Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Neurosci Lett ; 773: 136518, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35150776

RESUMEN

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.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Lordosis , Área Preóptica , Animales , Estradiol/farmacología , Estrógenos/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/patología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Lordosis/inducido químicamente , Área Preóptica/efectos de los fármacos , Área Preóptica/patología , Progesterona/farmacología , Ratas , Conducta Sexual Animal/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/patología
2.
Sci Rep ; 11(1): 3148, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542258

RESUMEN

Delayed emergence from anesthesia was previously reported in a case study of a child with Glycine Encephalopathy. To investigate the neural basis of this delayed emergence, we developed a zebrafish glial glycine transporter (glyt1 - / -) mutant model. We compared locomotor behaviors; dose-response curves for tricaine, ketamine, and 2,6-diisopropylphenol (propofol); time to emergence from these anesthetics; and time to emergence from propofol after craniotomy in glyt1-/- mutants and their siblings. To identify differentially active brain regions in glyt1-/- mutants, we used pERK immunohistochemistry as a proxy for brain-wide neuronal activity. We show that glyt1-/- mutants initiated normal bouts of movement less frequently indicating lethargy-like behaviors. Despite similar anesthesia dose-response curves, glyt1-/- mutants took over twice as long as their siblings to emerge from ketamine or propofol, mimicking findings from the human case study. Reducing glycine levels rescued timely emergence in glyt1-/- mutants, pointing to a causal role for elevated glycine. Brain-wide pERK staining showed elevated activity in hypnotic brain regions in glyt1-/- mutants under baseline conditions and a delay in sensorimotor integration during emergence from anesthesia. Our study links elevated activity in preoptic brain regions and reduced sensorimotor integration to lethargy-like behaviors and delayed emergence from propofol in glyt1-/- mutants.


Asunto(s)
Retraso en el Despertar Posanestésico/genética , Proteínas de Transporte de Glicina en la Membrana Plasmática/genética , Glicina/metabolismo , Hiperglicinemia no Cetósica/genética , Neuronas/metabolismo , Área Preóptica/metabolismo , Proteínas de Pez Cebra/genética , Aminobenzoatos , Anestesia General , Anestésicos , Animales , Animales Modificados Genéticamente , Craneotomía , Retraso en el Despertar Posanestésico/metabolismo , Retraso en el Despertar Posanestésico/fisiopatología , Retraso en el Despertar Posanestésico/prevención & control , Modelos Animales de Enfermedad , Expresión Génica , Glicina/farmacología , Proteínas de Transporte de Glicina en la Membrana Plasmática/deficiencia , Hiperglicinemia no Cetósica/tratamiento farmacológico , Hiperglicinemia no Cetósica/metabolismo , Hiperglicinemia no Cetósica/fisiopatología , Ketamina , Locomoción/fisiología , Neuronas/efectos de los fármacos , Neuronas/patología , Área Preóptica/efectos de los fármacos , Área Preóptica/patología , Propofol , Pez Cebra , Proteínas de Pez Cebra/deficiencia , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
3.
EMBO J ; 40(5): e104267, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33491217

RESUMEN

Impairments in social relationships and awareness are features observed in autism spectrum disorders (ASDs). However, the underlying mechanisms remain poorly understood. Shank2 is a high-confidence ASD candidate gene and localizes primarily to postsynaptic densities (PSDs) of excitatory synapses in the central nervous system (CNS). We show here that loss of Shank2 in mice leads to a lack of social attachment and bonding behavior towards pubs independent of hormonal, cognitive, or sensitive deficits. Shank2-/- mice display functional changes in nuclei of the social attachment circuit that were most prominent in the medial preoptic area (MPOA) of the hypothalamus. Selective enhancement of MPOA activity by DREADD technology re-established social bonding behavior in Shank2-/- mice, providing evidence that the identified circuit might be crucial for explaining how social deficits in ASD can arise.


Asunto(s)
Trastorno Autístico/tratamiento farmacológico , Modelos Animales de Enfermedad , Relaciones Interpersonales , Conducta Materna/efectos de los fármacos , Proteínas del Tejido Nervioso/fisiología , Piperazinas/farmacología , Área Preóptica/efectos de los fármacos , Animales , Trastorno Autístico/etiología , Trastorno Autístico/metabolismo , Trastorno Autístico/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Área Preóptica/metabolismo , Área Preóptica/patología , Sinapsis
4.
Am J Physiol Heart Circ Physiol ; 318(1): H34-H48, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31675258

RESUMEN

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.


Asunto(s)
Apoptosis , Presión Arterial , Caspasa 3/metabolismo , Hipertensión/prevención & control , Hipoxia/complicaciones , Núcleo Hipotalámico Paraventricular/enzimología , Área Preóptica/enzimología , Animales , Caspasa 3/genética , Ritmo Circadiano , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Hipertensión/enzimología , Hipertensión/patología , Hipertensión/fisiopatología , Hipoxia/enzimología , Hipoxia/patología , Hipoxia/fisiopatología , Masculino , Estrés Oxidativo , Núcleo Hipotalámico Paraventricular/patología , Núcleo Hipotalámico Paraventricular/fisiopatología , Área Preóptica/patología , Área Preóptica/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Transducción de Señal
5.
Horm Behav ; 112: 65-76, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30959023

RESUMEN

Sex differences in the anatomy and physiology of the vertebrate preoptic area (POA) arise during development, and influence sex-specific reproductive functions later in life. Relative to masculinization, mechanisms for feminization of the POA are not well understood. The purpose of this study was to induce sex change from male to female in the anemonefish Amphiprion ocellaris, and track the timing of changes in POA cytoarchitecture, composition of the gonads and circulating sex steroid levels. Reproductive males were paired together and then sampled after 3 weeks, 6 months, 1 year and 3 years. Results show that as males change sex into females, number of medium cells in the anterior POA (parvocellular region) approximately double to female levels over the course of several months to 1 year. Feminization of gonads, and plasma sex steroids occur independently, on a variable timescale, up to years after POA sex change has completed. Findings suggest the process of POA feminization is orchestrated by factors originating from within the brain as opposed to being cued from the gonads, consistent with the dominant hypothesis in mammals. Anemonefish provide an opportunity to explore active mechanisms responsible for female brain development in an individual with male gonads and circulating sex steroid levels.


Asunto(s)
Feminización/etiología , Feminización/patología , Gónadas/fisiología , Perciformes/fisiología , Área Preóptica/fisiología , Animales , Encéfalo/patología , Recuento de Células , Femenino , Feminización/sangre , Feminización/veterinaria , Hormonas Esteroides Gonadales/sangre , Gónadas/patología , Masculino , Perciformes/metabolismo , Área Preóptica/patología , Caracteres Sexuales , Diferenciación Sexual/fisiología , Testículo/patología
6.
Sci Rep ; 9(1): 4837, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30886382

RESUMEN

Sexual differentiation is the early life process by which the brain is prepared for male or female typical behaviors, and is directed by sex chromosomes, hormones and early life experiences. We have recently found that innate immune cells residing in the brain, including microglia and mast cells, are more numerous in the male than female rat brain. Neuroimmune cells are also key participants in the sexual differentiation process, specifically organizing the synaptic development of the preoptic area and leading to male-typical sexual behavior in adulthood. Mast cells are known for their roles in allergic responses, thus in this study we sought to determine if exposure to an allergic response of the pregnant female in utero would alter the sexual differentiation of the preoptic area of offspring and resulting sociosexual behavior in later life. Pregnant rats were sensitized to ovalbumin (OVA), bred, and challenged intranasally with OVA on gestational day 15, which produced robust allergic inflammation, as measured by elevated immunoglobulin E. Offspring of these challenged mother rats were assessed relative to control rats in the early neonatal period for mast cell and microglia activation within their brains, downstream dendritic spine patterning on POA neurons, or grown to adulthood to assess behavior and dendritic spines. In utero exposure to allergic inflammation increased mast cell and microglia activation in the neonatal brain, and led to masculinization of dendritic spine density in the female POA. In adulthood, OVA-exposed females showed an increase in male-typical mounting behavior relative to control females. In contrast, OVA-exposed males showed evidence of dysmasculinization, including reduced microglia activation, reduced neonatal dendritic spine density, decreased male-typical copulatory behavior, and decreased olfactory preference for female-typical cues. Together these studies show that early life allergic events may contribute to natural variations in both male and female sexual behavior, potentially via underlying effects on brain-resident mast cells.


Asunto(s)
Alérgenos/inmunología , Neuroinmunomodulación/fisiología , Efectos Tardíos de la Exposición Prenatal/inmunología , Diferenciación Sexual/inmunología , Conducta Sexual Animal/fisiología , Animales , Técnicas de Observación Conductual , Señales (Psicología) , Espinas Dendríticas/inmunología , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Masculino , Mastocitos/inmunología , Exposición Materna/efectos adversos , Microglía/inmunología , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/psicología , Área Preóptica/citología , Área Preóptica/inmunología , Área Preóptica/patología , Área Preóptica/fisiopatología , Ratas , Conducta Social
7.
Reproduction ; 154(2): 145-152, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28559473

RESUMEN

Paracetamol/acetaminophen (N-Acetyl-p-Aminophenol; APAP) is the preferred analgesic for pain relief and fever during pregnancy. It has therefore caused concern that several studies have reported that prenatal exposure to APAP results in developmental alterations in both the reproductive tract and the brain. Genitals and nervous system of male mammals are actively masculinised during foetal development and early postnatal life by the combined actions of prostaglandins and androgens, resulting in the male-typical reproductive behaviour seen in adulthood. Both androgens and prostaglandins are known to be inhibited by APAP. Through intrauterine exposure experiments in C57BL/6 mice, we found that exposure to APAP decreased neuronal number in the sexually dimorphic nucleus (SDN) of the preoptic area (POA) in the anterior hypothalamus of male adult offspring. Likewise, exposure to the environmental pollutant and precursor of APAP, aniline, resulted in a similar reduction. Decrease in neuronal number in the SDN-POA is associated with reductions in male sexual behaviour. Consistent with the changes, male mice exposed in uteri to APAP exhibited changes in urinary marking behaviour as adults and had a less aggressive territorial display towards intruders of the same gender. Additionally, exposed males had reduced intromissions and ejaculations during mating with females in oestrus. Together, these data suggest that prenatal exposure to APAP may impair male sexual behaviour in adulthood by disrupting the sexual neurobehavioral programming. These findings add to the growing body of evidence suggesting the need to limit the widespread exposure and use of APAP by pregnant women.


Asunto(s)
Acetaminofén/toxicidad , Compuestos de Anilina/toxicidad , Neuronas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Área Preóptica/efectos de los fármacos , Caracteres Sexuales , Conducta Sexual Animal/efectos de los fármacos , Agresión/efectos de los fármacos , Animales , Eyaculación/efectos de los fármacos , Femenino , Edad Gestacional , Masculino , Ratones Endogámicos C57BL , Neuronas/patología , Embarazo , Área Preóptica/crecimiento & desarrollo , Área Preóptica/patología , Medición de Riesgo , Territorialidad , Micción/efectos de los fármacos
8.
J Endocrinol ; 231(2): 167-180, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27613338

RESUMEN

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.


Asunto(s)
Deshidratación/fisiopatología , Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Neuronas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Supraóptico/efectos de los fármacos , Desequilibrio Hidroelectrolítico/prevención & control , Animales , Arginina Vasopresina/agonistas , Arginina Vasopresina/análisis , Arginina Vasopresina/metabolismo , Conducta Animal/efectos de los fármacos , Deshidratación/terapia , Ingestión de Líquidos/efectos de los fármacos , Terapia de Reemplazo de Estrógeno , Femenino , Fluidoterapia , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Ovariectomía/efectos adversos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/patología , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Área Preóptica/patología , Ratas Wistar , Órgano Subfornical/efectos de los fármacos , Órgano Subfornical/metabolismo , Órgano Subfornical/patología , Núcleo Supraóptico/metabolismo , Núcleo Supraóptico/patología , Núcleo Vestibular Lateral/efectos de los fármacos , Núcleo Vestibular Lateral/metabolismo , Núcleo Vestibular Lateral/patología , Desequilibrio Hidroelectrolítico/sangre , Desequilibrio Hidroelectrolítico/etiología , Desequilibrio Hidroelectrolítico/fisiopatología
9.
Neuroendocrinology ; 101(2): 120-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25613345

RESUMEN

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.


Asunto(s)
Estradiol/análogos & derivados , Estrógenos/farmacología , Locus Coeruleus/efectos de los fármacos , Norepinefrina/metabolismo , Extractos Vegetales/farmacología , Área Preóptica/efectos de los fármacos , Animales , Cimicifuga , Dopamina beta-Hidroxilasa/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Inmunohistoquímica , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Técnicas de Trazados de Vías Neuroanatómicas , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ovariectomía , Área Preóptica/metabolismo , Área Preóptica/patología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Acta Med Iran ; 53(1): 1-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25597598

RESUMEN

In Wistar rats, reproductive behavior is controlled in a neural circuit of ventral forebrain including the medial amygdala (Me), bed nucleus of the stria terminalis (BNST) and medial preoptic area (MPOA) via perception of social odors. Diabetes Mellitus (DM) is a widespread metabolic disease that affects many organs in a variety of levels. DM can cause central neuropathies such as neuronal apoptosis, dendritic atrophy, neurochemical alterations and also causes reproductive dysfunctions. So we hypothesized damage to the nuclei of this circuit can cause reproductive dysfunctions. Therefore in this project we assessed diabetic effect on these nuclei. For this purpose neuron tracing technique and TUNEL assay were used. We injected HRP in the MPOA and counted labeled cells in the Me and BNST to evaluate the reduction of neurons in diabetic animals. Also, coronal sections were analyzed with the TMB histochemistry method. Animals in this study were adult male Wistar rats (230 ± 8g) divided to control and 10-week streptozotocin-induced diabetic groups. After data analysis by SPSS 16 software, a significant reduction of HRP-labeled neurons was shown in both Me and BNST nuclei in the diabetic group. Moreover, apoptotic cells were significantly observed in diabetic animals in contrast to control the group. In conclusion, these alterations of the circuit as a result of diabetes might be one of the reasons for reproductive dysfunctions.


Asunto(s)
Amígdala del Cerebelo/patología , Diabetes Mellitus Experimental/fisiopatología , Hiperglucemia/patología , Vías Nerviosas/fisiopatología , Área Preóptica/patología , Núcleos Septales/patología , Amígdala del Cerebelo/fisiopatología , Animales , Apoptosis , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Hiperglucemia/etiología , Hiperglucemia/fisiopatología , Masculino , Vías Nerviosas/patología , Neuronas/patología , Área Preóptica/fisiopatología , Ratas , Ratas Wistar , Núcleos Septales/fisiopatología
11.
Brain ; 137(Pt 10): 2847-61, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25142380

RESUMEN

Fragmented sleep is a common and troubling symptom in ageing and Alzheimer's disease; however, its neurobiological basis in many patients is unknown. In rodents, lesions of the hypothalamic ventrolateral preoptic nucleus cause fragmented sleep. We previously proposed that the intermediate nucleus in the human hypothalamus, which has a similar location and neurotransmitter profile, is the homologue of the ventrolateral preoptic nucleus, but physiological data in humans were lacking. We hypothesized that if the intermediate nucleus is important for human sleep, then intermediate nucleus cell loss may contribute to fragmentation and loss of sleep in ageing and Alzheimer's disease. We studied 45 older adults (mean age at death 89.2 years; 71% female; 12 with Alzheimer's disease) from the Rush Memory and Aging Project, a community-based study of ageing and dementia, who had at least 1 week of wrist actigraphy proximate to death. Upon death a median of 15.5 months later, we used immunohistochemistry and stereology to quantify the number of galanin-immunoreactive intermediate nucleus neurons in each individual, and related this to ante-mortem sleep fragmentation. Individuals with Alzheimer's disease had fewer galaninergic intermediate nucleus neurons than those without (estimate -2872, standard error = 829, P = 0.001). Individuals with more galanin-immunoreactive intermediate nucleus neurons had less fragmented sleep, after adjusting for age and sex, and this association was strongest in those for whom the lag between actigraphy and death was <1 year (estimate -0.0013, standard error = 0.0005, P = 0.023). This association did not differ between individuals with and without Alzheimer's disease, and similar associations were not seen for two other cell populations near the intermediate nucleus. These data are consistent with the intermediate nucleus being the human homologue of the ventrolateral preoptic nucleus. Moreover, they demonstrate that a paucity of galanin-immunoreactive intermediate nucleus neurons is accompanied by sleep fragmentation in older adults with and without Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Neuronas/patología , Área Preóptica/patología , Trastornos del Sueño-Vigilia/patología , Sueño/fisiología , Actigrafía , Anciano de 80 o más Años , Envejecimiento/fisiología , Enfermedad de Alzheimer/fisiopatología , Recuento de Células , Estudios de Cohortes , Interpretación Estadística de Datos , Femenino , Galanina/metabolismo , Humanos , Inmunohistoquímica , Masculino , Área Preóptica/fisiopatología , Descanso/fisiología , Privación de Sueño/patología , Privación de Sueño/fisiopatología , Trastornos del Sueño-Vigilia/fisiopatología , Núcleo Supraquiasmático/crecimiento & desarrollo , Núcleo Supraquiasmático/patología
12.
Adv Gerontol ; 27(1): 81-6, 2014.
Artículo en Ruso | MEDLINE | ID: mdl-25051762

RESUMEN

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.


Asunto(s)
Astrocitos , Hipotálamo , Kisspeptinas , Melatonina , Neuronas , Factores de Edad , Animales , Astrocitos/metabolismo , Astrocitos/patología , Biotransformación , Senescencia Celular/fisiología , Vías de Administración de Medicamentos , Hipotálamo/metabolismo , Hipotálamo/patología , Kisspeptinas/agonistas , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/metabolismo , Kisspeptinas/farmacología , Masculino , Melatonina/metabolismo , Melatonina/farmacología , Neuronas/metabolismo , Neuronas/patología , Área Preóptica/metabolismo , Área Preóptica/patología , Ratas , Ratas Wistar , Resultado del Tratamiento
13.
Mol Cell Endocrinol ; 382(1): 107-119, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24064031

RESUMEN

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.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Inflamación/etiología , Síndrome Metabólico/complicaciones , Neuronas/patología , Área Preóptica/metabolismo , Área Preóptica/patología , Animales , Anticuerpos Monoclonales/farmacología , Biomarcadores/metabolismo , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacología , Dieta Alta en Grasa , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hormona Liberadora de Gonadotropina/sangre , Hormona Liberadora de Gonadotropina/genética , Inmunohistoquímica , Inflamación/sangre , Infliximab , Masculino , Síndrome Metabólico/sangre , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Área Preóptica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Receptores Acoplados a Proteínas G/metabolismo
14.
Horm Behav ; 65(3): 254-63, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24333848

RESUMEN

Behaviors associated with breeding are seasonally modulated in a variety of species. These changes in behavior are mediated by sex steroids, levels of which likewise vary with season. The effects of androgens on behaviors associated with breeding may in turn be partly mediated by the nonapeptides vasopressin (VP) and oxytocin (OT) in mammals, and vasotocin (VT) in birds. The effects of testosterone (T) on production of these neuropeptides have been well-studied; however, the regulation of VT receptors by T is not well understood. In this study, we investigated steroid-dependent regulation of VT receptor (VTR) mRNA in a seasonally breeding songbird, the white-throated sparrow (Zonotrichia albicollis). We focused on VTR subtypes that have been most strongly implicated in social behavior: V1a and oxytocin-like receptor (OTR). Using in situ hybridization, we show that T-treatment of non-breeding males altered V1a and OTR mRNA expression in several regions associated with seasonal reproductive behaviors. For example, T-treatment increased V1a mRNA expression in the medial preoptic area, bed nucleus of the stria terminalis, and ventromedial hypothalamus. T-treatment also affected both V1a and OTR mRNA expression in nuclei of the song system; some of these effects depended on the presence or absence of a chromosomal rearrangement that affects singing behavior, plasma T, and VT immunolabeling in this species. Overall, our results strengthen evidence that VT helps mediate the behavioral effects of T in songbirds, and suggest that the chromosomal rearrangement in this species may affect the sensitivity of the VT system to seasonal changes in T.


Asunto(s)
Área Preóptica/metabolismo , Receptores de Vasopresinas/metabolismo , Núcleos Septales/metabolismo , Gorriones/fisiología , Testosterona/fisiología , Núcleo Hipotalámico Ventromedial/metabolismo , Vocalización Animal/fisiología , Animales , Cruzamiento , Masculino , Área Preóptica/patología , ARN Mensajero/metabolismo , Receptores de Oxitocina/metabolismo , Estaciones del Año , Núcleos Septales/patología , Conducta Sexual Animal/fisiología , Testosterona/sangre , Núcleo Hipotalámico Ventromedial/patología
15.
Neuroscience ; 248: 427-35, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-23806712

RESUMEN

Neuronal losses have been shown to occur in the brainstem following a neonatal hypoxic-ischaemic (HI) insult. In particular serotonergic neurons, situated in the dorsal raphé nuclei, appear to be vulnerable to HI injury. Nonetheless the mechanisms contributing to losses of serotonergic neurons in the brainstem remain to be elucidated. One possible mechanism is that disruption of neural projections from damaged forebrain areas to dorsal raphé nuclei may play a role in the demise of serotonergic neurons. To test this, postnatal day 3 (P3) rat pups underwent unilateral common carotid artery ligation followed by hypoxia (6% O2 for 30 min). On P38 a retrograde tracer, fluorescent-coupled choleratoxin b, was deposited in the dorsal raphé dorsal (DR dorsal) nucleus or the dorsal raphé ventral (DR ventral) nucleus. Compared to control animals, P3 HI animals had significant losses of retrogradely labelled neurons in the medial prefrontal cortex, preoptic area and lateral habenula after tracer deposit in the DR dorsal nucleus. On the other hand, after tracer deposit in the DR ventral nucleus, we found significant reductions in numbers of retrogradely labelled neurons in the hypothalamus, preoptic area and medial amygdala in P3 HI animals compared to controls. Since losses of descending inputs are associated with decreases in serotonergic neurons in the brainstem raphé nuclei, we propose that disruption of certain descending neural inputs from the forebrain to the DR dorsal and the DR ventral nuclei may contribute to losses of serotonergic neurons after P3 HI. It is important to delineate the phenotypes of different neuronal networks affected by neonatal HI, and the mechanisms underpinning this damage, so that interventions can be devised to target and protect axons from the harmful effects of neonatal HI.


Asunto(s)
Muerte Celular , Núcleo Dorsal del Rafe/patología , Vías Eferentes/patología , Hipoxia-Isquemia Encefálica/patología , Prosencéfalo/patología , Neuronas Serotoninérgicas/patología , Animales , Animales Recién Nacidos , Hipotálamo/patología , Trazadores del Tracto Neuronal/química , Corteza Prefrontal/patología , Área Preóptica/patología , Ratas
16.
J Cereb Blood Flow Metab ; 33(2): 183-90, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23093068

RESUMEN

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.


Asunto(s)
5'-Nucleotidasa/metabolismo , Adenosina Monofosfato/metabolismo , Regulación de la Temperatura Corporal , Fiebre/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Área Preóptica/metabolismo , Receptor de Adenosina A1/metabolismo , Adenosina Monofosfato/farmacología , Inhibidores de Captación Adrenérgica/efectos adversos , Inhibidores de Captación Adrenérgica/farmacología , Animales , Dinoprostona/efectos adversos , Dinoprostona/farmacología , Fiebre/inducido químicamente , Fiebre/tratamiento farmacológico , Humanos , Hipotermia Inducida , Masculino , Ratones , N-Metil-3,4-metilenodioxianfetamina/efectos adversos , N-Metil-3,4-metilenodioxianfetamina/farmacología , Neuronas/metabolismo , Oxitócicos/efectos adversos , Oxitócicos/farmacología , Área Preóptica/patología , Mal Uso de Medicamentos de Venta con Receta , Ratas , Ratas Wistar
17.
Eur Neurol ; 67(5): 268-71, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22472481

RESUMEN

BACKGROUND: Hypothermia is a rare condition in patients with multiple sclerosis (MS). Only 17 patients have been reported so far. CASE REPORT: We report 2 patients with a long history of MS who presented with severe acute hypothermia associated with thrombocytopenia and hepatic cytolysis. Both patients presented with an aggravation of their neurological status, psychomotor slowing and confusion. There was no clinical or biological sign of infection. Magnetic resonance imaging (1.5 T) revealed bilateral preoptic T(2)-weighted lesions for 1 patient. The neurological status of one patient was worse after the episode of hypothermia than before, which suggests that hypothermia could be a factor of poor prognosis concerning disease progression.


Asunto(s)
Hipotermia/complicaciones , Esclerosis Múltiple/complicaciones , Área Preóptica/patología , Adulto , Humanos , Hipotermia/diagnóstico , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/diagnóstico
18.
Neurotoxicol Teratol ; 34(3): 331-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22507915

RESUMEN

Perinatal treatment with relatively high doses of bisphenol A (BPA) appears to have little effect on volume of the rodent sexually dimorphic nucleus of the preoptic area (SDN-POA). However, doses more relevant to human exposures have not been examined. Here, effects of pre- and post-natal treatment with low BPA doses on SDN-POA volume of postnatal day (PND) 21 Sprague-Dawley rats were evaluated. Pregnant rats were orally gavaged with vehicle, 2.5 or 25.0 µg/kg BPA, or 5.0 or 10.0 µg/kg ethinyl estradiol (EE2) on gestational days 6-21. Beginning on the day after birth, offspring were orally treated with the same dose their dam had received. On PND 21, offspring (n=10-15/sex/group; 1/sex/litter) were perfused and volume evaluation was conducted blind to treatment. SDN-POA outline was delineated using calbindin D28K immunoreactivity. Pairwise comparisons of the significant treatment by sex interaction indicated that neither BPA dose affected female volume. However, females treated with 5.0 or 10.0 µg/kg EE2 exhibited volumes that were larger than same-sex controls, respectively (p<0.001). Males treated with either BPA dose or 10.0 µg/kg/day EE2 had larger volumes than same-sex controls (p<0.006). These data indicate that BPA can have sex-specific effects on SDN-POA volume and that these effects manifest as larger volumes in males. Sensitivity of the methodology as well as the treatment paradigm was confirmed by the expected EE2-induced increase in female volume. These treatment effects might lead to organizational changes within sexually dimorphic neuroendocrine pathways which, if persistent, could theoretically alter adult reproductive physiology and socio-sexual behavior in rats.


Asunto(s)
Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Área Preóptica/efectos de los fármacos , Caracteres Sexuales , Administración Oral , Animales , Animales Recién Nacidos , Compuestos de Bencidrilo , Relación Dosis-Respuesta a Droga , Femenino , Edad Gestacional , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Área Preóptica/embriología , Área Preóptica/crecimiento & desarrollo , Área Preóptica/patología , Ratas , Ratas Sprague-Dawley
19.
Dev Psychobiol ; 54(1): 16-27, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21656762

RESUMEN

Prenatal stress disrupts normal sexual differentiation and behavior with concomitant alterations in brain development; however, its effects on the cytoarchitecture of neurons in the sexually dimorphic medial preoptic area (mPOA) of the hypothalamus is not known. Morphometric analysis of the mPOA of adult rats showed sex differences as neurons from control females had significantly greater numbers of basal dendritic branches and cumulative basal dendritic length as compared to control male neurons. Prenatal stress significantly altered these sexual dimorphisms, as prenatally stressed (P-S) males had increased measures of cell body area, perimeter, cumulative basal dendritic length, and branch point numbers as compared to control males. Prenatal stress also altered the cytoarchitecture in the female mPOA neurons as P-S female neurons had significantly greater measures for primary dendritic branch number and a trend towards significance for several additional measures as compared to control females. Therefore, there are significant effects of both sex and prenatal stress on neuronal architecture in the mPOA that may help to explain the well-documented alterations in reproductive behaviors observed in P-S animals.


Asunto(s)
Neuronas/patología , Efectos Tardíos de la Exposición Prenatal/patología , Área Preóptica/crecimiento & desarrollo , Estrés Fisiológico/fisiología , Animales , Forma de la Célula/fisiología , Dendritas/patología , Femenino , Masculino , Embarazo , Área Preóptica/patología , Ratas , Ratas Sprague-Dawley , Tinción con Nitrato de Plata
20.
Exp Neurol ; 232(2): 176-84, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21906594

RESUMEN

The ventromedial nucleus of the hypothalamus (VMH) is a key cell group in the medial-basal hypothalamus that participates in the regulation of energy balance. Previous studies have shown that the cellular organization of the VMH is altered in mice with a disruption of the steroidogenic factor-1 (NR5a1) gene (SF-1 KO mice). The present study examined orexigenic/anorexigenic peptides (neuropeptide Y (NPY), agouti-related peptide (AgRP) and cocaine- and amphetamine-regulated transcript (CART)) and neural connections to and from the VMH in SF1 KO mice. NeuroVue tracing and Golgi staining were used to evaluate connections between the preoptic area (POA) and VMH and the orientation of dendrites in the VMH, respectively. Results of this study reveal changes in the cytoarchitecture of the region of the VMH with respect to the distribution of immunoreactive NPY, AgRP and CART. In WT mice projections from the POA normally surround the VMH while in SF-1 KO mice, projections from the POA stream through the region that would otherwise be VMH. Golgi impregnation of the region revealed fewer dendrites with ventrolateral orientations and in general, more variable dendritic orientations in SF-1 KO mice providing additional evidence that the connectivity of cells in the region is likely altered due to the cellular rearrangements consequent to disruption of the NR5a1 gene. In conclusion, this study greatly extends the data showing that the morphology of the regions containing the VMH is disrupted in SF-1 KO mice and suggests that changes in the location of cells or fibers containing NPY, AgRP and CART may, in part, account for changes in body weight homeostasis in these mice.


Asunto(s)
Dendritas/patología , Metabolismo Energético/fisiología , Neuronas/patología , Factor Esteroidogénico 1/genética , Núcleo Hipotalámico Ventromedial/patología , Núcleo Hipotalámico Ventromedial/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Peso Corporal/fisiología , Dendritas/metabolismo , Homeostasis/fisiología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Área Preóptica/metabolismo , Área Preóptica/patología , Área Preóptica/fisiología , Factor Esteroidogénico 1/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA