Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Neurosci ; 27(5): 862-872, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38528203

RESUMEN

The mammalian telencephalon contains distinct GABAergic projection neuron and interneuron types, originating in the germinal zone of the embryonic basal ganglia. How genetic information in the germinal zone determines cell types is unclear. Here we use a combination of in vivo CRISPR perturbation, lineage tracing and ChIP-sequencing analyses and show that the transcription factor MEIS2 favors the development of projection neurons by binding enhancer regions in projection-neuron-specific genes during mouse embryonic development. MEIS2 requires the presence of the homeodomain transcription factor DLX5 to direct its functional activity toward the appropriate binding sites. In interneuron precursors, the transcription factor LHX6 represses the MEIS2-DLX5-dependent activation of projection-neuron-specific enhancers. Mutations of Meis2 result in decreased activation of regulatory enhancers, affecting GABAergic differentiation. We propose a differential binding model where the binding of transcription factors at cis-regulatory elements determines differential gene expression programs regulating cell fate specification in the mouse ganglionic eminence.


Asunto(s)
Desarrollo Embrionario , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Factores de Transcripción , Animales , Ratones , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Desarrollo Embrionario/fisiología , Elementos de Facilitación Genéticos/genética , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Diferenciación Celular/fisiología , Interneuronas/metabolismo , Interneuronas/fisiología , Proteínas con Homeodominio LIM/metabolismo , Proteínas con Homeodominio LIM/genética , Neurogénesis/fisiología , Proteínas del Tejido Nervioso
2.
Sleep ; 47(5)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38314840

RESUMEN

Restless legs syndrome (RLS) is a neurological disorder characterized by uncomfortable or unpleasant sensations in the legs during rest periods. To relieve these sensations, patients move their legs, causing sleep disruption. While the pathogenesis of RLS has yet to be resolved, there is a strong genetic association with the MEIS1 gene. A missense variant in MEIS1 is enriched sevenfold in people with RLS compared to non-affected individuals. We generated a mouse line carrying this mutation (p.Arg272His/c.815G>A), referred to herein as Meis1R272H/R272H (Meis1 point mutation), to determine whether it would phenotypically resemble RLS. As women are more prone to RLS, driven partly by an increased risk of developing RLS during pregnancy, we focused on female homozygous mice. We evaluated RLS-related outcomes, particularly sensorimotor behavior and sleep, in young and aged mice. Compared to noncarrier littermates, homozygous mice displayed very few differences. Significant hyperactivity occurred before the lights-on (rest) period in aged female mice, reflecting the age-dependent incidence of RLS. Sensory experiments involving tactile feedback (rotarod, wheel running, and hotplate) were only marginally different. Overall, RLS-like phenomena were not recapitulated except for the increased wake activity prior to rest. This is likely due to the focus on young mice. Nevertheless, the Meis1R272H mouse line is a potentially useful RLS model, carrying a clinically relevant variant and showing an age-dependent phenotype.


Asunto(s)
Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Fenotipo , Síndrome de las Piernas Inquietas , Animales , Síndrome de las Piernas Inquietas/genética , Síndrome de las Piernas Inquietas/fisiopatología , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Ratones , Femenino , Modelos Animales de Enfermedad , Masculino , Proteínas de Homeodominio/genética , Mutación Missense/genética , Humanos , Proteínas de Neoplasias/genética , Mutación Puntual/genética , Ratones Endogámicos C57BL , Factores de Edad , Sueño/genética , Sueño/fisiología
3.
J Neurosci ; 42(8): 1557-1573, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34965974

RESUMEN

Collagen VI is a key component of muscle basement membranes, and genetic variants can cause monogenic muscular dystrophies. Conversely, human genetic studies recently implicated collagen VI in central nervous system function, with variants causing the movement disorder dystonia. To elucidate the neurophysiological role of collagen VI, we generated mice with a truncation of the dystonia-related collagen α3 VI (COL6A3) C-terminal domain (CTD). These Col6a3CTT mice showed a recessive dystonia-like phenotype in both sexes. We found that COL6A3 interacts with the cannabinoid receptor 1 (CB1R) complex in a CTD-dependent manner. Col6a3CTT mice of both sexes have impaired homeostasis of excitatory input to the basal pontine nuclei (BPN), a motor control hub with dense COL6A3 expression, consistent with deficient endocannabinoid (eCB) signaling. Aberrant synaptic input in the BPN was normalized by a CB1R agonist, and motor performance in Col6a3CTT mice of both sexes was improved by CB1R agonist treatment. Our findings identify a readily therapeutically addressable synaptic mechanism for motor control.SIGNIFICANCE STATEMENT Dystonia is a movement disorder characterized by involuntary movements. We previously identified genetic variants affecting a specific domain of the COL6A3 protein as a cause of dystonia. Here, we created mice lacking the affected domain and observed an analogous movement disorder. Using a protein interaction screen, we found that the affected COL6A3 domain mediates an interaction with the cannabinoid receptor 1 (CB1R). Concordantly, our COL6A3-deficient mice showed a deficit in synaptic plasticity linked to a deficit in cannabinoid signaling. Pharmacological cannabinoid augmentation rescued the motor impairment of the mice. Thus, cannabinoid augmentation could be a promising avenue for treating dystonia, and we have identified a possible molecular mechanism mediating this.


Asunto(s)
Cannabinoides , Colágeno Tipo VI , Distonía , Trastornos Distónicos , Neuronas Motoras , Plasticidad Neuronal , Animales , Cannabinoides/metabolismo , Cannabinoides/farmacología , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Distonía/genética , Distonía/metabolismo , Trastornos Distónicos/genética , Trastornos Distónicos/metabolismo , Femenino , Masculino , Ratones , Neuronas Motoras/efectos de los fármacos , Mutación , Plasticidad Neuronal/efectos de los fármacos , Receptores de Cannabinoides/genética , Receptores de Cannabinoides/metabolismo
4.
Hum Mol Genet ; 31(11): 1733-1746, 2022 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34888668

RESUMEN

A highly evolutionarily conserved myeloid ecotropic viral integration site 1 (MEIS1) intronic region is strongly associated with restless legs syndrome (RLS) and insomnia. To understand its regulatory function, we dissected the region by analyzing chromatin accessibility, enhancer-promoter contacts, DNA methylation and expression quantitative trait locus (eQTLs) in different human neural cell types and tissues. We observed specific activity with respect to cell type and developmental maturation, indicating a prominent role for distinct highly conserved intronic elements in forebrain inhibitory neuron differentiation. Two elements were hypomethylated in neural cells with higher MEIS1 expression, suggesting a role of enhancer demethylation in gene regulation. MEIS1 eQTLs showed a striking modular chromosomal distribution, with forebrain eQTLs clustering in intron 8/9. Clustered regularly interspersed short palindromic repeats interference targeting of individual elements in this region attenuated MEIS1 expression, revealing a complex regulatory interplay of distinct elements. In summary, we found that MEIS1 regulation is organized in a modular pattern. Disease-associated intronic regulatory elements control MEIS1 expression with cell type and maturation stage specificity, particularly in the inhibitory neuron lineage. The precise spatiotemporal activity of these elements likely contributes to the pathogenesis of insomnia and RLS.


Asunto(s)
Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Síndrome de las Piernas Inquietas , Trastornos del Inicio y del Mantenimiento del Sueño , Epigénesis Genética , Humanos , Intrones/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Síndrome de las Piernas Inquietas/genética , Trastornos del Inicio y del Mantenimiento del Sueño/genética
5.
J Sleep Res ; 30(5): e13311, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34008292

RESUMEN

Restless legs syndrome (RLS) is a common neurological disorder in which sensorimotor symptoms lead to sleep disturbances with substantial impact on life quality. RLS is caused by a combination of genetic and environmental factors, and Meis homeobox 1 (MEIS1) was identified as the main genetic risk factor. The efficacy of dopaminergic agonists, including dopamine D2 receptor (DRD2) agonists, for treating RLS led to the hypothesis of dopaminergic impairment. However, it remains unclear whether it is directly involved in the disease aetiology and what the role of MEIS1 is considering its developmental and postnatal expression in the striatum, a critical structure in motor control. We addressed the role of MEIS1 in striatal dopaminergic signalling in Meis1+/- mice, a valid animal model of RLS, and in Meis1Drd2-/- mice carrying a somatic null mutation of Meis1 in Drd2+ neurones. Motor behaviours, pharmacological exploration of DRD2 signalling, and quantitative analyses of DRD2+ and DRD1+ expressing neurones were investigated. Although Meis1+/- mice displayed an RLS-like phenotype, including motor hyperactivity at the beginning of the rest phase, no reduction of dopaminoceptive neurones was observed in the striatum. Moreover, the null mutation of Meis1 in DRD2+ cells did not lead to RLS-like symptoms and dysfunction of the DRD2 pathway. These data indicate that MEIS1 does not modulate DRD2-dependent signalling in a cell-autonomous manner. Thus, the efficiency of D2 -like agonists may reflect the involvement of other dopaminergic receptors or normalisation of motor circuit abnormalities downstream from defects caused by MEIS1 dysfunction.


Asunto(s)
Síndrome de las Piernas Inquietas , Animales , Modelos Animales de Enfermedad , Dopamina , Genes Homeobox , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Síndrome de las Piernas Inquietas/genética
6.
PLoS One ; 15(12): e0244793, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33382813

RESUMEN

Enhancer redundancy has been postulated to provide a buffer for gene expression against genetic and environmental perturbations. While work in Drosophila has identified functionally overlapping enhancers, work in mammalian models has been limited. Recently, we have identified two partially redundant enhancers, nPE1 and nPE2, that drive proopiomelanocortin gene expression in the hypothalamus. Here we demonstrate that deletion of nPE1 produces mild obesity while knockout of nPE2 has no discernible metabolic phenotypes. Additionally, we show that acute leptin administration has significant effects on nPE1 knockout mice, with food intake and body weight change significantly impacted by peripheral leptin treatment. nPE1 knockout mice became less responsive to leptin treatment over time as percent body weight change increased over 2 week exposure to peripheral leptin. Both Pomc and Agrp mRNA were not differentially affected by chronic leptin treatment however we did see a decrease in Pomc and Agrp mRNA in both nPE1 and nPE2 knockout calorie restricted mice as compared to calorie restricted PBS-treated WT mice. Collectively, these data suggest dynamic regulation of Pomc by nPE1 such that mice with nPE1 knockout become less responsive to the anorectic effects of leptin treatment over time. Our results also support our earlier findings in which nPE2 may only be critical in adult mice that lack nPE1, indicating that these neural enhancers work synergistically to influence metabolism.


Asunto(s)
Depresores del Apetito/farmacología , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Elementos de Facilitación Genéticos , Hipotálamo/efectos de los fármacos , Leptina/farmacología , Neuronas/efectos de los fármacos , Proopiomelanocortina/genética , Animales , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo
7.
Curr Neurol Neurosci Rep ; 19(11): 92, 2019 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-31768667

RESUMEN

PURPOSE OF REVIEW: To summarize the molecular and clinical findings of KMT2B-related dystonia (DYT-KMT2B), a newly identified genetic dystonia syndrome. RECENT FINDINGS: Since first described in 2016, 66 different KMT2B-affecting variants, encompassing a set of frameshift, nonsense, splice-site, missense, and deletion mutations, have been reported in 76 patients. Most mutations are de novo and expected to mediate epigenetic dysregulation by inducing KMT2B haploinsufficiency. DYT-KMT2B is characterized phenotypically by limb-onset childhood dystonia that tends to spread progressively, resulting in generalized dystonia with cranio-cervical involvement. Co-occuring signs such as intellectual disability are frequently observed. Sustained response to deep brain stimulation (DBS), including restoration of independent ambulation, is seen in 93% (27/29) of patients. DYT-KMT2B is emerging as a prevalent monogenic dystonia. Childhood-onset dystonia presentations should prompt a search for KMT2B mutations, preferentially via next-generation-sequencing and genomic-array technologies, to enable specific counseling and treatment. Prospective multicenter studies are desirable to establish KMT2B mutational status as a DBS outcome predictor.


Asunto(s)
Trastornos Distónicos/diagnóstico , Trastornos Distónicos/genética , N-Metiltransferasa de Histona-Lisina/genética , Mutación/genética , Niño , Estimulación Encefálica Profunda/métodos , Estimulación Encefálica Profunda/tendencias , Distonía/diagnóstico , Distonía/genética , Distonía/terapia , Trastornos Distónicos/terapia , Pruebas Genéticas/métodos , Pruebas Genéticas/tendencias , Genómica/métodos , Genómica/tendencias , Humanos , Fenotipo , Estudios Prospectivos
8.
Adv Pharmacol ; 84: 175-184, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31229170

RESUMEN

MEIS1 is a transcription factor playing an important role in the development of several organs, including central and peripheral nervous systems. A genetic locus spanning the MEIS1 coding region has been associated with the risk of RLS in genome-wide association studies, with increasing evidence that MEIS1 is the causal RLS gene. The RLS-linked genetic signal has been mapped to an intronic regulatory element within MEIS1. This element plays a role in the ganglionic eminences of the developing forebrain, with the RLS risk allele related to a reduced activation of the enhancer. This suggests that the ganglionic eminences play an important role in the development of genetic susceptibility to RLS. In addition, rare variants within MEIS1 have been shown to contribute to the disease risk. These variants were identified first in RLS families and later found in further RLS cases by targeted sequencing. Some of these variants alone are sufficient to suppress MEIS1 function in neural development, providing further evidence of the importance of neurodevelopmental processes in the pathological mechanism of MEIS1 in RLS. Heterozygous Meis1 inactivation in mice causes hyperactivity at the onset of the inactive period, consistent with human RLS. In addition, these mice revealed an effect of MEIS1 on the dopaminergic system at both the spinal and supraspinal level. More studies are needed in human genetics to determine the exact role of MEIS1 variants in the risk of RLS, as well as in functional genetics and animal studies to further elucidate the pathological mechanism of MEIS1 in RLS.


Asunto(s)
Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Síndrome de las Piernas Inquietas/genética , Animales , Modelos Animales de Enfermedad , Dopamina/metabolismo , Humanos , Ratones
9.
Genet Med ; 21(11): 2532-2542, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31036918

RESUMEN

PURPOSE: The purpose of this study was to expand the genetic architecture of neurodevelopmental disorders, and to characterize the clinical features of a novel cohort of affected individuals with variants in ZNF142, a C2H2 domain-containing transcription factor. METHODS: Four independent research centers used exome sequencing to elucidate the genetic basis of neurodevelopmental phenotypes in four unrelated families. Following bioinformatic filtering, query of control data sets, and secondary variant confirmation, we aggregated findings using an online data sharing platform. We performed in-depth clinical phenotyping in all affected individuals. RESULTS: We identified seven affected females in four pedigrees with likely pathogenic variants in ZNF142 that segregate with recessive disease. Affected cases in three families harbor either nonsense or frameshifting likely pathogenic variants predicted to undergo nonsense mediated decay. One additional trio bears ultrarare missense variants in conserved regions of ZNF142 that are predicted to be damaging to protein function. We performed clinical comparisons across our cohort and noted consistent presence of intellectual disability and speech impairment, with variable manifestation of seizures, tremor, and dystonia. CONCLUSION: Our aggregate data support a role for ZNF142 in nervous system development and add to the emergent list of zinc finger proteins that contribute to neurocognitive disorders.


Asunto(s)
Discapacidades del Desarrollo/genética , Trastornos del Neurodesarrollo/genética , Transactivadores/genética , Adolescente , Adulto , Niño , Estudios de Cohortes , Biología Computacional/métodos , Distonía/genética , Familia , Femenino , Humanos , Discapacidad Intelectual/genética , Mutación , Mutación Missense , Linaje , Fenotipo , Convulsiones/genética , Trastornos del Habla/genética , Transactivadores/metabolismo , Secuenciación del Exoma
10.
Artículo en Inglés | MEDLINE | ID: mdl-30262571

RESUMEN

Calcium/calmodulin-dependent protein kinases (CaMKs) are key mediators of calcium signaling and underpin neuronal health. Although widely studied, the contribution of CaMKs to Mendelian disease is rather enigmatic. Here, we describe an unusual neurodevelopmental phenotype, characterized by milestone delay, intellectual disability, autism, ataxia, and mixed hyperkinetic movement disorder including severe generalized dystonia, in a proband who remained etiologically undiagnosed despite exhaustive testing. We performed trio whole-exome sequencing to identify a de novo essential splice-site variant (c.981+1G>A) in CAMK4, encoding CaMKIV. Through in silico evaluation and cDNA analyses, we demonstrated that c.981+1G>A alters CAMK4 pre-mRNA processing and results in a stable mRNA transcript containing a 77-nt out-of-frame deletion and a premature termination codon within the last exon. The expected protein, p.Lys303Serfs*28, exhibits selective loss of the carboxy-terminal regulatory domain of CaMKIV and bears striking structural resemblance to previously reported synthetic mutants that confer constitutive CaMKIV activity. Biochemical studies in proband-derived cells confirmed an activating effect of c.981+1G>A and indicated that variant-induced excessive CaMKIV signaling is sensitive to pharmacological manipulation. Additionally, we found that variants predicted to cause selective depletion of CaMKIV's regulatory domain are unobserved in diverse catalogs of human variation, thus revealing that c.981+1G>A is a unique molecular event. We propose that our proband's phenotype is explainable by a dominant CAMK4 splice-disrupting mutation that acts through a gain-of-function mechanism. Our findings highlight the importance of CAMK4 in human neurodevelopment, provide a foundation for future clinical research of CAMK4, and suggest the CaMKIV signaling pathway as a potential drug target in neurological disease.


Asunto(s)
Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/genética , Hipercinesia/genética , Discapacidad Intelectual/genética , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Ataxia Cerebelosa/genética , Codón sin Sentido/genética , Exoma , Exones/genética , Femenino , Mutación del Sistema de Lectura/genética , Mutación con Ganancia de Función/genética , Humanos , Masculino , Mutación , Linaje , Fenotipo , Empalme del ARN/genética , Secuenciación del Exoma
11.
Curr Opin Pharmacol ; 37: 100-106, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29107871

RESUMEN

Obesity results from the consumption of food in excess of bodily energy requirements, with the excess energy stored as adipose tissue. Sequelae of obesity, such as heart disease and type 2 diabetes, consistently rank among the top causes of death worldwide. The global prevalence of obesity highlights the urgency of understanding the mechanisms regulating hunger and satiety. Appetite, defined as the motivational drive to obtain food, is regulated by a complex neurocircuitry which integrates a variety of interoceptive signals to gauge nutritional state and guide appropriate levels of food-seeking. Here we review key recent developments in the identification of cell groups, neural circuits, endogenous and exogenous substances, and intracellular signaling pathways which drive hunger and satiety. We also consider particularly promising pharmacological targets for appetite modulation.


Asunto(s)
Encéfalo/fisiología , Hambre/fisiología , Saciedad/fisiología , Animales , Depresores del Apetito/farmacología , Humanos , Transducción de Señal
12.
Mol Metab ; 5(3): 245-252, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26977396

RESUMEN

OBJECTIVE: Obesity is one of the primary healthcare challenges of the 21st century. Signals relaying information regarding energy needs are integrated within the brain to influence body weight. Central among these integration nodes are the brain pro-opiomelanocortin (POMC) peptides, perturbations of which disrupt energy balance and promote severe obesity. However, POMC neurons are neurochemically diverse and the crucial source of POMC peptides that regulate energy homeostasis and body weight remains to be fully clarified. METHODS: Given that a 5-hydroxytryptamine 2c receptor (5-HT2CR) agonist is a current obesity medication and 5-HT2CR agonist's effects on appetite are primarily mediated via POMC neurons, we hypothesized that a critical source of POMC regulating food intake and body weight is specifically synthesized in cells containing 5-HT2CRs. To exclusively manipulate Pomc synthesis only within 5-HT2CR containing cells, we generated a novel 5-HT 2C R (CRE) mouse line and intercrossed it with Cre recombinase-dependent and hypothalamic specific reactivatable Pomc (NEO) mice to restrict Pomc synthesis to the subset of hypothalamic cells containing 5-HT2CRs. This provided a means to clarify the specific contribution of a defined subgroup of POMC peptides in energy balance and body weight. RESULTS: Here we transform genetically programed obese and hyperinsulinemic male mice lacking hypothalamic Pomc with increased appetite, reduced physical activity and compromised brown adipose tissue (BAT) into lean, healthy mice via targeted restoration of Pomc function only within 5-HT2CR expressing cells. Remarkably, the same metabolic transformation does not occur in females, who despite corrected feeding behavior and normalized insulin levels remain physically inactive, have lower energy expenditure, compromised BAT and develop obesity. CONCLUSIONS: These data provide support for the functional heterogeneity of hypothalamic POMC neurons, revealing that Pomc expression within 5-HT2CR expressing neurons is sufficient to regulate energy intake and insulin sensitivity in male and female mice. However, an unexpected sex difference in the function of this subset of POMC neurons was identified with regard to energy expenditure. We reveal that a large sex difference in physical activity, energy expenditure and the development of obesity is driven by this subpopulation, which constitutes approximately 40% of all POMC neurons in the hypothalamic arcuate nucleus. This may have broad implications for strategies utilized to combat obesity, which at present largely ignore the sex of the obese individual.

13.
Diabetes ; 65(3): 660-72, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26467632

RESUMEN

Hypothalamic proopiomelanocortin (POMC) is essential for the physiological regulation of energy balance; however, its role in glucose homeostasis remains less clear. We show that hypothalamic arcuate nucleus (Arc)POMC-deficient mice, which develop severe obesity and insulin resistance, unexpectedly exhibit improved glucose tolerance and remain protected from hyperglycemia. To explain these paradoxical phenotypes, we hypothesized that an insulin-independent pathway is responsible for the enhanced glucose tolerance. Indeed, the mutant mice demonstrated increased glucose effectiveness and exaggerated glycosuria relative to wild-type littermate controls at comparable blood glucose concentrations. Central administration of the melanocortin receptor agonist melanotan II in mutant mice reversed alterations in glucose tolerance and glycosuria, whereas, conversely, administration of the antagonist Agouti-related peptide (Agrp) to wild-type mice enhanced glucose tolerance. The glycosuria of ArcPOMC-deficient mice was due to decreased levels of renal GLUT 2 (rGLUT2) but not sodium-glucose cotransporter 2 and was associated with reduced renal catecholamine content. Epinephrine treatment abolished the genotype differences in glucose tolerance and rGLUT2 levels, suggesting that reduced renal sympathetic nervous system (SNS) activity is the underlying mechanism for the observed glycosuria and improved glucose tolerance in ArcPOMC-deficient mice. Therefore, the ArcPOMC-SNS-rGLUT2 axis is potentially an insulin-independent therapeutic target to control diabetes.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Glucemia/metabolismo , Transportador de Glucosa de Tipo 2/genética , Glucosuria Renal/genética , Resistencia a la Insulina/genética , Riñón/metabolismo , Obesidad/genética , Proopiomelanocortina/genética , Sistema Nervioso Simpático/metabolismo , Proteína Relacionada con Agouti/farmacología , Animales , Glucemia/efectos de los fármacos , Western Blotting , Epinefrina/metabolismo , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 2/metabolismo , Glucosuria Renal/metabolismo , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Ratones , Ratones Noqueados , Norepinefrina/metabolismo , Obesidad/metabolismo , Péptidos Cíclicos/farmacología , Proopiomelanocortina/deficiencia , Proopiomelanocortina/metabolismo , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inhibidores , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
14.
Am J Hum Genet ; 96(6): 883-93, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26004199

RESUMEN

Isolated dystonia is a disorder characterized by involuntary twisting postures arising from sustained muscle contractions. Although autosomal-dominant mutations in TOR1A, THAP1, and GNAL have been found in some cases, the molecular mechanisms underlying isolated dystonia are largely unknown. In addition, although emphasis has been placed on dominant isolated dystonia, the disorder is also transmitted as a recessive trait, for which no mutations have been defined. Using whole-exome sequencing in a recessive isolated dystonia-affected kindred, we identified disease-segregating compound heterozygous mutations in COL6A3, a collagen VI gene associated previously with muscular dystrophy. Genetic screening of a further 367 isolated dystonia subjects revealed two additional recessive pedigrees harboring compound heterozygous mutations in COL6A3. Strikingly, all affected individuals had at least one pathogenic allele in exon 41, including an exon-skipping mutation that induced an in-frame deletion. We tested the hypothesis that disruption of this exon is pathognomonic for isolated dystonia by inducing a series of in-frame deletions in zebrafish embryos. Consistent with our human genetics data, suppression of the exon 41 ortholog caused deficits in axonal outgrowth, whereas suppression of other exons phenocopied collagen deposition mutants. All recessive mutation carriers demonstrated early-onset segmental isolated dystonia without muscular disease. Finally, we show that Col6a3 is expressed in neurons, with relevant mRNA levels detectable throughout the adult mouse brain. Taken together, our data indicate that loss-of-function mutations affecting a specific region of COL6A3 cause recessive isolated dystonia with underlying neurodevelopmental deficits and highlight the brain extracellular matrix as a contributor to dystonia pathogenesis.


Asunto(s)
Colágeno Tipo VI/genética , Trastornos Distónicos/genética , Trastornos Distónicos/patología , Variación Genética , Animales , Secuencia de Bases , Biología Computacional , Análisis Mutacional de ADN , Exoma/genética , Genes Recesivos , Pruebas Genéticas , Hibridación in Situ , Imagen por Resonancia Magnética , Ratones , Datos de Secuencia Molecular , Músculo Esquelético , Mutación/genética , Linaje , Pez Cebra/genética
15.
PLoS Genet ; 11(2): e1004935, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25671638

RESUMEN

Cell-specific expression of many genes is conveyed by multiple enhancers, with each individual enhancer controlling a particular expression domain. In contrast, multiple enhancers drive similar expression patterns of some genes involved in embryonic development, suggesting regulatory redundancy. Work in Drosophila has indicated that functionally overlapping enhancers canalize development by buffering gene expression against environmental and genetic disturbances. However, little is known about regulatory redundancy in vertebrates and in genes mainly expressed during adulthood. Here we study nPE1 and nPE2, two phylogenetically conserved mammalian enhancers that drive expression of the proopiomelanocortin gene (Pomc) to the same set of hypothalamic neurons. The simultaneous deletion of both enhancers abolished Pomc expression at all ages and induced a profound metabolic dysfunction including early-onset extreme obesity. Targeted inactivation of either nPE1 or nPE2 led to very low levels of Pomc expression during early embryonic development indicating that both enhancers function synergistically. In adult mice, however, Pomc expression is controlled additively by both enhancers, with nPE1 being responsible for ∼80% and nPE2 for ∼20% of Pomc transcription. Consequently, nPE1 knockout mice exhibit mild obesity whereas nPE2-deficient mice maintain a normal body weight. These results suggest that nPE2-driven Pomc expression is compensated by nPE1 at later stages of development, essentially rescuing the earlier phenotype of nPE2 deficiency. Together, these results reveal that cooperative interactions between the enhancers confer robustness of Pomc expression against gene regulatory disturbances and preclude deleterious metabolic phenotypes caused by Pomc deficiency in adulthood. Thus, our study demonstrates that enhancer redundancy can be used by genes that control adult physiology in mammals and underlines the potential significance of regulatory sequence mutations in common diseases.


Asunto(s)
Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Proopiomelanocortina/biosíntesis , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Secuencia Conservada , Femenino , Regulación del Desarrollo de la Expresión Génica , Mamíferos/genética , Ratones , Neuronas/metabolismo , Filogenia , Embarazo , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética
16.
Endocrinology ; 156(4): 1292-302, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25594696

RESUMEN

Peptides derived from the proopiomelanocortin (POMC) precursor are critical for the normal regulation of many physiological parameters, and POMC deficiency results in severe obesity and metabolic dysfunction. Conversely, augmentation of central nervous system melanocortin function is a promising therapeutic avenue for obesity and diabetes but is confounded by detrimental cardiovascular effects including hypertension. Because the hypothalamic population of POMC-expressing neurons is neurochemically and neuroanatomically heterogeneous, there is interest in the possible dissociation of functionally distinct POMC neuron subpopulations. We used a Cre recombinase-dependent and hypothalamus-specific reactivatable PomcNEO allele to restrict Pomc expression to hypothalamic neurons expressing leptin receptor (Lepr) in mice. In contrast to mice with total hypothalamic Pomc deficiency, which are severely obese, mice with Lepr-restricted Pomc expression displayed fully normal body weight, food consumption, glucose homeostasis, and locomotor activity. Thus, Lepr+ POMC neurons, which constitute approximately two-thirds of the total POMC neuron population, are sufficient for normal regulation of these parameters. This functional dissociation approach represents a promising avenue for isolating therapeutically relevant POMC neuron subpopulations.


Asunto(s)
Metabolismo Energético/fisiología , Homeostasis/fisiología , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptores de Leptina/metabolismo , Animales , Glucemia/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Proopiomelanocortina/genética , Receptores de Leptina/genética
17.
Diabetes ; 61(2): 321-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22210318

RESUMEN

As patients decline from health to type 2 diabetes, glucose-stimulated insulin secretion (GSIS) typically becomes impaired. Although GSIS is driven predominantly by direct sensing of a rise in blood glucose by pancreatic ß-cells, there is growing evidence that hypothalamic neurons control other aspects of peripheral glucose metabolism. Here we investigated the role of the brain in the modulation of GSIS. To examine the effects of increasing or decreasing hypothalamic glucose sensing on glucose tolerance and insulin secretion, glucose or inhibitors of glucokinase, respectively, were infused into the third ventricle during intravenous glucose tolerance tests (IVGTTs). Glucose-infused rats displayed improved glucose handling, particularly within the first few minutes of the IVGTT, with a significantly lower area under the excursion curve within the first 10 min (AUC0-10). This was explained by increased insulin secretion. In contrast, infusion of the glucokinase inhibitors glucosamine or mannoheptulose worsened glucose tolerance and decreased GSIS in the first few minutes of IVGTT. Our data suggest a role for brain glucose sensors in the regulation of GSIS, particularly during the early phase. We propose that pharmacological agents targeting hypothalamic glucose-sensing pathways may represent novel therapeutic strategies for enhancing early phase insulin secretion in type 2 diabetes.


Asunto(s)
Glucosa/metabolismo , Hipotálamo/fisiología , Insulina/metabolismo , Páncreas/metabolismo , Animales , Glucoquinasa/fisiología , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Secreción de Insulina , Masculino , Manoheptulosa/farmacología , Ratas , Ratas Sprague-Dawley
18.
Cell Metab ; 13(5): 584-91, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21531340

RESUMEN

Serotonin (5-HT) and leptin play important roles in the modulation of energy balance. Here we investigated mechanisms by which leptin might interact with CNS 5-HT pathways to influence appetite. Although some leptin receptor (LepRb) neurons lie close to 5-HT neurons in the dorsal raphe (DR), 5-HT neurons do not express LepRb. Indeed, while leptin hyperpolarizes some non-5-HT DR neurons, leptin does not alter the activity of DR 5-HT neurons. Furthermore, 5-HT depletion does not impair the anorectic effects of leptin. The serotonin transporter-cre allele (Sert(cre)) is expressed in 5-HT (and developmentally in some non-5-HT) neurons. While Sert(cre) promotes LepRb excision in a few LepRb neurons in the hypothalamus, it is not active in DR LepRb neurons, and neuron-specific Sert(cre)-mediated LepRb inactivation in mice does not alter body weight or adiposity. Thus, leptin does not directly influence 5-HT neurons and does not meaningfully modulate important appetite-related determinants via 5-HT neuron function.


Asunto(s)
Apetito , Encéfalo/efectos de los fármacos , Leptina/farmacología , Neuronas/efectos de los fármacos , Receptores de Leptina/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Serotonina/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/citología , Encéfalo/metabolismo , Electrofisiología , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vías Nerviosas , Neuronas/citología , Neuronas/metabolismo , Obesidad/etiología , Obesidad/metabolismo
19.
Behav Brain Res ; 222(1): 274-8, 2011 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-21440571

RESUMEN

Maintaining glucose levels within the appropriate physiological range is necessary for survival. The identification of specific neuronal populations, within discreet brain regions, sensitive to changes in glucose concentration has led to the hypothesis of a central glucose-sensing system capable of directly modulating feeding behaviour. Glucokinase (GK) has been identified as a glucose-sensor responsible for detecting such changes both within the brain and the periphery. We previously reported that antagonism of centrally expressed GK by administration of glucosamine (GSN) was sufficient to induce protective glucoprivic feeding in rats. Here we examine a neurochemical mechanism underlying this effect and report that GSN stimulated food intake is highly correlated with the induction of the neuronal activation marker cFOS within two nuclei with a demonstrated role in central glucose sensing and appetite, the arcuate nucleus of the hypothalamus (ARC) and lateral hypothalamic area (LHA). Furthermore, GSN stimulated cFOS within the ARC was observed in orexigenic neurons expressing the endogenous melanocortin receptor antagonist agouti-related peptide (AgRP) and neuropeptide Y (NPY), but not those expressing the anorectic endogenous melanocortin receptor agonist alpha-melanocyte stimulating hormone (α-MSH). In the LHA, GSN stimulated cFOS was found within arousal and feeding associated orexin/hypocretin (ORX), but not orexigenic melanin-concentrating hormone (MCH) expressing neurons. Our data suggest that GK within these specific feeding and arousal related populations of AgRP/NPY and ORX neurons may play a modulatory role in the sensing of and appetitive response to hypoglycaemia.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucosamina/farmacología , Hipotálamo/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Neuropéptidos/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Orexinas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Estadística como Asunto
20.
Pharmacol Biochem Behav ; 97(1): 84-91, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20837046

RESUMEN

An inverse relationship between brain serotonin and food intake and body weight has been known for more than 30 years. Specifically, augmentation of brain serotonin inhibits food intake, while depletion of brain serotonin promotes hyperphagia and weight gain. Through the decades, serotonin receptors have been identified and their function in the serotonergic regulation of food intake clarified. Recent refined genetic studies now indicate that a primary mechanism through which serotonin influences appetite and body weight is via serotonin 2C receptor (5-HT(2C)R) and serotonin 1B receptor (5-HT(1B)R) influencing the activity of endogenous melanocortin receptor agonists and antagonists at the melanocortin 4 receptor (MC4R). However, other mechanisms are also possible and the challenge of future research is to delineate them in the complete elucidation of the complex neurocircuitry underlying the serotonergic control of appetite and body weight.


Asunto(s)
Encéfalo/metabolismo , Ingestión de Alimentos/fisiología , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Animales , Peso Corporal/fisiología , Ingestión de Alimentos/psicología , Conducta Alimentaria/fisiología , Conducta Alimentaria/psicología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA