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










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 10(1): 10954, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616776

RESUMO

Children diagnosed with brain tumors have the lowest overall survival of all pediatric cancers. Recent molecular studies have resulted in the discovery of recurrent driver mutations in many pediatric brain tumors. However, despite these molecular advances, the clinical outcomes of high grade tumors, including H3K27M diffuse midline glioma (H3K27M DMG), remain poor. To address the paucity of tissue for biological studies, we have established a comprehensive protocol for the coordination and processing of donated specimens at postmortem. Since 2010, 60 postmortem pediatric brain tumor donations from 26 institutions were coordinated and collected. Patient derived xenograft models and cell cultures were successfully created (76% and 44% of attempts respectively), irrespective of postmortem processing time. Histological analysis of mid-sagittal whole brain sections revealed evidence of treatment response, immune cell infiltration and the migratory path of infiltrating H3K27M DMG cells into other midline structures and cerebral lobes. Sequencing of primary and disseminated tumors confirmed the presence of oncogenic driver mutations and their obligate partners. Our findings highlight the importance of postmortem tissue donations as an invaluable resource to accelerate research, potentially leading to improved outcomes for children with aggressive brain tumors.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Glioma/patologia , Histonas/genética , Mutação , Adolescente , Adulto , Animais , Autopsia , Neoplasias Encefálicas/genética , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Humanos , Lactente , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
2.
Endocrinology ; 160(4): 803-816, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30753503

RESUMO

We have proposed that arcuate neurons coexpressing kisspeptin, neurokinin B, and dynorphin (KNDy neurons) contribute to hot flushes via projections to neurokinin 3 receptor (NK3R)-expressing neurons in the median preoptic nucleus (MnPO). To characterize the thermoregulatory role of MnPO NK3R neurons in female mice, we ablated these neurons using injections of saporin toxin conjugated to a selective NK3R agonist. Loss of MnPO NK3R neurons increased the core temperature (TCORE) during the light phase, with the frequency distributions indicating a regulated shift in the balance point. The increase in TCORE in the ablated mice occurred despite changes in the ambient temperature and regardless of estrogen status. We next determined whether an acute increase in ambient temperature or higher TCORE would induce Fos in preoptic enhanced green fluorescent protein (EGFP)-immunoreactive neurons in Tacr3-EGFP mice. Fos activation was increased in the MnPO but no induction of Fos was found in NK3R (EGFP-immunoreactive) neurons. Thus, MnPO NK3R neurons are not activated by warm thermosensors in the skin or viscera and are not warm-sensitive neurons. Finally, RNAscope was used to determine whether Tacr3 (NK3R) mRNA was coexpressed with vesicular glutamate transporter 2 or vesicular γ-aminobutyric acid (GABA) transporter mRNA, markers of glutamatergic and GABAergic neurotransmission, respectively. In the MnPO, 94% of NK3R neurons were glutamatergic, but in the adjacent medial preoptic area, 97% of NK3R neurons were GABAergic. Thus, NK3R neurons in the MnPO are glutamatergic and play a role in reducing TCORE but are not activated by warm thermal stimuli (internal or external). These findings suggest that KNDy neurons modulate thermosensory pathways for heat defense indirectly via a subpopulation of glutamatergic MnPO neurons that express NK3R.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Área Pré-Óptica/metabolismo , Receptores da Neurocinina-3/metabolismo , Animais , Dinorfinas/metabolismo , Feminino , Fogachos/metabolismo , Temperatura Alta , Kisspeptinas/metabolismo , Camundongos , Neurocinina B/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
3.
Int J Obes (Lond) ; 43(9): 1759-1768, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30568267

RESUMO

BACKGROUND/OBJECTIVES: Inadequate sleep increases obesity and environmental noise contributes to poor sleep. However, women may be more vulnerable to noise and hence more susceptible to sleep disruption-induced weight gain than men. In male rats, exposure to environmental (i.e. ambient) noise disrupts sleep and increases feeding and weight gain. However, the effects of environmental noise on sleep and weight gain in female rats are unknown. Thus, this study was designed to determine whether noise exposure would disturb sleep, increase feeding and weight gain and alter the length of the estrous cycle in female rats. SUBJECTS/METHODS: Female rats (12 weeks old) were exposed to noise for 17d (8 h/d during the light period) to determine the effects of noise on weight gain and food intake. In a separate set of females, estrous cycle phase and length, EEG, EMG, spontaneous physical activity and energy expenditure were recorded continuously for 27d during baseline (control, 9d), noise exposure (8 h/d, 9d) and recovery (9d) from sleep disruption. RESULTS: Noise exposure significantly increased weight gain and food intake compared to females that slept undisturbed. Noise also significantly increased wakefulness, reduced sleep and resulted in rebound sleep during the recovery period. Total energy expenditure was significantly lower during both noise exposure and recovery due to lower energy expenditure during spontaneous physical activity and sleep. Notably, noise did not alter the estrous cycle length. CONCLUSIONS: As previously observed in male rats, noise exposure disrupted sleep and increased weight gain in females but did not alter the length of the estrous cycle. This is the first demonstration of weight gain in female rats during sleep disruption. We conclude that the sleep disruption caused by exposure to environmental noise is a significant tool for determining how sleep loss contributes to obesity in females.


Assuntos
Metabolismo Energético/efeitos da radiação , Ruído/efeitos adversos , Privação do Sono/etiologia , Aumento de Peso/efeitos da radiação , Animais , Feminino , Hiperfagia/etiologia , Ratos , Ratos Sprague-Dawley , Sono/efeitos da radiação
4.
Temperature (Austin) ; 5(1): 56-69, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29687044

RESUMO

Hot flushes are due to estrogen withdrawal and characterized by the episodic activation of heat dissipation effectors. Recent studies (in humans and rats) have implicated neurokinin 3 (NK3) receptor signaling in the genesis of hot flushes. Although transgenic mice are increasingly used for biomedical research, there is limited information on how 17ß-estradiol and NK3 receptor signaling alters thermoregulation in the mouse. In this study, a method was developed to measure tail skin temperature (TSKIN) using a small data-logger attached to the surface of the tail, which, when combined with a telemetry probe for core temperature (TCORE), allowed us to monitor thermoregulation in freely-moving mice over long durations. We report that estradiol treatment of ovariectomized mice reduced TCORE during the light phase (but not the dark phase) while having no effect on TSKIN or activity. Estradiol also lowered TCORE in mice exposed to ambient temperatures ranging from 20 to 36°C. Unlike previous studies in the rat, estradiol treatment of ovariectomized mice did not reduce TSKIN during the dark phase. Subcutaneous injections of an NK3 receptor agonist (senktide) in ovariectomized mice caused an acute increase in TSKIN and a reduction in TCORE, consistent with the activation of heat dissipation effectors. These changes were reduced by estradiol, suggesting that estradiol lowers the sensitivity of central thermoregulatory pathways to NK3 receptor activation. Overall, we show that estradiol treatment of ovariectomized mice decreases TCORE during the light phase, reduces the thermoregulatory effects of senktide and modulates thermoregulation differently than previously described in the rat.

5.
Endocrinology ; 157(5): 2015-27, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26937713

RESUMO

In the human infundibular (arcuate) nucleus, a subpopulation of neurons coexpress kisspeptin and neurokinin B (NKB), 2 peptides required for normal reproductive function. A homologous group of neurons exists in the arcuate nucleus of rodents, termed KNDy neurons based on the coexpression of kisspeptin, NKB, and dynorphin. To study their function, we recently developed a method to selectively ablate KNDy neurons using NK3-SAP, a neurokinin 3 receptor agonist conjugated to saporin (SAP). Here, we ablated KNDy neurons in female rats to determine whether these neurons are required for estrous cyclicity and the steroid induced LH surge. NK3-SAP or Blank-SAP (control) was microinjected into the arcuate nucleus using stereotaxic surgery. After monitoring vaginal smears for 3-4 weeks, rats were ovariectomized and given 17ß-estradiol and progesterone in a regimen that induced an afternoon LH surge. Rats were killed at the time of peak LH levels, and brains were harvested for NKB and dual labeled GnRH/Fos immunohistochemistry. In ovary-intact rats, ablation of KNDy neurons resulted in hypogonadotropic hypogonadism, characterized by low levels of serum LH, constant diestrus, ovarian atrophy with increased follicular atresia, and uterine atrophy. Surprisingly, the 17ß-estradiol and progesterone-induced LH surge was 3 times higher in KNDy-ablated rats. Despite the marked increase in the magnitude of the LH surge, the number of GnRH or anterior ventral periventricular nucleus neurons expressing Fos was not significantly different between groups. Our studies show that KNDy neurons are essential for tonic levels of serum LH and estrous cyclicity and may play a role in limiting the magnitude of the LH surge.


Assuntos
Dinorfinas/metabolismo , Hipogonadismo/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Neurocinina B/metabolismo , Neurônios/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/efeitos dos fármacos , Dinorfinas/genética , Estradiol/farmacologia , Ciclo Estral/metabolismo , Feminino , Hipogonadismo/genética , Kisspeptinas/genética , Neurocinina B/genética , Neurônios/efeitos dos fármacos , Progesterona/farmacologia , Ratos , Ratos Sprague-Dawley
6.
Endocrinology ; 156(7): 2552-62, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25825817

RESUMO

KNDy neurons facilitate tail skin vasodilation and modulate the effects of estradiol on thermoregulation. We hypothesize that KNDy neurons influence cutaneous vasodilation via projections to neurons in the median preoptic nucleus (MnPO) that express the neurokinin 3 receptor (NK3R). In support of this hypothesis, focal microinjections of senktide, an NK3R agonist, into the MnPO lowers core temperature (TCORE) in the female rat. To further study the role of MnPO NK3R neurons in thermoregulation, these neurons were specifically ablated using a conjugate of a selective NK3R agonist and saporin (NK3-SAP). NK3-SAP or blank-SAP (control) was injected into the MnPO/medial septum. Tail skin temperature (TSKIN) and TCORE were measured in ovariectomized rats exposed to 3 ambient temperatures (TAMBIENT) before and after estradiol-17ß (E2) treatment. Before killing, we injected senktide (sc), monitored TCORE for 70 minutes, and harvested brains for Fos immunohistochemistry. Ablation of MnPO NK3R neurons lowered TSKIN at neutral and subneutral TAMBIENT regardless of E2 treatment. However, ablation did not prevent the effects of E2 on TCORE and TSKIN. In control rats, senktide injections induced hypothermia with numerous Fos-immunoreactive cells in the MnPO. In contrast, in NK3-SAP rats, senktide did not alter TCORE and minimal Fos-immunoreactive neurons were identified in the MnPO. These data show that NK3R neurons in the MnPO are required for the hypothermic effects of senktide but not for the E2 modulation of thermoregulation. The lower TSKIN in NK3-SAP-injected rats suggests that MnPO NK3R neurons, like KNDy neurons, facilitate cutaneous vasodilation, an important heat-dissipation effector.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Neurônios/fisiologia , Área Pré-Óptica/fisiologia , Receptores da Neurocinina-3/fisiologia , Vasodilatação/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Regulação da Temperatura Corporal/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Fogachos/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Área Pré-Óptica/citologia , Área Pré-Óptica/efeitos dos fármacos , Ratos , Receptores da Neurocinina-3/agonistas , Pele , Substância P/análogos & derivados , Substância P/farmacologia , Vasodilatação/efeitos dos fármacos
7.
Endocrinology ; 155(7): 2555-65, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24735328

RESUMO

Neurons in the arcuate nucleus that coexpress kisspeptin, neurokinin B (NKB), and dynorphin (KNDy neurons) play an important role in the modulation of reproduction by estrogens. Here, we study the anatomical and electrophysiological properties of arcuate NKB neurons in heterozygous female transgenic mice with enhanced green fluorescent protein (EGFP) under the control of the Tac2 (NKB) promoter (Tac2-EGFP mice). The onset of puberty, estrous cyclicity, and serum LH were comparable between Tac2-EGFP and wild-type mice. The location of EGFP-immunoreactive neurons was consistent with previous descriptions of Tac2 mRNA-expressing neurons in the rodent. In the arcuate nucleus, nearly 80% of EGFP neurons expressed pro-NKB-immunoreactivity. Moreover, EGFP fluorescent intensity in arcuate neurons was increased by ovariectomy and reduced by 17ß-estradiol (E2) treatment. Electrophysiology of single cells in tissue slices was used to examine the effects of chronic E2 treatment on Tac2-EGFP neurons in the arcuate nucleus of ovariectomized mice. Whole-cell recordings revealed arcuate NKB neurons to be either spontaneously active or silent in both groups. E2 had no significant effect on the basic electrophysiological properties or spontaneous firing frequencies. Arcuate NKB neurons exhibited either tonic or phasic firing patterns in response to a series of square-pulse current injections. Notably, E2 reduced the number of action potentials evoked by depolarizing current injections. This study demonstrates the utility of the Tac2-EGFP mouse for electrophysiological and morphological studies of KNDy neurons in tissue slices. In parallel to E2 negative feedback on LH secretion, E2 decreased the intensity of the EGFP signal and reduced the excitability of NKB neurons in the arcuate nucleus of ovariectomized Tac2-EGFP mice.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Neurocinina B/metabolismo , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Estradiol/sangue , Estradiol/farmacologia , Estrogênios/sangue , Estrogênios/farmacologia , Feminino , Proteínas de Fluorescência Verde/genética , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hormônio Luteinizante/sangue , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Neurocinina B/genética , Neurônios/metabolismo , Ovariectomia , Técnicas de Patch-Clamp , Regiões Promotoras Genéticas/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo
8.
Front Neuroendocrinol ; 34(3): 211-27, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23872331

RESUMO

Despite affecting millions of individuals, the etiology of hot flushes remains unknown. Here we review the physiology of hot flushes, CNS pathways regulating heat-dissipation effectors, and effects of estrogen on thermoregulation in animal models. Based on the marked changes in hypothalamic kisspeptin, neurokinin B and dynorphin (KNDy) neurons in postmenopausal women, we hypothesize that KNDy neurons play a role in the mechanism of flushes. In the rat, KNDy neurons project to preoptic thermoregulatory areas that express the neurokinin 3 receptor (NK3R), the primary receptor for NKB. Furthermore, activation of NK3R in the median preoptic nucleus, part of the heat-defense pathway, reduces body temperature. Finally, ablation of KNDy neurons reduces cutaneous vasodilatation and partially blocks the effects of estrogen on thermoregulation. These data suggest that arcuate KNDy neurons relay estrogen signals to preoptic structures regulating heat-dissipation effectors, supporting the hypothesis that KNDy neurons participate in the generation of flushes.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Dinorfinas/fisiologia , Fogachos/fisiopatologia , Hipotálamo/metabolismo , Kisspeptinas/fisiologia , Neurocinina B/fisiologia , Animais , Regulação da Temperatura Corporal/efeitos dos fármacos , Estradiol/farmacologia , Ciclo Estral/efeitos dos fármacos , Feminino , Humanos , Hormônio Luteinizante/metabolismo , Modelos Biológicos , Neurônios/fisiologia , Ovariectomia , Pós-Menopausa/fisiologia , Área Pré-Óptica/metabolismo , Ratos , Receptores da Neurocinina-3/metabolismo , Transdução de Sinais , Pele/irrigação sanguínea , Cauda/irrigação sanguínea , Vasodilatação
9.
Proc Natl Acad Sci U S A ; 109(48): 19846-51, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-23150555

RESUMO

Estrogen withdrawal in menopausal women leads to hot flushes, a syndrome characterized by the episodic activation of heat dissipation effectors. Despite the extraordinary number of individuals affected, the etiology of flushes remains an enigma. Because menopause is accompanied by marked alterations in hypothalamic kisspeptin/neurokinin B/dynorphin (KNDy) neurons, we hypothesized that these neurons could contribute to the generation of flushes. To determine if KNDy neurons participate in the regulation of body temperature, we evaluated the thermoregulatory effects of ablating KNDy neurons by injecting a selective toxin for neurokinin-3 expressing neurons [NK(3)-saporin (SAP)] into the rat arcuate nucleus. Remarkably, KNDy neuron ablation consistently reduced tail-skin temperature (T(SKIN)), indicating that KNDy neurons facilitate cutaneous vasodilatation, an important heat dissipation effector. Moreover, KNDy ablation blocked the reduction of T(SKIN) by 17ß-estradiol (E(2)), which occurred in the environmental chamber during the light phase, but did not affect the E(2) suppression of T(SKIN) during the dark phase. At the high ambient temperature of 33 °C, the average core temperature (T(CORE)) of ovariectomized (OVX) control rats was significantly elevated, and this value was reduced by E(2) replacement. In contrast, the average T(CORE) of OVX, KNDy-ablated rats was lower than OVX control rats at 33 °C, and not altered by E(2) replacement. These data provide unique evidence that KNDy neurons promote cutaneous vasodilatation and participate in the E(2) modulation of body temperature. Because cutaneous vasodilatation is a cardinal sign of a hot flush, these results support the hypothesis that KNDy neurons could play a role in the generation of flushes.


Assuntos
Regulação da Temperatura Corporal , Kisspeptinas/fisiologia , Neurocinina B/fisiologia , Neurônios/fisiologia , Pele/irrigação sanguínea , Vasodilatação/fisiologia , Animais , Feminino , Ratos , Ratos Sprague-Dawley , Cauda/irrigação sanguínea
10.
Endocrinology ; 153(6): 2800-12, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22508514

RESUMO

Estrogen withdrawal increases gonadotropin secretion and body weight, but the critical cell populations mediating these effects are not well understood. Recent studies have focused on a subpopulation of hypothalamic arcuate neurons that coexpress estrogen receptor α, neurokinin 3 receptor (NK(3)R), kisspeptin, neurokinin B, and dynorphin for the regulation of reproduction. To investigate the function of kisspeptin/neurokinin B/dynorphin (KNDy) neurons, a novel method was developed to ablate these cells using a selective NK(3)R agonist conjugated to the ribosome-inactivating toxin, saporin (NK(3)-SAP). Stereotaxic injections of NK(3)-SAP in the arcuate nucleus ablated KNDy neurons, as demonstrated by the near-complete loss of NK(3)R, NKB, and kisspeptin-immunoreactive (ir) neurons and depletion of the majority of arcuate dynorphin-ir neurons. Selectivity was demonstrated by the preservation of proopiomelanocortin, neuropeptide Y, and GnRH-ir elements in the arcuate nucleus and median eminence. In control rats, ovariectomy (OVX) markedly increased serum LH, FSH, and body weight, and these parameters were subsequently decreased by treatment with 17ß-estradiol. KNDy neuron ablation prevented the rise in serum LH after OVX and attenuated the rise in serum FSH. KNDy neuron ablation did not completely block the suppressive effects of E(2) on gonadotropin secretion, a finding consistent with redundant pathways for estrogen negative feedback. However, regardless of estrogen status, KNDy-ablated rats had lower levels of serum gonadotropins compared with controls. Surprisingly, KNDy neuron ablation prevented the dramatic effects of OVX and 17ß-estradiol (E(2)) replacement on body weight and abdominal girth. These data provide evidence that arcuate KNDy neurons are essential for tonic gonadotropin secretion, the rise in LH after removal of E(2), and the E(2) modulation of body weight.


Assuntos
Peso Corporal/fisiologia , Dinorfinas/metabolismo , Gonadotropinas/metabolismo , Kisspeptinas/metabolismo , Neurocinina B/metabolismo , Neurônios/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/sangue , Imuno-Histoquímica , Imunotoxinas/química , Imunotoxinas/farmacologia , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Ovariectomia , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Inativadoras de Ribossomos Tipo 1/química , Saporinas
11.
Endocrinology ; 152(12): 4894-905, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22028440

RESUMO

Estrogens have pronounced effects on thermoregulation, as illustrated by the occurrence of hot flushes secondary to estrogen withdrawal in menopausal women. Because neurokinin B (NKB) gene expression is markedly increased in the infundibular (arcuate) nucleus of postmenopausal women, and is modulated by estrogen withdrawal and replacement in multiple species, we have hypothesized that NKB neurons could play a role in the generation of flushes. There is no information, however, on whether the primary NKB receptor [neurokinin 3 receptor (NK(3)R)] modulates body temperature in any species. Here, we determine the effects of microinfusion of a selective NK(3)R agonist (senktide) into the rat median preoptic nucleus (MnPO), an important site in the heat-defense pathway. Senktide microinfusion into the rat MnPO decreased core temperature in a dose-dependent manner. The hypothermia induced by senktide was similar in ovariectomized rats with and without 17ß-estradiol replacement. The hypothermic effect of senktide was prolonged in rats exposed to an ambient temperature of 29.0 C, compared with 21.5 C. Senktide microinfusion also altered tail skin vasomotion in rats exposed to an ambient temperature of 29.0 but not 21.5 C. Comparisons of the effects of senktide at different ambient temperatures indicated that the hypothermia was not secondary to thermoregulatory failure or a reduction in cold-induced thermogenesis. Other than a very mild increase in drinking, senktide microinfusion did not affect behavior. Terminal fluorescent dextran microinfusion showed targeting of the MnPO and adjacent septum, and immunohistochemical studies revealed that senktide induced a marked increase in Fos-activation in the MnPO. Because MnPO neurons expressed NK(3)R-immunoreactivity, the induction of MnPO Fos by senktide is likely a direct effect. By demonstrating that NK(3)R activation in the MnPO modulates body temperature, these studies support the hypothesis that hypothalamic NKB neurons could be involved in the generation of menopausal flushes.


Assuntos
Temperatura Corporal , Área Pré-Óptica/metabolismo , Receptores da Neurocinina-3/agonistas , Animais , Regulação da Temperatura Corporal , Estradiol/farmacologia , Feminino , Fogachos/etiologia , Fogachos/metabolismo , Neurocinina B , Neurônios/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Receptores da Neurocinina-3/metabolismo , Substância P/análogos & derivados , Substância P/farmacologia
12.
Endocrinology ; 152(7): 2750-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21521752

RESUMO

Estrogen has pronounced effects on thermoregulation, but the anatomic sites of integration between the reproductive and thermoregulatory axes are unknown. In this study, we tested whether estradiol-17ß (E(2)) treatment would alter the activity of thermoregulatory brain regions responding to mild changes in ambient temperature (T(AMBIENT)). Core and tail skin temperatures were recorded at the ambient temperatures of 20, 24, or 31 C in ovariectomized (OVX) rats with and without E(2). Neuronal activity was evaluated by counting the number of Fos-immunoreactive cells in the brains of rats killed 90 min after exposure to one of the three ambient temperatures. Of 14 brain areas examined, the median preoptic nucleus (MnPO) was the only site that exhibited increased Fos immunoreactivity at the high T(AMBIENT) of 31 C. At 24 C, OVX rats exhibited increased numbers of MnPO Fos-immunoreactive cells, compared with OVX + E(2) rats. Interestingly, tail skin vasomotion and MnPO Fos expression were affected in a similar manner by T(AMBIENT) and E(2) treatment. In the arcuate nucleus and anteroventral periventricular nucleus (AVPV), Fos immunoreactivity was highest at the low T(AMBIENT) of 20 C, with inhibitory (arcuate nucleus) and stimulatory (AVPV) effects of E(2). No other areas responded to both T(AMBIENT) and E(2) treatment. These results implicate the MnPO, the arcuate nucleus, and the AVPV as sites of integration between the reproductive and thermoregulatory axes. Combined with studies showing the importance of MnPO neurons in heat-defense pathways, the MnPO emerges as a likely site for E(2) modulation of thermoregulatory vasomotion.


Assuntos
Regulação da Temperatura Corporal , Estradiol/fisiologia , Estrogênios/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Pele/irrigação sanguínea , Temperatura , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Mapeamento Encefálico , Feminino , Hemodinâmica , Fogachos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Ovariectomia , Área Pré-Óptica/citologia , Área Pré-Óptica/metabolismo , Ratos , Ratos Sprague-Dawley , Pele/metabolismo , Transmissão Sináptica , Terceiro Ventrículo/citologia , Terceiro Ventrículo/metabolismo , Vasodilatação
14.
Endocrinology ; 151(11): 5389-94, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20861232

RESUMO

In the rat, tail skin vasomotion is a primary heat loss mechanism that can be monitored by changes in tail skin temperature (T(SKIN)). Previous studies showed that ovariectomy and estrogen replacement modify T(SKIN) in the rat. Based on these findings, the ovariectomized (OVX) rat has been used as a model to study the mechanisms and treatment of menopausal hot flushes. It is not known, however, if T(SKIN) changes across the estrous cycle in intact rats. Here, we describe an improved method for monitoring T(SKIN) in freely moving rats using a SubCue Mini datalogger mounted on the ventral surface of the tail. This method is noninvasive, cost-effective, and does not require restraints or tethering. We observed a distinct pattern of T(SKIN) across the estrous cycle characterized by low T(SKIN) on proestrous night. To determine whether this pattern was secondary to secretion of ovarian steroids, we monitored the thermoregulatory effects of 17ß-estradiol (E(2)) and E(2) plus progesterone, administered via SILASTIC capsules to OVX rats. E(2) treatment of OVX rats significantly reduced T(SKIN) in the dark phase from 2 to 21 d after hormone treatment. The T(SKIN) of E(2)-treated OVX animals was not significantly different from OVX rats receiving E(2) plus progesterone. These data provide evidence that the reduction in T(SKIN) on proestrous night was secondary to elevated levels of ovarian estrogens. This study provides the first description of T(SKIN) changes with the estrous cycle and supports the role of estrogens in normal thermoregulation in the rat.


Assuntos
Estradiol/farmacologia , Ciclo Estral/fisiologia , Progesterona/farmacologia , Temperatura Cutânea/efeitos dos fármacos , Temperatura Cutânea/fisiologia , Termômetros , Análise de Variância , Animais , Feminino , Ovariectomia , Radioimunoensaio , Ratos
15.
Brain Res ; 1364: 116-28, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20800582

RESUMO

Loss-of-function mutations in the genes encoding either neurokinin B (NKB) or its receptor, NK3 (NK3R), result in hypogonadotropic hypogonadism, characterized by an absence of pubertal development and low circulating levels of LH and gonadal steroids. These studies implicate NKB and NK3R as essential elements of the human reproductive axis. Studies over the last two decades provide evidence that a group of neurons in the hypothalamic infundibular/arcuate nucleus form an important component of this regulatory circuit. These neurons are steroid-responsive and coexpress NKB, kisspeptin, dynorphin, NK3R, and estrogen receptor α (ERα) in a variety of mammalian species. Compelling evidence in the human indicates these neurons function in the hypothalamic circuitry regulating estrogen negative feedback on gonadotropin-releasing hormone (GnRH) secretion. Moreover, in the rat, they form a bilateral, interconnected network that projects to NK3R-expressing GnRH terminals in the median eminence. This network provides an anatomical framework to explain how coordination among NKB/kisspeptin/dynorphin/NK3R/ERα neurons could mediate feedback information from the gonads to modulate pulsatile GnRH secretion. There is substantial (but indirect) evidence that this network may be part of the neural circuitry known as the "GnRH pulse generator," with NK3R signaling as an important component. This theory provides a compelling explanation for the occurrence of hypogonadotropic hypogonadism in patients with inactivating mutations in the TAC3 or TACR3 genes. Future studies will be needed to determine whether NKB signaling plays a permissive role in the onset of puberty or is part of the driving force initiating the maturation of reproductive function.


Assuntos
Hipotálamo/fisiologia , Neurocinina B/fisiologia , Reprodução/fisiologia , Envelhecimento/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/fisiologia , Dinorfinas/fisiologia , Estrogênios/fisiologia , Feminino , Expressão Gênica/genética , Humanos , Kisspeptinas , Macaca mulatta , Masculino , Neurônios/fisiologia , Pós-Menopausa/fisiologia , Gravidez , Receptores da Neurocinina-3/agonistas , Receptores da Neurocinina-3/genética , Receptores da Neurocinina-3/fisiologia , Caracteres Sexuais , Transdução de Sinais/fisiologia , Terminologia como Assunto , Proteínas Supressoras de Tumor/fisiologia
16.
Endocrinology ; 151(3): 1187-93, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20051485

RESUMO

Hot flushes represent a disorder of central thermoregulation characterized by the episodic activation of heat loss mechanisms. Although flushes are associated with estrogen withdrawal, there is little understanding of the effects of estrogen on thermoregulation in any species. It has been proposed that hormone withdrawal increases the sensitivity of hypothalamic neural pathways that control heat dissipation effectors. If so, we predicted that ovariectomized rats without estradiol treatment would activate tail skin vasodilatation (a major heat loss effector) at lower ambient temperatures and thereby lower the thermoneutral zone. The thermoneutral zone, defined as the range of ambient temperatures in which thermoregulation is achieved only by sensible (dry) heat loss, was evaluated based on properties of skin vasomotion. Core and tail skin temperatures were recorded in ovariectomized rats (with and without estradiol-17beta) exposed to ambient temperatures from 13 to 34 C in an environmental chamber. Rats without estradiol exhibited increased skin vasodilatation and a shift in the thermoneutral zone to lower ambient temperatures. Moreover, the ambient temperature threshold for skin vasodilatation was significantly lower in rats without estradiol treatment. At most ambient temperatures, average core temperature was unaffected by estradiol. However, at ambient temperatures of 32.5 C and above, untreated ovariectomized rats exhibited higher core temperatures compared with estradiol-treated rats. Thus, estradiol-17beta treatment enhanced the maintenance of core temperature during heat exposure. These findings support the hypothesis that estrogen withdrawal increases the sensitivity of thermoregulatory neural pathways and modifies the activation of heat loss mechanisms.


Assuntos
Regulação da Temperatura Corporal , Estradiol/fisiologia , Fogachos/fisiopatologia , Vasodilatação , Sistema Vasomotor/fisiopatologia , Animais , Feminino , Ovariectomia , Ratos , Ratos Sprague-Dawley
17.
Peptides ; 30(1): 111-22, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18614256

RESUMO

Menopause is characterized by depletion of ovarian follicles, a reduction of ovarian hormones to castrate levels and elevated levels of serum gonadotropins. Rather than degenerating, the reproductive neuroendocrine axis in postmenopausal women is intact and responds robustly to the removal of ovarian hormones. Studies in both human and non-human primates provide evidence that the gonadotropin hypersecretion in postmenopausal women is secondary to increased gonadotropin-releasing hormone (GnRH) secretion from the hypothalamus. In addition, menopause is accompanied by hypertrophy of neurons in the infundibular (arcuate) nucleus expressing KiSS-1, neurokinin B (NKB), substance P, dynorphin and estrogen receptor alpha (ERalpha) mRNA. Ovariectomy in experimental animals induces nearly identical findings, providing evidence that these changes are a compensatory response to ovarian failure. The anatomical site of the hypertrophied neurons, as well as the extensive data implicating kisspeptin, NKB and dynorphin in the regulation of GnRH secretion, provide compelling evidence that these neurons are part of the neural network responsible for the increased levels of serum gonadotropins in postmenopausal women. We propose that neurons expressing KiSS-1, NKB, substance P, dynorphin and ERalpha mRNA in the infundibular nucleus play an important role in sex-steroid feedback on gonadotropin secretion in the human.


Assuntos
Estrogênios/metabolismo , Retroalimentação Fisiológica , Hipotálamo/fisiologia , Neurocinina B/metabolismo , Neurônios/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Envelhecimento/fisiologia , Animais , Dinorfinas/genética , Dinorfinas/metabolismo , Feminino , Humanos , Hipotálamo/citologia , Kisspeptinas , Menopausa , Pessoa de Meia-Idade , Neurocinina B/genética , Neurônios/citologia , Sistemas Neurossecretores/fisiologia , Ovário/fisiologia , Ratos , Substância P/genética , Substância P/metabolismo , Proteínas Supressoras de Tumor/genética
18.
J Clin Endocrinol Metab ; 92(7): 2744-50, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17488799

RESUMO

CONTEXT: Human menopause is characterized by ovarian failure, gonadotropin hypersecretion, and neuronal hypertrophy in the hypothalamic infundibular (arcuate) nucleus. Recent studies have demonstrated a critical role for kisspeptins in reproductive regulation, but it is not known whether menopause is accompanied by changes in hypothalamic kisspeptin neurons. OBJECTIVES: Our objective was to map the location of neurons expressing kisspeptin gene (KiSS-1) transcripts in the human hypothalamus and determine whether menopause is associated with changes in the size and gene expression of kisspeptin neurons. In monkeys, our objective was to evaluate the effects of ovariectomy and hormone replacement on neurons expressing KiSS-1 mRNA in the infundibular nucleus. SUBJECTS: Hypothalamic tissues were collected at autopsy from eight premenopausal and nine postmenopausal women and from 42 young cynomolgus monkeys in various endocrine states. METHODS: We used hybridization histochemistry, quantitative autoradiography, and computer-assisted microscopy. RESULTS: Examination of human hypothalamic sections revealed that KiSS-1 neurons were located predominantly in the infundibular nucleus. In the infundibular nucleus of postmenopausal women, there was a significant increase in the size of neurons expressing KiSS-1 mRNA and the number of labeled cells and autoradiographic grains per neuron. Similar to postmenopausal women, ovariectomy induced neuronal hypertrophy and increased KiSS-1 gene expression in the monkey infundibular nucleus. Conversely, in ovariectomized monkeys, estrogen replacement markedly reduced KiSS-1 gene expression. CONCLUSIONS: The cynomolgus monkey experiments provide strong evidence that the increase in KiSS-1 neuronal size and gene expression in postmenopausal women is secondary to ovarian failure. These studies suggest that kisspeptin neurons regulate estrogen negative feedback in the human.


Assuntos
Neuro-Hipófise/fisiologia , Pós-Menopausa/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/fisiologia , Terapia de Reposição de Estrogênios , Estrogênios/metabolismo , Estrogênios/farmacologia , Estrogênios/uso terapêutico , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Humanos , Hipertrofia , Kisspeptinas , Macaca fascicularis , Pessoa de Meia-Idade , Neurônios/fisiologia , Ovariectomia , Neuro-Hipófise/citologia , Progesterona/farmacologia , Progesterona/uso terapêutico , RNA Mensageiro/metabolismo
19.
Mol Cell Endocrinol ; 260-262: 40-8, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17079072

RESUMO

Human FSH exists as two major glycoforms designated, tetra-glycosylated and di-glycosylated hFSH. The former possesses both alpha- and beta-subunit carbohydrates while the latter possesses only alpha-subunit carbohydrate. Western blotting differentiated the glycosylated, 24,000 M(r) hFSHbeta band from the non-glycosylated 21,000 M(r) FSHbeta band. Postmenopausal urinary hFSH preparations possessed 75-95% 24,000 M(r) hFSHbeta, while pituitary hFSH immunopurified from 21- to 43-year-old females and 21-43-year-old males possessed only 35-40% 24,000 M(r) hFSHbeta. The pituitary hFSH from a postmenopausal woman on estrogen replacement was 75% 21,000 M(r) hFSHbeta. Other immunopurified postmenopausal pituitary hFSH preparations possessed 50-60% 21,000 M(r) hFSHbeta. Gel filtration removed predominantly 21,000 M(r) free hFSHbeta and reduced its abundance to 13-22% in postmenopausal pituitary hFSH heterodimer preparations. A major regulatory mechanism for FSH glycosylation involves control of beta-subunit N-glycosylation, possibly by inhibition of oligosaccharyl transferase. Two primate species exhibited the same all-or-none pattern of pituitary FSHbeta glycosylation.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/química , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Macaca/metabolismo , Adulto , Idoso , Animais , Western Blotting , Cromatografia em Gel , Feminino , Subunidade beta do Hormônio Folículoestimulante/análise , Glicosilação , Humanos , Compostos Organofosforados/metabolismo , Ovário/cirurgia , Hipófise/química , Isoformas de Proteínas/análise , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
20.
J Comp Neurol ; 498(5): 712-26, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-16917850

RESUMO

Considerable evidence suggests that dynorphin and neurokinin B (NKB) neurons in the hypothalamic arcuate nucleus participate in the sex-steroid regulation of reproduction. In the present study, we used dual-label immunofluorescence to explore the distribution of prodynorphin and proNKB immunoreactivity in the rat hypothalamus. Additionally, we investigated whether arcuate prodynorphin-ir (immunoreactive) neurons expressed the neurokinin 3 receptor (NK3R) or nuclear estrogen receptor-alpha (ERalpha). We found that the majority of prodynorphin-ir neurons in the rat arcuate nucleus expressed proNKB, whereas nearly all (99%) of the proNKB neurons were immunoreactive for prodynorphin. The arcuate nucleus was the only site in the hypothalamus where neuronal somata coexpressing prodynorphin and proNKB-immunoreactivity were identified. A dense plexus of double-labeled prodynorphin/proNKB-ir fibers was found within the arcuate nucleus extending to the median eminence and throughout the periventricular zone of the hypothalamus. Prodynorphin/proNKB fibers were also identified in the paraventricular nucleus, anterior hypothalamic area, medial preoptic area, median preoptic nucleus, anteroventral periventricular nucleus, and bed nucleus of the stria terminalis in a distribution consistent with previously described arcuate nucleus projections. Interestingly, the majority of prodynorphin-ir neurons in the arcuate nucleus expressed NK3R, and nearly 100% of the prodynorphin-ir neurons contained nuclear ERalpha. Our results suggest that there is a close functional relationship between dynorphin and NKB peptides within the arcuate nucleus of the rat, which may include an autofeedback loop mediated through NK3R. The diverse hypothalamic projections of fibers expressing both prodynorphin and proNKB provide evidence that these neurons may participate in a variety of homeostatic and neuroendocrine processes.


Assuntos
Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/fisiologia , Encefalinas/metabolismo , Neurocinina B/metabolismo , Neurônios/metabolismo , Precursores de Proteínas/metabolismo , Animais , Contagem de Células/métodos , Feminino , Imuno-Histoquímica/métodos , Vias Neurais/citologia , Vias Neurais/metabolismo , Neurônios/citologia , Ratos , Ratos Sprague-Dawley , Receptores de Taquicininas/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...