Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurosci Lett ; 773: 136518, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35150776

RESUMO

In normal hormonal conditions, increased neuronal activity in the ventromedial hypothalamus (VMH) induces lordosis whereas activation of the preoptic area (POA) exerts an opposite effect. In the present work, we explored the effect of bilateral infusion of different doses of the apelin-13 (0.37, 0.75, 1.5, and 15 µg) in both brain areas on the expression of lordosis behavior. Lordosis quotient and lordosis reflex score were performed at 30, 120, and 240 min. Weak lordosis was observed following the 0.37 µg dose of apelin-13 at 30 min in the VMH of EB-primed rats; however, the rest of the doses induced significant lordosis relative to the control group. At 120 min, all doses induced lordosis behavior, while at 240 min, the highest dose of 15 µg did not induce significant differences. Interestingly, only the 0.75 µg infusion of apelin in the POA induced significant lordosis at 120 and 240 min. These results indicate that apelin-13 acts preferably in HVM and slightly in POA to initiate lordosis behavior in estrogen-primed rats.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular , Lordose , Área Pré-Óptica , Animais , Estradiol/farmacologia , Estrogênios/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Lordose/induzido quimicamente , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/patologia , Progesterona/farmacologia , Ratos , Comportamento Sexual Animal/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/patologia
2.
Am J Physiol Heart Circ Physiol ; 318(1): H34-H48, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31675258

RESUMO

Obstructive sleep apnea is characterized by interrupted breathing that leads to cardiovascular sequelae including chronic hypertension that can persist into the waking hours. Chronic intermittent hypoxia (CIH), which models the hypoxemia associated with sleep apnea, is sufficient to cause a sustained increase in blood pressure that involves the central nervous system. The median preoptic nucleus (MnPO) is an integrative forebrain region that contributes to blood pressure regulation and neurogenic hypertension. The MnPO projects to the paraventricular nucleus (PVN), a preautonomic region. We hypothesized that pathway-specific lesions of the projection from the MnPO to the PVN would attenuate the sustained component of chronic intermittent hypoxia-induced hypertension. Adult male Sprague-Dawley rats (250-300 g) were anesthetized with isoflurane and stereotaxically injected bilaterally in the PVN with a retrograde Cre-containing adeno-associated virus (AAV; AAV9.CMV.HI.eGFP-Cre.WPRE.SV40) and injected in the MnPO with caspase-3 (AAV5-flex-taCasp3-TEVp) or control virus (AAV5-hSyn-DIO-mCherry). Three weeks after the injections the rats were exposed to a 7-day intermittent hypoxia protocol. During chronic intermittent hypoxia, controls developed a diurnal hypertension that was blunted in rats with caspase lesions. Brain tissue processed for FosB immunohistochemistry showed decreased staining with caspase-induced lesions of MnPO and downstream autonomic-regulating nuclei. Chronic intermittent hypoxia significantly increased plasma levels of advanced oxidative protein products in controls, but this increase was blocked in caspase-lesioned rats. The results indicate that PVN-projecting MnPO neurons play a significant role in blood pressure regulation in the development of persistent chronic intermittent hypoxia hypertension.NEW & NOTEWORTHY Chronic intermittent hypoxia associated with obstructive sleep apnea increases oxidative stress and leads to chronic hypertension. Sustained hypertension may be mediated by angiotensin II-induced neural plasticity of excitatory median preoptic neurons in the forebrain that project to the paraventricular nucleus of the hypothalamus. Selective caspase lesions of these neurons interrupt the drive for sustained hypertension and cause a reduction in circulating oxidative protein products. This indicates that a functional connection between the forebrain and hypothalamus is necessary to drive diurnal hypertension associated with intermittent hypoxia. These results provide new information about central mechanisms that may contribute to neurogenic hypertension.


Assuntos
Apoptose , Pressão Arterial , Caspase 3/metabolismo , Hipertensão/prevenção & controle , Hipóxia/complicações , Núcleo Hipotalâmico Paraventricular/enzimologia , Área Pré-Óptica/enzimologia , Animais , Caspase 3/genética , Ritmo Circadiano , Modelos Animais de Doenças , Frequência Cardíaca , Hipertensão/enzimologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Hipóxia/enzimologia , Hipóxia/patologia , Hipóxia/fisiopatologia , Masculino , Estresse Oxidativo , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Área Pré-Óptica/patologia , Área Pré-Óptica/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais
3.
J Endocrinol ; 231(2): 167-180, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27613338

