Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 233
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Intervalo de año de publicación
1.
Prog Mol Biol Transl Sci ; 189(1): 1-34, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35595346

RESUMEN

Animal models remain essential to understand the fundamental mechanisms of physiology and pathology. Particularly, the complex and dynamic nature of neuroendocrine cells of the hypothalamus make them difficult to study. The neuroendocrine systems of the hypothalamus are critical for survival and reproduction, and are highly conserved throughout vertebrate evolution. Their roles in controlling body metabolism, growth and body composition, stress, electrolyte balance, and reproduction, have been intensively studied, and have yielded groundbreaking discoveries. Many of these discoveries would not have been feasible without the use of the domestic sheep (Ovis aries). The sheep has been used for decades to study the neuroendocrine systems of the hypothalamus and has become a model for human neuroendocrinology. The aim of this chapter is to review some of the profound biomedical discoveries made possible by the use of sheep. The advantages and limitations of sheep as a neuroendocrine model will be discussed. While no animal model can perfectly recapitulate a human disease or condition, sheep are invaluable for enabling manipulations not possible in human subjects and isolating physiologic variables to garner insight into neuroendocrinology and associated pathologies.


Asunto(s)
Hipotálamo , Neuroendocrinología , Animales , Humanos , Hipotálamo/metabolismo , Sistemas Neurosecretores/metabolismo , Reproducción , Ovinos
2.
J Neuroendocrinol ; 34(6): e13133, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35474620

RESUMEN

Links between the regulation of growth and energy balance are clear; to fuel growth, there must be consumption of energy. Therefore, it is perhaps intuitive that interactions between the hypothalamic - pituitary - growth hormone axis (growth axis) and pathways that drive metabolic processes exist. Overproduction of growth hormone has been associated with diabetes and metabolic disease for decades and the opposing effects of growth hormone and insulin have been studied since early experiments almost a century ago. The relationship between neuroendocrine axes can be complex and the growth axis is no exception, interacting with energy balance in several organ systems, both in the periphery and centrally in hypothalamic nuclei. Much is known about peripheral interactions between growth axis hormones and processes such as glucose homeostasis and adipogenesis. More is still being learned about the molecular actions of growth axis hormones in adipose and other metabolically active tissues, and recent findings are discussed in this perspective. However, less is known about interactions with central energy balance pathways in the hypothalamus. This perspective aims to summarise what is known about these interactions, taking lessons from human studies and animal genetic and seasonal models, and discusses what this may mean in an evolving landscape of personalised medicine.


Asunto(s)
Apetito , Hormona de Crecimiento Humana , Animales , Metabolismo Energético/fisiología , Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/metabolismo , Hipotálamo/metabolismo , Sistemas Neurosecretores/metabolismo
3.
Int J Mol Sci ; 23(3)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35163554

RESUMEN

Lactation is a physiological state of hyperprolactinemia and associated amenorrhea. Despite the fact that exact mechanisms standing behind the hypothalamus-pituitary-ovarian axis during lactation are still not clear, a general overview of events leading to amenorrhea may be suggested. Suckling remains the most important stimulus maintaining suppressive effect on ovaries after pregnancy. Breastfeeding is accompanied by high levels of prolactin, which remain higher than normal until the frequency and duration of daily suckling decreases and allows normal menstrual function resumption. Hyperprolactinemia induces the suppression of hypothalamic Kiss1 neurons that directly control the pulsatile release of GnRH. Disruption in the pulsatile manner of GnRH secretion results in a strongly decreased frequency of corresponding LH pulses. Inadequate LH secretion and lack of pre-ovulatory surge inhibit the progression of the follicular phase of a menstrual cycle and result in anovulation and amenorrhea. The main consequences of lactational amenorrhea are connected with fertility issues and increased bone turnover. Provided the fulfillment of all the established conditions of its use, the lactational amenorrhea method (LAM) efficiently protects against pregnancy. Because of its accessibility and lack of additional associated costs, LAM might be especially beneficial in low-income, developing countries, where modern contraception is hard to obtain. Breastfeeding alone is not equal to the LAM method, and therefore, it is not enough to successfully protect against conception. That is why LAM promotion should primarily focus on conditions under which its use is safe and effective. More studies on larger study groups should be conducted to determine and confirm the impact of behavioral factors, like suckling parameters, on the LAM efficacy. Lactational bone loss is a physiologic mechanism that enables providing a sufficient amount of calcium to the newborn. Despite the decline in bone mass during breastfeeding, it rebuilds after weaning and is not associated with a postmenopausal decrease in BMD and osteoporosis risk. Therefore, it should be a matter of concern only for lactating women with additional risk factors or with low BMD before pregnancy. The review summarizes the effect that breastfeeding exerts on the hypothalamus-pituitary axis as well as fertility and bone turnover aspects of lactational amenorrhea. We discuss the possibility of the use of lactation as contraception, along with this method's prevalence, efficacy, and influencing factors. We also review the literature on the topic of lactational bone loss: its mechanism, severity, and persistence throughout life.


Asunto(s)
Amenorrea/metabolismo , Remodelación Ósea , Lactancia , Sistemas Neurosecretores/metabolismo , Anticoncepción/métodos , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Prolactina/metabolismo , Regulación hacia Arriba
4.
Brain Behav Immun ; 101: 146-152, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34973397

RESUMEN

BACKGROUND: The study of neural-endocrine-immune system interactions has led to substantial advances in our understanding of neuropsychiatric disorders. Growing evidence reveals the pivotal roles of inflammatory cytokines signalling the brain to produce neurochemical, neuroendocrine, and neuroimmune changes which affect mood and behaviour. Ageing is accompanied by the development of low-grade systemic inflammation which may promote changes in the neural systems predisposing to geriatric depression via the hypothalamic-pituitary-adrenal (HPA) axis. The aim of this study was to investigate the longitudinal associations between baseline values and conditional changes (independent of baseline) in immune-endocrine biomarkers and mental health status in a population-based cohort of older adults. METHODS: Data from 347 subjects (200 men, 147 women) who participated in the Hertfordshire Ageing Study at baseline (1994/5, mean age 67.3 years) and at 9-year follow-up were analysed. Serum samples for analysis of inflammatory and endocrinological measures were collected at baseline and follow-up. At follow-up, depression (Hospital Anxiety and Depression Scale) and mental health (Short Form-36 questionnaire) were assessed. Baseline values and changes in biomarkers in relation to risk of high depression scores (top sex-specific third) and low mental health scores (bottom sex-specific third) were examined using logistic regression. RESULTS: Lower baseline cortisol was related to greater risk of high depression scores; higher baseline cortisol: dehydroepiandrosterone sulphate ratio (men only) and higher baseline C-reactive protein (CRP) (women only) were related to greater risk of poor mental health scores. In addition, greater decline in cortisol was related to increased risk of high depression scores among men. These relationships were robust (p < 0.05) after controlling for sex, age, BMI, smoking, alcohol consumption and number of systems medicated. CONCLUSION: This study provides further evidence of the role of the HPA axis and inflammation in older adults with poor mental health. In addition, the findings highlight sex differences where increased inflammation in women and declines in cortisol in men were linked to poorer mental health. Further research is warranted to confirm these findings. This could lead to the search for potential biomarkers to stratify medications as well as developing novel intervention targets to improve mental health at older age.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Anciano , Envejecimiento , Biomarcadores , Depresión , Femenino , Humanos , Hidrocortisona , Sistema Hipotálamo-Hipofisario/metabolismo , Inflamación/metabolismo , Masculino , Salud Mental , Sistemas Neurosecretores/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo
5.
Int J Mol Sci ; 22(12)2021 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-34201099

RESUMEN

Astrocytes are a type of glial cell anatomically and functionally integrated into the neuronal regulatory circuits for the neuroendocrine control of metabolism. Being functional integral compounds of synapses, astrocytes are actively involved in the physiological regulatory aspects of metabolic control, but also in the pathological processes that link neuronal dysfunction and obesity. Between brain areas, the hypothalamus harbors specialized functional circuits that seem selectively vulnerable to metabolic damage, undergoing early cellular rearrangements which are thought to be at the core of the pathogenesis of diet-induced obesity. Such changes in the hypothalamic brain region consist of a rise in proinflammatory cytokines, the presence of a reactive phenotype in astrocytes and microglia, alterations in the cytoarchitecture and synaptology of hypothalamic circuits, and angiogenesis, a phenomenon that cannot be found elsewhere in the brain. Increasing evidence points to the direct involvement of hypothalamic astrocytes in such early metabolic disturbances, thus moving the study of these glial cells to the forefront of obesity research. Here we provide a comprehensive review of the most relevant findings of molecular and pathophysiological mechanisms by which hypothalamic astrocytes might be involved in the pathogenesis of obesity.


Asunto(s)
Astrocitos/patología , Hipotálamo/patología , Sistemas Neurosecretores/patología , Obesidad/patología , Animales , Astrocitos/metabolismo , Humanos , Hipotálamo/metabolismo , Sistemas Neurosecretores/metabolismo , Obesidad/metabolismo
6.
J Neuroendocrinol ; 33(7): e12975, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33942400

RESUMEN

The Wistar audiogenic rat (WAR) strain is used as an animal model of epilepsy, which when submitted to acute acoustic stimulus presents tonic-clonic seizures, mainly dependent on brainstem (mesencephalic) structures. However, when WARs are exposed to chronic acoustic stimuli (audiogenic kindling-AK), they usually present tonic-clonic seizures, followed by limbic seizures, after recruitment of forebrain structures such as the cortex, hippocampus and amygdala. Although some studies have reported that hypothalamic-hypophysis function is also altered in WAR through modulating vasopressin (AVP) and oxytocin (OXT) secretion, the role of these neuropeptides in epilepsy still is controversial. We analyzed the impact of AK and consequent activation of mesencephalic neurocircuits and the recruitment of forebrain limbic (LiR) sites on the hypothalamic-neurohypophysial system and expression of Avpr1a and Oxtr in these structures. At the end of the AK protocol, nine out of 18 WARs presented LiR. Increases in both plasma vasopressin and oxytocin levels were observed in WAR when compared to Wistar rats. These results were correlated with an increase in the expressions of heteronuclear (hn) and messenger (m) RNA for Oxt in the paraventricular nucleus (PVN) in WARs submitted to AK that presented LiR. In the paraventricular nucleus, the hnAvp and mAvp expressions increased in WARs with and without LiR, respectively. There were no significant differences in Avp and Oxt expression in supraoptic nuclei (SON). Also, there was a reduction in the Avpr1a expression in the central nucleus of the amygdala and frontal lobe in the WAR strain. In the inferior colliculus, Avpr1a expression was lower in WARs after AK, especially those without LiR. Our results indicate that both AK and LiR in WARs lead to changes in the hypothalamic-neurohypophysial system and its receptors, providing a new molecular basis to better understaind epilepsy.


Asunto(s)
Epilepsia Refleja , Hipotálamo/metabolismo , Excitación Neurológica/fisiología , Sistemas Neurosecretores/metabolismo , Neurohipófisis/metabolismo , Estimulación Acústica , Animales , Modelos Animales de Enfermedad , Epilepsia Refleja/genética , Epilepsia Refleja/metabolismo , Epilepsia Refleja/patología , Epilepsia Refleja/fisiopatología , Regulación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Hipotálamo/patología , Hipotálamo/fisiopatología , Excitación Neurológica/patología , Masculino , Sistemas Neurosecretores/patología , Sistemas Neurosecretores/fisiopatología , Oxitocina/sangre , Oxitocina/genética , Oxitocina/metabolismo , Neurohipófisis/patología , Neurohipófisis/fisiopatología , Ratas , Ratas Wistar , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/fisiopatología , Convulsiones/psicología , Vasopresinas/sangre , Vasopresinas/genética , Vasopresinas/metabolismo
7.
Sci Rep ; 11(1): 2335, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33504846

RESUMEN

Long-standing clinical findings report a dramatic surge of vasopressin in umbilical cord blood of the human neonate, but the neural underpinnings and function(s) of this phenomenon remain obscure. We studied neural activation in perinatal mice and rats, and found that birth triggers activation of the suprachiasmatic, supraoptic, and paraventricular nuclei of the hypothalamus. This was seen whether mice were born vaginally or via Cesarean section (C-section), and when birth timing was experimentally manipulated. Neuronal phenotyping showed that the activated neurons were predominantly vasopressinergic, and vasopressin mRNA increased fivefold in the hypothalamus during the 2-3 days before birth. Copeptin, a surrogate marker of vasopressin, was elevated 30-to 50-fold in plasma of perinatal mice, with higher levels after a vaginal than a C-section birth. We also found an acute decrease in plasma osmolality after a vaginal, but not C-section birth, suggesting that the difference in vasopressin release between birth modes is functionally meaningful. When vasopressin was administered centrally to newborns, we found an ~ 50% reduction in neuronal cell death in specific brain areas. Collectively, our results identify a conserved neuroendocrine response to birth that is sensitive to birth mode, and influences peripheral physiology and neurodevelopment.


Asunto(s)
Hipotálamo/metabolismo , Sistemas Neurosecretores/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Vasopresinas/metabolismo , Animales , Biomarcadores/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Osmorregulación/genética , Osmorregulación/fisiología , Vasopresinas/genética
8.
Sci Rep ; 11(1): 1996, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479437

RESUMEN

Female puberty is subject to Polycomb Group (PcG)-dependent transcriptional repression. Kiss1, a puberty-activating gene, is a key target of this silencing mechanism. Using a gain-of-function approach and a systems biology strategy we now show that EED, an essential PcG component, acts in the arcuate nucleus of the hypothalamus to alter the functional organization of a gene network involved in the stimulatory control of puberty. A central node of this network is Kdm6b, which encodes an enzyme that erases the PcG-dependent histone modification H3K27me3. Kiss1 is a first neighbor in the network; genes encoding glutamatergic receptors and potassium channels are second neighbors. By repressing Kdm6b expression, EED increases H3K27me3 abundance at these gene promoters, reducing gene expression throughout a gene network controlling puberty activation. These results indicate that Kdm6b repression is a basic mechanism used by PcG to modulate the biological output of puberty-activating gene networks.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/genética , Kisspeptinas/genética , Complejo Represivo Polycomb 2/genética , Pubertad/genética , Animales , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Neuronas/metabolismo , Sistemas Neurosecretores/crecimiento & desarrollo , Sistemas Neurosecretores/metabolismo , Proteínas del Grupo Polycomb/genética , Regiones Promotoras Genéticas/genética , Pubertad/fisiología , Ratas , Biología de Sistemas
9.
Cell Mol Life Sci ; 78(1): 1-16, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32564094

RESUMEN

Research into the physiological actions of anti-Müllerian hormone (AMH) has rapidly expanded from its classical role in male sexual differentiation to the regulation of ovarian function, routine clinical use in reproductive health and potential use as a biomarker in the diagnosis of polycystic ovary syndrome (PCOS). During the past 10 years, the notion that AMH could act exclusively at gonadal levels has undergone another paradigm shift as several exciting studies reported unforeseen AMH actions throughout the Hypothalamic-Pituitary-Gonadal (HPG) axis. In this review, we will focus on these findings reporting novel AMH actions across the HPG axis and we will discuss their potential impact and significance to better understand human reproductive disorders characterized by either developmental alterations of neuroendocrine circuits regulating fertility and/or alterations of their function in adult life. Finally, we will summarize recent preclinical studies suggesting that elevated levels of AMH may potentially be a contributing factor to the central pathophysiology of PCOS and other reproductive diseases.


Asunto(s)
Hormona Antimülleriana/metabolismo , Gónadas/metabolismo , Hipotálamo/metabolismo , Hipófisis/metabolismo , Femenino , Humanos , Sistemas Neurosecretores/crecimiento & desarrollo , Sistemas Neurosecretores/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Reproducción , Transducción de Señal
10.
Sci Rep ; 10(1): 16947, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046791

RESUMEN

Protein 4.1N, a member of the protein 4.1 family, is highly expressed in the brain. But its function remains to be fully defined. Using 4.1N-/- mice, we explored the function of 4.1N in vivo. We show that 4.1N-/- mice were born at a significantly reduced Mendelian ratio and exhibited high mortality between 3 to 5 weeks of age. Live 4.1N-/- mice were smaller than 4.1N+/+ mice. Notably, while there were no significant differences in organ/body weight ratio for most of the organs, the testis/body and ovary/body ratio were dramatically decreased in 4.1N-/- mice, demonstrating selective effects of 4.1N deficiency on the development of the reproductive systems. Histopathology of the reproductive organs showed atrophy of both testis and ovary. Specifically, in the testis there is a lack of spermatogenesis, lack of leydig cells and lack of mature sperm. Similarly, in the ovary there is a lack of follicular development and lack of corpora lutea formation, as well as lack of secretory changes in the endometrium. Examination of pituitary glands revealed that the secretory granules were significantly decreased in pituitary glands of 4.1N-/- compared to 4.1N+/+. Moreover, while GnRH was expressed in both neuronal cell body and axons in the hypothalamus of 4.1N+/+ mice, it was only expressed in the cell body but not the axons of 4.1N-/- mice. Our findings uncover a novel role for 4.1N in the axis of hypothalamus-pituitary gland-reproductive system.


Asunto(s)
Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/fisiología , Genitales/metabolismo , Genitales/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/fisiología , Neuropéptidos/deficiencia , Neuropéptidos/fisiología , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/patología , Animales , Proteínas del Citoesqueleto/genética , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Proteínas de la Membrana/genética , Ratones Noqueados , Neuropéptidos/genética , Tamaño de los Órganos , Ovario/patología , Hipófisis/metabolismo , Hipófisis/patología , Espermatogénesis/genética , Testículo/patología
11.
Mol Cell Endocrinol ; 518: 110996, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32860862

RESUMEN

The hypothalamic-pituitary-gonadal axis is controlled by gonadotropin-releasing hormone (GnRH) released by the hypothalamus. Disruption of this system leads to impaired reproductive maturation and function, a condition known as hypogonadotropic hypogonadism (HH). Most studies to date have focused on genetic causes of HH that impact neuronal development and function. However, variants may also impact the functioning of non-neuronal cells known as glia. Glial cells make up 50% of brain cells of humans, primates, and rodents. They include radial glial cells, microglia, astrocytes, tanycytes, oligodendrocytes, and oligodendrocyte precursor cells. Many of these cells influence the hypothalamic neuroendocrine system controlling fertility. Indeed, glia regulate GnRH neuronal activity and secretion, acting both at their cell bodies and their nerve endings. Recent work has also made clear that these interactions are an essential aspect of how the HPG axis integrates endocrine, metabolic, and environmental signals to control fertility. Recognition of the clinical importance of interactions between glia and the GnRH network may pave the way for the development of new treatment strategies for dysfunctions of puberty and adult fertility.


Asunto(s)
Células Endocrinas/fisiología , Hipogonadismo/etiología , Animales , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Neuronas/fisiología , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/fisiología , Reproducción/fisiología
12.
Endocrinology ; 161(8)2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32609838

RESUMEN

Disorders of androgen imbalance, such as hyperandrogenism in females or hypoandrogenism in males, increase risk of visceral adiposity, type 2 diabetes, and infertility. Androgens act upon androgen receptors (AR) which are expressed in many tissues. In the brain, AR are abundant in hypothalamic nuclei involved in regulation of reproduction and energy homeostasis, yet the role of androgens acting via AR in specific neuronal populations has not been fully elucidated. Leptin receptor (LepRb)-expressing neurons coexpress AR predominantly in hypothalamic arcuate and ventral premammillary nuclei (ARH and PMv, respectively), with low colocalization in other LepRb neuronal populations, and very low colocalization in the pituitary gland and gonads. Deletion of AR from LepRb-expressing cells (LepRbΔAR) has no effect on body weight, energy expenditure, and glucose homeostasis in male and female mice. However, LepRbΔAR female mice show increased body length later in life, whereas male LepRbΔAR mice show an increase in spontaneous ambulatory activity. LepRbΔAR mice display typical pubertal timing, estrous cycles, and fertility, but increased testosterone levels in males. Removal of sex steroid negative feedback action induced an exaggerated rise in luteinizing hormone in LepRbΔAR males and follicle-stimulating hormone in LepRbΔAR females. Our findings show that AR can directly affect a subset of ARH and PMv neurons in a sex-specific manner and demonstrate specific androgenic actions in the neuroendocrine hypothalamus.


Asunto(s)
Sistemas Neurosecretores/fisiopatología , Receptores Androgénicos/genética , Receptores de Leptina/genética , Caminata/fisiología , Animales , Metabolismo Energético/genética , Epistasis Genética , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/patología , Receptores Androgénicos/deficiencia , Caracteres Sexuales , Transducción de Señal/genética
13.
Psychoneuroendocrinology ; 119: 104749, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32554173

RESUMEN

Posttraumatic stress disorder (PTSD) is associated with dysregulation of the neuroendocrine system, including cortisol, allopregnanolone, and pregnanolone. Preliminary evidence from animal models suggests that baseline levels of these biomarkers may predict response to PTSD treatment. We report the change in biomarkers over the course of PTSD treatment. Biomarkers were sampled from individuals participating in (1) a randomized controlled trial comparing a web-version of Prolonged Exposure (Web-PE) therapy to in-person Present-Centered Therapy (PCT) and (2) from individuals participating in a nonrandomized effectiveness study testing PE delivered in-person as part of an intensive outpatient PTSD program. We found that higher cortisol reactivity during script-driven imagery was associated with higher baseline PTSD severity and that baseline allopregnanolone, pregnanolone, and cortisol reactivity were associated with PTSD treatment responder status over the course of intensive outpatient treatment. These findings demonstrate that peripherally assessed biomarkers are associated with PTSD severity and likelihood of successful treatment outcome of PE delivered daily over two weeks. These assessments could be used to determine which patients are likely to respond to treatment and which patients require augmentation to increase the likelihood of optimal response to PTSD treatment.


Asunto(s)
Biomarcadores/metabolismo , Terapia Implosiva , Personal Militar , Sistemas Neurosecretores/metabolismo , Trastornos por Estrés Postraumático/terapia , Adulto , Campaña Afgana 2001- , Biomarcadores/análisis , Femenino , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Hidrocortisona/análisis , Hidrocortisona/metabolismo , Terapia Implosiva/métodos , Guerra de Irak 2003-2011 , Masculino , Persona de Mediana Edad , Personal Militar/psicología , Saliva/química , Saliva/metabolismo , Trastornos por Estrés Postraumático/diagnóstico , Trastornos por Estrés Postraumático/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos
14.
J Endocrinol ; 246(2): R33-R50, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32380471

RESUMEN

In most species, survival relies on the hypothalamic control of endocrine axes that regulate critical functions such as reproduction, growth, and metabolism. For decades, the complexity and inaccessibility of the hypothalamic-pituitary axis has prevented researchers from elucidating the relationship between the activity of endocrine hypothalamic neurons and pituitary hormone secretion. Indeed, the study of central control of endocrine function has been largely dominated by 'traditional' techniques that consist of studying in vitro or ex vivo isolated cell types without taking into account the complexity of regulatory mechanisms at the level of the brain, pituitary and periphery. Nowadays, by exploiting modern neuronal transfection and imaging techniques, it is possible to study hypothalamic neuron activity in situ, in real time, and in conscious animals. Deep-brain imaging of calcium activity can be performed through gradient-index lenses that are chronically implanted and offer a 'window into the brain' to image multiple neurons at single-cell resolution. With this review, we aim to highlight deep-brain imaging techniques that enable the study of neuroendocrine neurons in awake animals whilst maintaining the integrity of regulatory loops between the brain, pituitary and peripheral glands. Furthermore, to assist researchers in setting up these techniques, we discuss the equipment required and include a practical step-by-step guide to performing these deep-brain imaging studies.


Asunto(s)
Estado de Conciencia/fisiología , Hipotálamo/diagnóstico por imagen , Sistemas Neurosecretores/diagnóstico por imagen , Animales , Encéfalo , Humanos , Hipotálamo/citología , Neuroendocrinología/métodos , Sistemas Neurosecretores/metabolismo
15.
Mol Cell Endocrinol ; 514: 110876, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32473184

RESUMEN

Seasonal rhythms in energy balance are well documented across temperate and equatorial zones animals. The long-term regulated changes in seasonal physiology consists of a rheostatic system that is essential to successful time annual cycles in reproduction, hibernation, torpor, and migration. Most animals use the annual change in photoperiod as a reliable and robust environmental cue to entrain endogenous (i.e. circannual) rhythms. Research over the past few decades has predominantly examined the role of first order neuroendocrine peptides for the rheostatic changes in energy balance. These anorexigenic and orexigenic neuropeptides in the arcuate nucleus include neuropeptide y (Npy), agouti-related peptide (Agrp), cocaine and amphetamine related transcript (Cart) and pro-opiomelanocortin (Pomc). Recent studies also indicate that VGF nerve growth factor inducible (Vgf) in the arcuate nucleus is involved in the seasonal regulation of energy balance. In situ hybridization, qPCR and RNA-sequencing studies have identified that Pomc expression across fish, avian and mammalian species, is a neuroendocrine marker that reflects seasonal energetic states. Here we highlight that long-term changes in arcuate Pomc and Vgf expression is conserved across species and may provide rheostatic regulation of seasonal energy balance.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Factores de Crecimiento Nervioso/farmacología , Neuropéptidos/metabolismo , Proopiomelanocortina/farmacología , Proteína Relacionada con Agouti/farmacología , Proteína Relacionada con Agouti/fisiología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/fisiología , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/farmacología , Proteínas del Tejido Nervioso/fisiología , Neuropéptido Y/farmacología , Neuropéptido Y/fisiología , Neuropéptidos/efectos de los fármacos , Sistemas Neurosecretores/efectos de los fármacos , Sistemas Neurosecretores/metabolismo
16.
Gen Comp Endocrinol ; 295: 113519, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32470473

RESUMEN

Wild animals are brought into captivity for many reasons. However, unlike laboratory-bred animals, wild caught animals often respond to the dramatic shift in their environment with physiological changes in the stress and reproductive pathways. Using wild-caught male and female house sparrows (Passer domesticus) we examined how time in captivity affects the expression of reproductive and stress-associated genes in the brain, specifically, the hypothalamus. We quantified relative mRNA expression of a neurohormone involved in the stress response (corticotropin releasing hormone [CRH]), a hypothalamic inhibitor of reproduction (gonadotropin inhibitory hormone [GnIH]), and the glucocorticoid receptor (GR), which is important in terminating the stress response. To understand potential shifts at the cellular level, we also examined the presence of hypothalamic GnIH (GnIH-ir) using immunohistochemistry. We hypothesized that expression of these genes and the abundance of cells immunoreactive for GnIH would change in response to time in captivity as compared to free-living individuals. We found that GR mRNA expression and GnIH-ir cell abundance increased after 24 and 45 days in captivity, as compared to wild-caught birds. At 66 days in captivity, GR expression and GnIH cell abundance did not differ from wild-caught birds, suggesting birds had acclimated to captivity. Evaluation of CRH and GnIH mRNA expression yielded similar trends, though they were not statistically significant. In addition, although neuroendocrine factors appeared to acclimate to captivity, a previous study indicated that corticosterone release and immune responses of these same birds did not acclimate to captivity, suggesting that neuroendocrine endpoints may adapt more rapidly to captivity than downstream physiological measures. These data expand our understanding of the physiological shifts occurring when wild animals are brought into captivity.


Asunto(s)
Hipotálamo/fisiología , Sistemas Neurosecretores/metabolismo , Reproducción/fisiología , Gorriones/fisiología , Estrés Fisiológico , Animales , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Regulación de la Expresión Génica , Gónadas/anatomía & histología , Hormonas Hipotalámicas/metabolismo , Masculino , Tamaño de los Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Gorriones/genética
17.
Animal ; 14(S1): s65-s77, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32024569

RESUMEN

Improvements in feed intake of dairy cows entering the early lactation period potentially decrease the risk of metabolic disorders, but before developing approaches targeting the intake level, mechanisms controlling and dysregulating energy balance and feed intake need to be understood. This review focuses on different inflammatory pathways interfering with the neuroendocrine system regulating feed intake of periparturient dairy cows. Subacute inflammation in various peripheral organs often occurs shortly before or after calving and is associated with increased pro-inflammatory cytokine levels. These cytokines are released into the circulation and sensed by neurons located in the hypothalamus, the key brain region regulating energy balance, to signal reduction in feed intake. Besides these peripheral humoral signals, glia cells in the brain may produce pro-inflammatory cytokines independent of peripheral inflammation. Preliminary results show intensive microglia activation in early lactation, suggesting their involvement in hypothalamic inflammation and the control of feed intake of dairy cows. On the other hand, pro-inflammatory cytokine-induced activation of the vagus nerve transmits signalling to the brain, but this pathway seems not exclusively necessary to signal feed intake reduction. Yet, less studied in dairy cows so far, the endocannabinoid system links inflammation and the hypothalamic control of feed intake. Distinct endocannabinoids exert anti-inflammatory action but also stimulate the posttranslational cleavage of neuronal proopiomelanocortin towards ß-endorphin, an orexigen promoting feed intake. Plasma endocannabinoid concentrations and hypothalamic ß-endorphin levels increase from late pregnancy to early lactation, but less is known about the regulation of the hypothalamic endocannabinoid system during the periparturient period of dairy cows. Dietary fatty acids may modulate the formation of endocannabinoids, which opens new avenues to improve metabolic health and immune status of dairy cows.


Asunto(s)
Bovinos/fisiología , Citocinas/metabolismo , Metabolismo Energético , Inflamación/veterinaria , Leche/metabolismo , Transducción de Señal , Animales , Dieta/veterinaria , Femenino , Hipotálamo/metabolismo , Lactancia , Lipopolisacáridos/metabolismo , Sistemas Neurosecretores/metabolismo , Embarazo
18.
Psychoneuroendocrinology ; 113: 104549, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31884322

RESUMEN

Endogenous cannabinoids (endocannabinoids, eCB) are expressed throughout the body and contribute to regulation of the hypothalamo-pituitary-adrenal (HPA) axis and general stress reactivity. This study assessed the contributions of CB1 receptors (CB1R) in the modulation of basal and stress-induced neural and HPA axis activities. Catheterized adult male rats were placed in chambers to acclimate overnight, with their catheters connected and exteriorized from the chambers for relatively stress-free remote injections. The next morning, the CB1R antagonist AM251 (1 or 2 mg/kg) or vehicle was administered, and 30 min later, rats were exposed to loud noise stress (30 min) or no noise (basal condition). Blood, brains, pituitary and adrenal glands were collected immediately after the procedures for analysis of c-fos and CB1R mRNAs, corticosterone (CORT) and adrenocorticotropin hormone (ACTH) plasma levels. Basally, CB1R antagonism induced c-fos mRNA in the basolateral amygdala (BLA) and auditory cortex (AUD) and elevated plasma CORT, indicating disruption of eCB-mediated constitutive inhibition of activity. CB1R blockade also potentiated stress-induced hormone levels and c-fos mRNA in several regions such as the bed nucleus of the stria terminalis (BST), lateral septum (LS), and basolateral amygdala (BLA) and the paraventricular nucleus of the hypothalamus (PVN). CB1R mRNA was detected in all central tissues investigated, and the adrenal cortex, but at very low levels in the anterior pituitary gland. Interestingly, CB1R mRNA was rapidly and bidirectionally regulated in response to stress and/or antagonist treatment in some regions. eCBs therefore modulate the HPA axis by regulating both constitutive and activity-dependent inhibition at multiple levels.


Asunto(s)
Células Neuroendocrinas/fisiología , Receptor Cannabinoide CB1/fisiología , Corteza Suprarrenal/metabolismo , Glándulas Suprarrenales/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Corticosterona/sangre , Endocannabinoides/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Masculino , Células Neuroendocrinas/efectos de los fármacos , Células Neuroendocrinas/metabolismo , Sistemas Neurosecretores/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Piperidinas/farmacología , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas Proto-Oncogénicas c-fos/sangre , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Restricción Física/psicología , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología
19.
Gen Comp Endocrinol ; 285: 113250, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31445009

RESUMEN

Seasonally breeding animals initiate gonadal recrudescence when mechanisms that suppress reproduction give way to mechanisms that stimulate it. However, knowledge of mechanistic changes in hormonal regulation during this transition is limited. Further, most studies of reproductive timing have focused on males, despite the critical role of females in determining breeding phenology. Closely related populations that live in the same environment but differ in reproductive timing provide an opportunity to examine differences in mechanisms during the transition from the pre-reproductive to reproductive state. We studied closely related migrant and resident populations of dark-eyed juncos (Junco hyemalis) that reside in the same environment in spring but differ in breeding phenology. Residents initiate breeding earlier than migrants, which do not breed until after they have migrated. To directly study differences in the hypothalamic mechanisms of reproduction, we captured 16 migrant and 13 resident females from the field on March 25-April 11. We quantified expression of mRNA transcripts and show that resident females had higher abundance of gonadotropin-releasing hormone transcripts than migrant females, indicating greater reproductive development in resident than migrant females living in the same environment. We also found higher transcript abundance of estrogen receptor and androgen receptor in migrant than resident females, suggesting that negative feedback may delay reproductive development in migrant females until after they migrate. These differences in hypothalamic mechanisms may help to explain differences in reproductive timing in populations that differ in migratory strategy.


Asunto(s)
Migración Animal/fisiología , Sistemas Neurosecretores/metabolismo , Estaciones del Año , Pájaros Cantores/fisiología , Simpatría/fisiología , Animales , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Modelos Lineales , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Eur J Neurosci ; 51(1): 509-530, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30472752

RESUMEN

Reproduction, like many other biological functions, exhibits marked daily and seasonal rhythms in order to anticipate and adapt breeding activity to environmental challenges. In recent years, studies investigating the neuroendocrine mechanisms driving rhythms in reproduction have unveiled the pivotal role of hypothalamic neurons expressing kisspeptin in integrating and forwarding daily and seasonal cues to the reproductive system. The objective of this review is to summarize the knowledge on the effect and role of this neuropeptide on the mammalian hypothalamo-pituitary-gonadal axis and describe how it is involved in the daily control of ovulation in females and long-term adaptation of reproduction in seasonal breeders.


Asunto(s)
Neuropéptidos , Reproducción , Animales , Femenino , Hipotálamo/metabolismo , Neuropéptidos/metabolismo , Sistemas Neurosecretores/metabolismo , Estaciones del Año
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA