Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Endocrinology ; 165(7)2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38728240

RESUMO

GH acts in numerous organs expressing the GH receptor (GHR), including the brain. However, the mechanisms behind the brain's permeability to GH and how this hormone accesses different brain regions remain unclear. It is well-known that an acute GH administration induces phosphorylation of the signal transducer and activator of transcription 5 (pSTAT5) in the mouse brain. Thus, the pattern of pSTAT5 immunoreactive cells was analyzed at different time points after IP or intracerebroventricular GH injections. After a systemic GH injection, the first cells expressing pSTAT5 were those near circumventricular organs, such as arcuate nucleus neurons adjacent to the median eminence. Both systemic and central GH injections induced a medial-to-lateral pattern of pSTAT5 immunoreactivity over time because GH-responsive cells were initially observed in periventricular areas and were progressively detected in lateral brain structures. Very few choroid plexus cells exhibited GH-induced pSTAT5. Additionally, Ghr mRNA was poorly expressed in the mouse choroid plexus. In contrast, some tanycytes lining the floor of the third ventricle expressed Ghr mRNA and exhibited GH-induced pSTAT5. The transport of radiolabeled GH into the hypothalamus did not differ between wild-type and dwarf Ghr knockout mice, indicating that GH transport into the mouse brain is GHR independent. Also, single-photon emission computed tomography confirmed that radiolabeled GH rapidly reaches the ventral part of the tuberal hypothalamus. In conclusion, our study provides novel and valuable information about the pattern and mechanisms behind GH transport into the mouse brain.


Assuntos
Encéfalo , Hormônio do Crescimento , Receptores da Somatotropina , Fator de Transcrição STAT5 , Animais , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/genética , Encéfalo/metabolismo , Hormônio do Crescimento/metabolismo , Camundongos , Receptores da Somatotropina/metabolismo , Receptores da Somatotropina/genética , Masculino , Camundongos Knockout , Camundongos Endogâmicos C57BL , Fosforilação , Plexo Corióideo/metabolismo , Hipotálamo/metabolismo , Injeções Intraventriculares
2.
BMJ Case Rep ; 17(1)2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38171637

RESUMO

BACKGROUND: Osmotic demyelination syndrome (ODS) with cerebral cortical involvement is a rare complication of severe hyponatremia correction. Careful management of hyponatremia is crucial, particularly in patients with risk factors, such as alcohol use disorder and diabetes insipidus. CASE: A patient in his 40s with a history of alcohol use disorder and central diabetes insipidus developed ODS after a 24 mEq/L osmolar increase during the treatment of hyponatremia. The patient's condition progressed into locked-in syndrome and then improved to spastic tetraparesis after cortical basal ganglia ODS improved. DISCUSSION: The differential diagnosis of cortical demyelination includes laminar cortical necrosis, being the interpretation of Apparent Diffusion Coefficient (ADC) MRI sequence is a useful tool.This case underscores the need to investigate and improve diagnosis and treatment strategies in patients with ODS. It also emphasises the significance of careful hyponatremia correction and frequent monitoring, particularly in patients with known risk factors for ODS.


Assuntos
Alcoolismo , Doenças Desmielinizantes , Diabetes Insípido Neurogênico , Diabetes Mellitus , Hiponatremia , Humanos , Hiponatremia/diagnóstico , Diabetes Insípido Neurogênico/complicações , Diabetes Insípido Neurogênico/diagnóstico , Alcoolismo/complicações , Doenças Desmielinizantes/complicações , Doenças Desmielinizantes/diagnóstico por imagem , Fatores de Risco
3.
Pract Neurol ; 24(1): 66-69, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-37852755

RESUMO

Late-onset Pompe disease manifests predominantly in the proximal lower limbs and may be mistaken for an inflammatory myopathy. A 46-year-old man with acromegaly had an 8-year history of progressive weakness. His myopathy was initially attributed to the acromegaly, but severe progression prompted a muscle biopsy, which suggested an inflammatory myopathy. However, his weakness progressed despite treatment for polymyositis. His muscle ultrasound scan pattern was more suggestive of Pompe disease than polymyositis, and Pompe disease was confirmed by genetic and enzymatic testing. Patients with apparent polymyositis, which persists despite treatment, require reconsideration of the diagnosis, with particular attention to treatable genetic causes.


Assuntos
Acromegalia , Doença de Depósito de Glicogênio Tipo II , Miosite , Polimiosite , Masculino , Humanos , Pessoa de Meia-Idade , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Polimiosite/diagnóstico , Polimiosite/patologia , Erros de Diagnóstico
5.
Adv Neurobiol ; 27: 131-176, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36169815

RESUMO

Rabbit maternal behavior (MB) impacts meat and fur production on the farm, survival of the species in the wild, and pet welfare. Specific characteristics of rabbit MB (i.e., three-step nest building process; single, brief, daily nursing bout) have been used as models for exploring particular themes in neuroscience, like obsessive-compulsive actions, circadian rhythms, and cognition. Particular hormonal combinations regulate nest building by acting on brain regions controlling MB in other mammals. Nonhormonal factors like type of lodging and the doe's social rank influence nursing and milk production. The concurrency of pregnancy and lactation, the display of nonselective nursing, and the rapid growth of altricial young - despite a minimal effort of maternal care - have prompted the study of mother-young affiliation, neurodevelopment, and weaning. Neurohormonal mechanisms, common to other mammals, plus additional strategies (perhaps unique to rabbits) allow the efficient, adaptive display of MB in multiple settings.


Assuntos
Comportamento Materno , Neuroendocrinologia , Animais , Ritmo Circadiano/fisiologia , Feminino , Humanos , Lactação/fisiologia , Mamíferos , Comportamento Materno/fisiologia , Gravidez , Coelhos
6.
Endocrinology ; 163(11)2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36099517

RESUMO

Growth hormone (GH) secretion is controlled by short and long negative feedback loops. In this regard, both GH (short-loop feedback) and insulin-like growth factor 1 (IGF-1; long-loop feedback) can target somatotropic cells of the pituitary gland and neuroendocrine hypothalamic neurons to regulate the GH/IGF-1 axis. GH-releasing hormone (GHRH)-expressing neurons play a fundamental role in stimulating pituitary GH secretion. However, it is currently unknown whether IGF-1 action on GHRH-expressing cells is required for the control of the GH/IGF-1/growth axis. In the present study, we investigated the phenotype of male and female mice carrying ablation of IGF-1 receptor (IGF1R) exclusively in GHRH cells. After weaning, both male and female GHRHΔIGF1R mice exhibited increases in body weight, lean body mass, linear growth, and length of long bones (tibia, femur, humerus, and radius). In contrast, the percentage of body fat was similar between control and GHRHΔIGF1R mice. The higher body growth of GHRHΔIGF1R mice can be explained by increases in mean GH levels, GH pulse amplitude, and pulse frequency, calculated from 36 blood samples collected from each animal at 10-minute intervals. GHRHΔIGF1R mice also showed increased hypothalamic Ghrh mRNA levels, pituitary Gh mRNA expression, hepatic Igf1 expression, and serum IGF-1 levels compared with control animals. Furthermore, GHRHΔIGF1R mice displayed significant alterations in the sexually dimorphic hepatic gene expression profile, with a prevailing feminization in most genes analyzed. In conclusion, our findings indicate that GHRH neurons represent a key and necessary site for the long-loop negative feedback that controls the GH/IGF-1 axis and body growth.


Assuntos
Hormônio do Crescimento , Fator de Crescimento Insulin-Like I , Animais , Feminino , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/metabolismo
7.
Endocrinology ; 163(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35803590

RESUMO

Growth hormone (GH) acts in several hypothalamic neuronal populations to modulate metabolism and the autoregulation of GH secretion via negative-feedback loops. However, few studies have investigated whether GH receptor (GHR) expression in specific neuronal populations is required for the homeostatic control of GH secretion and energy homeostasis. In the present study, we investigated the consequences of the specific GHR ablation in GABAergic (VGAT-expressing) or glutamatergic (VGLUT2-expressing) cells. GHR ablation in GABAergic neurons led to increased GH secretion, lean mass, and body growth in male and female mice. VGAT-specific GHR knockout (KO) male mice also showed increased serum insulin-like growth factor-1, hypothalamic Ghrh, and hepatic Igf1 messenger RNA levels. In contrast, normal GH secretion, but reduced lean body mass, was observed in mice carrying GHR ablation in glutamatergic neurons. GHR ablation in GABAergic cells increased weight loss and led to decreased blood glucose levels during food restriction, whereas VGLUT2-specific GHR KO mice showed blunted feeding response to 2-deoxy-D-glucose both in males and females, and increased relative food intake, oxygen consumption, and serum leptin levels in male mice. Of note, VGLUT2-cre female mice, independently of GHR ablation, exhibited a previously unreported phenotype of mild reduction in body weight without further metabolic alterations. The autoregulation of GH secretion via negative-feedback loops requires GHR expression in GABAergic cells. Furthermore, GHR ablation in GABAergic and glutamatergic neuronal populations leads to distinct metabolic alterations. These findings contribute to the understanding of the neuronal populations responsible for mediating the neuroendocrine and metabolic effects of GH.


Assuntos
Neurônios GABAérgicos , Receptores da Somatotropina , Animais , Feminino , Hormônio do Crescimento/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptores para Leptina/metabolismo , Receptores da Somatotropina/genética , Receptores da Somatotropina/metabolismo
8.
Poult Sci ; 101(8): 101919, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35704954

RESUMO

There is increasing societal concern regarding the negative impact of intensive poultry production on animal welfare, human health, and on the environment. This is leading to the inclusion of animal welfare as an imperative aspect for sustainable production. Certain environmental factors may challenge domesticated birds, resulting in poor health and welfare status. Resilience is the capacity to rapidly return to prechallenge status after coping with environmental stressors, thus resilient individuals have better chances to maintain good health and welfare. Immune-neuroendocrine system, thoroughly characterized in the domestic bird species, is the physiological scaffold for stress coping and health maintenance, influencing resilience and linking animal welfare status to these vital responses. Modern domestic bird lines have undergone specific genetic selective pressures for fast-growing, or high egg-production, leading to a diversity of birds that differ in their coping capacities and resilience. Deepening the knowledge on pro/anti-inflammatory milieus, humoral/cell-mediated immune responses, hormonal regulations, intestinal microbial communities and mediators that define particular immune and neuroendocrine configurations will shed light on coping strategies at the individual and population level. The understanding of the profiles leading to differential coping and resilience potential will be highly relevant for improving bird health and welfare in a wider range of challenging scenarios and, therefore, crucial to scientifically tackle long term sustainability.


Assuntos
Galinhas , Aves Domésticas , Adaptação Psicológica , Bem-Estar do Animal , Animais , Humanos , Sistemas Neurossecretores , Aves Domésticas/fisiologia
9.
Endocrinology ; 163(5)2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35395079

RESUMO

Hypophysiotropic somatostatin (SST) neurons in the periventricular hypothalamic area express growth hormone (GH) receptor (GHR) and are frequently considered as the key neuronal population that mediates the negative feedback loop controlling the hypothalamic-GH axis. Additionally, insulin-like growth factor-1 (IGF-1) may also act at the hypothalamic level to control pituitary GH secretion via long-loop negative feedback. However, to the best of our knowledge, no study so far has tested whether GHR or IGF-1 receptor (IGF1R) signaling specifically in SST neurons is required for the homeostatic control of GH secretion. Here we show that GHR ablation in SST neurons did not impact the negative feedback mechanisms that control pulsatile GH secretion or body growth in male and female mice. The sex difference in hepatic gene expression profile was only mildly affected by GHR ablation in SST neurons. Similarly, IGF1R ablation in SST neurons did not affect pulsatile GH secretion, body growth, or hepatic gene expression. In contrast, simultaneous ablation of both GHR and IGF1R in SST-expressing cells increased mean GH levels and pulse amplitude in male and female mice, and partially disrupted the sex differences in hepatic gene expression. Despite the increased GH secretion in double knockout mice, no alterations in body growth and serum or liver IGF-1 levels were observed. In summary, GHR and IGF1R signaling in SST neurons play a redundant role in the control of GH secretion. Furthermore, our results reveal the importance of GH/IGF-1 negative feedback mechanisms on SST neurons for the establishment of sex differences in hepatic gene expression profile.


Assuntos
Hormônio do Crescimento , Hormônio do Crescimento Humano , Animais , Feminino , Hormônio do Crescimento/metabolismo , Hormônio do Crescimento Humano/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptores da Somatotropina/genética , Receptores da Somatotropina/metabolismo , Somatostatina/metabolismo
10.
Neurosci Lett ; 770: 136402, 2022 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-34929316

RESUMO

Growth hormone (GH) receptor (GHR) signaling induces the phosphorylation of the signal transducer and activator of transcription 5 (pSTAT5) in the cells of several tissues including in the hypothalamus. During pregnancy, several STAT5-recruiting hormones (e.g., prolactin, GH and placental lactogens) are highly secreted. However, the precise contribution of GHR signaling to the surge of pSTAT5 immunoreactive neurons that occurs in the hypothalamus of pregnant mice is currently unknown. Thus, the objective of the present study was to determine whether GHR expression in neurons is required for inducing pSTAT5 expression in several hypothalamic nuclei during pregnancy. Initially, we demonstrated that late pregnant C57BL/6 mice (gestational day 14 to 18) exhibited increased pulsatile GH secretion compared to virgin females. Next, we confirmed that neuron-specific GHR ablation robustly reduces hypothalamic Ghr mRNA levels and prevents GH-induced pSTAT5 in the arcuate, paraventricular and ventromedial hypothalamic nuclei. Subsequently, the number of pSTAT5 immunoreactive cells was determined in the hypothalamus of late pregnant mice. Although neuron-specific GHR ablation did not affect the number of pSTAT5 immunoreactive cells in the paraventricular nucleus of the hypothalamus, reduced pSTAT5 expression was observed in the arcuate and ventromedial nuclei of pregnant neuron-specific GHR knockouts, compared to control pregnant mice. In summary, a subset of hypothalamic neurons requires GHR signaling to express pSTAT5 during pregnancy. These findings contribute to the understanding of the endocrine factors that affect the activation of transcription factors in the brain during pregnancy.


Assuntos
Proteínas de Transporte/metabolismo , Hipotálamo/metabolismo , Prenhez/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Proteínas de Transporte/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Gravidez
11.
Int J Mol Sci ; 22(18)2021 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-34576072

RESUMO

Corticotropin-releasing hormone (CRH) cells are the dominant neuronal population responsive to the growth hormone (GH) in the paraventricular nucleus of the hypothalamus (PVH). However, the physiological importance of GH receptor (GHR) signaling in CRH neurons is currently unknown. Thus, the main objective of the present study was to investigate the consequences of GHR ablation in CRH-expressing cells of male and female mice. GHR ablation in CRH cells did not cause significant changes in body weight, body composition, food intake, substrate oxidation, locomotor activity, glucose tolerance, insulin sensitivity, counterregulatory response to 2-deoxy-D-glucose and ghrelin-induced food intake. However, reduced energy expenditure was observed in female mice carrying GHR ablation in CRH cells. The absence of GHR in CRH cells did not affect anxiety, circadian glucocorticoid levels or restraint-stress-induced corticosterone secretion and activation of PVH neurons in both male and female mice. In summary, GHR ablation, specifically in CRH-expressing neurons, does not lead to major alterations in metabolism, hypothalamic-pituitary-adrenal axis, acute stress response or anxiety in mice. Considering the previous studies showing that central GHR signaling regulates homeostasis in situations of metabolic stress, future studies are still necessary to identify the potential physiological importance of GH action on CRH neurons.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Neurônios/metabolismo , Receptores da Somatotropina/metabolismo , Animais , Ansiedade/metabolismo , Ritmo Circadiano/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Feminino , Grelina/farmacologia , Glucose/metabolismo , Hormônio do Crescimento/farmacologia , Homeostase/efeitos dos fármacos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Estresse Fisiológico/efeitos dos fármacos
12.
J Comp Neurol ; 529(6): 1228-1239, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32844436

RESUMO

Multiple neuroendocrine, autonomic and behavioral responses are regulated by the paraventricular nucleus of the hypothalamus (PVH). Previous studies have shown that PVH neurons express the growth hormone (GH) receptor (GHR), although the role of GH signaling on PVH neurons is still unknown. Given the great heterogeneity of cell types located in the PVH, we performed a detailed analysis of the neurochemical identity of GH-responsive cells to understand the possible physiological importance of GH action on PVH neurons. GH-responsive cells were detected via the phosphorylated form of the signal transducer and activator of transcription-5 (pSTAT5) in adult male mice that received an intraperitoneal GH injection. Approximately 51% of GH-responsive cells in the PVH co-localized with the vesicular glutamate transporter 2. Rare co-localizations between pSTAT5 and vesicular GABA transporter or vasopressin were observed, whereas approximately 20% and 38% of oxytocin and tyrosine hydroxylase (TH) cells, respectively, were responsive to GH in the PVH. Approximately 55%, 35% and 63% of somatostatin, thyrotropin-releasing hormone (TRH) and corticotropin-releasing hormone (CRH) neurons expressed GH-induced pSTAT5, respectively. Additionally, 8%, 49% and 75% of neuroendocrine TH, TRH and CRH neurons, and 67%, 32% and 74% of nonneuroendocrine TH, TRH and CRH neurons were responsive to GH in the PVH of Fluoro-Gold-injected mice. Our findings suggest that GH action on PVH neurons is involved in the regulation of the thyroid, somatotropic and adrenal endocrine axes, possibly influencing homeostatic and stress responses.


Assuntos
Hormônio do Crescimento/metabolismo , Núcleo Hipotalâmico Paraventricular/química , Núcleo Hipotalâmico Paraventricular/metabolismo , Fenótipo , Receptores da Somatotropina/metabolismo , Animais , Hormônio do Crescimento/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Paraventricular/citologia , Receptores da Somatotropina/análise
13.
Rev. argent. dermatol ; Rev. argent. dermatol;101(4): 41-50, dic. 2020. graf
Artigo em Espanhol | LILACS-Express | LILACS | ID: biblio-1251055

RESUMO

Resumen El folículo piloso es una estructura compleja que presenta diversas características morfológicas macroscópicas, microscópicas e inmunológicas especiales que permiten el adecuado funcionamiento de la misma, en algunasenfermedades estos mecanismos de regulación inmunológica se ven alteradose incluso exacerbados por factores como el estrés emocional. El objetivo de esta revisión es conocer los mecanismos inmunobiológicos específicos del folículo pilosoanalizando el papel que juegan diversos factores como la pérdida delinmunoprivilegio y el estrés emocional en el desarrollo de la alopecia areata.


Abstract The hair follicle is a complex structure that presents diverse morphologicaland immunological characteristics that allow the proper functioning of the unit.In some diseases as alopecia areata these mechanisms of immune regulation are disrupted by external factorssuch as emotional stress preventing the growth of the hair shaft. The objective of this review is to recognize the specific immunobiological mechanisms of the hair follicle, analyzing the role played by the loss of immunoprivilege and emotional stress in the development of alopecia areata.

14.
Artigo em Inglês | MEDLINE | ID: mdl-32849267

RESUMO

Lactation is a complex physiological process, depending on orchestrated central and peripheral events, including substantial brain plasticity. Among these events is a novel expression of pro-melanin-concentrating hormone (Pmch) mRNA in the rodent hypothalamus, such as the ventral part of the medial preoptic area (vmMPOA). This expression reaches its highest levels around postpartum day 19 (PPD19), when dams transition from lactation to the weaning period. The appearance of this lactation-related Pmch expression occurs simultaneously with the presence of one of the Pmch products, melanin-concentrating hormone (MCH), in the serum. Given the relevance of the MPOA to maternal physiology and the contemporaneity between Pmch expression in this structure and the weaning period, we hypothesized that MCH has a role in the termination of lactation, acting as a mediator between central and peripheral changes. To test this, we investigated the presence of the MCH receptor 1 (MCHR1) and its gene expression in the mammary gland of female rats in different stages of the reproductive cycle. To that end, in situ hybridization, RT-PCR, RT-qPCR, nucleotide sequencing, immunohistochemistry, and Western blotting were employed. Although Mchr1 expression was detected in the epidermis and dermis of both diestrus and lactating rats, parenchymal expression was exclusively found in the functional mammary gland of lactating rats. The expression of Mchr1 mRNA oscillated through the lactation period and reached its maximum in PPD19 dams. Presence of MCHR1 was confirmed with immunohistochemistry with preferential location of MCHR1 immunoreactive cells in the alveolar secretory cells. As was the case for gene expression, the MCHR1 protein levels were significantly higher in PPD19 than in other groups. Our data demonstrate the presence of an anatomical basis for the participation of MCH peptidergic system on the control of lactation through the mammary gland, suggesting that MCH could modulate a prolactation action in early postpartum days and the opposite role at the end of the lactation.


Assuntos
Lactação , Glândulas Mamárias Animais/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Hormônio Hipofisário/genética , Receptores do Hormônio Hipofisário/metabolismo , Animais , Feminino , Imuno-Histoquímica , Masculino , Glândulas Mamárias Animais/crescimento & desenvolvimento , Ratos , Ratos Long-Evans
15.
J Pineal Res ; 69(3): e12675, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32598502

RESUMO

Studying communities at different stages of urbanisation and industrialisation can teach us how timing and intensity of light affect the circadian clock under real-life conditions. We have previously described a strong tendency towards morningness in the Baependi Heart Study, located in a small rural town in Brazil. Here, we tested the hypothesis that this morningness tendency is associated with early circadian phase based on objective measurements (as determined by dim light melatonin onset, DLMO, and activity) and light exposure. We also analysed how well the previously collected chronotype questionnaire data were able to predict these DLMO values. The average DLMO observed in 73 participants (40 female) was 20:03 ± 01:21, SD, with an earlier average onset in men (19:38 ± 01:16) than in women (20:24 ± 01:21; P ≤ .01). However, men presented larger phase angle between DLMO and sleep onset time as measured by actigraphy (4.11 hours vs 3.16 hours; P ≤ .01). Correlational analysis indicated associations between light exposure, activity rhythms and DLMO, such that early DLMO was observed in participants with higher exposure to light, higher activity and earlier light exposure. The strongest significant predictor of DLMO was morningness-eveningness questionnaire (MEQ) (beta=-0.35, P ≤ .05), followed by age (beta = -0.47, P ≤ .01). Sex, light exposure and variables derived from the Munich chronotype questionnaire were not significant predictors. Our observations demonstrate that both early sleep patterns and earlier circadian phase have been retained in this small rural town in spite of availability of electrification, in contrast to metropolitan postindustrial areas.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Melatonina/metabolismo , População Rural , Sono/fisiologia , Inquéritos e Questionários , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
16.
Rev. argent. neurocir ; 34(1): 15-35, mar. 2020. ilus, grafs
Artigo em Espanhol | LILACS, BINACIS | ID: biblio-1151244

RESUMO

Introducción: Los tumores de hipófisis representan aproximadamente un 10% de los tumores intracraneales. Los adenomas hipofisarios son ampliamente la lesión selar más frecuente. Es fundamental que se conformen equipos capaces de tratar holísticamente esta patología en las distintas regiones de nuestro vasto territorio. El propósito del presente trabajo es presentar nuestra experiencia en el manejo quirúrgico de los adenomas hipofisarios en el Noroeste Argentino. Materiales y métodos: Estudio retrospectivo de pacientes operados por adenomas selares, desde enero de 2013 hasta abril de 2019, en la provincia de Tucumán, por el autor Senior. Resultados: Se analizaron 211 pacientes operados de adenomas pituitarios. En el 75% (n=158) las resecciones fueron totales y en el 25% (n=53) fueron resecciones subtotales. De los 88 pacientes con macroadenomas no funcionantes y déficit visual, el 94% (n=83) presentó mejoría visual en el postoperatorio. De los 52 pacientes con acromegalia, un 75% (n=39) presentó remisión bioquímica completa postquirúrgica. De los 26 pacientes con enfermedad de Cushing, un 81% (n=21) presentó remisión bioquímica completa postquirúrgica. De los 21 pacientes con prolactinomas, un 71% (n=15) normalizaron los niveles de prolactina en el postoperatorio. Conclusión: Los resultados en el tratamiento quirúrgico de los adenomas hipofisarios de la presente serie son comparables a los reportados por centros especializados internacionales. Debemos formar equipos capaces de tratar esta patología en las distintas regiones de la Argentina.


Background: Pituitary tumors account for 10% of intracranial tumors. Pituitary adenomas are the most common selar lesion. It is essential that teams be formed capable of treating this pathology holistically in the different regions of our vast territory. The purpose of this article is to present our experience in the surgical management of pituitary adenomas in the Northwest of Argentine. Materials and methods: A retrospective analysis was done studding patients operated from pituitary adenomas, from January 2013 to April 2019, in Tucumán, by the Senior Author. Results: We analyzed 211 patients operated from pituitary adenomas, of which 75% (n=158) had gross total resections and 25% (n=53) had subtotal resections. Of the 88 patients with non-functioning macroadenomas and visual deficit, 94% (n=83) presented visual improvement after surgery. Of the 52 patients with acromegaly, 75% (n=39) presented complete biochemical remission after surgery. Of the 26 patients with Cushing's disease, 81% (n=21) presented complete biochemical remission after surgery. Of the 21 patients with prolactinomas, 71% (n=15) normalized prolactin levels after surgery. Conclusion: Our results in the surgical treatment of pituitary adenomas are comparable to those reported by international specialized centers. We must form teams capable of treating this pathology in the different regions of Argentina


Assuntos
Adenoma , Hipófise , Neoplasias Hipofisárias , Crânio , Neuroendocrinologia
17.
J Endocrinol ; 244(1): 71-82, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31557728

RESUMO

The mTOR/S6Ks signaling is one of the intracellular pathways important for metabolic control, acting both peripherally and centrally. In the hypothalamus, mTOR/S6Ks axis mediates the action of leptin and insulin and can modulate the expression of neuropeptides. We analyzed the role of different S6Ks isoforms in the hypothalamic regulation of metabolism. We observed decreased food intake and decreased expression of agouti-related peptide (AgRP) following intracerebroventricular (icv) injections of adenoviral-mediated overexpression of three different S6Ks isoforms. Moreover, mice overexpressing p70-S6K1 in undefined periventricular hypothalamic neurons presented changes in glucose metabolism, as an increase in gluconeogenesis. To further evaluate the hypothalamic role of a less-studied S6K isoform, p54-S6K2, we used a Cre-LoxP approach to specifically overexpress it in AgRP neurons. Our findings demonstrate the potential participation of S6K2 in AgRP neurons regulating feeding behavior.


Assuntos
Comportamento Alimentar/efeitos dos fármacos , Glucose/metabolismo , Isoformas de Proteínas/farmacologia , Proteínas Quinases S6 Ribossômicas 90-kDa/farmacologia , Proteínas Quinases S6 Ribossômicas/farmacologia , Proteína Relacionada com Agouti/metabolismo , Animais , Ingestão de Alimentos/genética , Hipotálamo/metabolismo , Camundongos , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo
18.
Eur J Neurosci ; 51(1): 429-453, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30408249

RESUMO

Adult rabbits show robust circadian rhythms of: nursing, food and water intake, hard faeces excretion, locomotion, body temperature, blood and intraocular pressure, corticosteroid secretion, and sleep. Control of several circadian rhythms involves a light-entrained circadian clock and a food-entrained oscillator. Nursing periodicity, however, relies on a suckling stimulation threshold. Brain structures regulating this activity include the paraventricular nucleus and preoptic area, as determined by lesions and quantification of cFOS- and PER1 clock gene-immunoreactive proteins. Melatonin synthesis in the rabbit pineal gland shows a diurnal rhythm, with highest values at night and lowest ones during the day. In kits the main zeitgeber is milk intake, which synchronizes locomotor activity, body temperature, and corticosterone secretion. Brain regions involved in these effects include the median preoptic nucleus and several olfactory structures. As models for particular human illnesses rabbits have been valuable for studying glaucoma and cardiovascular disease. Circadian variations in intraocular pressure (main risk factor for glaucoma) have been found, with highest values at night, which depend on sympathetic innervation. Rabbits fed a high fat diet develop cholesterol plaques and high blood pressure, as do humans, and such increased fat intake directly modulates cardiovascular homeostasis and circadian patterns, independently of white adipose tissue accumulation. Rabbits have also been useful to investigate the characteristics of sleep across the day and its modulation by infections, cytokines and other endogenous humoral factors. Rabbit circadian biology warrants deeper investigation of the role of the suprachiasmatic nucleus in regulating most behavioral and physiological rhythms described above.


Assuntos
Relógios Circadianos , Núcleo Supraquiasmático , Animais , Biologia , Proteínas CLOCK , Ritmo Circadiano , Feminino , Masculino , Coelhos
19.
Ann N Y Acad Sci ; 1455(1): 81-97, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31008525

RESUMO

Ghrelin is a peptide mainly produced and secreted by the stomach. Since its discovery, the impact of ghrelin on the regulation of food intake has been the most studied function of this hormone; however, ghrelin affects a wide range of physiological systems, many of which are controlled by the hypothalamic paraventricular nucleus (PVN). Several pathways may mediate the effects of ghrelin on PVN neurons, such as direct or indirect effects mediated by circumventricular organs and/or the arcuate nucleus. The ghrelin receptor is expressed in PVN neurons, and the peripheral or intracerebroventricular administration of ghrelin affects PVN neuronal activity. Intra-PVN application of ghrelin increases food intake and decreases fat oxidation, which chronically contribute to the increased adiposity. Additionally, ghrelin modulates the neuroendocrine axes controlled by the PVN, increasing the release of vasopressin and oxytocin by magnocellular neurons and corticotropin-releasing hormone by neuroendocrine parvocellular neurons, while possibly inhibiting the release of thyrotropin-releasing hormone. Thus, the PVN is an important target for the actions of ghrelin. Our review discusses the mechanisms of ghrelin actions in the PVN, and its potential implications for energy balance, neuroendocrine, and integrative physiological control.


Assuntos
Grelina/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Animais , Metabolismo Energético , Humanos
20.
Front Cell Neurosci ; 12: 275, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30210300

RESUMO

The paraventricular nucleus (PVN) is involved in the control of sympathetic tone and the secretion of hormones, both functions known to be influenced by ghrelin, suggesting direct effect of ghrelin in this nucleus. However, the effects of ghrelin on the excitability of different PVN neuronal populations have not been demonstrated. This study assessed the effects of ghrelin on the activity of PVN neurons, correlating the responses to subpopulations of PVN neurons. We used a 64 multielectrode array to examine the effects of ghrelin administration on extracellular spike frequency in PVN neurons recorded in brain slices obtained from male Sprague-Dawley rats. Bath administration of 10 nM ghrelin increased (29/97, 30%) or decreased (37/97, 38%) spike frequency in PVN neurons. The GABAA and glutamate receptors antagonists abolish the decrease in spike frequency, without changes in the proportion of increases in spike frequency (23/53, 43%) induced by ghrelin. The results indicate a direct effect of ghrelin increasing PVN neurons activity and a synaptic dependent effect decreasing PVN neurons activity. The patch clamp recordings showed similar proportions of PVN neurons influenced by 10 nM ghrelin (33/95, 35% depolarized; 29/95, 30% hyperpolarized). Using electrophysiological fingerprints to identify specific subpopulations of PVN neurons we observed that the majority of pre-autonomic neurons (11/18 -61%) were depolarized by ghrelin, while both neuroendocrine (29% depolarizations, 40% hyperpolarizations), and magnocellular neurons (29% depolarizations, 21% hyperpolarizations) showed mixed responses. Finally, to correlate the electrophysiological response and the neurochemical phenotype of PVN neurons, cell cytoplasm was collected after recordings and RT-PCR performed to assess the presence of mRNA for vasopressin, oxytocin, thyrotropin (TRH) and corticotropin (CRH) releasing hormones. The single-cell RT-PCR showed that most TRH-expressing (4/5) and CRH-expressing (3/4) neurons are hyperpolarized in response to ghrelin. In conclusion, ghrelin either directly increases or indirectly decreases the activity of PVN neurons, this suggests that ghrelin acts on inhibitory PVN neurons that, in turn, decrease the activity of TRH-expressing and CRH-expressing neurons in the PVN.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA