Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Lancet Diabetes Endocrinol ; 9(4): 235-246, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33647242

RESUMEN

Prader-Willi syndrome is a rare genetic neurodevelopmental disorder resulting from the loss of expression of maternally imprinted genes located in the paternal chromosomal region, 15q11-13. Impaired hypothalamic development and function is the cause of most of the phenotypes comprising the developmental trajectory of Prader-Willi syndrome: from anorexia at birth to excessive weight gain preceding hyperphagia, and early severe obesity with hormonal deficiencies, behavioural problems, and dysautonomia. Growth hormone deficiency, hypogonadism, hypothyroidism, premature adrenarche, corticotropin deficiency, precocious puberty, and glucose metabolism disorders are the main endocrine dysfunctions observed. Additionally, as a result of hypothalamic dysfunction, oxytocin and ghrelin systems are impaired in most patients. Standard pituitary and gonadal hormone replacement therapies are required. In this Review, we discuss Prader-Willi syndrome as a model of hypothalamic dysfunction, and provide a comprehensive description of the accumulated knowledge on genetics, pathophysiology, and treatment approaches of this rare disorder.


Asunto(s)
Enfermedades del Sistema Endocrino/fisiopatología , Hipotálamo/fisiopatología , Síndrome de Prader-Willi/fisiopatología , Animales , Enfermedades del Sistema Endocrino/genética , Enfermedades del Sistema Endocrino/terapia , Humanos , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/terapia , Proteínas/genética
2.
JCI Insight ; 5(17)2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32879135

RESUMEN

Prader-Willi syndrome (PWS) is a developmental disorder caused by loss of maternally imprinted genes on 15q11-q13, including melanoma antigen gene family member L2 (MAGEL2). The clinical phenotypes of PWS suggest impaired hypothalamic neuroendocrine function; however, the exact cellular defects are unknown. Here, we report deficits in secretory granule (SG) abundance and bioactive neuropeptide production upon loss of MAGEL2 in humans and mice. Unbiased proteomic analysis of Magel2pΔ/m+ mice revealed a reduction in components of SG in the hypothalamus that was confirmed in 2 PWS patient-derived neuronal cell models. Mechanistically, we show that proper endosomal trafficking by the MAGEL2-regulated WASH complex is required to prevent aberrant lysosomal degradation of SG proteins and reduction of mature SG abundance. Importantly, loss of MAGEL2 in mice, NGN2-induced neurons, and human patients led to reduced neuropeptide production. Thus, MAGEL2 plays an important role in hypothalamic neuroendocrine function, and cellular defects in this pathway may contribute to PWS disease etiology. Moreover, these findings suggest unanticipated approaches for therapeutic intervention.


Asunto(s)
Antígenos de Neoplasias/fisiología , Hipotálamo/patología , Neuronas/patología , Neuropéptidos/metabolismo , Síndrome de Prader-Willi/fisiopatología , Proteínas/metabolismo , Proteínas/fisiología , Vesículas Secretoras/patología , Animales , Femenino , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Fenotipo , Transporte de Proteínas , Proteínas/genética , Proteoma/análisis , Proteoma/metabolismo , Vesículas Secretoras/metabolismo
3.
Endocr J ; 67(10): 1029-1037, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-32565499

RESUMEN

Nutritional intervention for maintaining an appropriate body composition is central to the management of Prader-Willi syndrome (PWS). Despite evidence that visceral adipose tissue (VAT) is associated with increased metabolic risks, the effects of nutritional intervention on fat distribution have not been evaluated for PWS children. We herein investigated fat distribution in 20 genetically diagnosed PWS children (9 males and 11 females); 17 of which received nutritional intervention with or without growth hormone (GH) treatment [GH-treated group (n = 8), GH-untreated group (n = 9)]. GH treatment continued for median of 4.9 years. GH treatment significantly increased height standard deviation score (SDS) whereas body weight SDS and body mass index SDS were not affected in GH-treated group. In GH-untreated group, height SDS significantly decreased during approximately 5 years of follow-up. Fat distribution was evaluated at the median age of 6.93 years in GH-treated group and 7.01 years in GH-untreated group. VAT was maintained within the reference range in both groups. Subcutaneous adipose tissue (SAT) was elevated in GH-untreated groups compared to reference values whereas it was not in GH-treated group. The remaining three subjects, who had never received nutritional intervention or GH treatment, showed increased VAT and SAT. In conclusion, nutritional intervention is beneficial in maintaining VAT within the reference range during childhood, although excessive nutritional intervention may cause unfavorable effect on linear growth.


Asunto(s)
Distribución de la Grasa Corporal , Dietoterapia , Hormona de Crecimiento Humana/uso terapéutico , Grasa Intraabdominal , Obesidad/prevención & control , Síndrome de Prader-Willi/terapia , Grasa Subcutánea , Adolescente , Índice de Masa Corporal , Restricción Calórica , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/fisiopatología
4.
JCI Insight ; 5(12)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32365348

RESUMEN

Imprinted genes are highly expressed in the hypothalamus; however, whether specific imprinted genes affect hypothalamic neuromodulators and their functions is unknown. It has been suggested that Prader-Willi syndrome (PWS), a neurodevelopmental disorder caused by lack of paternal expression at chromosome 15q11-q13, is characterized by hypothalamic insufficiency. Here, we investigate the role of the paternally expressed Snord116 gene within the context of sleep and metabolic abnormalities of PWS, and we report a significant role of this imprinted gene in the function and organization of the 2 main neuromodulatory systems of the lateral hypothalamus (LH) - namely, the orexin (OX) and melanin concentrating hormone (MCH) - systems. We observed that the dynamics between neuronal discharge in the LH and the sleep-wake states of mice with paternal deletion of Snord116 (PWScrm+/p-) are compromised. This abnormal state-dependent neuronal activity is paralleled by a significant reduction in OX neurons in the LH of mutant mice. Therefore, we propose that an imbalance between OX- and MCH-expressing neurons in the LH of mutant mice reflects a series of deficits manifested in the PWS, such as dysregulation of rapid eye movement (REM) sleep, food intake, and temperature control.


Asunto(s)
Conducta Animal/fisiología , Área Hipotalámica Lateral/metabolismo , Hipotálamo/metabolismo , Orexinas/metabolismo , ARN Nucleolar Pequeño/genética , Sueño/fisiología , Animales , Modelos Animales de Enfermedad , Conducta Alimentaria , Área Hipotalámica Lateral/fisiopatología , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Ratones , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatología
5.
J Neuroendocrinol ; 31(7): e12728, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31046160

RESUMEN

Subsequent to the discovery of ghrelin as the endogenous ligand of growth hormone secretagogue receptor 1a, this unique gut peptide has been found to exert numerous physiological effects, such as appetite stimulation and lipid accumulation via the central regulating mechanisms in the hypothalamus, stimulation of gastric motility, regulation of glucose metabolism and brown fat thermogenesis, and modulation of stress, anxiety, taste sensation, reward-seeking behaviour and the sleep/wake cycle. Prader-Willi syndrome (PWS) has been described as a unique pathological state characterised by severe obesity and high circulating levels of ghrelin. It was hypothesised that hyperghrelinaemia would explain at least a part of the feeding behaviour and body composition of PWS patients, who are characterised by hyperphagia, an obsession with food and food-seeking, and increased adiposity. Initially, the link between hyperghrelinaemia and growth hormone deficiency, which is observed in 90% of the children with PWS, was not fully understood. Over the years, however, the increasing knowledge on ghrelin, PWS features and the natural history of the disease has led to a more comprehensive description of the abnormal ghrelin system and its role in the pathophysiology of this rare and complex neurodevelopmental genetic disease. In the present study, we (a) present the current view of PWS; (b) explain its natural history, including recent data on the ghrelin system in PWS patients; and (c) discuss the therapeutic approach of modulating the ghrelin system in these patients and the first promising results.


Asunto(s)
Encéfalo/fisiopatología , Ghrelina/fisiología , Síndrome de Prader-Willi/fisiopatología , Animales , Sistema Endocrino/fisiopatología , Conducta Alimentaria/fisiología , Humanos , Hiperfagia/fisiopatología , Hipotálamo/fisiopatología , Neuronas/fisiología
6.
Neuroimage Clin ; 21: 101662, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30639180

RESUMEN

OBJECTIVE: To investigate, based on a putative abnormal neural processing of disgusting signals in Prader Willi syndrome (PWS) patients, the brain response to visual representations of disgusting food in PWS using functional MRI (fMRI). METHODS: Twenty-one genetically-confirmed PWS patients, 30 age- and sex-matched and 28 BMI-matched control subjects viewed a movie depicting disgusting food-related scenes interspersed with scenes of appetizing food while fMRI was acquired. Brain activation maps were compared between groups and correlated with disgust and hunger ratings. RESULTS: At the cortical level, the response to disgusting food representations in PWS patients was qualitatively similar to that of control subjects, albeit less extensive, and engaged brain regions typically related to visually-evoked disgust, such as the anterior insula/frontal operculum, the lateral frontal cortex and visual areas. By contrast, activation was almost absent in limbic structures directly concerned with the regulation of instinctive behavior robustly activated in control subjects, such as the hypothalamus, amygdala/hippocampus and periaqueductal gray. CONCLUSIONS: Our study provides novel insights into the neural substrates of appetite control in a genetically-mediated cause of obesity. The presence of significant cortical changes further indicates that PWS patients consciously process disgusting stimuli, but the virtual absence of response in deep, limbic structures suggests that disgusting signals do not adequately reach the primary brain system for the appetite control.


Asunto(s)
Encéfalo/fisiopatología , Corteza Cerebral/fisiopatología , Hipotálamo/fisiopatología , Síndrome de Prader-Willi/fisiopatología , Adolescente , Adulto , Mapeo Encefálico/métodos , Femenino , Humanos , Hipotálamo/patología , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Obesidad/fisiopatología , Adulto Joven
7.
Hum Mol Genet ; 27(18): 3129-3136, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29878108

RESUMEN

Prader-Willi Syndrome (PWS) is a neurodevelopmental disorder causing social and learning deficits, impaired satiety and severe childhood obesity. Genetic underpinning of PWS involves deletion of a chromosomal region with several genes, including MAGEL2, which is abundantly expressed in the hypothalamus. Of appetite regulating hypothalamic cell types, both AGRP and POMC-expressing neurons contain Magel2 transcripts but the functional impact of its deletion on these cells has not been fully characterized. Here, we investigated these key neurons in Magel2-null mice in terms of the activity levels at different energy states as well as their behavioral function. Using cell type specific ex vivo electrophysiological recordings and in vivo chemogenetic activation approaches we evaluated impact of Magel2 deletion on AGRP and POMC-neuron induced changes in appetite. Our results suggest that POMC neuron activity profile as well as its communication with downstream targets is significantly compromised, while AGRP neuron function with respect to short term feeding is relatively unaffected in Magel2 deficiency.


Asunto(s)
Proteína Relacionada con Agouti/genética , Antígenos de Neoplasias/genética , Apetito/genética , Síndrome de Prader-Willi/genética , Proopiomelanocortina/genética , Proteínas/genética , Animales , Apetito/fisiología , Deleción Cromosómica , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Ratones , Ratones Noqueados , Neuronas/patología , Obesidad/complicaciones , Obesidad/genética , Obesidad/fisiopatología , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/fisiopatología
8.
Diabetes Obes Metab ; 19(12): 1751-1761, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28556449

RESUMEN

AIMS: There are no treatments for the extreme hyperphagia and obesity in Prader-Willi syndrome (PWS). The bestPWS clinical trial assessed the efficacy, safety and tolerability of the methionine aminopeptidase 2 (MetAP2) inhibitor, beloranib. MATERIALS AND METHODS: Participants with PWS (12-65 years old) were randomly assigned (1:1:1) to biweekly placebo, 1.8 mg beloranib or 2.4 mg beloranib injection for 26 weeks at 15 US sites. Co-primary endpoints were the changes in hyperphagia [measured by Hyperphagia Questionnaire for Clinical Trials (HQ-CT); possible score 0-36] and weight by intention-to-treat. ClinicalTrials.gov registration: NCT02179151. RESULTS: One-hundred and seven participants were included in the intention-to-treat analysis: placebo (n = 34); 1.8 mg beloranib (n = 36); or 2.4 mg beloranib (n = 37). Improvement (reduction) in HQ-CT total score was greater in the 1.8 mg (mean difference -6.3, 95% CI -9.6 to -3.0; P = .0003) and 2.4 mg beloranib groups (-7.0, 95% CI -10.5 to -3.6; P = .0001) vs placebo. Compared with placebo, weight change was greater with 1.8 mg (mean difference - 8.2%, 95% CI -10.8 to -5.6; P < .0001) and 2.4 mg beloranib (-9.5%, 95% CI -12.1 to -6.8; P < .0001). Injection site bruising was the most frequent adverse event with beloranib. Dosing was stopped early due to an imbalance in venous thrombotic events in beloranib-treated participants (2 fatal events of pulmonary embolism and 2 events of deep vein thrombosis) compared with placebo. CONCLUSIONS: MetAP2 inhibition with beloranib produced statistically significant and clinically meaningful improvements in hyperphagia-related behaviours and weight loss in participants with PWS. Although investigation of beloranib has ceased, inhibition of MetAP2 is a novel mechanism for treating hyperphagia and obesity.


Asunto(s)
Aminopeptidasas/antagonistas & inhibidores , Depresores del Apetito/uso terapéutico , Cinamatos/uso terapéutico , Ciclohexanos/uso terapéutico , Compuestos Epoxi/uso terapéutico , Glicoproteínas/antagonistas & inhibidores , Hiperfagia/prevención & control , Obesidad/prevención & control , Síndrome de Prader-Willi/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Sesquiterpenos/uso terapéutico , Adolescente , Adulto , Aminopeptidasas/metabolismo , Depresores del Apetito/administración & dosificación , Depresores del Apetito/efectos adversos , Índice de Masa Corporal , Cinamatos/administración & dosificación , Cinamatos/efectos adversos , Ciclohexanos/administración & dosificación , Ciclohexanos/efectos adversos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Terminación Anticipada de los Ensayos Clínicos , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/efectos adversos , Femenino , Glicoproteínas/metabolismo , Humanos , Hiperfagia/etiología , Hiperfagia/fisiopatología , Análisis de Intención de Tratar , Masculino , Metionil Aminopeptidasas , Obesidad/etiología , Síndrome de Prader-Willi/fisiopatología , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/efectos adversos , Sesquiterpenos/administración & dosificación , Sesquiterpenos/efectos adversos , Índice de Severidad de la Enfermedad , Trombosis de la Vena/inducido químicamente , Trombosis de la Vena/fisiopatología , Pérdida de Peso/efectos de los fármacos , Adulto Joven
9.
Hum Mol Genet ; 21(21): 4703-17, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22865874

RESUMEN

MAGED1, NECDIN and MAGEL2 are members of the MAGE gene family. The latter two of these genes have been involved in Prader-Willi syndrome (PWS), which includes hyperphagia, repetitive and compulsive behaviors, and cognitive impairment. Here, we show that Maged1-deficient mice develop progressive obesity associated with hyperphagia and reduced motor activity. Loss of Maged1 also results in a complex behavioral syndrome that includes reduced social interactions and memory, deficient sexual behavior, as well as increased anxiety and self-grooming. Oxytocin (OT), which is produced in the hypothalamus, can act as a neurotransmitter that reduces anxiety, promotes social behaviors and regulates food intake. Growing evidences indicate that OT is involved in autism. We found that Maged1 mutants showed a severe reduction in the levels of mature OT, but not of its precursors, in the hypothalamus. Moreover, the administration of OT rescued the deficit in social memory of these mice. We conclude that Maged1 is required for OT processing or stability. A decrease in mature OT levels in Maged1 mutants affects social interactions and possibly other behavioral processes. Our observations suggest that, in human, MAGED1 could play a role in autism or cause a neurodevelopmental condition that is reminiscent of the PWS.


Asunto(s)
Proteínas de Neoplasias , Oxitocina , Síndrome de Prader-Willi , Conducta Sexual Animal , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ansiedad/fisiopatología , Trastorno Autístico/metabolismo , Trastorno Autístico/fisiopatología , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Relaciones Interpersonales , Ratones , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Oxitocina/administración & dosificación , Oxitocina/biosíntesis , Oxitocina/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatología , Conducta Sexual Animal/efectos de los fármacos , Conducta Sexual Animal/fisiología
10.
J Clin Endocrinol Metab ; 96(1): E173-80, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20980432

RESUMEN

CONTEXT: Prader-Willi syndrome (PWS) is a genetic disease associated with hypogonadism and partial GH insufficiency, possibly explained in part by a hypothalamic dysfunction. Partial insufficiency of the hypothalamic-pituitary-adrenal (HPA) axis has recently been suggested. OBJECTIVE: The objective of the study was to further explore the HPA axis in PWS by use of routine tests. DESIGN: Nonselected PWS patients were examined with a standard high-dose synacthen test or the insulin tolerance test (ITT). A random serum (s) cortisol was measured in case of acute illness. SETTING: The study was conducted at university hospitals in Denmark and Sweden. PATIENTS: Sixty-five PWS patients with a confirmed genetic diagnosis participated in the study. MAIN OUTCOME MEASURES: A s-cortisol value above 500 nmol/liter as well as an increase of 250 nmol/liter or greater was considered a normal response. RESULTS: Fifty-seven PWS patients (median age 22 yr, total range 0.5-48 yr) were examined with the high-dose synacthen test. The median s-cortisol at the time of 30 min was 699 (474-1578) nmol/liter. Only one patient had a s-cortisol level below 500 nmol/liter but an increase of 359 nmol/liter. This patient subsequently showed a normal ITT response. Two patients had increases less than 250 nmol/liter but a time of 30-min s-cortisol values of 600 nmol/liter or greater. These three patients were interpreted as normal responders. Eight patients [aged 26 (16-36) yr] examined with the ITT had a median peak s-cortisol of 668 (502-822) nmol/liter. Four children admitted for acute illnesses had s-cortisol values ranging from 680 to 1372 nmol/liter. CONCLUSION: In this PWS cohort, the function of the HPA axis was normal, suggesting that clinically significant adrenal insufficiency in PWS is rare.


Asunto(s)
Insuficiencia Suprarrenal/diagnóstico , Cosintropina/administración & dosificación , Hidrocortisona/sangre , Sistema Hipotálamo-Hipofisario/fisiopatología , Sistema Hipófiso-Suprarrenal/fisiopatología , Síndrome de Prader-Willi/fisiopatología , Adolescente , Insuficiencia Suprarrenal/sangre , Insuficiencia Suprarrenal/fisiopatología , Adulto , Niño , Preescolar , Femenino , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Inmunoensayo , Lactante , Insulina/administración & dosificación , Masculino , Persona de Mediana Edad , Pruebas de Función Hipofisaria , Sistema Hipófiso-Suprarrenal/metabolismo , Síndrome de Prader-Willi/sangre
11.
Curr Opin Pulm Med ; 16(6): 568-73, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20814307

RESUMEN

PURPOSE OF REVIEW: Although several studies in the last years have evaluated obesity, obstructive sleep apnea (OSAS), and excessive daytime sleepiness (EDS) in patients with Prader-Willi syndrome (PWS), their pathophysiologies and interactions and the role of treatment with growth hormone are not completely understood. The present review analyzes the contributing role of obesity, OSAS, and sleep structure abnormalities in determining the EDS and the role of specific treatment in improving the clinical outcome. RECENT FINDINGS: The studies on sleep structure of PWS patients show abnormalities of rapid eye movement (REM) sleep and a decrease in non-REM sleep instability, corroborating the hypothesis of the presence of a primary disorder of vigilance and the similarities with narcolepsy. These sleep alterations might also be linked to the action of mediators of inflammation (i.e. adiponectin or cytokines) determined by obesity. Obesity and hypothalamic dysfunction could be responsible for the primary abnormalities of ventilation during sleep that, in turn, might contribute to EDS. Although EDS seems to resemble narcolepsy, PWS patients do not present the other typical symptoms of narcolepsy. SUMMARY: The most consistent hypothesis for linking the three different symptoms of PWS is a primary central hypothalamic dysfunction. Further research is needed to evaluate the contribution of the upper airway resistance syndrome in the pathogenesis of EDS, the role of the alterations of sleep microstructure, the relationships between PWS and narcoleptic phenotype, the involvement of orexin/hypocretin, and the effects of drugs acting on REM sleep and/or wakefulness.


Asunto(s)
Trastornos de Somnolencia Excesiva/fisiopatología , Obesidad/fisiopatología , Síndrome de Prader-Willi/fisiopatología , Apnea Obstructiva del Sueño/fisiopatología , Comorbilidad , Trastornos de Somnolencia Excesiva/epidemiología , Trastornos de Somnolencia Excesiva/etiología , Humanos , Hipotálamo/fisiopatología , Obesidad/epidemiología , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/epidemiología , Apnea Obstructiva del Sueño/epidemiología
12.
Neurobiol Dis ; 39(2): 169-80, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20394819

RESUMEN

RNA transcripts encoding the 2C-subtype of serotonin (5HT(2C)) receptor undergo up to five adenosine-to-inosine editing events to encode twenty-four protein isoforms. To examine the effects of altered 5HT(2C) editing in vivo, we generated mutant mice solely expressing the fully-edited (VGV) isoform of the receptor. Mutant animals present phenotypic characteristics of Prader-Willi syndrome (PWS) including a failure to thrive, decreased somatic growth, neonatal muscular hypotonia, and reduced food consumption followed by post-weaning hyperphagia. Though previous studies have identified alterations in both 5HT(2C) receptor expression and 5HT(2C)-mediated behaviors in both PWS patients and mouse models of this disorder, to our knowledge the 5HT(2C) gene is the first locus outside the PWS imprinted region in which mutations can phenocopy numerous aspects of this syndrome. These results not only strengthen the link between the molecular etiology of PWS and altered 5HT(2C) expression, but also demonstrate the importance of normal patterns of 5HT(2C) RNA editing in vivo.


Asunto(s)
Regulación de la Expresión Génica/genética , Síndrome de Prader-Willi/genética , Edición de ARN/genética , Receptor de Serotonina 5-HT2C/genética , Animales , Animales Recién Nacidos , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Conducta Alimentaria/fisiología , Femenino , Fuerza de la Mano/fisiología , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Síndrome de Prader-Willi/patología , Síndrome de Prader-Willi/fisiopatología , Desempeño Psicomotor/fisiología , ARN Mensajero/metabolismo
13.
Obes Rev ; 11(10): 709-21, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20233310

RESUMEN

Hypothalamic obesity (HyOb) was first defined as the significant polyphagia and weight gain that occurs after extensive suprasellar operations for excision of hypothalamic tumours. However, polyphagia and weight gain complicate other disorders related to the hypothalamus, including those that cause structural damage to the hypothalamus like tumours, trauma, radiotherapy; genetic disorders such as Prader-Willi syndrome; side effects of psychotropic drugs; and mutations in several genes involved in hypothalamic satiety signalling. Moreover, 'simple' obesity is associated with polymorphisms in several genes involved in hypothalamic weight-regulating pathways. Thus, understanding HyOb may enhance our understanding of 'simple' obesity. This review will claim that HyOb is a far wider phenomenon than hitherto understood by the narrow definition of post-surgical weight gain. It will emphasize the similarity in clinical characteristics and therapeutic approaches for HyOb, as well as its mechanisms. HyOb, regardless of its aetiology, is a result of impairment in hypothalamic regulatory centres of body weight and energy expenditure. The pathophysiology includes loss of sensitivity to afferent peripheral humoral signals, such as, leptin on the one hand and dysfunctional afferent signals, on the other hand. The most important afferent signals deranged are energy regulation by the sympathetic nervous system and regulation of insulin secretion. Dys-regulation of 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) activity and melatonin may also have a role in the development of HyOb. The complexity of the syndrome requires simultaneous targeting of several mechanisms that are deranged in the HyOb patient. We review the studies evaluating possible treatment strategies, including sympathomimetics, somatostatin analogues, triiodothyronine, sibutramine, and surgery.


Asunto(s)
Metabolismo Energético/fisiología , Homeostasis/fisiología , Hipotálamo/fisiopatología , Obesidad/etiología , Ingestión de Alimentos/fisiología , Humanos , Hipotálamo/fisiología , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/terapia , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/fisiopatología , Psicotrópicos/efectos adversos , Aumento de Peso
14.
Horm Res ; 72(3): 153-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19729946

RESUMEN

BACKGROUND/AIMS: To investigate the etiology of hypogonadism in women with Prader-Willi Syndrome (PWS). METHODS: Ten women aged 23 +/- 5.5 years with PWS and 10 age- and BMI-matched controls were included. Blood samples were drawn and abdominal ultrasounds were performed on days 2-4 of spontaneous cycles or at random from amenorrheic women. Anti-Mullerian hormone (AMH), inhibin B (INB), gonadotropins, sex steroids, TSH, prolactin, ovarian volume and antral follicle count (AFC) in PWS women were compared with results from controls and the reference ranges. RESULTS: Compared to controls, PWS women had lower INB (mean +/- SD = 17.6 +/- 12.8 pg/ml vs. 110.6 +/- 54.5; p = 0.0002) and AMH levels (1.18 +/- 0.86 ng/ml vs. 3.53 +/- 2.42; p = 0.01). INB levels were exceptionally low in all PWS women, but individual AMH levels overlapped with the levels in the controls. Ovarian volume (mean +/- SD = 3.7 +/- 2.3 ml vs. 30.5 +/- 28.8; p = 0.03) and AFC (6.4 +/- 6.9 vs. 14.0 +/- 8.2; p = 0.01) were lower in the PWS group compared to the controls. Three PWS patients had abnormally high follicle-stimulating hormone levels, while only 1 had hypogonadotropic hypogonadism. CONCLUSIONS: Our results suggest a unique follicular stage-specific insult in women with PWS. Thus, primary ovarian dysfunction is a major component of hypogonadism in PWS.


Asunto(s)
Hipogonadismo/etiología , Enfermedades del Ovario/complicaciones , Síndrome de Prader-Willi/fisiopatología , Adolescente , Adulto , Andrógenos/sangre , Hormona Antimülleriana/sangre , Femenino , Hormona Folículo Estimulante/sangre , Humanos , Hipotálamo/fisiopatología , Inhibinas/sangre , Ovario/diagnóstico por imagen , Hipófisis/fisiopatología , Estudios Prospectivos , Ultrasonografía , Útero/diagnóstico por imagen
15.
Pediatr Phys Ther ; 21(1): 68-78, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19214079

RESUMEN

PURPOSE: The purpose of this case series is to describe the implementation of an aquatic physical therapy (PT) program at a pediatric hospital and to document improvements in participants' abilities after PT intervention. METHODS: Four patients with cerebral palsy, juvenile idiopathic arthritis, or Prader-Willi syndrome participated in aquatic and land-based PT intervention. Three of the patients had orthopedic conditions which required limited weight-bearing or low-joint impact during motor activities. A wide range of outcomes were used to assess changes in participation, activity, and body function. When available, minimal detectable change and minimal important difference values were used to interpret data. RESULTS: Clinically significant improvements were documented in functional mobility, walking endurance, range of motion, muscle strength, and/or pain reduction for all 4 patients. CONCLUSIONS: Aquatic PT used as an adjunct to land-based PT interventions may be effective in improving outcomes in patients with physical disabilities.


Asunto(s)
Artritis Juvenil/rehabilitación , Parálisis Cerebral/rehabilitación , Hidroterapia/métodos , Síndrome de Prader-Willi/rehabilitación , Adolescente , Artritis Juvenil/fisiopatología , Parálisis Cerebral/fisiopatología , Niño , Evaluación de la Discapacidad , Femenino , Hospitales Pediátricos , Humanos , Masculino , Síndrome de Prader-Willi/fisiopatología , Psicometría , Rango del Movimiento Articular , Resultado del Tratamiento
16.
J Autism Dev Disord ; 38(9): 1642-53, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18311513

RESUMEN

Prader-Willi syndrome (PWS) is a neurodevelopmental disorder characterized by hyperphagia and food preoccupations. Although dysfunction of the hypothalamus likely has a critical role in hyperphagia, it is only one of several regions involved in the regulation of eating. The purpose of this research was to examine food-related neural circuitry using functional magnetic resonance imaging in individuals with PWS and matched controls. Individuals with PWS showed increased activation in neural circuitry known to mediate hunger and motivation (hypothalamus, OFC) in response to high- versus low-calorie foods and in comparison to controls. This suggests neural circuitry for PWS is abnormally activated during hunger, particularly for high-calorie foods, and may mediate abnormally strong hunger states, therefore playing a significant role in PWS-induced hyperphagia.


Asunto(s)
Ingestión de Energía , Alimentos , Red Nerviosa/fisiopatología , Obesidad/epidemiología , Síndrome de Prader-Willi/epidemiología , Síndrome de Prader-Willi/fisiopatología , Adolescente , Adulto , Niño , Humanos , Hipotálamo/fisiopatología , Cariotipificación , Imagen por Resonancia Magnética , Masculino , Corteza Prefrontal/fisiopatología
17.
Sleep Med Rev ; 12(1): 65-75, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18201664

RESUMEN

Prader Willi Syndrome (PWS) is a rare genetic disorder characterized by a range of physical, psychological and physiological abnormalities. PWS patients may also demonstrate a range of abnormalities of sleep architecture and of breathing during sleep, and excessive daytime sleepiness (EDS). In the general population EDS is associated with Obstructive Sleep Apnea Syndrome (OSAS). In PWS, by contrast, OSAS is unlikely to fully explain EDS and other factors, including hypothalamic dysfunction are likely to contribute. The present review examines OSAS and hypothalamic dysfunction and other contributing factors to EDS in PWS.


Asunto(s)
Trastornos de Somnolencia Excesiva/diagnóstico , Síndrome de Prader-Willi/diagnóstico , Apnea Obstructiva del Sueño/diagnóstico , Adolescente , Adulto , Edad de Inicio , Índice de Masa Corporal , Niño , Trastornos de Somnolencia Excesiva/fisiopatología , Humanos , Hipotálamo/fisiopatología , Polisomnografía , Síndrome de Prader-Willi/fisiopatología , Factores de Riesgo , Apnea Obstructiva del Sueño/fisiopatología
18.
Pediatrics ; 118(2): e442-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16882785

RESUMEN

OBJECTIVE: The purpose of this work was to detect brain developmental abnormalities in Prader-Willi syndrome by using diffusion tensor imaging based on a high-field MRI system. METHODS: Eight patients with Prader-Willi syndrome and 8 age- and gender-matched normal control subjects were examined using a high-field (3.0 T) MRI system. Trace value and fractional anisotropy were assessed simultaneously in multiple representative brain regions: the deep gray matter (putamen, caudate head, and dorsomedial thalamus) and the white matter structures (frontal and parietal white matter, posterior limb of internal capsule, and corpus callosum). RESULTS: In Prader-Willi syndrome patients, trace value was found to be significantly higher in the left frontal white matter and the left dorsomedial thalamus, whereas fractional anisotropy was significantly reduced in the posterior limb of the internal capsule bilaterally, the right frontal white matter, and the splenium of the corpus callosum. The observed diffusivity characteristics indicate developmental abnormalities in these areas, which are highly consistent with the clinical features of Prader-Willi syndrome. CONCLUSIONS: The study provides the first objective evidence that Prader-Willi syndrome patients indeed have developmental abnormalities in specific areas of the brain, providing a new window toward understanding the pathophysiology of Prader-Willi syndrome.


Asunto(s)
Encéfalo/anomalías , Imagen de Difusión por Resonancia Magnética , Síndrome de Prader-Willi/patología , Adolescente , Adulto , Anisotropía , Niño , Cuerpo Calloso/patología , Femenino , Lóbulo Frontal/patología , Humanos , Cápsula Interna/patología , Masculino , Síndrome de Prader-Willi/fisiopatología , Tálamo/patología
19.
Sleep Med Rev ; 9(4): 269-310, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16006155

RESUMEN

Human narcolepsy is a chronic sleep disorder affecting 1:2000 individuals. The disease is characterized by excessive daytime sleepiness, cataplexy and other abnormal manifestations of REM sleep, such as sleep paralysis and hypnagogic hallucinations. Recently, it was discovered that the pathophysiology of (idiopathic) narcolepsy-cataplexy is linked to hypocretin ligand deficiency in the brain and cerebrospinal fluid (CSF), as well as the positivity of the human leukocyte antigen (HLA) DR2/DQ6 (DQB1*0602). The symptoms of narcolepsy can also occur during the course of other neurological conditions (i.e. symptomatic narcolepsy). We define symptomatic narcolepsy as those cases that meet the International Sleep Disorders Narcolepsy Criteria, and which are also associated with a significant underlying neurological disorder that accounts for excessive daytime sleepiness (EDS) and temporal associations. To date, we have counted 116 symptomatic cases of narcolepsy reported in literature. As, several authors previously reported, inherited disorders (n=38), tumors (n=33), and head trauma (n=19) are the three most frequent causes for symptomatic narcolepsy. Of the 116 cases, 10 are associated with multiple sclerosis, one case of acute disseminated encephalomyelitis, and relatively rare cases were reported with vascular disorders (n=6), encephalitis (n=4) and degeneration (n=1), and hererodegenerative disorder (three cases in a family). EDS without cataplexy or any REM sleep abnormalities is also often associated with these neurological conditions, and defined as symptomatic cases of EDS. Although it is difficult to rule out the comorbidity of idiopathic narcolepsy in some cases, review of the literature reveals numerous unquestionable cases of symptomatic narcolepsy. These include cases with HLA negative and/or late onset, and cases in which the occurrences of the narcoleptic symptoms are parallel with the rise and fall of the causative disease. A review of these cases (especially those with brain tumors), illustrates a clear picture that the hypothalamus is most often involved. Several cases of symptomatic cataplexy (without EDS) were also reported and in contrast, these cases appear to be often associated with non-hypothalamic structures. CSF hypocretin-1 measurement were also carried out in a limited number of symptomatic cases of narcolepsy/EDS, including narcolepsy/EDS associated with tumors (n=5), head trauma (n=3), vascular disorders (n=5), encephalopathies (n=3), degeneration (n=30), demyelinating disorder (n=7), genetic/congenital disorders (n=11) and others (n=2). Reduced CSF hypocretin-1 levels were seen in most symptomatic narcolepsy cases of EDS with various etiologies and EDS in these cases is sometimes reversible with an improvement of the causative neurological disorder and an improvement of the hypocretin status. It is also noted that some symptomatic EDS cases (with Parkinson diseases and the thalamic infarction) appeared, but they are not linked with hypocretin ligand deficiency. In contrast to idiopathic narcolepsy cases, an occurrence of cataplexy is not tightly associated with hypocretin ligand deficiency in symptomatic cases. Since CSF hypocretin measures are still experimental, cases with sleep abnormalities/cataplexy are habitually selected for CSF hypocretin measures. Therefore, it is still not known whether all or a large majority of cases with low CSF hypocretin-1 levels with CNS interventions, exhibit EDS/cataplexy. It appears that further studies of the involvement of the hypocretin system in symptomatic narcolepsy and EDS are helpful to understand the pathophysiological mechanisms for the occurrence of EDS and cataplexy.


Asunto(s)
Trastornos de Somnolencia Excesiva/metabolismo , Trastornos de Somnolencia Excesiva/fisiopatología , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Narcolepsia/líquido cefalorraquídeo , Narcolepsia/fisiopatología , Neuropéptidos/metabolismo , Receptores de Neuropéptido/metabolismo , Niño , Enfermedad Crónica , Síndrome de Coffin-Lowry/líquido cefalorraquídeo , Síndrome de Coffin-Lowry/fisiopatología , Traumatismos Craneocerebrales/líquido cefalorraquídeo , Traumatismos Craneocerebrales/fisiopatología , Trastornos de Somnolencia Excesiva/inmunología , Antígenos HLA-DQ/inmunología , Antígenos HLA-DR/inmunología , Humanos , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/fisiopatología , Distrofia Miotónica/líquido cefalorraquídeo , Distrofia Miotónica/inmunología , Distrofia Miotónica/fisiopatología , Narcolepsia/inmunología , Enfermedades Neurodegenerativas/líquido cefalorraquídeo , Enfermedades Neurodegenerativas/fisiopatología , Enfermedades de Niemann-Pick/líquido cefalorraquídeo , Enfermedades de Niemann-Pick/inmunología , Enfermedades de Niemann-Pick/fisiopatología , Receptores de Orexina , Orexinas , Enfermedad de Parkinson Posencefalítica/líquido cefalorraquídeo , Enfermedad de Parkinson Posencefalítica/fisiopatología , Síndrome de Prader-Willi/líquido cefalorraquídeo , Síndrome de Prader-Willi/inmunología , Síndrome de Prader-Willi/fisiopatología , Receptores Acoplados a Proteínas G , Enfermedades Vasculares/líquido cefalorraquídeo , Enfermedades Vasculares/fisiopatología
20.
Int Rev Cytol ; 240: 305-75, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15548416

RESUMEN

A few examples of hypothalamic, peptidergic disorders leading to clinical signs and symptoms are presented in this review. Increased activity of corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus (PVN) and decreased activity of the vasopressin neurons in the biological clock and of the thyroxine-releasing hormone (TRH) neurons in the PVN contribute to the signs and symptoms of depression. In men, the central nucleus of the bed nucleus of the stria terminalis (BSTc) is about twice as large and contains twice as many somatostatin neurons as in women. In transsexuals this sex difference is reversed, pointing to a role of this structure in gender. Luteinizing hormone-releasing hormone (LHRH) neurons are formed in the fetal olfactory placade and migrate along the terminal nerve fibers into the hypothalamus. In Kallmann's syndrome the migration process of the LHRH (gonadotropin-releasing hormone) neurons is aborted, which explains the joint occurrence of hypogonadotropic hypogonadism and anosmia in this syndrome. In postmenopausal women, the neurons of the infundibular nucleus hypertrophy and become hyperactive because of the disappearance of the estrogen feedback and contain hyperactive peptidergic neurons. Climacteric flushes may be caused by hyperactivity of the neurokinin-B or LHRH neurons in this nucleus. The hypocretin (orexin) neurons in the perifornical area are involved in sleep. In narcolepsy with cataplexy, a loss of these neurons, probably due to an autoimmune process, is found. Obese subjects with a mutation in the gene that encodes for leptin, the preproghrelin gene, or the alpha-melanocyte-stimulating hormone (alpha-MSH) gene have been described. Decreased numbers and activity of the oxytocin neurons in the PVN may be responsible for the absence of satiety in Prader-Willi syndrome. Moreover, a glucocorticoid receptor polymorphism is associated with obesitas and dysregulation of the hypothalamus-pituitary-adrenal axis. In contrast, two single nucleotide polymorphisms (SNPs) of the AGRP gene have been associated with anorexia nervosa.


Asunto(s)
Enfermedades Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Vías Nerviosas/metabolismo , Neuropéptidos/metabolismo , Animales , Trastorno Depresivo/metabolismo , Trastorno Depresivo/fisiopatología , Humanos , Enfermedades Hipotalámicas/fisiopatología , Hipotálamo/anatomía & histología , Hipotálamo/fisiopatología , Narcolepsia/metabolismo , Narcolepsia/fisiopatología , Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiopatología , Sistemas Neurosecretores/fisiopatología , Obesidad/metabolismo , Obesidad/fisiopatología , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA