Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Mol Metab ; 57: 101439, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35007790

RESUMEN

OBJECTIVE: In Huntington's disease (HD), the disease-causing huntingtin (HTT) protein is ubiquitously expressed and causes both central and peripheral pathology. In clinical HD, a higher body mass index has been associated with slower disease progression, indicating the role of metabolic changes in disease pathogenesis. Underlying mechanisms of metabolic changes in HD remain poorly understood, but recent studies suggest the involvement of hypothalamic dysfunction. The present study aimed to investigate whether modulation of hypothalamic HTT levels would affect metabolic phenotype and disease features in HD using mouse models. METHODS: We used the R6/2 and BACHD mouse models that express different lengths of mutant HTT to develop lean- and obese phenotypes, respectively. We utilized adeno-associated viral vectors to overexpress either mutant or wild-type HTT in the hypothalamus of R6/2, BACHD, and their wild-type littermates. The metabolic phenotype was assessed by body weight measurements over time and body composition analysis using dual-energy x-ray absorptiometry at the endpoint. R6/2 mice were further characterized using behavioral analyses, including rotarod, nesting-, and hindlimb clasping tests during early- and late-time points of disease progression. Finally, gene expression analysis was performed in R6/2 mice and wild-type littermates in order to assess transcriptional changes in the hypothalamus and adipose tissue. RESULTS: Hypothalamic overexpression of mutant HTT induced significant gender-affected body weight gain in all models, including wild-type mice. In R6/2 females, early weight gain shifted to weight loss during the corresponding late stage of disease despite increased fat accumulation. Body weight changes were accompanied by behavioral alterations. During the period of early weight gain, R6/2 mice displayed a comparable locomotor capacity to wild-type mice. When assessing behavior just prior to weight loss onset in R6/2 mice, decreased locomotor performance was observed in R6/2 females with hypothalamic overexpression of mutant HTT. Transcriptional downregulation of beta-3 adrenergic receptor (B3AR), adipose triglyceride lipase (ATGL), and peroxisome proliferator-activated receptor-gamma (PPARγ) in gonadal white adipose tissue was accompanied by distinct alterations in hypothalamic gene expression profiles in R6/2 females after mutant HTT overexpression. No significant effect on metabolic phenotype in R6/2 was seen in response to wild-type HTT overexpression. CONCLUSIONS: Taken together, our findings provide further support for the role of HTT in metabolic control via hypothalamic neurocircuits. Understanding the specific central neurocircuits and their peripheral link underlying metabolic imbalance in HD may open up avenues for novel therapeutic interventions.


Asunto(s)
Enfermedad de Huntington , Animales , Modelos Animales de Enfermedad , Femenino , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Hipotálamo/metabolismo , Ratones , Ratones Transgénicos , Fenotipo
2.
Neuropathol Appl Neurobiol ; 47(7): 979-989, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33755993

RESUMEN

AIMS: To determine the underlying cellular changes and clinical correlates associated with pathology of the hypothalamus in amyotrophic lateral sclerosis (ALS), as hypothalamic atrophy occurs in the preclinical phase of the disease. METHODS: The hypothalamus was pathologically examined in nine patients with amyotrophic lateral sclerosis in comparison to eight healthy control subjects. The severity of regional atrophy (paraventricular nucleus: PVN, fornix and total hypothalamus) and peptidergic neuronal loss (oxytocin, vasopressin, cocaine- and amphetamine-regulating transcript: CART, and orexin) was correlated with changes in eating behaviour, sleep function, cognition, behaviour and disease progression. RESULTS: Tar DNA-binding protein 43 (TDP-43) inclusions were present in the hypothalamus of all patients with amyotrophic lateral sclerosis. When compared to controls, there was atrophy of the hypothalamus (average 21% atrophy, p = 0.004), PVN (average 30% atrophy p = 0.014) and a loss of paraventricular oxytocin-producing neurons (average 49% loss p = 0.02) and lateral hypothalamic orexin-producing neurons (average 37% loss, significance p = 0.02). Factor analysis identified strong relationships between abnormal eating behaviour, hypothalamic atrophy and loss of orexin-producing neurons. With increasing disease progression, abnormal sleep behaviour and cognition associated with atrophy of the fornix. CONCLUSIONS: Substantial loss of hypothalamic oxytocin-producing neurons occurs in ALS, with regional atrophy and the loss of orexin neurons relating to abnormal eating behaviour in ALS. Oxytocin- and orexin neurons display TDP43 inclusions. Our study points to significant pathology in the hypothalamus that may play a key role in metabolic and pathogenic changes in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Hipotálamo/metabolismo , Orexinas/metabolismo , Oxitocina/metabolismo , Sueño/fisiología , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/patología , Atrofia/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/patología
3.
J Huntingtons Dis ; 8(4): 375-391, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31594240

RESUMEN

Huntington's disease (HD) is a fatal genetic neurodegenerative disorder. It has mainly been considered a movement disorder with cognitive symptoms and these features have been associated with pathology of the striatum and cerebral cortex. Importantly, individuals with the mutant huntingtin gene suffer from a spectrum of non-motor features often decades before the motor disorder manifests. These symptoms and signs include a range of psychiatric symptoms, sleep problems and metabolic changes with weight loss particularly in later stages. A higher body mass index at diagnosis is associated with slower disease progression. The common psychiatric symptom of apathy progresses with the disease. The fact that non-motor features are present early in the disease and that they show an association to disease progression suggest that unravelling the underlying neurobiological mechanisms may uncover novel targets for early disease intervention and better symptomatic treatment. The hypothalamus and the limbic system are important brain regions that regulate emotion, social cognition, sleep and metabolism. A number of studies using neuroimaging, postmortem human tissue and genetic manipulation in animal models of the disease has collectively shown that the hypothalamus and the limbic system are affected in HD. These findings include the loss of neuropeptide-expressing neurons such as orexin (hypocretin), oxytocin, vasopressin, somatostatin and VIP, and increased levels of SIRT1 in distinct nuclei of the hypothalamus. This review provides a summary of the results obtained so far and highlights the potential importance of these changes for the understanding of non-motor features in HD.


Asunto(s)
Síntomas Conductuales , Enfermedad de Huntington , Hipotálamo , Enfermedades Metabólicas , Animales , Síntomas Conductuales/etiología , Síntomas Conductuales/fisiopatología , Humanos , Enfermedad de Huntington/complicaciones , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Hipotálamo/metabolismo , Hipotálamo/patología , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo
4.
J Neurol Neurosurg Psychiatry ; 88(12): 1033-1041, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28596251

RESUMEN

OBJECTIVE: Our objective was to study the hypothalamic volume in a cohort of patients with amyotrophic lateral sclerosis (ALS) including symptomatic and presymptomatic ALS mutation carriers. METHODS: High-resolution three-dimensional T1-weighted MRI datasets from 251 patients with sporadic ALS, 19 symptomatic and 32 presymptomatic ALS mutation carriers and 112 healthy controls (HC) were retrospectivally registered for manual delineation of the hypothalamus. The volume of the hypothalamus, in total or subdivided, was normalised to the intracranial volume and adjusted to age. Correlation analyses were performed with clinical and metabolic outcomes. Pathologically defined ALS stages were determined in vivo by diffusion tensor imaging (DTI). RESULTS: We observed a severe atrophy of the hypothalamus both in patients with sporadic ALS (-21.8%, p<0.0001) and symptomatic ALS mutation carriers (-13.4%, p<0.001). The atrophy in patients with sporadic ALS was observed in both the anterior (-27.6% p<0.0001) and the posterior parts of the hypothalamus (-17.7%, p<0.0001). Notably, this atrophy was also observed in presymptomatic ALS mutation carriers (-15.5%, p<0.001) and was unrelated to whole brain volume atrophy or disease stage as assessed using DTI or functional status. Hypothalamic volume was correlated with body mass index (BMI) in patients with sporadic ALS (p=0.0434, ρ=+0.1579), and this correlation was much stronger in patients with familial ALS (fALS) (p=0.0060, ρ=+0.6053). Anterior hypothalamic volume was correlated with age at onset, but not with survival after MRI. CONCLUSIONS: Hypothalamus is atrophied in ALS, even in premorbid stages, and correlates with BMI, especially in fALS. Decreased anterior hypothalamic volume is associated with earlier onset of disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Hipotálamo/patología , Adolescente , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/diagnóstico por imagen , Atrofia , Índice de Masa Corporal , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Estudios de Cohortes , Imagen de Difusión Tensora , Femenino , Humanos , Hipotálamo/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Clin Endocrinol (Oxf) ; 87(4): 359-366, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28502079

RESUMEN

OBJECTIVE: Metabolic complications are frequent in childhood leukaemia (ALL) survivors treated with cranial radiotherapy (CRT). These complications are potentially mediated by damage to the hypothalamus (HT), as childhood onset (CO) craniopharyngioma (CP) survivors without HT involvement are spared overt obesity. Diffusion tensor imaging (DTI) shows brain tissue microstructure alterations, by fractional anisotrophy (FA), mean diffusivity (MD), axial diffusivity (AD) and radial diffusivity (RD). We used DTI to determine the integrity of the microstructure of the HT in ALL survivors. DESIGN: Case-control study. PATIENTS: Three groups were included: (i) 27 CRT treated ALL survivors on hormone supplementation, (ii) 17 CO-CP survivors on hormone supplementation but without HT involvement and (iii) 27 matched controls. MEASUREMENTS: DTI parameters of the HT were measured and body composition. RESULTS: Microstructural alterations in the HT were more severe in ALL survivors with a BMI ≥25 than with BMI <25. Compared to controls, ALL survivors had reduced FA (P=.04), increased MD (P<.001), AD (P<.001) and RD (P<.001) in the right and left HT. In the right HT, ALL survivors with a BMI ≥25 showed elevated MD (P=.03) and AD (P=.02) compared to ALL survivors with BMI <25. In contrast, DTI parameters did not differ between CP survivors and controls. CONCLUSIONS: Long-term follow-up after CRT for ALL DTI measures were affected in the HT despite complete hormone replacement. The present data suggest that ALL survivors have demyelination and axonal loss in the HT.


Asunto(s)
Encéfalo/patología , Craneofaringioma/patología , Imagen de Difusión Tensora/métodos , Hipotálamo/patología , Leucemia/patología , Adulto , Composición Corporal/fisiología , Índice de Masa Corporal , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
6.
J Neurol Sci ; 364: 148-53, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-27084236

RESUMEN

Subjective reports of sleep disturbance are a common feature of Huntington's disease (HD); however, there is limited research investigating the relationship between sleep problems with changes in brain and behaviour. This study aimed to investigate whether subjective reports of sleep problems in HD are associated with brain volume, neurocognitive decline, and neuropsychiatric symptoms. This retrospective pilot study used brain volume, neurocognitive and neuropsychiatric data from premanifest (pre-HD) and symptomatic HD (symp-HD). Subjective sleep problem was measured using the sleep item of the Beck's Depression Inventory-II (BDI-II). Pre-HD individuals reporting sleep problems had significantly poorer neuropsychiatric outcomes compared to those not reporting sleep problems. In the symp-HD group, those with sleep problems had significantly accelerated thalamic degeneration and poorer neuropsychiatric outcomes compared to those without sleep problems. There was no relationship between subjective sleep problems and neurocognitive measures. These findings suggest an association between subjective sleep disturbance, neuropathology, and development of neuropsychiatric symptoms in HD. Further studies using quantitative EEG-based monitoring of sleep in HD and changes in the brain and behaviour will be necessary to establish the causal nature of this relationship.


Asunto(s)
Encéfalo/diagnóstico por imagen , Trastornos del Conocimiento/etiología , Enfermedad de Huntington/complicaciones , Trastornos del Sueño-Vigilia/etiología , Adulto , Trastornos del Conocimiento/diagnóstico por imagen , Estudios Transversales , Femenino , Humanos , Enfermedad de Huntington/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Proyectos Piloto , Escalas de Valoración Psiquiátrica , Estudios Retrospectivos , Trastornos del Sueño-Vigilia/diagnóstico por imagen , Tálamo/diagnóstico por imagen
7.
PLoS One ; 11(1): e0147575, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26824435

RESUMEN

Metabolic complications are prevalent in individuals treated with cranial radiotherapy (CRT) for childhood acute lymphoblastic leukemia (ALL). The hypothalamus is a master regulator of endocrine and metabolic control. The aim of this study was to investigate whether the hypothalamic volume would be associated to metabolic parameters in ALL survivors. Thirty-eight (21 women) survivors participated in this study 34 years after diagnosis and with a median age of 38 (27-46) years. All were treated with a median CRT dose of 24 Gy and 11 years (3-13) of complete hormone supplementation. Comparisons were made to 31 matched controls. We performed analyses of fat mass, fat free mass, plasma (p)-glucose, p-insulin, Homa-Index (a measure of insulin resistance), serum (s)-leptin, s-ghrelin and of the hypothalamic volume in scans obtained by magnetic resonance imaging (MRI) at 3 Tesla. Serum leptin/kg fat mass (r = -0.4, P = 0.04) and fat mass (r = -0.4, P = 0.01) were negatively correlated with the HT volume among ALL survivors, but not among controls. We also detected significantly higher BMI, waist, fat mass, p-insulin, Homa-Index, leptin/kg fat mass and s-ghrelin and significantly lower fat free mass specifically among female ALL survivors (all P<0.01). Interestingly, s-ghrelin levels increased with time since diagnosis and with low age at diagnosis for childhood ALL. Our results showed that leptin/kg fat mass and fat mass were associated with a reduced HT volume 34 years after ALL diagnosis and that women treated with CRT after ALL are at high risk of metabolic abnormalities. Taken together our data suggest that the hypothalamus is involved in the metabolic consequences after CRT in ALL survivors.


Asunto(s)
Irradiación Craneana , Terapia de Reemplazo de Hormonas , Hipotálamo/metabolismo , Hipotálamo/efectos de la radiación , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Sobrevivientes , Adulto , Glucemia/metabolismo , Composición Corporal , Índice de Masa Corporal , Estudios de Casos y Controles , Femenino , Ghrelina/sangre , Hormona del Crecimiento/uso terapéutico , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/patología , Insulina/sangre , Resistencia a la Insulina , Leptina/sangre , Estudios Longitudinales , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/radioterapia , Riesgo
8.
Sci Rep ; 5: 14598, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26419281

RESUMEN

Expression of mutant huntingtin (htt) protein has been shown to cause metabolic imbalance in animal models of Huntington disease (HD). The pathways involved are not fully understood but dysfunction of both the hypothalamus and brown adipose tissue (BAT) has been implicated. Here we show that targeted expression of mutant HTT in the hypothalamus leads to loss of the A13 dopaminergic cell group located in the zona incerta and reduced mRNA expression of neuropeptide Y1 receptor in the hypothalamus. Furthermore, this is accompanied by downregulation of uncoupling protein 1 expression and PPARγ coactivator-1 alpha in BAT and a rapid body weight gain. Taken together, our data might provide a mechanistic link between expression of mutant HTT, reduced activity of a hypothalamic dopaminergic pathway and dysfunction of BAT and in part explain the development of an obese phenotype in HD mouse models.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Expresión Génica , Hipotálamo/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Animales , Recuento de Células , Dependovirus/genética , Neuronas Dopaminérgicas/metabolismo , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Proteína Huntingtina , Hipotálamo/patología , Inmunohistoquímica , Ratones , Neuronas/metabolismo , Fenotipo , Factores de Tiempo , Transgenes , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
10.
PLoS One ; 10(2): e0117593, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25659157

RESUMEN

Huntington disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin gene. Non-motor symptoms and signs such as psychiatric disturbances, sleep problems and metabolic dysfunction are part of the disease manifestation. These aspects may relate to changes in the hypothalamus, an area of the brain involved in the regulation of emotion, sleep and metabolism. Neuropathological and imaging studies using both voxel-based morphometry (VBM) of magnetic resonance imaging (MRI) as well as positron emission tomography (PET) have demonstrated pathological changes in the hypothalamic region during early stages in symptomatic HD. In this investigation, we aimed to establish a robust method for measurements of the hypothalamic volume in MRI in order to determine whether the hypothalamic dysfunction in HD is associated with the volume of this region. Using T1-weighted imaging, we describe a reproducible delineation procedure to estimate the hypothalamic volume which was based on the same landmarks used in histologically processed postmortem hypothalamic tissue. Participants included 36 prodromal HD (pre-HD), 33 symptomatic HD (symp-HD) and 33 control participants who underwent MRI scanning at baseline and 18 months follow-up as part of the IMAGE-HD study. We found no evidence of cross-sectional or longitudinal changes between groups in hypothalamic volume. Our results suggest that hypothalamic pathology in HD is not associated with volume changes.


Asunto(s)
Enfermedad de Huntington/diagnóstico por imagen , Hipotálamo/diagnóstico por imagen , Tomografía de Emisión de Positrones , Adulto , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Radiografía
11.
PLoS One ; 8(12): e83050, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376631

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expanded polyglutamine repeat in the huntingtin protein. Neuropathology in the basal ganglia and in the cerebral cortex has been linked to the motor and cognitive symptoms whereas recent work has suggested that the hypothalamus might be involved in the metabolic dysfunction. Several mouse models of HD that display metabolic dysfunction have hypothalamic pathology, and expression of mutant huntingtin in the hypothalamus has been causally linked to the development of metabolic dysfunction in mice. Although the pathogenic mechanisms by which mutant huntingtin exerts its toxic functions in the HD brain are not fully known, several studies have implicated a role for the lysososomal degradation pathway of autophagy. Interestingly, changes in autophagy in the hypothalamus have been associated with the development of metabolic dysfunction in wild-type mice. We hypothesized that expression of mutant huntingtin might lead to changes in the autophagy pathway in the hypothalamus in mice with metabolic dysfunction. We therefore investigated whether there were changes in basal levels of autophagy in a mouse model expressing a fragment of 853 amino acids of mutant huntingtin selectively in the hypothalamus using a recombinant adeno-associate viral vector approach as well as in the transgenic BACHD mice. We performed qRT-PCR and Western blot to investigate the mRNA and protein expression levels of selected autophagy markers. Our results show that basal levels of autophagy are maintained in the hypothalamus despite the presence of metabolic dysfunction in both mouse models. Furthermore, although there were no major changes in autophagy in the striatum and cortex of BACHD mice, we detected modest, but significant differences in levels of some markers in mice at 12 months of age. Taken together, our results indicate that overexpression of mutant huntingtin in mice do not significantly perturb basal levels of autophagy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Autofagia/genética , Enfermedad de Huntington/genética , Hipotálamo/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Adenoviridae/genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Vectores Genéticos , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Hipotálamo/patología , Lisosomas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
12.
Hum Mol Genet ; 22(17): 3485-97, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23697793

RESUMEN

Psychiatric symptoms such as depression and anxiety are important clinical features of Huntington's disease (HD). However, the underlying neurobiological substrate for the psychiatric features is not fully understood. In order to explore the biological origin of depression and anxiety in HD, we used a mouse model that expresses the human full-length mutant huntingtin, the BACHD mouse. We found that the BACHD mice displayed depressive- and anxiety-like features as early as at 2 months of age as assessed using the Porsolt forced swim test (FST), the sucrose preference test and the elevated plus maze (EPM). BACHD mice subjected to chronic treatment with the anti-depressant sertraline were not different to vehicle-treated BACHD mice in the FST and EPM. The behavioral manifestations occurred in the absence of reduced hippocampal cell proliferation/neurogenesis or upregulation of the hypothalamic-pituitary-adrenal axis. However, alterations in anxiety- and depression-regulating genes were present in the hypothalamus of BACHD mice including reduced mRNA expression of neuropeptide Y, tachykinin receptor 3 and vesicular monoamine transporter type 2 as well as increased expression of cocaine and amphetamine regulated transcript. Interestingly, the orexin neuronal population in the hypothalamus was increased and showed cellular atrophy in old BACHD mice. Furthermore, inactivation of mutant huntingtin in a subset of the hypothalamic neurons prevented the development of the depressive features. Taken together, our data demonstrate that the BACHD mouse recapitulates clinical HD with early psychiatric aspects and point to the role of hypothalamic dysfunction in the development of depression and anxiety in the disease.


Asunto(s)
Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/genética , Ansiedad/fisiopatología , Conducta Animal , Depresión/tratamiento farmacológico , Depresión/genética , Depresión/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/fisiopatología , Hipotálamo/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Neuropéptido Y/genética , Receptores de Taquicininas/genética , Sertralina/uso terapéutico , Proteínas de Transporte Vesicular de Monoaminas/genética
13.
Eur J Neurosci ; 36(6): 2789-800, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22731249

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin (htt) gene. Neuropathology is most severe in the striatum and cerebral cortex. As mutant htt is ubiquitously expressed, it has not been possible to establish clear structure-to-function relationships for the clinical aspects. In the present study, we have injected recombinant adeno-associated viral vectors of serotype 5 (rAAV5) expressing an 853-amino-acid fragment of htt with either 79 (mutant) or 18 (wild-type) glutamines (Q) in the dorsal striatum of neonatal rats to achieve expression of htt in the forebrain. Rats were followed for 6 months and compared with control rats. Neuropathological assessment showed long-term expression of the green fluorescent protein (GFP) transgene (used as a marker protein) and accumulation of htt inclusions in the cerebral cortex with the rAAV5-htt-79Q vectors. We estimated that around 10% of NeuN-positive cells in the cerebral cortex and 2% of DARPP-32 neurons in the striatum were targeted with the GFP-expressing vector. Formation of intracellular htt inclusions was not associated with neuronal loss, gliosis or microglia activation and did not lead to altered motor activity or changes in body weight. However, the same mutant htt vector caused orexin loss in the hypothalamus - another area known to be affected in HD. In conclusion, our results demonstrate that widespread forebrain expression of mutant htt can be achieved using rAAV5-vectors and suggest that this technique can be further explored to study region-specific effects of mutant htt or other disease-causing genes in the brain.


Asunto(s)
Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Prosencéfalo/patología , Ratas , Animales , Peso Corporal , Factor Neurotrófico Derivado del Encéfalo/análisis , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Glutamina/genética , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Hipotálamo/química , Cuerpos de Inclusión/patología , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Actividad Motora , Mutación , Proteínas del Tejido Nervioso/análisis , Neuropéptidos/análisis , Neuropéptidos/genética , Proteínas Nucleares/análisis , Orexinas , Prosencéfalo/química , Prosencéfalo/fisiopatología , Ratas Sprague-Dawley
14.
J Huntingtons Dis ; 1(1): 5-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-25063186

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin gene. Today, the clinical diagnosis of the disease requires unequivocal signs of typical motor disturbances, which is thought to be due to pathology in the striatum of the basal ganglia. Increasing numbers of studies have emphasized that also non-motor symptoms and signs are common and occur early in HD. These include psychiatric disturbances and cognitive impairment as well as sleep disturbances with disrupted circadian rhythm, autonomic dysfunction and metabolic changes. Several of the non-motor features may be results of dysfunction of the hypothalamus and the limbic system, which are interconnected structures central in the regulation of emotion, sleep and metabolism. In fact, recent studies using postmortem tissue, magnetic resonance imaging and positron emission tomography have shown that hypothalamic and limbic system changes occur early in clinical HD. This review summarizes the current state of knowledge in this area based on clinical studies as well as experiments in animal models of the disease and establishes that hypothalamic and limbic system changes are part of the HD pathology.


Asunto(s)
Enfermedad de Huntington/fisiopatología , Hipotálamo/fisiopatología , Sistema Límbico/fisiopatología , Animales , Investigación Biomédica , Humanos , Ratones , Ratones Transgénicos
15.
Neuropeptides ; 45(6): 401-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21871662

RESUMEN

The Flinders Sensitive Line (FSL) rat is a genetic animal model of depression that displays characteristics similar to those of depressed patients including lower body weight, decreased appetite and reduced REM sleep latency. Hypothalamic neuropeptides such as orexin/hypocretin, melanin-concentrating hormone (MCH) and cocaine and amphetamine regulated transcript (CART), that are involved in the regulation of both energy metabolism and sleep, have recently been implicated also in depression. We therefore hypothesized that alterations in these neuropeptide systems may play a role in the development of the FSL phenotype with both depressive like behavior, metabolic abnormalities and sleep disturbances. In this study, we first confirmed that the FSL rats displayed increased immobility in the Porsolt forced swim test compared to their control strain, the Flinders Resistant Line (FRL), which is indicative of depressive-like behavior. We then examined the number of orexin-, MCH- and CART-immunopositive neurons in the hypothalamus using stereological analyses. We found that the total number of orexin-positive neurons was higher in the hypothalamus of female FSL rats compared to female FRL rats, whereas no changes in the MCH or CART populations could be detected between the strains. Chronic treatment with the selective serotonin reuptake inhibitor (SSRI) escitalopram reduced immobility only in the FRL rats where it also increased the number of MCH positive neurons compared to untreated rats. These findings support the view that orexin may be involved in depression and strengthen the notion that the "depressed" brain responds differently to pharmacological interventions than the normal brain.


Asunto(s)
Depresión/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Animales , Antidepresivos de Segunda Generación/farmacología , Antidepresivos de Segunda Generación/uso terapéutico , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Citalopram/farmacología , Citalopram/uso terapéutico , Depresión/tratamiento farmacológico , Depresión/fisiopatología , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Femenino , Humanos , Hormonas Hipotalámicas/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Melaninas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Pruebas Neuropsicológicas , Orexinas , Hormonas Hipofisarias/metabolismo , Ratas , Ratas Endogámicas
16.
Cell Metab ; 13(4): 428-439, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21459327

RESUMEN

In Huntington's disease (HD), the mutant huntingtin protein is ubiquitously expressed. The disease was considered to be limited to the basal ganglia, but recent studies have suggested a more widespread pathology involving hypothalamic dysfunction. Here we tested the hypothesis that expression of mutant huntingtin in the hypothalamus causes metabolic abnormalities. First, we showed that bacterial artificial chromosome-mediated transgenic HD (BACHD) mice developed impaired glucose metabolism and pronounced insulin and leptin resistance. Selective hypothalamic expression of a short fragment of mutant huntingtin using adeno-associated viral vectors was sufficient to recapitulate these metabolic disturbances. Finally, selective hypothalamic inactivation of the mutant gene prevented the development of the metabolic phenotype in BACHD mice. Our findings establish a causal link between mutant huntingtin expression in the hypothalamus and metabolic dysfunction, and indicate that metabolic parameters are powerful readouts to assess therapies aimed at correcting dysfunction in HD by silencing huntingtin expression in the brain.


Asunto(s)
Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Femenino , Regulación de la Expresión Génica , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Hipotálamo/fisiopatología , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , Proteínas Nucleares/genética , Orexinas , Fenotipo
17.
Ann Neurol ; 69(2): 312-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21387376

RESUMEN

OBJECTIVE: Behavioral-variant frontotemporal dementia (bvFTD) is a progressive neurodegenerative brain disorder, clinically characterized by changes in cognition, personality, and behavior. Marked disturbances in eating behavior, such as overeating and preference for sweet foods, are also commonly reported. The hypothalamus plays a critical role in feeding regulation, yet the relation between pathology in this region and eating behavior in FTD is unknown. This study aimed to address this issue using 2 complementary approaches. METHODS: First, 18 early stage bvFTD patients and 16 healthy controls underwent high-resolution structural magnetic resonance imaging and assessment of eating behavior. Hypothalamic volumes were traced manually on coronal images. Second, postmortem analyses of 12 bvFTD cases and 6 matched controls were performed. Fixed hypothalamic tissue sections were stained for a cell marker and for peptides regulating feeding behaviors using immunohistochemistry. Stereological estimates of the hypothalamic volume and the number of neurons and glia were performed. RESULTS: Significant atrophy of the hypothalamus in bvFTD was present in both analyses. Patients with high feeding disturbance exhibited significant atrophy of the posterior hypothalamus. Neuronal loss, which was observed only in bvFTD cases with Tar DNA protein-43 deposition, was also predominant posteriorly. In contrast, orexin (hypocretin), neuropeptide Y, cocaine- and amphetamine-regulating transcript, and vasopressin-containing neurons that regulate appetite were spared in posterior nuclei known to participate in feeding regulation. INTERPRETATION: Degeneration and consequent dysregulation within the hypothalamus relates to significant feeding disturbance in bvFTD. These findings provide a basis for the development of therapeutic models.


Asunto(s)
Conducta Alimentaria/fisiología , Demencia Frontotemporal/patología , Hipotálamo/patología , Anciano , Apetito/fisiología , Atrofia/patología , Recuento de Células , Femenino , Preferencias Alimentarias/fisiología , Demencia Frontotemporal/fisiopatología , Humanos , Hipotálamo/fisiopatología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Neuroglía/patología , Neuronas/patología , Tamaño de los Órganos
18.
Acta Neuropathol ; 120(6): 777-88, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20821223

RESUMEN

Huntington disease (HD) is a fatal neurodegenerative disorder caused by expansion of a CAG repeat in the HD gene. Degeneration concentrating in the basal ganglia has been thought to account for the characteristic psychiatric symptoms, cognitive decline and motor dysfunction. However, the homeostatic control of emotions and metabolism are disturbed early in HD, and focused studies have identified a loss of orexin (hypocretin) neurons in the lateral hypothalamus in HD patients. There has been limited assessment of other hypothalamic cell populations that may be involved. In this study, we quantified the neuropeptide-expressing hypothalamic neurons known to regulate metabolism and emotion in patients with HD compared to healthy controls using unbiased stereological methods. We confirmed the loss of orexin-expressing neurons in HD and revealed substantial differences in the peptide expression of other neuronal populations in the same patients. Both oxytocin- and vasopressin-expressing neurons were decreased by 45 and 24%, respectively, while the number of cocaine- and amphetamine-regulated transcript (CART)-expressing neurons was increased by 30%. The increased expression of CART in the hypothalamus is consistent with a previous study showing increased CART levels in cerebrospinal fluid from HD patients. There was no difference in the numbers of neuropeptide Y-expressing neurons. These results show significant and specific alterations in the peptide expression of hypothalamic neurons known to regulate metabolism and emotion. They may be important in the development of psychiatric symptoms and metabolic disturbances in HD, and may provide potential targets for therapeutic interventions.


Asunto(s)
Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Hipotálamo/metabolismo , Hipotálamo/patología , Neuropéptidos/metabolismo , Humanos , Enfermedad de Huntington/diagnóstico , Degeneración Nerviosa/diagnóstico , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología
19.
Neurobiol Dis ; 40(3): 531-43, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20682340

RESUMEN

Huntington disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG repeat. Its length can be used to estimate the time of clinical diagnosis, which is defined by overt motor symptoms. Non-motor symptoms begin before motor onset, and involve changes in hypothalamus-regulated functions such as sleep, emotion and metabolism. Therefore we hypothesized that hypothalamic changes occur already prior to the clinical diagnosis. We performed voxel-based morphometry and logistic regression analyses of cross-sectional MR images from 220 HD gene carriers and 75 controls in the Predict-HD study. We show that changes in the hypothalamic region are detectable before clinical diagnosis and that its grey matter contents alone are sufficient to distinguish HD gene carriers from control cases. In conclusion, our study shows, for the first time, that alterations in grey matter contents in the hypothalamic region occur at least a decade before clinical diagnosis in HD using MRI.


Asunto(s)
Enfermedad de Huntington/patología , Hipotálamo/patología , Adulto , Diagnóstico Precoz , Femenino , Heterocigoto , Humanos , Enfermedad de Huntington/genética , Estudios Longitudinales , Imagen por Resonancia Magnética , Masculino
20.
Behav Brain Res ; 205(1): 175-82, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-19573560

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the HD gene. Besides psychiatric, motor and cognitive symptoms, HD patients suffer from sleep disturbances. In order to screen a rat model transgenic for HD (tgHD rats) for sleep-wake cycle dysregulation, we monitored their circadian activity peaks in the present study. TgHD rats of both sexes showed hyperactivity during the dark cycle and more frequent light cycle activity peaks indicative for a disturbed sleep-wake cycle. Focusing on males at the age of 4 and 14 months, analyses of receptor levels in the hypothalamus and the basal forebrain revealed that 5-HT(2A)- and adrenergic alpha(2)-receptor densities in these regions were significantly altered in tgHD rats compared to their wild-type littermates. Adrenergic receptor densities correlated negatively with the light cycle hyperactivity peaks at later stages of the disease in male tgHD rats. Furthermore, reduced leptin levels, a feature associated with circadian misalignment, were present. Our study demonstrates that the male tgHD rat is a suitable model to investigate HD associated sleep alterations. Further studies are warranted to elucidate the role of adrenergic- and 5-HT(2A)-receptors as therapeutic targets for dysregulation of the circadian activity in HD.


Asunto(s)
Encéfalo/fisiopatología , Enfermedad de Huntington/fisiopatología , Parasomnias/fisiopatología , Fotoperiodo , Receptores Adrenérgicos alfa 2/metabolismo , Animales , Colina O-Acetiltransferasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipotálamo/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leptina/metabolismo , Masculino , Actividad Motora/fisiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Orexinas , Prosencéfalo/fisiopatología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Receptor de Serotonina 5-HT2A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA