHEA) to old ovary-intact female rhesus macaques for ∼2.5 months. Using computerized touch screen memory tasks, including delayed response (DR) and delayed matching-to-sample (DMS), we observed improved performance with time in all of the animals but failed to detect a significant effect of DHEA. On the other hand, gene expression profiling disclosed a significant correlation between cognitive performance and the expression of several steroidogenic and steroid-responsive genes. The DR performance was positively correlated with hippocampal expression of AKR1C3 and STAR and negatively correlated with the expression of SDRD5A1. A positive correlation was also found between DMS performance and prefrontal cortical expression of AKR1C3 and a negative correlation with STAR, as well as a negative correlation with the hippocampal expression of HSD11B1 and NR3C1. Taken together, the results suggest that steroidogenic gene regulation within the brain may help to maintain cognitive function during the perimenopausal transition period, despite a decline in sex-steroid levels in the circulation.


Assuntos
Cognição/efeitos dos fármacos , Cognição/fisiologia , Desidroepiandrosterona/farmacologia , Fatores Etários , Animais , Estradiol/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Macaca mulatta , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Menopausa/efeitos dos fármacos , Menopausa/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo
3.
Sci Rep ; 5: 12820, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26333415

RESUMO

Male fertility depends on spermatogenesis, which takes place in the seminiferous tubules of the testis. This compartment is devoid of blood vessels, which are however found in the wall of the seminiferous tubules. Our proteomic study using cultured human testicular peritubular cells (HTPCs) i.e. the cells, which form this wall, revealed that they constitutively secrete pigment epithelium-derived factor, PEDF, which is known to exert anti-angiogenic actions. Immunohistochemistry supports its presence in vivo, in the human tubular wall. Co-culture studies and analysis of cell migration patterns showed that human endothelial cells (HUVECs) are repulsed by HTPCs. The factor involved is likely PEDF, as a PEDF-antiserum blocked the repulsing action. Thus testicular peritubular cells, via PEDF, may prevent vascularization of human seminiferous tubules. Dihydrotestosterone (DHT) increased PEDF (qPCR) in HTPCs, however PEDF expression in the testis of a non-human primate occurs before puberty. Thus PEDF could be involved in the establishment of the avascular nature of seminiferous tubules and after puberty androgens may further reinforce this feature. Testicular microvessels and blood flow are known to contribute to the spermatogonial stem cell niche. Hence HTPCs via control of testicular microvessels may contribute to the regulation of spermatogonial stem cells, as well.


Assuntos
Proteínas do Olho/metabolismo , Neovascularização Fisiológica/fisiologia , Fatores de Crescimento Neural/metabolismo , Túbulos Seminíferos/irrigação sanguínea , Túbulos Seminíferos/metabolismo , Serpinas/metabolismo , Testículo/irrigação sanguínea , Testículo/metabolismo , Adulto , Células Cultivadas , Humanos , Masculino , Adulto Jovem
4.
Andrology ; 2(5): 695-701, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25052196

RESUMO

Besides the two nuclear oestrogen receptors (ESR1/ESR2), the G protein-coupled oestrogen receptor (GPER) was described in the human testis but little is known about testicular GPER during development or male infertility. We performed an immunohistochemical analysis using human and rhesus monkey testicular samples. The results obtained in adult primate testes showed GPER in interstitial and vascular cells as well as in smooth muscle-like peritubular cells, which build the wall of seminiferous tubules. Expression of GPER was also found in cultured human testicular peritubular cells (HPTCs) by Western blotting and RT-PCR/sequencing. Furthermore, as seen in time-lapse videos of cultured cells, addition of a specific GPER agonist (G1) significantly reduced the numbers of HTPCs within 24 h. A GPER antagonist (G15) prevented this action, implying a role for GPER related to the control of cell proliferation or cell death of peritubular cells. Peritubular cell functions and their phenotype change, for example, during post-natal development and in the cases of male infertility. The study of non-human primate samples revealed that GPER in peritubular cells was detectable only from the time of puberty onwards, while in samples from infantile and prepubertal monkeys only interstitial cells showed immunopositive staining. In testicular biopsies of men with mixed atrophy, a reduction or loss of immunoreactive GPER was found in peritubular cells surrounding those tubules, in which spermatogenesis was impaired. In other cases of impaired spermatogenesis, namely when the tubular wall was fibrotically remodelled, a complete loss of GPER was seen. Thus, the observed inverse relation between the state of fertility and GPER expression by peritubular cells implies that the regulation of primate testicular peritubular cells by oestrogens is mediated by GPER in both, health and disease.


Assuntos
Infertilidade Masculina/metabolismo , Células Intersticiais do Testículo/metabolismo , Receptores de Estrogênio/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Túbulos Seminíferos/metabolismo , Células de Sertoli/metabolismo , Animais , Células Cultivadas , Fertilidade , Humanos , Células Intersticiais do Testículo/citologia , Macaca mulatta , Masculino , Túbulos Seminíferos/citologia , Células de Sertoli/citologia , Maturidade Sexual , Espermatogênese
5.
J Neuroendocrinol ; 25(11): 1062-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23796387

RESUMO

Similar to humans, rhesus macaques (Macaca mulatta) are large, long-lived diurnal primates, and show similar age-related changes in the secretion of many steroid hormones, including oestradiol, testosterone, cortisol and dehydroepiandrosterone (DHEA). Consequently, they represent a pragmatic animal model in which to examine the mechanisms by which these steroidal changes contribute to perturbed sleep-wake cycles and cognitive decline in the elderly. Using remote serial blood sampling, we have found the circulating levels of DHEA sulphate, as well as oestradiol and testosterone, decline markedly in old monkeys. Furthermore, using the real-time polymerase chain reaction, we have shown that the genes for the enzymes associated with the conversion of DHEA to oestradiol and testosterone (3ß-hydroxysteroid dehydrogenase, 17ß-hydroxysteroid dehydrogenase, and aromatase) are highly expressed in brain areas associated with cognition and behaviour, including the hippocampus, prefrontal cortex and amygdala. Taken together, these findings suggest that the administration of supplementary DHEA in the elderly may have therapeutic potential for cognitive and behavioural disorders, although with fewer negative side effects outside of the central nervous system. To test this, we have developed a novel steroid supplementation paradigm for use in old animals; this involves the oral administration of DHEA and testosterone at physiologically relevant times of the day to mimic the circadian hormone patterns observed in young adults. We are currently evaluating the efficacy of this steroid supplementation paradigm with respect to reversing age-associated disorders, including perturbed sleep-wake cycles and cognitive decline, as well as an impaired immune response.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Ritmo Circadiano/fisiologia , Desidroepiandrosterona/metabolismo , Estradiol/metabolismo , Macaca mulatta/fisiologia , Testosterona/metabolismo , Envelhecimento/sangue , Animais , Desidroepiandrosterona/farmacologia , Estradiol/sangue , Macaca mulatta/metabolismo , Testosterona/sangue
6.
Rejuvenation Res ; 15(2): 128-31, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22533414

RESUMO

Circulating levels of dehydroepiandrosterone, a major adrenal steroid, show a marked age-related decrease in both humans and nonhuman primates. Because this decrease has been implicated in age-related cognitive decline, we administered supplementary dehydroepiandrosterone to perimenopausal rhesus macaques (Macaca mulatta) to test for cognitive benefits. Although recognition memory improved, there was no benefit to spatial working memory. To address the limitations of this study we developed a hormone supplementation regimen in aged male macaques that more accurately replicates the 24-hr androgen profiles of young animals. We hypothesize that this more comprehensive physiological hormone replacement paradigm will enhance cognitive function in the elderly.


Assuntos
Envelhecimento , Desidroepiandrosterona/uso terapêutico , Hormônios/uso terapêutico , Esteroides/uso terapêutico , Androgênios/metabolismo , Animais , Cognição/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Macaca mulatta , Masculino , Transtornos da Memória/tratamento farmacológico , Memória de Curto Prazo/efeitos dos fármacos , Testosterona/uso terapêutico
7.
Int J Androl ; 35(4): 550-61, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22413766

RESUMO

Decorin (DCN), a component of the extracellular matrix of the peritubular wall and the interstitial areas of the human testis, can interact with growth factor (GF) signalling, thereby blocking downstream actions of GFs. In the present study the expression and regulation of DCN using both human testes and two experimental animal models, namely the rhesus monkey and mouse, were examined. DCN protein was present in peritubular and interstitial areas of adult human and monkey testes, while it was almost undetectable in adult wild type mice. Interestingly, the levels and sites of testicular DCN expression in the monkeys were inversely correlated with testicular maturation markers. A strong DCN expression associated with the abundant connective tissue of the interstitial areas in the postnatal through pre-pubertal phases was observed. In adult and old monkeys the DCN pattern was similar to the one in normal human testes, presenting strong expression at the peritubular region. In the testes of both infertile men and in a mouse model of inflammation associated infertility (aromatase-overexpressing transgenic mice), the fibrotic changes and increased numbers of tumour necrosis factor (TNF)-α-producing immune cells were shown to be associated with increased production of DCN. Furthermore, studies with human testicular peritubular cells isolated from fibrotic testis indicated that TNF-α significantly increased DCN production. The data, thus, show that an increased DCN level is associated with impaired testicular function, supporting our hypothesis that DCN interferes with paracrine signalling of the testis in health and disease.


Assuntos
Decorina/metabolismo , Infertilidade Masculina/patologia , Testículo/metabolismo , Testículo/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/metabolismo , Fibrose , Humanos , Inflamação , Macaca mulatta , Masculino , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Testículo/citologia , Fator de Necrose Tumoral alfa/biossíntese
8.
Exp Neurol ; 217(1): 55-62, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19416674

RESUMO

In rodents, spatial learning and memory tests require navigation, whereas in nonhuman primates these tests generally do not involve a navigational component, thus assessing nonhomologous neural systems. To allow closer parallels between rodent and primate studies, we developed a navigational spatial learning and memory task for nonhuman primates and assessed the performance of elderly (19-25 years) female rhesus monkeys (Macaca mulatta). The animals were allowed to navigate in a room containing a series of food ports. After they learned to retrieve food from the ports, a single port was repeatedly baited and the animals were tested until they learned the correct location. The location of the baited port was then changed (shift position). We also determined whether test performance was associated with circadian activity measured with accelerometers. Performance measures included trials to criterion, search strategies, and several indices of circadian activity. Animals learned the task as reflected in their search strategies. Correlations were found between the number of initial or shift trials and circadian activity parameters including day activity, dark:light activity ratio, sleep latency, and wake bouts. Thus, disruptions in circadian rhythms in nonhuman primates are associated with poorer performance on this novel test. These data support the usefulness of this spatial navigational test to assess spatial learning and memory in rhesus monkeys and the importance of circadian activity in performance.


Assuntos
Envelhecimento/fisiologia , Aprendizagem por Associação/fisiologia , Ritmo Circadiano/fisiologia , Macaca mulatta/fisiologia , Memória/fisiologia , Comportamento Espacial/fisiologia , Animais , Discriminação Psicológica , Feminino , Estatística como Assunto
9.
J Mol Endocrinol ; 33(1): 145-53, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15291749

RESUMO

The hypothalamus of rhesus macaques expresses two molecular forms of gonadotropin-releasing hormone (GnRH-I and GnRH-II). However, it is unclear whether these two neuropeptides play similar roles in the control of reproductive neuroendocrine function, especially in the context of positive and negative estrogen feedback. To address this issue, in situ hybridization histochemistry was used to compare the effect of 17beta-estradiol (E) on the expression of GnRH-I and GnRH-II mRNA in the medial basal hypothalamus (MBH) of adult female macaques. GnRH-I mRNA expression was found to be significantly (P<0.01) more abundant in ovariectomized (ovx) animals compared with ovariectomized E-treated (ovx+E) animals. In marked contrast, GnRH-II mRNA expression was found to be significantly (P<0.05) more abundant in ovx+E animals than in the ovx animals. To help elucidate how E exerts this stimulatory action on GnRH-II gene expression, hypothalamic sections were subsequently double labeled using a combination of immunohistochemisty for estrogen receptor (ER) -alpha or -beta and in situ hybridization histochemistry for GnRH-II. Approximately 50% of the GnRH-II positive cells in the MBH were found to express ERbeta, but none expressed ERalpha. Taken together, these data give credence to a novel pathway by which E may control the primate neuroendocrine reproductive axis, one that involves stimulation of GnRH-II release via an ERbeta-mediated mechanism.


Assuntos
Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/efeitos dos fármacos , Animais , Feminino , Hipotálamo/metabolismo , Macaca mulatta , RNA Mensageiro/genética
10.
Ann N Y Acad Sci ; 1019: 443-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15247063

RESUMO

Although dietary caloric restriction (CR) can retard aging in laboratory rats and mice, it is unclear whether CR can exert similar effects in long-lived species, such as primates. Therefore, we tested the effect of CR on plasma levels of dehydroepiandrosterone sulfate (DHEAS), a reliable endocrine marker of aging. The study included six young (approximately 10 years) and ten old (approximately 25 years) male rhesus macaques, approximately half of the animals in each age group having undergone >4 years of 30% CR. Hourly blood samples were collected remotely for 24 hours, through a vascular catheter, and assayed for DHEAS and cortisol. Both of these adrenal steroids showed a pronounced diurnal plasma pattern, with peaks occurring in late morning, but only DHEAS showed an aging-related decline. More importantly, there was no significant difference in plasma DHEAS concentrations between the CR animals and age-matched controls. These data fail to support the hypothesis that CR can attenuate the aging-related decline in plasma DHEAS concentrations, at least not when initiated after puberty.


Assuntos
Envelhecimento , Restrição Calórica , Sulfato de Desidroepiandrosterona/sangue , Hidrocortisona/metabolismo , Animais , Macaca mulatta , Masculino , Fatores de Tempo
11.
Neuroreport ; 14(4): 613-8, 2003 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-12657897

RESUMO

Hypothalamic GnRH (gonadotropin-releasing hormone) neurons play a critical role in the initiation and maintenance of reproduction competence. Using the mouse GnRH neuronal cell line, GT1-7, we have characterized the expression of the gene mPer1, a recognized key element of the mammalian circadian clockwork. Both mPer1 transcripts and the 136 kDa mPER1 gene product could be detected in these cells. Immunocytochemical analysis also confirmed expression of mPER1 both in vitro and in vivo in GnRH neurons. Activation of cyclic AMP signalling pathways in vitro elevated GnRH secretion as well as mPer1 expression and nuclear mPER1 immunoreactivity. As mPER1 is known to feedback on transcriptional activities in many cell models, the data presented here point to a role for mPER1 in the regulation of gene expression in GnRH neurons, and thus in the control of neuroendocrine activities.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Animais , Proteínas de Ciclo Celular , Células Cultivadas , Colforsina/farmacologia , Hormônio Liberador de Gonadotropina/análise , Immunoblotting/métodos , Imuno-Histoquímica/métodos , Camundongos , Fármacos Neuroprotetores/farmacologia , Proteínas Nucleares/genética , Proteínas Circadianas Period , Área Pré-Óptica/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Peptídeo Intestinal Vasoativo/farmacologia
12.
Trends Endocrinol Metab ; 12(10): 428-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11701332

RESUMO

Leptin is thought to relay metabolic information to the hypothalamic-pituitary- gonadal axis and to participate in the neuroendocrine control of puberty. To help elucidate the underlying mechanism, Cheung et al. recently performed a diverse series of experiments, the results of which undermine the prevailing hypothesis that leptin acts as a metabolic trigger for the initiation of puberty. Instead, their results suggest that leptin is one of many permissive metabolic factors that allow pubertal development to proceed.


Assuntos
Proteínas de Transporte/metabolismo , Leptina/administração & dosagem , Leptina/sangue , Puberdade/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores de Superfície Celular , Animais , Proteínas de Transporte/genética , Feminino , Humanos , Masculino , Camundongos , Puberdade/fisiologia , RNA Mensageiro/genética , Ratos , Receptores para Leptina
13.
J Reprod Immunol ; 51(2): 109-29, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11543851

RESUMO

In recent years, several forms of gonadotrophin releasing hormone (GnRH) molecules have been isolated from primate brain. These molecules are very similar in sequence and this raises the question of whether previously developed neutralisation vaccines based on GnRH (now termed GnRH-I) would remove other forms of GnRH (namely GnRH-II) as well. As the function of these other molecules has not yet been clearly defined, potential health risks could exist by their ablation. In view of the high sequence homology between the molecules, this paper describes the production of highly specific polyclonal antibodies against GnRH-I and GnRH-II, with negligible cross-reactivity. The ultimate aim of this is to develop an anti-fertility vaccine which does not present any inappropriate side-effects, caused by neutralisation of a GnRH molecule which may or may not be directly involved in reproduction. Several formulations were investigated, based on analogues of the following molecules, conjugated to tetanus toxoid: 1. GnRH-I pGlu-His-Trp-Ser-Try-Gly-Leu-Arg-Pro-Gly-NH2 and 2. GnRH-II pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH2. The specificity of the antibodies produced was examined, together with effects on fertility and any inappropriate side-effects. Immunostaining of hypothalamic sections was carried out, using the generated antisera, to determine the regional distribution of GnRH-I and GnRH-II neurones, as well as to further evaluate the specificity of the antibodies.


Assuntos
Especificidade de Anticorpos , Hormônio Liberador de Gonadotropina/imunologia , Hormônio Liberador de Gonadotropina/metabolismo , Vacinas Anticoncepcionais/imunologia , Animais , Química Encefálica , Anticoncepcionais Masculinos/efeitos adversos , Reações Cruzadas/imunologia , Hormônio Liberador de Gonadotropina/análogos & derivados , Humanos , Esquemas de Imunização , Isotipos de Imunoglobulinas/sangue , Macaca mulatta , Masculino , Ratos , Ratos Sprague-Dawley , Testículo/anatomia & histologia , Testículo/patologia , Toxoide Tetânico/efeitos adversos , Toxoide Tetânico/imunologia , Vacinas Conjugadas/efeitos adversos , Vacinas Conjugadas/imunologia , Vacinas Anticoncepcionais/efeitos adversos
15.
Brain Res Mol Brain Res ; 89(1-2): 119-25, 2001 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-11311982

RESUMO

Male Syrian hamsters (Mesocricetus auratus) are seasonal breeders. They show marked testicular regression when exposed to short autumnal photoperiods, and then remain sexually quiescent for several months. By mid-winter, however, they show a loss in responsiveness to the inhibitory influence of short photoperiods and their testes begin to recrudesce. To shed light on the neuroendocrine mechanism responsible for mediating these reproductive changes, we examined the influence of photoperiod on the expression of GnRH mRNA in the hamster forebrain. Adult males were either exposed to short photoperiods (6L:18D) for 16 weeks or were maintained under long photoperiods (14L:10D); additional animals were exposed to short or long photoperiods for 22 weeks. As expected, exposure to short photoperiods for 12 weeks resulted in a marked decrease (P<0.01) in testicular mass and serum testosterone levels, but after 22 weeks these reproductive parameters were once again significantly elevated (P<0.01). In contrast, quantitative in situ hybridization histochemistry revealed no difference (P>0.05) between the GnRH mRNA levels of the short-photoperiod hamsters and their aged-matched long-photoperiod controls, although an age-related decrease (P<0.05) was evident in both photoperiod-treatment groups. These data emphasize that GnRH mRNA is highly expressed in hamsters even when their reproductive axis has been rendered sexually quiescent by exposure to short photoperiods, and that photoperiod-induced changes in GnRH secretion, rather than synthesis, are more likely to regulate the timing of the breeding season. On the other hand, the data indicate that GnRH mRNA levels show an aging-related decrease, regardless of photoperiod, suggesting that in the long term a decrease in GnRH gene expression may contribute to the reduced fertility of old hamsters.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Fotoperíodo , Estações do Ano , Testículo/fisiologia , Envelhecimento/fisiologia , Animais , Cricetinae , Expressão Gênica/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Mesocricetus , RNA Mensageiro/metabolismo , Reprodução/fisiologia , Testosterona/fisiologia
16.
J Clin Endocrinol Metab ; 86(1): 324-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11232019

RESUMO

GnRH-I is thought to represent the primary neuroendocrine link between the brain and the reproductive axis. Recently, however, a second molecular form of this decapeptide (GnRH-II) was found to be highly expressed in the brains of humans and nonhuman primates. In this study, in situ hybridization was used to examine the regional expression of GnRH-II messenger ribonucleic acid in the hypothalamus of immature (0.6 yr) and adult (10-15 yr) male and female rhesus macaques (Macaca mulatta). Overall, no sex-related differences were observed. In all of the animals (n = 3 animals/group), intense hybridization of a monkey GnRH-II riboprobe was evident in the paraventricular nucleus and supraoptic nucleus and to a lesser extent in the suprachiasmatic nucleus, but no age- or sex-related differences were apparent. Intense hybridization of the riboprobe also occurred in the mediobasal hypothalamus, and this was markedly greater in the adults than in the immature animals. These data show that the expression of GnRH-II messenger ribonucleic acid increases developmentally in a key neuroendocrine center of the brain. Moreover, because GnRH-II can stimulate LH release in vivo, it is plausible that changes in its gene expression represent an important component of the mechanism by which the hypothalamus controls reproductive function.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Macaca mulatta/metabolismo , RNA Mensageiro/metabolismo , Animais , Autorradiografia , Feminino , Masculino , Isoformas de Proteínas/genética , Prata
17.
Endocrinology ; 141(11): 4226-38, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11089557

RESUMO

Energy dissipating mechanisms and their regulatory components represent key elements of metabolism and may offer novel targets in the treatment of metabolic disorders, such as obesity and diabetes. Recent studies have shown that a mitochondrial uncoupling protein (UCP2), which uncouples mitochondrial oxidation from phosphorylation, is expressed in the rodent brain by neurons that are known to regulate autonomic, metabolic, and endocrine processes. To help establish the relevance of these rodent data to primate physiology, we now examined UCP2 messenger RNA and peptide expressions in the brain and pituitary gland of nonhuman primates. In situ hybridization histochemistry showed that UCP2 messenger RNA is expressed in the paraventricular, supraoptic, suprachiasmatic, and arcuate nuclei of the primate hypothalamus and also in the anterior lobe of the pituitary gland. Immunocytochemistry revealed abundant UCP2 expression in cell bodies and axonal processes in the aforementioned nuclei as well as in other hypothalamic and brain stem regions and all parts of the pituitary gland. In the hypothalamus, UCP2 was coexpressed with neuropeptide Y, CRH, oxytocin, and vasopressin. In the pituitary, vasopressin and oxytocin-producing axonal processes in the posterior lobe and POMC cells in the intermediate and anterior lobes expressed UCP2. On the other hand, none of the GH-producing cells of the anterior pituitary was found to produce UCP2. The abundance and distribution pattern of UCP2 in the primate brain and pituitary suggest that this protein is evolutionary conserved and may relate to central autonomic, endocrine and metabolic regulation.


Assuntos
Química Encefálica , Proteínas de Membrana Transportadoras , Proteínas Mitocondriais , Hipófise/química , Proteínas/análise , Animais , Chlorocebus aethiops , Hormônio Liberador da Corticotropina/análise , Expressão Gênica , Hipotálamo/química , Imuno-Histoquímica , Hibridização In Situ , Canais Iônicos , Sistema Límbico/química , Macaca fascicularis , Macaca mulatta , Microscopia de Fluorescência , Neuropeptídeo Y/análise , Ocitocina/análise , Adeno-Hipófise/química , Neuro-Hipófise/química , Proteínas/genética , RNA Mensageiro/análise , Proteína Desacopladora 2 , Vasopressinas/análise
18.
Brain Res Mol Brain Res ; 76(2): 191-204, 2000 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-10762694

RESUMO

This study used in situ hybridization (ISH) to examine the distribution of estrogen receptor beta (ERbeta) mRNA in hypothalamic, limbic, and midbrain regions of monkey brain and its regulation by estrogen (E) and progesterone (P). Monkey-specific ERbeta cDNAs were developed with human primers and reverse transcription and polymerase chain reaction (RT-PCR) using mRNA extracted from a rhesus monkey prostate gland. ERbeta 5' (262 bases) and 3' (205 bases) riboprobes were used in combination for ISH. Ovariectomized and hysterectomized (spayed) pigtail macaques (Macaca nemestrina; four per treatment group) were either untreated spayed-controls, treated with E (28 days), or treated with E plus P (14 days E+14 days E and P). Dense ERbeta hybridization signal was seen in the preoptic area, paraventricular nucleus, and ventromedial nucleus of the hypothalamus; the substantia nigra, caudal linear, dorsal raphe, and pontine nuclei of the midbrain; the dentate gyrus, CA1, CA2, CA3, CA4, and the prosubiculum/subiculum areas of the hippocampus. Expression in the suprachiasmatic region, supraoptic nucleus, arcuate nucleus, and amygdala was less intense. Image analysis of the dense areas showed no significant difference in the hybridization signal in individual regions of the hypothalamus, midbrain, or hippocampus between any of the treatment groups. However, P treatment decreased overall ERbeta signal in the hypothalamus and hippocampus when several different subregions were combined. The localization of ERbeta in monkey brain by ISH is in general agreement with that previously described in rodents. The presence of monkey ERbeta mRNA in brain regions that lack ERalpha should help to clarify the molecular mechanisms by which E acts in the central nervous system to influence hormone secretion, mood disorders, cognition, and neuroprotection.


Assuntos
Encéfalo/metabolismo , Estradiol/farmacologia , Terapia de Reposição de Estrogênios , Regulação da Expressão Gênica , Progesterona/farmacologia , Receptores de Estrogênio/genética , Tonsila do Cerebelo/metabolismo , Animais , Sequência de Bases , Encéfalo/efeitos dos fármacos , Receptor beta de Estrogênio , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Histerectomia , Macaca nemestrina , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Dados de Sequência Molecular , Ovariectomia , Próstata/metabolismo , RNA Mensageiro/análise , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Lobo Temporal/efeitos dos fármacos , Lobo Temporal/metabolismo
19.
Brain Res Mol Brain Res ; 75(2): 287-92, 2000 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-10686350

RESUMO

Gonadotropin-releasing hormone represents the primary neuroendocrine link between the brain and the reproductive axis, and at least two distinct molecular forms of this decapeptide (GnRH-I and GnRH-II) are known to be expressed in the forebrain of rhesus macaques (Macaca mulatta). Although the distribution pattern of the two corresponding mRNAs is largely dissimilar, their expression appears to show some overlap in specific regions of the hypothalamus; this raises the possibility that some cells express both molecular forms of GnRH. To resolve this issue, double-label histochemistry was performed on hypothalamic sections from six male rhesus macaques, using a monoclonal antibody to GnRH-I and a riboprobe to monkey GnRH-II mRNA. In total, more than 2000 GnRH neurons were examined but in no instance were GnRH-I peptide and GnRH-II mRNA found to be coexpressed. This finding emphasizes that GnRH-I and GnRH-II are synthesized by two distinct populations of hypothalamic neurons, and suggests that they may be regulated by different neuroendocrine pathways.


Assuntos
Hormônio Liberador de Gonadotropina/análogos & derivados , Hipotálamo/metabolismo , Animais , Hormônio Liberador de Gonadotropina/biossíntese , Hipotálamo/química , Imuno-Histoquímica , Técnicas In Vitro , Macaca mulatta , Masculino , Isoformas de Proteínas/biossíntese , RNA Mensageiro/biossíntese
20.
Neuroendocrinology ; 71(1): 43-50, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10644898

RESUMO

Intrinsic neuron-like cells expressing the catecholamine-biosynthetic enzyme tyrosine hydroxylase (TH) were recently identified in the testis of the prepubertal rhesus monkey. In this study, we characterized the neuron-like nature of these cells and examined distribution and frequency of neuronal elements in the testes of monkeys during postnatal development, puberty and adulthood. Using immunohistochemical methods, we detected both nerve fibers and cell bodies, immunoreactive for the neuronal markers neurofilament 200 (NF-200) and synaptosomal associated protein of 25 kDa (SNAP-25), TH and neuropeptide Y (NPY) in perivascular locations, intermingled with interstitial cells and close to the wall of seminiferous tubules. Marked age-related differences in the numbers of these neuronal elements became apparent, when we quantified NF-200-immunoreactive neuronal elements. Thus, intrinsic neuron-like cell bodies were found only in the testes from immature animals (i.e. , until about 3 years of age). Conversely, nerve fibers, presumably representing mainly the extrinsic innervation, were observed at all ages although they became more prominent after the pubertal increase in LH and testosterone levels. Interestingly, another testicular cell type known to contain potent regulatory substances, mast cells, was found to be in close anatomical proximity to nerve fibers. The number of these cells, positively identified with an antibody to tryptase, increased significantly after puberty following the same pattern as nerve fibers. These results confirm that the testicular nervous system of the monkey is composed of two components, intrinsic nerve cells and extrinsic fibers, both of which are catecholaminergic and peptidergic in nature. Furthermore, both components show a marked degree of plasticity during development, especially around the time of puberty. The intratesticular locations of neuron-like cells and fibers suggest that catecholamines and neuropeptides are likely to have multiple sites of actions, and may affect Leydig cells, cells of the tubular wall and vascular cells directly and/or indirectly via intermediation of mast cells.


Assuntos
Proteínas de Membrana , Sistemas Neurossecretores/química , Sistemas Neurossecretores/citologia , Testículo/citologia , Testículo/inervação , Fatores Etários , Animais , Quimases , Hormônio Luteinizante/sangue , Macaca mulatta , Masculino , Mastócitos/citologia , Mastócitos/enzimologia , Fibras Nervosas/química , Fibras Nervosas/enzimologia , Proteínas do Tecido Nervoso/análise , Proteínas de Neurofilamentos/análise , Neuropeptídeo Y/análise , Sistemas Neurossecretores/crescimento & desenvolvimento , Serina Endopeptidases/análise , Maturidade Sexual/fisiologia , Proteína 25 Associada a Sinaptossoma , Testículo/crescimento & desenvolvimento , Testosterona/sangue , Triptases , Tirosina 3-Mono-Oxigenase/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...