RESUMO

Water deprivation (WD) induces changes in plasma volume and osmolality, which in turn activate several responses, including thirst, the activation of the renin-angiotensin system (RAS) and vasopressin (AVP) and oxytocin (OT) secretion. These systems seem to be influenced by oestradiol, as evidenced by the expression of its receptor in brain areas that control fluid balance. Thus, we investigated the effects of oestradiol treatment on behavioural and neuroendocrine changes of ovariectomized rats in response to WD. We observed that in response to WD, oestradiol treatment attenuated water intake, plasma osmolality and haematocrit but did not change urinary volume or osmolality. Moreover, oestradiol potentiated WD-induced AVP secretion, but did not alter the plasma OT or angiotensin II (Ang II) concentrations. Immunohistochemical data showed that oestradiol potentiated vasopressinergic neuronal activation in the lateral magnocellular PVN (PaLM) and supraoptic (SON) nuclei but did not induce further changes in Fos expression in the median preoptic nucleus (MnPO) or subfornical organ (SFO) or in oxytocinergic neuronal activation in the SON and PVN of WD rats. Regarding mRNA expression, oestradiol increased OT mRNA expression in the SON and PVN under basal conditions and after WD, but did not induce additional changes in the mRNA expression for AVP in the SON or PVN. It also did not affect the mRNA expression of RAS components in the PVN. In conclusion, our results show that oestradiol acts mainly on the vasopressinergic system in response to WD, potentiating vasopressinergic neuronal activation and AVP secretion without altering AVP mRNA expression.


Assuntos
Desidratação/fisiopatologia , Estradiol/uso terapêutico , Estrogênios/uso terapêutico , Neurônios/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Supraóptico/efeitos dos fármacos , Desequilíbrio Hidroeletrolítico/prevenção & controle , Animais , Arginina Vasopressina/agonistas , Arginina Vasopressina/análise , Arginina Vasopressina/metabolismo , Comportamento Animal/efeitos dos fármacos , Desidratação/terapia , Ingestão de Líquidos/efeitos dos fármacos , Terapia de Reposição de Estrogênios , Feminino , Hidratação , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Ovariectomia/efeitos adversos , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Área Pré-Óptica/patologia , Ratos Wistar , Órgão Subfornical/efeitos dos fármacos , Órgão Subfornical/metabolismo , Órgão Subfornical/patologia , Núcleo Supraóptico/metabolismo , Núcleo Supraóptico/patologia , Núcleo Vestibular Lateral/efeitos dos fármacos , Núcleo Vestibular Lateral/metabolismo , Núcleo Vestibular Lateral/patologia , Desequilíbrio Hidroeletrolítico/sangue , Desequilíbrio Hidroeletrolítico/etiologia , Desequilíbrio Hidroeletrolítico/fisiopatologia
4.
Neuroendocrinology ; 101(2): 120-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25613345

RESUMO

AIMS: We investigated the norepinephrine pathway changes from the locus coeruleus (LC) to the preoptic area of the hypothalamus (POAH) in the brain of ovariectomized rats under low estrogen levels and explored the therapeutic effects of estradiol valerate (E2) and Remifemin (ICR) on these changes. METHODS: 40 female Sprague-Dawley rats were randomly divided into the following groups: surgery with vehicle (SHAM), ovariectomy surgery with vehicle (OVX), ovariectomy with E2 treatment (OVX + E2), and ovariectomy with Remifemin (OVX + ICR). After 4 weeks of treatment, we observed the changes by immunohistochemistry. RESULTS: (1) The average optical density of DBH-ir fibers and the number of α1-adrenoreceptor- and estrogen receptor (ER)α-positive neurons in the main nuclei of POAH were all reduced in OVX rats compared with the SHAM group. The above changes were normalized in all nuclei of the POAH in the E2 group, while they were normalized in some nuclei in the ICR group. Coexpression of ERα and α1-adrenoreceptor was observed in the POAH. (2) The number of DBH- and ERα-positive neurons in the LC decreased in the OVX group compared with the SHAM group and increased after treatment with E2 and ICR. Coexpression of ERα and DBH was observed in the LC. CONCLUSION: Low estrogen (OVX) altered norepinephrine synthesis in the LC, the projection of norepinephrine fibers and α1-adrenoreceptor expression in the POAH. Both E2 and ICR normalized the norepinephrine pathway, but E2 achieved greater effects than ICR. ICR had different effects in different nuclei in the POAH and its therapeutic effect was better in the LC.


Assuntos
Estradiol/análogos & derivados , Estrogênios/farmacologia , Locus Cerúleo/efeitos dos fármacos , Norepinefrina/metabolismo , Extratos Vegetais/farmacologia , Área Pré-Óptica/efeitos dos fármacos , Animais , Cimicifuga , Dopamina beta-Hidroxilase/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Feminino , Imuno-Histoquímica , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Técnicas de Rastreamento Neuroanatômico , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ovariectomia , Área Pré-Óptica/metabolismo , Área Pré-Óptica/patologia , Distribuição Aleatória , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Adv Gerontol ; 27(1): 81-6, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25051762

RESUMO

Kisspeptin activates neurocytes and astrocytes of the preoptic hypothalamic nucleus of 1-, 3- and 24-month-old male rats. Kisspeptin antagonist (P-234) depresses the neurocytes, but not the astrocytes of the preoptic nucleus. Melatonin at a dose of 100 mkg/100 g b.w. inhibits the neurons of old male rats. During combined administration of melatonin and kisspeptin, as well as melatonin and P-234, the state of the kisspeptinergic system is crusial for the activity of the neurons in the preoptic nucleus of 1- and 3-month-old animals. However, in old rats melatonin significantly changes the neuron response of the preoptic nucleus to kisspeptin and its antagonist administration, while it's observed the neuron stimulation. Generally, the state of the kisspeptinergic system has a determining influence on the preoptic hypothalamic nucleus of the immature and young mature male rats. In old rats the cell functional state of the preoptic nucleus depends on the interaction of the kisspeptinergic system and melatonin level.


Assuntos
Astrócitos , Hipotálamo , Kisspeptinas , Melatonina , Neurônios , Fatores Etários , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Biotransformação , Senescência Celular/fisiologia , Vias de Administração de Medicamentos , Hipotálamo/metabolismo , Hipotálamo/patologia , Kisspeptinas/agonistas , Kisspeptinas/antagonistas & inibidores , Kisspeptinas/metabolismo , Kisspeptinas/farmacologia , Masculino , Melatonina/metabolismo , Melatonina/farmacologia , Neurônios/metabolismo , Neurônios/patologia , Área Pré-Óptica/metabolismo , Área Pré-Óptica/patologia , Ratos , Ratos Wistar , Resultado do Tratamento
6.
Mol Cell Endocrinol ; 382(1): 107-119, 2014 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-24064031

RESUMO

Rabbits with high fat diet (HFD)-induced metabolic syndrome (MetS) developed hypogonadotropic hypogonadism (HH) and showed a reduced gonadotropin-releasing hormone (GnRH) immunopositivity in the hypothalamus. This study investigated the relationship between MetS and hypothalamic alterations in HFD-rabbits. Gonadotropin levels decreased as a function of MetS severity, hypothalamic gene expression of glucose transporter 4 (GLUT4) and interleukin-6 (IL-6). HFD determined a low-grade inflammation in the hypothalamus, significantly inducing microglial activation, expression and immunopositivity of IL-6, as well as GLUT4 and reduced immunopositivity for KISS1 receptor, whose mRNA expression was negatively correlated to glucose intolerance. Correcting glucose metabolism with obetcholic acid improved hypothalamic alterations, reducing GLUT4 and IL-6 immunopositivity and significantly increasing GnRH mRNA, without, however, preventing HFD-related HH. No significant effects at the hypothalamic level were observed after systemic anti-inflammatory treatment (infliximab). Our results suggest that HFD-induced metabolic derangements negatively affect GnRH neuron function through an inflammatory injury at the hypothalamic level.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Inflamação/etiologia , Síndrome Metabólica/complicações , Neurônios/patologia , Área Pré-Óptica/metabolismo , Área Pré-Óptica/patologia , Animais , Anticorpos Monoclonais/farmacologia , Biomarcadores/metabolismo , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacologia , Dieta Hiperlipídica , Regulação da Expressão Gênica/efeitos dos fármacos , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Hormônio Liberador de Gonadotropina/sangue , Hormônio Liberador de Gonadotropina/genética , Imuno-Histoquímica , Inflamação/sangue , Infliximab , Masculino , Síndrome Metabólica/sangue , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Área Pré-Óptica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Receptores Acoplados a Proteínas G/metabolismo
7.
J Cereb Blood Flow Metab ; 33(2): 183-90, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23093068

RESUMO

Therapeutic hypothermia is of relevance to treatment of increased body temperature and brain injury, but drugs inducing selective, rapid, and safe cooling in humans are not available. Here, we show that injections of adenosine 5'-monophosphate (AMP), an endogenous nucleotide, promptly triggers hypothermia in mice by directly activating adenosine A1 receptors (A1R) within the preoptic area (POA) of the hypothalamus. Inhibition of constitutive degradation of brain extracellular AMP by targeting ecto 5'-nucleotidase, also suffices to prompt hypothermia in rodents. Accordingly, sensitivity of mice and rats to the hypothermic effect of AMP is inversely related to their hypothalamic 5'-nucleotidase activity. Single-cell electrophysiological recording indicates that AMP reduces spontaneous firing activity of temperature-insensitive neurons of the mouse POA, thereby retuning the hypothalamic thermoregulatory set point towards lower temperatures. Adenosine 5'-monophosphate also suppresses prostaglandin E2-induced fever in mice, having no effects on peripheral hyperthermia triggered by dioxymetamphetamine (ecstasy) overdose. Together, data disclose the role of AMP, 5'-nucleotidase, and A1R in hypothalamic thermoregulation, as well and their therapeutic relevance to treatment of febrile illness.


Assuntos
5'-Nucleotidase/metabolismo , Monofosfato de Adenosina/metabolismo , Regulação da Temperatura Corporal , Febre/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Área Pré-Óptica/metabolismo , Receptor A1 de Adenosina/metabolismo , Monofosfato de Adenosina/farmacologia , Inibidores da Captação Adrenérgica/efeitos adversos , Inibidores da Captação Adrenérgica/farmacologia , Animais , Dinoprostona/efeitos adversos , Dinoprostona/farmacologia , Febre/induzido quimicamente , Febre/tratamento farmacológico , Humanos , Hipotermia Induzida , Masculino , Camundongos , N-Metil-3,4-Metilenodioxianfetamina/efeitos adversos , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Neurônios/metabolismo , Ocitócicos/efeitos adversos , Ocitócicos/farmacologia , Área Pré-Óptica/patologia , Uso Indevido de Medicamentos sob Prescrição , Ratos , Ratos Wistar
8.
Mol Cells ; 32(2): 203-7, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21643849

RESUMO

NELL2, a protein containing epidermal growth factor-like repeat domains, is predominantly expressed in the nervous system. In the mammalian brain, NELL2 expression is mostly neuronal. Previously we found that NELL2 is involved in the onset of female puberty by regulating the release of gonadotropin-releasing hormone (GnRH), and in normal male sexual behavior by controlling the development of the sexually dimorphic nucleus of the preoptic area (POA). In this study we investigated the effect of NELL2 on the female rat estrous cycle. NELL2 expression in the POA was highest during the proestrous phase. NELL2 mRNA levels in the POA were increased by estrogen treatment in ovariectomized female rats. Blocking NELL2 synthesis in the female rat hypothalamus decreased the expression of kisspeptin 1, an important regulator of the GnRH neuronal apparatus, and resulted in disruption of the estrous cycle at the diestrous phase. These results indicate that NELL2 is involved in the maintenance of the normal female reproductive cycle in mammals.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Área Pré-Óptica/metabolismo , Animais , Estrogênios/administração & dosagem , Ciclo Estral/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Masculino , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Oligonucleotídeos/genética , Área Pré-Óptica/patologia , Ratos , Ratos Sprague-Dawley , Comportamento Sexual Animal
9.
J Clin Invest ; 120(10): 3668-72, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20940512

RESUMO

Patients with Kallmann syndrome (KS) have hypogonadotropic hypogonadism caused by a deficiency of gonadotropin-releasing hormone (GnRH) and a defective sense of smell related to olfactory bulb aplasia. Based on the findings in a fetus affected by the X chromosome­linked form of the disease, it has been suggested that hypogonadism in KS results from the failed embryonic migration of neuroendocrine GnRH1 cells from the nasal epithelium to the forebrain. We asked whether this singular observation might extend to other developmental disorders that also include arrhinencephaly. We therefore studied the location of GnRH1 cells in fetuses affected by different arrhinencephalic disorders, specifically X-linked KS, CHARGE syndrome, trisomy 13, and trisomy 18, using immunohistochemistry. Few or no neuroendocrine GnRH1 cells were detected in the preoptic and hypothalamic regions of all arrhinencephalic fetuses, whereas large numbers of these cells were present in control fetuses. In all arrhinencephalic fetuses, many GnRH1 cells were present in the frontonasal region, the first part of their migratory path, as were interrupted olfactory nerve fibers that formed bilateral neuromas. Our findings define a pathological sequence whereby a lack of migration of neuroendocrine GnRH cells stems from the primary embryonic failure of peripheral olfactory structures. This can occur either alone, as in isolated KS, or as part of a pleiotropic disease, such as CHARGE syndrome, trisomy 13, and trisomy 18.


Assuntos
Anormalidades Múltiplas/patologia , Cromossomos Humanos Par 13 , Cromossomos Humanos Par 18 , Hormônio Liberador de Gonadotropina/análise , Síndrome de Kallmann/patologia , Células Neuroendócrinas/fisiologia , Bulbo Olfatório/anormalidades , Trissomia , Movimento Celular , Humanos , Hipotálamo/patologia , Área Pré-Óptica/patologia
10.
Horm Behav ; 58(2): 306-16, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20223236

RESUMO

Reproductive aging in males is characterized by a diminution in sexual behavior beginning in middle age. We investigated the relationships among testosterone, androgen receptor (AR) and estrogen receptor alpha (ERalpha) cell numbers in the hypothalamus, and their relationship to sexual performance in male rats. Young (3months) and middle-aged (12months) rats were given sexual behavior tests, then castrated and implanted with vehicle or testosterone capsules. Rats were tested again for sexual behavior. Numbers of AR and ERalpha immunoreactive cells were counted in the anteroventral periventricular nucleus and the medial preoptic nucleus, and serum hormones were measured. Middle-aged intact rats had significant impairments of all sexual behavior measures compared to young males. After castration and testosterone implantation, sexual behaviors in middle-aged males were largely comparable to those in the young males. In the hypothalamus, AR cell density was significantly (5-fold) higher, and ERalpha cell density significantly (6-fold) lower, in testosterone- than vehicle-treated males, with no age differences. Thus, restoration of serum testosterone to comparable levels in young and middle-aged rats resulted in similar preoptic AR and ERalpha cell density concomitant with a reinstatement of most behaviors. These data suggest that age-related differences in sexual behavior cannot be due to absolute levels of testosterone, and further, the middle-aged brain retains the capacity to respond to exogenous testosterone with changes in hypothalamic AR and ERalpha expression. Our finding that testosterone replacement in aging males has profound effects on hypothalamic receptors and behavior has potential medical implications for the treatment of age-related hypogonadism in men.


Assuntos
Envelhecimento/metabolismo , Receptor alfa de Estrogênio/metabolismo , Hipotálamo/metabolismo , Receptores Androgênicos/metabolismo , Comportamento Sexual Animal/fisiologia , Testosterona/metabolismo , Envelhecimento/patologia , Animais , Contagem de Células , Hipotálamo/patologia , Masculino , Orquiectomia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Área Pré-Óptica/metabolismo , Área Pré-Óptica/patologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Testosterona/sangue
11.
Neurobiol Dis ; 36(1): 126-41, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19631751

RESUMO

Absence epilepsy (AE) in humans and the genetic AE model in WAG/Rij rats are both associated with abnormalities in sleep architecture that suggest insufficiency of the sleep-promoting mechanisms. In this study we compared the functionality of sleep-active neuronal groups within two well-established sleep-promoting sites, the ventrolateral and median preoptic nuclei (VLPO and MnPN, respectively), in WAG/Rij and control rats. Neuronal activity was assessed using c-Fos immunoreactivity and chronic single-unit recording techniques. We found that WAG/Rij rats exhibited a lack of sleep-associated c-Fos activation of GABAergic MnPN and VLPO neurons, a lower percentage of MnPN and VLPO cells increasing discharge during sleep and reduced firing rates of MnPN sleep-active neurons, compared to non-epileptic rats. The role of sleep-promoting mechanisms in pathogenesis of absence seizures was assessed in non-epileptic rats using electrical stimulation and chemical manipulations restricted to the MnPN. We found that fractional activation of the sleep-promoting system in waking was sufficient to elicit absence-like seizures. Given that reciprocally interrelated sleep-promoting and arousal neuronal groups control thalamocortical excitability, we hypothesize that malfunctioning of sleep-promoting system results in impaired ascending control over thalamocortical rhythmogenic mechanisms during wake-sleep transitions thus favoring aberrant thalamocortical oscillations. Our findings suggest a pathological basis for AE-associated sleep abnormalities and a mechanism underlying association of absence seizures with wake-sleep transitions.


Assuntos
Epilepsia Tipo Ausência/patologia , Epilepsia Tipo Ausência/fisiopatologia , Área Pré-Óptica/fisiopatologia , Sono/fisiologia , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Análise de Variância , Animais , Contagem de Células/métodos , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Eletroencefalografia/métodos , Eletromiografia , Epilepsia Tipo Ausência/genética , Glutamato Descarboxilase/metabolismo , Neurônios/fisiologia , Área Pré-Óptica/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Mutantes , Ratos Wistar , Sono/genética , Privação do Sono/fisiopatologia , Vigília/genética
12.
Am J Physiol Regul Integr Comp Physiol ; 295(4): R1020-30, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18685071

RESUMO

Previous data have consistently demonstrated an inhibitory effect of androgens on stress-induced hypothalamic-pituitary-adrenal (HPA) responses. Several brain regions may influence androgen-mediated inhibition of the HPA axis, including the medial preoptic area. To test the role of the medial preoptic nucleus (MPN) specifically, we examined in high- and low-testosterone-replaced gonadectomized rats bearing discrete bilateral lesions of the MPN basal and stress-induced indexes of HPA function, and the relative levels of corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP) mRNA in the amygdala. High testosterone replacement decreased plasma adrenocorticotropin hormone (ACTH) and paraventricular nucleus (PVN) Fos responses to restraint exposure in sham- but not in MPN-lesioned animals. AVP-, but not CRH-immunoreactivity staining in the external zone of the median eminence was increased by testosterone in sham animals, and MPN lesions blocked this increment in AVP. A similar interaction between MPN lesions and testosterone occurred on AVP mRNA levels in the medial nucleus of the amygdala. These findings support an involvement of MPN projections in mediating the AVP response to testosterone in both the medial parvocellular PVN and medial amygdala. We conclude that the MPN forms part of an integral circuit that mediates the central effects of gonadal status on neuroendocrine and central stress responses.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Área Pré-Óptica/fisiologia , Testosterona/farmacologia , Hormônio Adrenocorticotrópico/sangue , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/fisiologia , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Peso Corporal/efeitos dos fármacos , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Implantes de Medicamento , Expressão Gênica/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Ácido Ibotênico/administração & dosagem , Ácido Ibotênico/farmacologia , Masculino , Orquiectomia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Testosterona/administração & dosagem , Testosterona/sangue
13.
Neuroendocrinology ; 87(2): 113-20, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17934249

RESUMO

Recent studies implicate the inhibitory neurotransmitter, gamma-aminobutyric acid (GABA), in septopreoptic (SPO) mechanisms that suppress preovulatory pituitary luteinizing hormone (LH) secretion during neuroglucopenia. Since Fos immunolabeling of the SPO of rats treated by caudal fourth ventricular (CV4) administration of the glucose antimetabolite, 5-thioglucose (5TG), parallels the distribution of GABA neuronal perikarya, the current studies investigated the genomic responsiveness of neuroanatomically-defined populations of glutamate decarboxylase (GAD)-immunoreactive (-ir) neurons in this region of the brain to hindbrain glucoprivation. In lieu of reports that CV4 5TG enhances SPO GABA turnover via mu opioid receptor (mu-R)-dependent mechanisms and evidence that GAD- and mu-R-ir are codistributed within the SPO, patterns of cellular colocalization of these antigens were also evaluated here. Neural tissue was obtained from groups of steroid-primed ovariectomized female rats 2 h after CV4 injection of vehicle or 5TG. Neuronal cell bodies in the lateral and medial septum, medial (MPN) and median preoptic nuclei (MEPO), and rostral medial preoptic area (rMPO) were immunostained for cytoplasmic GAD-ir, but only GAD-reactive neurons in the rMPO and MEPO exhibited robust nuclear colabeling for Fos in response to 5TG. SPO GABA neurons in the vehicle-treated controls were uniformly Fos-ir-negative. Dual immunolabeling for GAD- and mu-R revealed approximately 52% and 36% colabeling of this phenotype in the MEPO and MPN, and colocalization of lesser magnitude (18%) in the rMPO. These results demonstrate site-specific genomic activation of GABAergic neurons in the female rat SPO by CV4 glucose antimetabolite administration, and implicate MEPO and rMPO GABA cell populations in neural pathways that mediate regulatory effects of hindbrain glucoprivic signaling on CNS functions, including inhibition of the steroid positive feedback-activated gonadotropin-releasing hormone/LH neuroendocrine axis. The current studies also support the view that a proportion of neuroglucoprivic-sensitive GABA neurons in the MEPO and rMPO may be direct substrates for mu-R ligand modulatory actions during this state of central substrate imbalance.


Assuntos
Glucose/metabolismo , Glutamato Descarboxilase/metabolismo , Neurônios/enzimologia , Área Pré-Óptica/metabolismo , Rombencéfalo/metabolismo , Animais , Feminino , Hormônio Luteinizante/metabolismo , Neurônios/patologia , Ovariectomia , Área Pré-Óptica/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Rombencéfalo/patologia , Ácido gama-Aminobutírico/metabolismo
14.
Endocrinology ; 149(2): 597-604, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18006629

RESUMO

The absolute requirement for reproduction implies that the hypothalamo-pituitary-gonadal axis, controlling fertility, is an evolutionary robust mechanism. The GnRH neurons of the hypothalamus represent the key cell type within the body dictating fertility. However, the level of functional redundancy within the GnRH neuron population is unknown. As a result of a fortuitous transgene insertion event, GNR23 mice exhibit a marked allele-dependent reduction in GnRH neuron number within their brain. Wild-type mice have approximately 600 GnRH neurons, compared with approximately 200 (34%) and approximately 70 (12%) in GNR23(+/-) and GNR23(-/-) mice, respectively. Using these mice, we examined the minimal GnRH neuron requirements for fertility. Male GNR23(-/-) mice exhibited normal fertility. In contrast, female GNR23(-/-) mice were markedly subfertile, failing to produce normal litters, have estrous cycles, or ovulate. The failure of ovulation resulted from an inability of the few existing GnRH neurons to generate the LH surge. This was not the case, however, for the first cycle at puberty that appeared normal. Together, these observations demonstrate that 12% of the GnRH neuron population is sufficient for pulsatile gonadotropin secretion and puberty onset, whereas between 12 and 34% are required for cyclical control in adult female mice. This indicates that substantial redundancy exists within the GnRH neuronal population and suggests that the great majority of GnRH neurons must be dysfunctional before fertility is affected.


Assuntos
Fertilidade/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Infertilidade Feminina/fisiopatologia , Ovulação/fisiologia , Maturidade Sexual/fisiologia , Animais , Contagem de Células , Ciclo Estral/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/genética , Infertilidade Feminina/patologia , Hormônio Luteinizante/sangue , Eminência Mediana/patologia , Eminência Mediana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Neurônios/patologia , Neurônios/fisiologia , Ovariectomia , Área Pré-Óptica/patologia , Área Pré-Óptica/fisiologia
15.
Adv Gerontol ; 20(4): 61-3, 2007.
Artigo em Russo | MEDLINE | ID: mdl-18383712

RESUMO

In investigations carried out with organotypic culture of mediobasal preoptic area (MPA) of hypothalamus it was established that pineal peptide preparations epitalon (2 ng/ml) and epithalamin (100 ng/ml) stimulate the development of proliferative activity of explants in 3 month and 24 months female rats. It has been shown that epithalamin is more effective in young rats in comparison with old animals, but epitalon as well as epithalamin have almost the same less pronounced inducing effect on growth zone in MPA explants from young and old animals. This effect is tissue specific and could be dependent on inhibition of apoptosis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Oligopeptídeos/farmacologia , Peptídeos/farmacologia , Glândula Pineal/química , Área Pré-Óptica/efeitos dos fármacos , Envelhecimento/patologia , Animais , Apoptose/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Oligopeptídeos/isolamento & purificação , Peptídeos/isolamento & purificação , Área Pré-Óptica/patologia , Ratos , Ratos Wistar , Técnicas de Cultura de Tecidos , Proteína Supressora de Tumor p53/biossíntese
16.
Neuroscience ; 135(3): 679-90, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16154283

RESUMO

Rapid eye movement sleep plays a vital role in the survival of animals. Its deprivation causes alterations in brain functions and behaviors including activities of important enzymes, neurotransmitter levels, impairment of neural excitability and memory consolidation. However, there was a lack of knowledge regarding the effects of rapid eye movement sleep deprivation on neuronal morphology that may get affected much earlier than any permanent damage to the neurons. In the present study, some of these issues have been addressed by studying the effects of rapid eye movement sleep deprivation on various morphological parameters viz. neuronal perimeter, area and shape of neurons located in brain areas known to regulate rapid eye movement sleep and as a control in other brain areas which do not regulate rapid eye movement sleep. The results showed that rapid eye movement sleep deprivation differentially affected neurons depending on their physiological correlates of rapid eye movement sleep and neurotransmitter content. The effects could be reversed if the animals were allowed to recover from rapid eye movement sleep loss or by applying alpha1-adrenergic antagonist, prazosin. The findings in rats support reported data and help explaining previous observations.


Assuntos
Encéfalo/patologia , Encéfalo/ultraestrutura , Neurônios/patologia , Neurônios/ultraestrutura , Privação do Sono/patologia , Sono REM/fisiologia , Antagonistas de Receptores Adrenérgicos alfa 1 , Antagonistas Adrenérgicos alfa/farmacologia , Animais , Química Encefálica/fisiologia , Tamanho Celular , Colina O-Acetiltransferase/metabolismo , Imuno-Histoquímica , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Masculino , Neurônios/metabolismo , Neurotransmissores/metabolismo , Área Pré-Óptica/metabolismo , Área Pré-Óptica/patologia , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
17.
J Toxicol Sci ; 30(1): 7-18, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15800398

RESUMO

Diethylstilbestrol (DES) was administered subcutaneously at 0.5, 1.5 or 4.5 microg/kg/day (DES 0.5, 1.5 and 4.5 groups, respectively) to pregnant SD rats daily on days 7-21 of gestation, to investigate its effects on the development and functions of the reproductive system in their male offspring. Of the 10 pregnant rats in the DES 4.5 group, only 1 delivered, and this rat could not suckle the pups. Rat pups in the DES 0.5 and 1.5 groups were autopsied at 1, 3, 6 and 15 weeks after birth. The testosterone concentrations in the DES 1.5 and 0.5 groups at 6 weeks were significantly decreased and the plasma LH concentrations were not altered. In the DES 1.5 group, DES treatment did not change the volume of the sexually dimorphic nucleus in the preoptic area (SDN-POA) in the male offspring, although this dose of DES increased the volume of SDN-POA in female offspring. The DES treatment altered frequencies in the cycles of the seminiferous tubules, and suppressed histological maturation in the epididymis and the prostate weight. These observations indicate that prenatally administered DES impairs testicular endocrine function continuously as well as pituitary function, but the induced low level of testosterone disrupts spermatogenesis and permanently inhibits the morphological development of epididymis and prostate.


Assuntos
Dietilestilbestrol/toxicidade , Genitália Masculina/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Área Pré-Óptica/efeitos dos fármacos , Diferenciação Sexual/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Epididimo/efeitos dos fármacos , Feminino , Masculino , Exposição Materna , Gravidez , Área Pré-Óptica/patologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores LHRH/genética , Espermatogênese/efeitos dos fármacos
18.
Toxicology ; 208(1): 35-48, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15664431

RESUMO

Steroid hormones are powerful regulators of gene transcription in the brain and have the potential to permanently alter the structure and function of the developing brain. Steroid-mediated altered gene expression may thus be responsible for the molecular cascade for sexual differentiation. In this study, to assess effects of maternal exposure to ethinylestradiol (EE) on brain sexual differentiation of offspring, region-specific mRNA expression of two estrogen-responsive genes, gamma-aminobutyric acid transporter type 1 (GAT-1) and anti-apoptotic bcl-xL was measured in the medial preoptic area (MPOA), including sexually dimorphic nucleus (SDN), at the late stage of brain sexual differentiation in rats. Pregnant Sprague-Dawley animals were fed diets containing EE at concentrations of 0, 0.02, 0.1, and 0.5 ppm from day 15 of pregnancy to day 9 after delivery. In another group, neonates were directly injected with estradiol benzoate (EB: 10 microg/pup, sc) on postnatal day (PND) 2. The MPOA on PND 9 was microdissected from methacarn-fixed paraffin-embedded brain sections to measure mRNA levels by competitive RT-PCR, followed by plate hybridization. EE-exposure decreased GAT-1 expression dose-dependently from 0.02 ppm in females and at 0.5 ppm in males, while EB-treatment caused reduction only in females. EE-exposure did not alter Bcl-xL levels. At week 11, EE-exposed females exhibited a similar spectrum of histopathological changes in endocrine-linked organs as with EB, evident from 0.1 ppm, while in males EE-exposure did not cause histopathological alteration despite clear change with EB-treatment. Measurement of SDN-POA dimensions at week 11 revealed volume reduction in males exposed to 0.5 ppm EE or EB. The results suggest that GAT-1 expression in the developing MPOA is a sensitive measure for the level of disruption of brain sexual differentiation due to maternal dietary exposure to estrogens, despite definite reproductive abnormalities may not be detectable in males with this exposure protocol.


Assuntos
Estrogênios/toxicidade , Etinilestradiol/toxicidade , Proteínas de Membrana Transportadoras/biossíntese , Efeitos Tardios da Exposição Pré-Natal , Área Pré-Óptica/efeitos dos fármacos , RNA Mensageiro/biossíntese , Animais , Peso Corporal/efeitos dos fármacos , Regulação para Baixo , Feminino , Proteínas da Membrana Plasmática de Transporte de GABA , Regulação da Expressão Gênica no Desenvolvimento , Técnicas In Vitro , Masculino , Proteínas de Membrana Transportadoras/genética , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Área Pré-Óptica/metabolismo , Área Pré-Óptica/patologia , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
19.
Mol Cell Neurosci ; 27(4): 466-76, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15555924

RESUMO

The aromatase knockout (ArKO) mouse is unable to synthesize estrogens. Immunohistochemical studies on active caspase-3 and tyrosine hydroxylase (TH) revealed apoptosis of dopaminergic neurons in the medial preoptic area (MPO) and arcuate nucleus (Arc) of the hypothalamus of 1-year-old (1yo) male ArKO mice while no active caspase-3 was detected in wild type (WT). Furthermore, the number of TH-positive cells in the MPO and caudal Arc was significantly decreased in 1yo ArKO compared to WT. RNase protection assays support the presence of apoptosis in 1yo ArKO hypothalamus, revealing an up-regulation of pro-apoptotic genes: FASL, FADD, and caspase-8. Concomitantly, the ratio of bcl-2-related anti-apoptotic genes to pro-apoptotic genes in the hypothalamus of 1yo ArKO mice was significantly down-regulated. Previously, we have reported that no such changes were observed in the hypothalamus of female ArKO mice. Thus, we have provided direct evidence that estrogen is required to maintain the survival and functional integrity of dopaminergic neurons in the MPO and Arc of male, but not female mice.


Assuntos
Apoptose/genética , Núcleo Arqueado do Hipotálamo/fisiopatologia , Dopamina/metabolismo , Estrogênios/deficiência , Degeneração Neural/metabolismo , Área Pré-Óptica/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Núcleo Arqueado do Hipotálamo/patologia , Aromatase/deficiência , Aromatase/genética , Caspase 3 , Caspase 8 , Caspases/genética , Caspases/metabolismo , Contagem de Células , Sobrevivência Celular/genética , Regulação para Baixo/genética , Estrogênios/biossíntese , Proteína Ligante Fas , Proteína de Domínio de Morte Associada a Fas , Genes bcl-2/genética , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Degeneração Neural/genética , Área Pré-Óptica/patologia , RNA Mensageiro/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Regulação para Cima/genética
20.
Toxicol Sci ; 71(1): 74-83, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12520077

RESUMO

The present studies report the effects on neonatal rats of oral exposure to genistein during the period from birth to postnatal day (PND) 21 to generate data for use in assessing human risk following oral ingestion of genistein. Failure to demonstrate significant exposure of the newborn pups via the mothers milk led us to subcutaneously inject genistein into the pups over the period PND 1-7, followed by daily gavage dosing to PND 21. The targeted doses throughout were 4 mg/kg/day genistein (equivalent to the average exposure of infants to total isoflavones in soy milk) and a dose 10 times higher than this (40 mg/kg genistein). The dose used during the injection phase of the experiment was based on plasma determinations of genistein and its major metabolites. Diethylstilbestrol (DES) at 10 micro g/kg was used as a positive control agent for assessment of changes in the sexually dimorphic nucleus of the preoptic area (SDN-POA). Administration of 40 mg/kg genistein increased uterus weights at day 22, advanced the mean day of vaginal opening, and induced permanent estrus in the developing female pups. Progesterone concentrations were also decreased in the mature females. There were no effects in females dosed with 4 mg/kg genistein, the predicted exposure level for infants drinking soy-based infant formulas. There were no consistent effects on male offspring at either dose level of genistein. Although genistein is estrogenic at 40 mg/kg/day, as illustrated by the effects described above, this dose does not have the same repercussions as DES in terms of the organizational effects on the SDN-POA.


Assuntos
Animais Recém-Nascidos/fisiologia , Genisteína/toxicidade , Inibidores do Crescimento/toxicidade , Crescimento/efeitos dos fármacos , Administração Oral , Animais , Estro/efeitos dos fármacos , Feminino , Genisteína/administração & dosagem , Inibidores do Crescimento/administração & dosagem , Injeções Subcutâneas , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/patologia , Progesterona/sangue , Ratos , Caracteres Sexuais , Maturidade Sexual/efeitos dos fármacos , Útero/efeitos dos fármacos , Útero/patologia , Vagina/efeitos dos fármacos , Vagina/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA