Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Diabetes Obes Metab ; 26 Suppl 2: 34-45, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38450938

RESUMEN

Hypothalamic obesity (HO) is a rare and complex disorder that confers substantial morbidity and excess mortality. HO is a unique subtype of obesity characterized by impairment in the key brain pathways that regulate energy intake and expenditure, autonomic nervous system function, and peripheral hormonal signalling. HO often occurs in the context of hypothalamic syndrome, a constellation of symptoms that follow from disruption of hypothalamic functions, for example, temperature regulation, sleep-wake circadian control, and energy balance. Genetic forms of HO, including the monogenic obesity syndromes, often impact central leptin-melanocortin pathways. Acquired forms of HO occur as a result of tumours impacting the hypothalamus, such as craniopharyngioma, surgery or radiation to treat those tumours, or other forms of hypothalamic damage, such as brain injury impacting the region. Risk for severe obesity following hypothalamic injury is increased with larger extent of hypothalamic damage or lesions that contain the medial and posterior hypothalamic nuclei that support melanocortin signalling pathways. Structural damage in these hypothalamic nuclei often leads to hyperphagia, central insulin and leptin resistance, decreased sympathetic activity, low energy expenditure, and increased energy storage in adipose tissue, the collective effect of which is rapid weight gain. Individuals with hyperphagia are perpetually hungry. They do not experience fullness at the end of a meal, nor do they feel satiated after meals, leading them to consume larger and more frequent meals. To date, most efforts to treat HO have been disappointing and met with limited, if any, long-term success. However, new treatments based on the distinct pathophysiology of disturbed energy homeostasis in acquired HO may hold promise for the future.


Asunto(s)
Craneofaringioma , Enfermedades Hipotalámicas , Neoplasias Hipofisarias , Humanos , Leptina/metabolismo , Enfermedades Hipotalámicas/complicaciones , Enfermedades Hipotalámicas/terapia , Enfermedades Hipotalámicas/metabolismo , Obesidad/complicaciones , Obesidad/terapia , Obesidad/genética , Hipotálamo/metabolismo , Craneofaringioma/complicaciones , Craneofaringioma/terapia , Craneofaringioma/metabolismo , Hiperfagia , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Melanocortinas/metabolismo , Metabolismo Energético/fisiología
2.
Neuroendocrinology ; 110(9-10): 767-779, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32580186

RESUMEN

Craniopharyngiomas (CP) are rare brain tumors managed primarily with surgery and radiotherapy. There are 2 phenotypes of CP, i.e., one with a rather good outcome without hypothalamic damage and another with hypothalamic damage. With hypothalamic damage, progressive disease with recurrent operations and additional cranial radiotherapy often result in hypothalamic obesity, an affected psychosocial life, and cognitive dysfunction. The morbidity and mortality are increased for particularly cerebrovascular diseases. Preoperative hypothalamic involvement to predict hypothalamic damage is important for decision making for hypothalamus-sparing surgery. Also a postoperative hypothalamic damage evaluation with the use of hypothalamus volume measurement can predict hypothalamic obesity, which is important for early treatment options. The morbidity of CP includes cognitive dysfunction with attention deficits and impaired episodic memory and processing speed. Again patients with hypothalamic damage are more affected. Treatment options of hypothalamic obesity in the chronic phase are scarce and not convincingly successful. The most optimal situation is to try to hinder or stop the evolution of hypothalamic obesity. Prevention of hypothalamic damage is recommended, with special regard to hypothalamus-sparing therapeutic approaches that respect the integrity of essential nuclei located in both the medial and the posterior hypothalamic areas.


Asunto(s)
Craneofaringioma , Enfermedades Hipotalámicas , Obesidad , Craneofaringioma/complicaciones , Craneofaringioma/diagnóstico , Craneofaringioma/patología , Craneofaringioma/cirugía , Humanos , Enfermedades Hipotalámicas/diagnóstico , Enfermedades Hipotalámicas/etiología , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/fisiopatología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/terapia
3.
J Pediatr Endocrinol Metab ; 33(3): 331-337, 2020 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-29389666

RESUMEN

Background Hypothalamic damage may alter glucagon-like peptide-1 (GLP-1) secretion and be involved in the pathogenesis of obesity. We aim to evaluate the metabolic features and the dynamic changes of GLP-1 levels during an oral glucose tolerance test (OGTT) in children with hypothalamic obesity (HO) compared with simple obesity controls. Methods Subjects included eight patients (six females, aged 9-16 years) with hypothalamo-pituitary tumors who later developed obesity and eight controls with simple obesity matched for age, body mass index (BMI), gender and puberty. We assessed the metabolic syndrome features, fat mass, severity of hyperphagia using a standardized questionnaire, and measured glucose, insulin and GLP-1 levels during a standard 75 g OGTT. Results Age, gender distribution, pubertal status and BMI-Z scores were not significantly different. Subjects with HO had higher fasting triglycerides (TG) than controls (128 vs. 94 mg/dL; p=0.05). Four HO subjects and three controls met the criteria for the metabolic syndrome. Fasting and 120 min post-glucose load GLP-1 levels were significantly higher in HO patients than in controls (21.9 vs. 19.7 pg/mL; p=0.025, 22.1 vs. 17.7 pg/mL; p=0.012). Patients with HO had significantly higher hyperphagia scores than in simple obese controls (13 vs. 2.5; p=0.012). Conclusions Patients with HO appear to have more metabolic complications and hyperphagia than controls with simple obesity. Impaired satiety may play an important role in HO. Fasting and glucose-induced serum GLP-1 concentrations seem to be altered in HO patients and could be a part of the pathogenesis of HO.


Asunto(s)
Péptido 1 Similar al Glucagón/sangre , Glucosa/farmacología , Enfermedades Hipotalámicas/metabolismo , Obesidad/metabolismo , Adolescente , Glucemia/metabolismo , Índice de Masa Corporal , Niño , Femenino , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Humanos , Hiperfagia/metabolismo , Enfermedades Hipotalámicas/sangre , Neoplasias Hipotalámicas/sangre , Neoplasias Hipotalámicas/metabolismo , Insulina/sangre , Masculino , Síndrome Metabólico/metabolismo , Obesidad/sangre
4.
Appl Physiol Nutr Metab ; 45(7): 707-714, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31815525

RESUMEN

The mechanism underlying oligo/amenorrhea in exercising women is often presumed as hypothalamic inhibition secondary to energy deficiency; however, hyperandrogenism may provide an alternative mechanism in some exercising women. Our purpose was to compare reproductive, metabolic, and androgen profiles of exercising women with eumenorrheic, ovulatory menstrual cycles (n = 91), oligo/amenorrhea without evidence of hyperandrogenism (Oligo/Amen; n = 83), and oligo/amenorrhea with evidence of hyperandrogenism (Oligo/Amen-HA; n = 17), and determine the prevalence of oligo/amenorrhea with evidence of hyperandrogenism in exercising women. Self-reported menstrual history and quantification of daily estrogen and progesterone urinary metabolites determined reproductive status. Resting energy expenditure, body composition, and metabolic hormone concentrations determined metabolic status. Serum androgens and calculated free androgen index (FAI) determined androgen status. Groups were similar in age (22.4 ± 0.3 years), height (165.1 ± 0.5 cm), resting energy expenditure (1198.4 ± 12.0 kcal/day), and total triiodothyronine (85.0 ± 1.5 ng/dL) concentration. Oligo/Amen-HA had greater weight (60.0 ± 1.6, 56.1 ± 0.7 kg), body mass index (22.3 ± 0.4, 20.6 ± 0.2 kg/m2), percentage body fat (27.3% ± 1.4%, 24.4% ± 0.6%), fat mass (16.2 ± 1.0, 13.8 ± 0.4 kg), insulin (5.8 ± 0.7, 4.2 ± 0.3 µIU/mL), leptin (12.2 ± 2.3, 6.6 ± 0.7 ng/mL), FAI (6.1 ± 0.3, 1.7 ± 0.1), and luteinizing hormone/follicle-stimulating hormone (1.9 ± 0.3, 1.3 ± 0.2) compared with Oligo/Amen, respectively. In our sample, 17% of those with oligo/amenorrhea had concurrent hyperandrogenism. This study supports that oligo/amenorrhea in some exercising women is related to hyperandrogenism. Novelty Caution must be utilized when discriminating hypothalamic oligo/amenorrhea from hyperandrogenic oligo/amenorrhea. In our sample, 17% of those with presumed hypothalamic oligo/amenorrhea had concurrent hyperandrogenism. Exercise and/or mild energy deficiency may be protective against developing severe hyperandrogenic symptoms.


Asunto(s)
Amenorrea/diagnóstico , Ejercicio Físico/fisiología , Hiperandrogenismo/diagnóstico , Enfermedades Hipotalámicas/diagnóstico , Oligomenorrea/diagnóstico , Adulto , Amenorrea/metabolismo , Amenorrea/fisiopatología , Diagnóstico Diferencial , Femenino , Humanos , Hiperandrogenismo/metabolismo , Hiperandrogenismo/fisiopatología , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/fisiopatología , Oligomenorrea/metabolismo , Oligomenorrea/fisiopatología , Adulto Joven
5.
Prog Neurobiol ; 184: 101720, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31715222

RESUMEN

Microglia, the primary regulators of inflammatory responses in the brain, suffer deterioration during aging culminating in their inability to generate adequate adaptive responses to maintain physiological homeostasis in brain tissue. Microglia affect the function of other glial cells and neurons, including those involved in the hypothalamic control of body homeostasis. Microglial dysfunction with aging in cognitive areas such as the hippocampus is known to associate with cognitive decline; more recently, microglial alterations in the hypothalamus during midlife was suggested to participate in changes in the endocrine and metabolic control exerted by this brain region. Consequently, the feed-back loops between endocrine glands and the hypothalamus are altered. This generates a vicious circle in which the plasma levels of key neuroprotective hormones, such as gonadal hormones, insulin-like growth factor-1, growth hormone and leptin and their hypothalamic signaling are decreased, which further enhances microglial alterations and deterioration of hypothalamic function. Hypothalamic dysfunction is a risk factor for neurodegenerative diseases and these diseases in turn promote additional alterations in hypothalamic microglial cells, which are unable to cope with the neurodegenerative process, resulting in permanent damage of the neuronal-glial circuits controlling endocrine homeostasis, food intake and body metabolism. Thus, a "vicious cycle" may such be initiated.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Hipotalámicas/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Sistemas Neurosecretores/metabolismo , Animales , Humanos
6.
BMJ Case Rep ; 12(7)2019 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-31289170

RESUMEN

Diencephalic syndrome is one of the rare causes of failure to thrive in infants and young children. It is caused by a tumour in diencephalon (thalamus and hypothalamus), characterised by profound emaciation with uniform loss of body weight despite normal or slightly decreased appetite, locomotor hyperactivity, euphoria and visual symptoms. Anaesthetic considerations due to decreased body fat include positioning to avoid pressure necrosis, measures to avoid hypothermia, proper drug dosing, treating electrolyte imbalances and delayed recovery. We report successful anaesthetic management of a child with diencephalic syndrome scheduled for an endoscopic biopsy of suprasellar space occupying lesion under general anaesthesia.


Asunto(s)
Anestesia/métodos , Diencéfalo/patología , Enfermedades Hipotalámicas/patología , Enfermedades de la Hipófisis/patología , Anestésicos/administración & dosificación , Biopsia , Niño , Insuficiencia de Crecimiento/etiología , Humanos , Enfermedades Hipotalámicas/diagnóstico por imagen , Enfermedades Hipotalámicas/metabolismo , Unidades de Cuidado Intensivo Pediátrico/normas , Cuidados Intraoperatorios , Masculino , Enfermedades de la Hipófisis/sangre , Enfermedades de la Hipófisis/diagnóstico por imagen , Cuidados Preoperatorios , Pronóstico , Tomografía Computarizada por Rayos X , Derivación Ventriculoperitoneal/métodos
7.
Metabolism ; 97: 32-39, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31103608

RESUMEN

OBJECTIVE: It remains unclear whether food deprivation induces changes in components of the GH/IGF-1/IGFBPs axis and if yes, which ones are mediated by leptin, an adipocyte secreted hormone regulating neuroendocrine response to energy deprivation in animals and humans. We aimed to investigate components of the axis that have not been studied to date, i.e. IGF-binding proteins (IGFBPs) and related proteases (total and intact IGFBP 3 and IGFBP 4, total IGFBP 5, PAPPA, PAPPA2 and Stanniocalcin-2), during acute (short-term fasting in healthy subjects) and chronic (women with hypothalamic amenorrhea [HA] due to excessive exercise) energy deprivation and whether metreleptin administration, in replacement, supraphysiologic or pharmacologic levels, may mediate any changes of circulating levels of the above molecules in healthy individuals and in women with hypothalamic amenorrhea. METHODS: We studied: 1) 11 healthy men and women during three four day admissions i.e. a baseline admission in the fed isocaloric state and two admissions in the complete food deprivation state for 72-h with either placebo (resulting in a hypoleptinemic state) or metreleptin administration in doses designed to normalize circulating leptin levels for the duration of the study, 2) 15 healthy men and women during three 72-hour long admissions in a complete food deprivation state receiving three escalating doses of metreleptin designed to bring circulating leptin levels to physiologic, supraphysiologic, or pharmacologic levels, and 3) 18 women with HA randomized to either metreleptin treatment in replacement doses or placebo for nine months. RESULTS: There were no significant changes in the circulating profiles of the above molecules in the fasting vs. fed state and/or with metreleptin administration during acute and chronic energy deprivation. CONCLUSIONS: The studied components of the GH/IGF-1/IGFBPs axis are not affected by energy deprivation, leptin deficiency associated with energy deprivation, or by metreleptin administration in physiologic, supraphysiologic or pharmacologic doses.


Asunto(s)
Privación de Alimentos/fisiología , Glicoproteínas/metabolismo , Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leptina/análogos & derivados , Proteína Plasmática A Asociada al Embarazo/metabolismo , Ayuno/sangre , Femenino , Humanos , Enfermedades Hipotalámicas/metabolismo , Leptina/administración & dosificación , Leptina/metabolismo , Masculino
8.
Pituitary ; 22(3): 249-260, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30684166

RESUMEN

PURPOSE: Clinical research studies over the last 15 years have reported a significant burden of hypopituitarism in survivors of traumatic brain injury (TBI). However, debate still exists about the true prevalence of hypopituitarism after head injury. METHODS: We have reviewed the literature describing the frequency of post-traumatic hypopituitarism and discuss the factors which may explain the variable frequency of the reported deficits in clinical studies including research methodology and the natural history of the disease. RESULTS: Pituitary hormone perturbations in the acute phase following injury are frequent but are difficult to attribute to traumatic pituitary damage due to physiological hormonal changes in acute illness, the confounding effect of medications, other co-morbidities and lack of appropriate control subjects. Nevertheless, a small number of studies have emphasised the clinical importance of acute, dynamic disturbance of the hypothalamic-pituitary-adrenal axis. There is a much larger evidence base examining the frequency of hypopituitarism in the chronic, recovery phase following head injury. These studies report a very broad prevalence of long-term pituitary hormone dysfunction in survivors of TBI. However, systematic review suggests the prevalence to be between 27 and 31%. CONCLUSION: Survivors of head injury are at risk of pituitary hormone dysfunction and we suggest an approach to the diagnosis of post-traumatic hypopituitarism in routine clinical practice.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/diagnóstico , Enfermedades Hipotalámicas/diagnóstico , Animales , Humanos , Hipopituitarismo/diagnóstico , Hipopituitarismo/metabolismo , Hipopituitarismo/patología , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/patología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/patología , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/patología
9.
Curr Eye Res ; 44(6): 583-589, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30624085

RESUMEN

Background: This mini-review addressed the question "what do we know about the association between the dysregulation of stress systems (HPA axis and SAM) and the onset and prognosis of CSC in adult populations?" Methods: The literature mini-review was conducted through electronic searches using the PubMed, Web of Science, and Scopus databases. All published human and animal studies with both observational and experimental designs from 1966 to October 2018 were included. Results: Our search identified 229 reports, of which 32 articles were ultimately identified to be reviewed in this paper. Among these key articles, twenty-three were related to exogenous and/or endogenous high glucocorticoids as risk factors for CSC, seven were related to Type-A behavior and chronic psychological distress as risk factors for idiopathic CSC, and two were related to stress-induced animal models of CSC. Nineteen out of twenty-three studies in the first group reported a consistent association between high circulating corticosteroids and the onset and prognosis of CSC. Six out of seven studies in the second group reported a consistent association between stress-induced allostatic (over)load and the appearance of more- or less-severe CSC disorders, assuming that elevated circulating steroids may constitute a kind of risk factor for the eye through dysregulation of the HPA axis. All the selected studies reported HPA axis dysregulation as a possible common factor to explain the association between high circulating corticosteroids and CSC. In contrast, the involvement of the SAM system is only indirectly taken into consideration through the PA and HR measures and/or plasma and 24-h urinary catecholamine levels. Therefore, information regarding the involvement of SAM system dysregulation in the onset and prognosis of CSC is lacking. This observation is particularly relevant in view of the fact that animal models of CSC in monkeys are primarily induced by adrenergic hypertonia and that the course of experimental CSC is not further exacerbated by the administration of corticosteroids.


Asunto(s)
Coriorretinopatía Serosa Central/fisiopatología , Enfermedades Hipotalámicas/fisiopatología , Sistema Hipotálamo-Hipofisario/fisiopatología , Sistema Hipófiso-Suprarrenal/fisiopatología , Estrés Psicológico/fisiopatología , Animales , Humanos , Enfermedades Hipotalámicas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Factores de Riesgo
10.
Pituitary ; 22(3): 296-304, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30334138

RESUMEN

Neurohypophysial dysfunction is common in the first days following traumatic brain injury (TBI), manifesting as dysnatremia in approximately 1 in 4 patients. Both hyponatremia and hypernatremia can impair recovery from TBI and in the case of hypernatremia, there is a significant association with excess mortality. Hyponatremia secondary to syndrome of inappropriate antidiuretic hormone secretion (SIAD) is the commonest electrolyte disturbance following TBI. Acute adrenocorticotropic hormone (ACTH)/cortisol deficiency occurs in 10-15% of TBI patients and can present with a biochemical picture identical to SIAD. For this reason, exclusion of glucocorticoid deficiency is of particular importance in post-TBI SIAD. Cerebral salt wasting is a rare cause of hyponatremia following TBI. Hyponatremia predisposes to seizures, reduced consciousness, and prolonged hospital stay. Diabetes insipidus (DI) occurs in 20% of cases following TBI; where diminished consciousness is present, appropriate fluid replacement of renal water losses is occasionally inadequate, leading to hypernatremia. Hypernatremia is strongly predictive of mortality following TBI. Most cases of DI are transient, but persistent DI is also predictive of mortality, irrespective of plasma sodium concentration. Persistent DI may herald rising intracranial pressure due to coning. True adipsic DI is rare following TBI, but patients are vulnerable to severe hypernatremic dehydration, exacerbation of neurologic deficits and hypothalamic complications, therefore clinicians should be aware of this possible variant of DI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Neurohipófisis/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/patología , Diabetes Insípida/metabolismo , Diabetes Insípida/patología , Humanos , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/patología , Neurohipófisis/patología
11.
Pediatr Blood Cancer ; 65(5): e26917, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29369501

RESUMEN

Young children with emaciation caused by a hypothalamic glioma are considered to have diencephalic syndrome (DS), which is often poorly controlled with conventional treatment. We describe an infant with DS whose tumor progressed following chemotherapy. Biopsy was performed for molecular testing and demonstrated a BRAF fusion. Treatment with the MEK inhibitor trametinib for 18 months resulted in reduction of tumor size, normalization of his weight curve, and marked neurodevelopmental improvement. Our results build on earlier reports of using targeted agents for low-grade glioma, and we review the evolving management strategy for such patients in the era of precision medicine.


Asunto(s)
Enfermedades Hipotalámicas/tratamiento farmacológico , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridonas/uso terapéutico , Pirimidinonas/uso terapéutico , Manejo de la Enfermedad , Humanos , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/patología , Lactante , Masculino , Pronóstico
12.
Metabolism ; 80: 91-104, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29107598

RESUMEN

Anorexia nervosa (AN) and hypothalamic amenorrhea (HA) are states of chronic energy deprivation associated with severely compromised bone health. Poor bone accrual during adolescence followed by increased bone loss results in lifelong low bone density, degraded bone architecture, and higher risk of fractures, despite recovery from AN/HA. Amenorrhea is only one of several compensatory responses to the negative energy balance. Other hypothalamic-pituitary hormones are affected and contribute to bone deficits, including activation of hypothalamic-pituitary-adrenal axis and growth hormone resistance. Adipokines, particularly leptin, provide information on fat/energy stores, and gut hormones play a role in the regulation of appetite and food intake. Alterations in all these hormones influence bone metabolism. Restricted in scope, current pharmacologic approaches to improve bone health have had overall limited success.


Asunto(s)
Amenorrea/genética , Anorexia Nerviosa/metabolismo , Huesos/metabolismo , Enfermedades Hipotalámicas/metabolismo , Amenorrea/tratamiento farmacológico , Amenorrea/patología , Anorexia Nerviosa/tratamiento farmacológico , Anorexia Nerviosa/patología , Huesos/patología , Femenino , Humanos , Enfermedades Hipotalámicas/tratamiento farmacológico , Masculino
13.
Endocrinology ; 159(1): 368-387, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29077836

RESUMEN

Astrocytes participate in both physiological and pathophysiological responses to metabolic and nutrient signals. Although most studies have focused on the astrocytic response to weight gain due to high-fat/high-carbohydrate intake, surplus intake of a balanced diet also induces excess weight gain. We have accessed the effects of neonatal overnutrition, which has both age- and sex-dependent effects on weight gain, on hypothalamic inflammation/gliosis. Although both male and female Wistar rats accumulate excessive fat mass as early as postnatal day (PND) 10 with neonatal overnutrition, no increase in hypothalamic cytokine levels, markers of astrocytes or microglia, or inflammatory signaling pathways were observed. At PND 50, no effect of neonatal overnutriton was found in either sex, whereas at PND 150, males again weighed significantly more than their controls, and this was coincident with an increase in markers of inflammation and astrogliosis in the hypothalamus. Circulating triglycerides and free fatty acids were also elevated in these males, but not in females or in either sex at PND 10. Thus, the effects of fatty acids and estrogens on astrocytes in vitro were analyzed. Our results indicate that changes in circulating fatty acid levels may be involved in the induction of hypothalamic inflammation/gliosis in excess weight gain, even on a normal diet, and that estrogens could participate in the protection of females from these processes. In conclusion, the interaction of developmental influences, dietary composition, age, and sex determines the central inflammatory response and the associated long-term outcomes of excess weight gain.


Asunto(s)
Astrocitos/metabolismo , Gliosis/etiología , Hiperfagia/fisiopatología , Enfermedades Hipotalámicas/etiología , Hipotálamo/metabolismo , Microglía/metabolismo , Adiposidad , Factores de Edad , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Astrocitos/patología , Biomarcadores/metabolismo , Células Cultivadas , Citocinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gliosis/inmunología , Gliosis/metabolismo , Gliosis/patología , Enfermedades Hipotalámicas/inmunología , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/patología , Hipotálamo/inmunología , Hipotálamo/patología , Mediadores de Inflamación/metabolismo , Masculino , Microglía/inmunología , Microglía/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas Wistar , Caracteres Sexuales , Transducción de Señal , Aumento de Peso
14.
Curr Opin Clin Nutr Metab Care ; 20(5): 396-401, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28708669

RESUMEN

PURPOSE OF REVIEW: In cancer patients, the development of cachexia (muscle wasting) is frequently aggravated by anorexia (loss of appetite). Their concurrence is often referred to as anorexia-cachexia syndrome. This review focusses on the recent evidence underlining hypothalamic inflammation as key driver of these processes. Special attention is given to the involvement of hypothalamic serotonin. RECENT FINDINGS: The anorexia-cachexia syndrome is directly associated with higher mortality in cancer patients. Recent reports confirm its severe impact on the quality of life of patients and their families.Hypothalamic inflammation has been shown to contribute to muscle and adipose tissue loss in cancer via central hypothalamic interleukine (IL)1ß-induced activation of the hypothalamic-pituitary-adrenal axis. The resulting release of glucocorticoids directly stimulates catabolic processes in these tissues via activation of the ubiquitin-proteosome pathway. Next to this, hypothalamic inflammation has been shown to reduce food intake in cancer by triggering changes in orexigenic and anorexigenic responses via upregulation of serotonin availability and stimulation of its signalling pathways in hypothalamic tissues. This combination of reduced food intake and stimulation of tissue catabolism represents a dual mechanism by which hypothalamic inflammation contributes to the development and maintenance of anorexia and cachexia in cancer. SUMMARY: Hypothalamic inflammation is a driving force in the development of the anorexia-cachexia syndrome via hypothalamic-pituitary-adrenal axis and serotonin pathway activation.


Asunto(s)
Anorexia/etiología , Caquexia/etiología , Enfermedades Hipotalámicas/etiología , Hipotálamo/inmunología , Modelos Neurológicos , Neoplasias/fisiopatología , Serotonina/metabolismo , Adiposidad , Animales , Anorexia/inmunología , Anorexia/metabolismo , Anorexia/fisiopatología , Caquexia/inmunología , Caquexia/metabolismo , Caquexia/fisiopatología , Humanos , Enfermedades Hipotalámicas/inmunología , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/fisiopatología , Sistema Hipotálamo-Hipofisario/inmunología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/metabolismo , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Neoplasias/sangre , Neoplasias/inmunología , Neoplasias/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Sistema Hipófiso-Suprarrenal/inmunología , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Serotonina/sangre
15.
Braz J Med Biol Res ; 50(5): e5858, 2017 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-28380195

RESUMEN

Modifications in life-style and/or pharmacotherapies contribute to weight loss and ameliorate the metabolic profile of diet-induced obese humans and rodents. Since these strategies fail to treat hypothalamic obesity, we have assessed the possible mechanisms by which duodenal-jejunal bypass (DJB) surgery regulates hepatic lipid metabolism and the morphophysiology of pancreatic islets, in hypothalamic obese (HyO) rats. During the first 5 days of life, male Wistar rats received subcutaneous injections of monosodium glutamate (4 g/kg body weight, HyO group), or saline (CTL). At 90 days of age, HyO rats were randomly subjected to DJB (HyO DJB group) or sham surgery (HyO Sham group). HyO Sham rats were morbidly obese, insulin resistant, hypertriglyceridemic and displayed higher serum concentrations of non-esterified fatty acids (NEFA) and hepatic triglyceride (TG). These effects were associated with higher expressions of the lipogenic genes and fatty acid synthase (FASN) protein content in the liver. Furthermore, hepatic genes involved in ß-oxidation and TG export were down-regulated in HyO rats. In addition, these rats exhibited hyperinsulinemia, ß-cell hypersecretion, a higher percentage of islets and ß-cell area/pancreas section, and enhanced nuclear content of Ki67 protein in islet-cells. At 2 months after DJB surgery, serum concentrations of TG and NEFA, but not hepatic TG accumulation and gene and protein expressions, were normalized in HyO rats. Insulin release and Ki67 positive cells were also normalized in HyO DJB islets. In conclusion, DJB decreased islet-cell proliferation, normalized insulinemia, and ameliorated insulin sensitivity and plasma lipid profile, independently of changes in hepatic metabolism.


Asunto(s)
Duodeno/cirugía , Hígado Graso/metabolismo , Derivación Gástrica/métodos , Enfermedades Hipotalámicas/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Yeyuno/cirugía , Obesidad/metabolismo , Animales , Animales Recién Nacidos , Glucemia/metabolismo , Proliferación Celular , Colesterol/sangre , Acido Graso Sintasa Tipo I/metabolismo , Ácidos Grasos/sangre , Hígado Graso/fisiopatología , Enfermedades Hipotalámicas/fisiopatología , Enfermedades Hipotalámicas/cirugía , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Islotes Pancreáticos/fisiopatología , Lipogénesis/genética , Hígado/metabolismo , Hígado/patología , Masculino , Obesidad/fisiopatología , Obesidad/cirugía , Páncreas/metabolismo , Páncreas/patología , Distribución Aleatoria , Ratas Wistar , Reproducibilidad de los Resultados , Factores de Tiempo , Triglicéridos/sangre
16.
Pituitary ; 20(4): 409-421, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28265842

RESUMEN

At the beginning of the twentieth century, the hypothalamus was known merely as an anatomical region of the brain lying beneath the thalamus. An increasing number of clinicopathological reports had shown the association of diabetes insipidus and adiposogenital dystrophy (Babinski-Fröhlich's syndrome), with pituitary tumors involving the infundibulum and tuber cinereum, two structures of the basal hypothalamus. The French physicians Jean Camus (1872-1924) and Gustave Roussy (1874-1948) were the first authors to undertake systematic, controlled observations of the effects of localized injuries to the basal hypothalamus in dogs and cats by pricking the infundibulo-tuberal region (ITR) with a heated needle. Their series of surgical procedures, performed between 1913 and 1922, allowed them to claim that both permanent polyuria and adiposogenital dystrophy were symptoms caused by damage to the ITR. Their results challenged the dominant doctrine of hypopituitarism as cause of diabetes insipidus and adiposogenital dystrophy that derived from the experiments performed by Paulescu and Cushing a decade earlier. With their pioneering research, Camus and Roussy influenced the experimental work on the hypothalamus performed by Percival Bailey and Frederic Bremer at Cushing's laboratory, confirming the hypothalamic origin of these symptoms in 1921. More importantly, they provided the foundations for the physiological paradigm of Neuroendocrinology, the hypothalamus' control over the endocrine secretions of the pituitary gland, as well as over water balance and fat metabolism. This article aims to credit Camus and Roussy for their groundbreaking, decisive contributions to postulate the hypothalamus being the brain region in control of endocrine homeostasis and energy metabolism.


Asunto(s)
Hipotálamo/metabolismo , Hipófisis/metabolismo , Animales , Gatos , Diabetes Insípida/metabolismo , Diabetes Insípida/patología , Perros , Sistema Endocrino/metabolismo , Sistema Endocrino/patología , Humanos , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/patología , Hipotálamo/patología , Neuroendocrinología , Hipófisis/patología , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología
17.
Braz. j. med. biol. res ; 50(5): e5858, 2017. tab, graf
Artículo en Inglés | LILACS | ID: biblio-839295

RESUMEN

Modifications in life-style and/or pharmacotherapies contribute to weight loss and ameliorate the metabolic profile of diet-induced obese humans and rodents. Since these strategies fail to treat hypothalamic obesity, we have assessed the possible mechanisms by which duodenal-jejunal bypass (DJB) surgery regulates hepatic lipid metabolism and the morphophysiology of pancreatic islets, in hypothalamic obese (HyO) rats. During the first 5 days of life, male Wistar rats received subcutaneous injections of monosodium glutamate (4 g/kg body weight, HyO group), or saline (CTL). At 90 days of age, HyO rats were randomly subjected to DJB (HyO DJB group) or sham surgery (HyO Sham group). HyO Sham rats were morbidly obese, insulin resistant, hypertriglyceridemic and displayed higher serum concentrations of non-esterified fatty acids (NEFA) and hepatic triglyceride (TG). These effects were associated with higher expressions of the lipogenic genes and fatty acid synthase (FASN) protein content in the liver. Furthermore, hepatic genes involved in β-oxidation and TG export were down-regulated in HyO rats. In addition, these rats exhibited hyperinsulinemia, β-cell hypersecretion, a higher percentage of islets and β-cell area/pancreas section, and enhanced nuclear content of Ki67 protein in islet-cells. At 2 months after DJB surgery, serum concentrations of TG and NEFA, but not hepatic TG accumulation and gene and protein expressions, were normalized in HyO rats. Insulin release and Ki67 positive cells were also normalized in HyO DJB islets. In conclusion, DJB decreased islet-cell proliferation, normalized insulinemia, and ameliorated insulin sensitivity and plasma lipid profile, independently of changes in hepatic metabolism.


Asunto(s)
Animales , Masculino , Duodeno/cirugía , Hígado Graso/metabolismo , Derivación Gástrica/métodos , Enfermedades Hipotalámicas/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Yeyuno/cirugía , Obesidad/metabolismo , Animales Recién Nacidos , Glucemia/metabolismo , Proliferación Celular , Colesterol/sangre , Acido Graso Sintasa Tipo I/metabolismo , Ácidos Grasos/sangre , Hígado Graso/fisiopatología , Enfermedades Hipotalámicas/fisiopatología , Enfermedades Hipotalámicas/cirugía , Resistencia a la Insulina , Insulina/metabolismo , Islotes Pancreáticos/fisiopatología , Lipogénesis/genética , Hígado/metabolismo , Hígado/patología , Obesidad/fisiopatología , Obesidad/cirugía , Páncreas/metabolismo , Páncreas/patología , Distribución Aleatoria , Ratas Wistar , Reproducibilidad de los Resultados , Factores de Tiempo , Triglicéridos/sangre
18.
EBioMedicine ; 8: 96-102, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27428422

RESUMEN

Human hypothalamic hamartoma (HH) is a rare subcortical lesion associated with treatment-resistant epilepsy. Cellular mechanisms responsible for epileptogenesis are unknown. We hypothesized that neuronal gap junctions contribute to epileptogenesis through synchronous activity within the neuron networks in HH tissue. We studied surgically resected HH tissue with Western-blot analysis, immunohistochemistry, electron microscopy, biocytin microinjection of recorded HH neurons, and microelectrode patch clamp recordings with and without pharmacological blockade of gap junctions. Normal human hypothalamus tissue was used as a control. Western blots showed increased expression of both connexin-36 (Cx36) and connexin-43 (Cx43) in HH tissue compared with normal human mammillary body tissue. Immunohistochemistry demonstrated that Cx36 and Cx43 are expressed in HH tissue, but Cx36 was mainly expressed within neuron clusters while Cx43 was mainly expressed outside of neuron clusters. Gap-junction profiles were observed between small HH neurons with electron microscopy. Biocytin injection into single recorded small HH neurons showed labeling of adjacent neurons, which was not observed in the presence of a neuronal gap-junction blocker, mefloquine. Microelectrode field recordings from freshly resected HH slices demonstrated spontaneous ictal/interictal-like discharges in most slices. Bath-application of gap-junction blockers significantly reduced ictal/interictal-like discharges in a concentration-dependent manner, while not affecting the action-potential firing of small gamma-aminobutyric acid (GABA) neurons observed with whole-cell patch-clamp recordings from the same patient's HH tissue. These results suggest that neuronal gap junctions between small GABAergic HH neurons participate in the genesis of epileptic-like discharges. Blockade of gap junctions may be a new therapeutic strategy for controlling seizure activity in HH patients.


Asunto(s)
Epilepsia/etiología , Epilepsia/metabolismo , Uniones Comunicantes/metabolismo , Hamartoma/complicaciones , Hamartoma/metabolismo , Enfermedades Hipotalámicas/complicaciones , Enfermedades Hipotalámicas/metabolismo , Adolescente , Adulto , Carbenoxolona/farmacología , Niño , Preescolar , Conexinas/genética , Conexinas/metabolismo , Fenómenos Electrofisiológicos , Femenino , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/ultraestructura , Expresión Génica , Hamartoma/cirugía , Humanos , Enfermedades Hipotalámicas/cirugía , Lactante , Masculino , Neuronas/metabolismo , Neuronas/ultraestructura , Adulto Joven
19.
Neuropathology ; 36(5): 480-484, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27090596

RESUMEN

Hypothalamic hamartomas are rare tumors that typically present in childhood, often with gelastic seizures, precocious puberty, or as a manifestation of Pallister-Hall syndrome. Neurofibrillary tangles are cytoplasmic aggregates of hyperphosphorylated tau that are best recognized in Alzheimer disease, other tau-associated neurodegenerative diseases, or as part of aging, but occasionally may be seen in low-grade neoplasms with a ganglion cell component as gangliocytoma or ganglioglioma. Herein, we report a case of hypothalamic hamartoma with neurofibrillary tangles.


Asunto(s)
Hamartoma/complicaciones , Hamartoma/patología , Enfermedades Hipotalámicas/complicaciones , Enfermedades Hipotalámicas/patología , Ovillos Neurofibrilares/patología , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Hamartoma/diagnóstico por imagen , Hamartoma/metabolismo , Humanos , Enfermedades Hipotalámicas/diagnóstico por imagen , Enfermedades Hipotalámicas/metabolismo , Masculino , Ovillos Neurofibrilares/metabolismo , Neuronas/metabolismo , Neuronas/patología , Proteínas tau/metabolismo
20.
Biol Aujourdhui ; 210(4): 211-225, 2016.
Artículo en Francés | MEDLINE | ID: mdl-28327280

RESUMEN

The hypothalamus is a key brain region in the regulation of energy balance. It especially controls food intake and both energy storage and expenditure through integration of humoral, neural and nutrient-related signals and cues. Hypothalamic neurons and glial cells act jointly to orchestrate, both spatially and temporally, regulated metabolic functions of the hypothalamus. Thus, the existence of a causal link between hypothalamic inflammation and deregulations of feeding behavior, such as involuntary weight-loss or obesity, has been suggested. Among the inflammatory mediators that could induce deregulations of hypothalamic control of the energy balance, chemokines represent interesting candidates. Indeed, chemokines, primarily known for their chemoattractant role of immune cells to the inflamed site, have also been suggested capable of neuromodulation. Thus, chemokines could disrupt cellular activity together with synthesis and/or secretion of multiple neurotransmitters/mediators that are involved in the maintenance of energy balance. Here, we relate, on one hand, recent results showing the primary role of the central chemokinergic signaling CCL2/CCR2 for metabolic and behavioral adaptation to high-grade inflammation, especially loss of appetite and weight, through its activity on hypothalamic neurons producing the orexigenic peptide Melanin-Concentrating Hormone (MCH) and, on the other hand, results that suggest that chemokines could also deregulate hypothalamic neuropeptidergic circuits to induce an opposite phenotype and eventually participate in the onset/development of obesity. In more details, we will emphasize a study recently showing, in a model of high-grade acute inflammation of LPS injection in mice, that central CCL2/CCR2 signaling is of primary importance for several aspects explaining weight loss associated with inflammation: after LPS injection, animals lose weight, reduce their food intake, increase their fat oxidation (thus energy consumption from fat storage)...These inflammation-induced metabolic and behavioral changes are reduced when central CCR2 signaling is disrupted either pharmacologically (by a specific inhibitor of CCR2) or genetically (in mice deficient for CCR2). This underlines the importance of this signaling in inflammation-related weight loss. We further determined that the LPS-induced and CCR2-mediated weight loss depends on the direct effect of CCR2 activation on MCH neurons activity. Indeed, the MCH neurons express CCR2, and the application of CCL2 on brain slices revealed that activation of CCR2 actually depolarizes MCH neurons and induces delays and/or failures of action potential emission. Furthermore, CCL2 is able to reduce KCl-evoked MCH secretion from hypothalamic explants. Taken together, these results demonstrate the role of the central CCL2/CCR2 signaling in metabolic and behavioral adaptation to inflammation. On the other hand, this first description of how the chemokinergic system can actually modulate the activity of the hypothalamic regulation of energy balance, but also some less advanced studies and some unpublished data, suggest that some other chemokines, such as CCL5, could participate in the development of the opposite phenotype, that is to say obesity.


Asunto(s)
Quimiocinas/fisiología , Enfermedades Hipotalámicas/complicaciones , Enfermedades Hipotalámicas/metabolismo , Enfermedades Metabólicas/etiología , Animales , Metabolismo Energético/fisiología , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Inflamación , Ratones , Obesidad/etiología , Obesidad/inmunología , Obesidad/metabolismo , Pérdida de Peso/genética , Pérdida de Peso/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...