Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Mol Psychiatry ; 29(4): 1192-1204, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38212372

RESUMEN

At the center of the hippocampal tri-synaptic loop are synapses formed between mossy fiber (MF) terminals from granule cells in the dentate gyrus (DG) and proximal dendrites of CA3 pyramidal neurons. However, the molecular mechanism regulating the development and function of these synapses is poorly understood. In this study, we showed that neurotrophin-3 (NT3) was expressed in nearly all mature granule cells but not CA3 cells. We selectively deleted the NT3-encoding Ntf3 gene in the DG during the first two postnatal weeks to generate a Ntf3 conditional knockout (Ntf3-cKO). Ntf3-cKO mice of both sexes had normal hippocampal cytoarchitecture but displayed impairments in contextual memory, spatial reference memory, and nest building. Furthermore, male Ntf3-cKO mice exhibited anxiety-like behaviors, whereas female Ntf3-cKO showed some mild depressive symptoms. As MF-CA3 synapses are essential for encoding of contextual memory, we examined synaptic transmission at these synapses using ex vivo electrophysiological recordings. We found that Ntf3-cKO mice had impaired basal synaptic transmission due to deficits in excitatory postsynaptic currents mediated by AMPA receptors but normal presynaptic function and intrinsic excitability of CA3 pyramidal neurons. Consistent with this selective postsynaptic deficit, Ntf3-cKO mice had fewer and smaller thorny excrescences on proximal apical dendrites of CA3 neurons and lower GluR1 levels in the stratum lucidum area where MF-CA3 synapses reside but normal MF terminals, compared with control mice. Thus, our study indicates that NT3 expressed in the dentate gyrus is crucial for the postsynaptic structure and function of MF-CA3 synapses and hippocampal-dependent memory.


Asunto(s)
Región CA3 Hipocampal , Giro Dentado , Ratones Noqueados , Fibras Musgosas del Hipocampo , Neurotrofina 3 , Sinapsis , Animales , Giro Dentado/metabolismo , Fibras Musgosas del Hipocampo/metabolismo , Sinapsis/metabolismo , Ratones , Neurotrofina 3/metabolismo , Neurotrofina 3/genética , Masculino , Femenino , Región CA3 Hipocampal/metabolismo , Células Piramidales/metabolismo , Células Piramidales/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Transmisión Sináptica/fisiología , Cognición/fisiología , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Memoria/fisiología , Receptores AMPA/metabolismo
2.
Biomolecules ; 14(1)2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38254691

RESUMEN

The brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase receptor B (TrkB) are widely expressed in the central nervous system. It is well documented that neurons express BDNF and full-length TrkB (TrkB.FL) as well as a lower level of truncated TrkB (TrkB.T). However, there are conflicting reports regarding the expression of BDNF and TrkB in glial cells, particularly microglia. In this study, we employed a sensitive and reliable genetic method to characterize the expression of BDNF and TrkB in glial cells in the mouse brain. We utilized three Cre mouse strains in which Cre recombinase is expressed in the same cells as BDNF, TrkB.FL, or all TrkB isoforms, and crossed them to Cre-dependent reporter mice to label BDNF- or TrkB-expressing cells with soma-localized EGFP. We performed immunohistochemistry with glial cell markers to examine the expression of BDNF and TrkB in microglia, astrocytes, and oligodendrocytes. Surprisingly, we found no BDNF- or TrkB-expressing microglia in examined CNS regions, including the somatomotor cortex, hippocampal CA1, and spinal cord. Consistent with previous studies, most astrocytes only express TrkB.T in the hippocampus of adult brains. Moreover, there are a small number of astrocytes and oligodendrocytes that express BDNF in the hippocampus, the function of which is to be determined. We also found that oligodendrocyte precursor cells, but not mature oligodendrocytes, express both TrkB.FL and TrkB.T in the hippocampus of adult mice. These results not only clarify the expression of BDNF and TrkB in glial cells but also open opportunities to investigate previously unidentified roles of BDNF and TrkB in astrocytes and oligodendrocytes.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Neuroglía , Receptor trkB , Animales , Ratones , Astrocitos , Factor Neurotrófico Derivado del Encéfalo/genética , Microglía , Oligodendroglía , Receptor trkB/genética
3.
bioRxiv ; 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37503044

RESUMEN

The brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase receptor B (TrkB) are widely expressed in the central nervous system. It is well documented that neurons express BDNF and full-length TrkB (TrkB.FL), and a lower level of truncated TrkB (TrkB.T). With conflicting results, glial cells also have been reported to express BDNF and TrkB. In the current study, we employed a more sensitive and reliable genetic method to characterize the expression of BDNF and TrkB in glial cells in the mouse brain. We utilized three Cre mouse strains in which Cre recombinase is expressed in the same cells as BDNF, TrkB.FL, or all TrkB isoforms, and crossed them to Cre-dependent EGFP reporter mice to label BDNF- or TrkB- expressing cells. We performed immunohistochemistry with glial cell markers to examine the expression of BDNF and TrkB in microglia, astrocytes, and oligodendrocytes. Surprisingly, we found no BDNF- or TrkB- expressing microglia in the brain and spinal cord. Consistent with previous studies, most astrocytes only express TrkB.T in the adult brain. Moreover, there are a small number of astrocytes and oligodendrocytes that express BDNF, the function of which is to be determined. We also found that oligodendrocyte precursor cells, but not mature oligodendrocytes, express both TrkB.FL and TrkB.T in the adult brain. These results not only clarify the expression of BDNF and TrkB in glial cells, but also open opportunities to investigate previously unidentified roles of BDNF and TrkB in glial cells.

4.
SLAS Discov ; 28(3): 88-94, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36842668

RESUMEN

Deficiencies in brain-derived neurotrophic factor (BDNF) have been linked to several brain disorders, making compounds that can boost neuronal BDNF synthesis attractive as potential therapeutics. However, a sensitive and quantitative BDNF assay for high-throughput screening (HTS) is still missing. Here we report the generation of a new mouse Bdnf allele, BdnfNLuc, in which the sequence encoding nano luciferase (NLuc) is inserted into the Bdnf locus immediately before the stop codon so that the allele will produce a BDNF-NLuc fusion protein. BDNF-NLuc protein appears to function like BDNF as BdnfNLuc/NLuc homozygous mice grew and behaved almost normally. We were able to establish and optimize cultures of cortical and hippocampal BdnfNLuc/+ neurons isolated from mouse embryos in 384-well plates. We used the cultures as a phenotypic assay to detect the ability of 10 mM KCl to stimulate BDNF synthesis and achieved a reproducible Z' factor > 0.50 for the assay, a measure considered suitable for HTS. We successfully scaled up the assay to screen the 1280-compound LOPAC library (Library of Pharmacologically Active Compounds). The screen identified several BDNF-boosting compounds, one of which is Bay K8644, a L-type voltage-gated calcium channel (L-VGCC) agonist, which was previously shown to stimulate BDNF synthesis. These results indicate that our phenotypic neuronal assay is ready for HTS to identify novel BDNF-boosting compounds.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Ensayos Analíticos de Alto Rendimiento , Ratones , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas/metabolismo , Canales de Calcio Tipo L/metabolismo , Encéfalo/metabolismo
5.
Endocrinology ; 164(3)2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36631165

RESUMEN

High prevalence of obesity is attributable in part to consumption of highly palatable, fat-rich foods. However, the mechanism controlling dietary fat intake is largely unknown. In this study we investigated the role of brain-derived neurotrophic factor (BDNF) in the control of dietary fat intake in a mouse model that mimics the common human Val-to-Met (Val66Met) polymorphism that impairs BDNF release via the regulated secretory pathway. BdnfMet/Met mice gained weight much faster than wild-type (WT) mice and developed severe obesity due to marked hyperphagia when they were fed HFD. Hyperphagia in these mice worsened when the fat content in their diet was increased. Conversely, mice lacking leptin exhibited similar hyperphagia on chow and HFD. When 2 diets were provided simultaneously, WT and BdnfMet/Met mice showed a comparable preference for the more palatable diet rich in either fat or sucrose, indicating that increased hyperphagia on fat-rich diets in BdnfMet/Met mice is not due to enhanced hedonic drive. In support of this interpretation, WT and BdnfMet/Met mice increased calorie intake to a similar extent during the first day after chow was switched to HFD; however, WT mice decreased HFD intake faster than BdnfMet/Met mice in subsequent days. Furthermore, we found that refeeding after fasting or nocturnal feeding with HFD activated TrkB more strongly than with chow in the hypothalamus of WT mice, whereas TrkB activation under these 2 conditions was greatly attenuated in BdnfMet/Met mice. These results indicate that satiety factors generated during HFD feeding induce BDNF release to suppress excess dietary fat intake.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Hiperfagia , Animales , Ratones , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Dieta , Dieta Alta en Grasa , Grasas de la Dieta/farmacología , Hiperfagia/genética , Obesidad/genética
6.
eNeuro ; 9(2)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35338053

RESUMEN

Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrkB), are implicit in causing obesity. Mutations that reduce BDNF and TrkB expression are associated with obesity in humans and mice. Recently, it was reported that Bdnf gene deletion in the neurons of the paraventricular hypothalamus (PVH) caused positive energy balance and severe obesity in the form of hyperphagia, impaired adaptive thermogenesis, and decreased energy expenditure. Thus, we hypothesize that activation of these neurons will have the opposite effect and provide an opportunity for long-lasting obesity treatment. To specifically activate BDNF-expressing PVH (PVHBDNF) neurons, we injected Cre-dependent adeno-associated virus (AAV) expressing the excitatory DREADD hM3Dq bilaterally into the PVH of Bdnf2A-Cre/+ knock-in mice and then administered clozapine-N-oxide (CNO). Using this technique, we demonstrated that acute activation of these neurons rapidly decreased normal nocturnal feeding and fasting-induced feeding in male and female mice. At thermoneutral temperatures, acute activation also rapidly increased adaptive thermogenesis, increased core body temperature, increased locomotion, increased energy expenditure, and decreased respiratory exchange ratio (RER) in male and female mice. These observations indicate that acute stimulation of PVHBDNF neurons promotes negative energy balance and weight loss. However, the rapid decrease in RER after activation of PVHBDNF neurons was followed by a delayed and prolonged increase in RER that remained elevated for 3 d in female mice. Thus, although acute activation of PVHBDNF neurons promotes negative energy balance in the short term, long-term effects of activation include sexually dimorphic overcompensatory mechanisms that may promote positive energy balance in female mice.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Núcleo Hipotalámico Paraventricular , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Metabolismo Energético/fisiología , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Neuronas/fisiología , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33468645

RESUMEN

Mutations in the TrkB neurotrophin receptor lead to profound obesity in humans, and expression of TrkB in the dorsomedial hypothalamus (DMH) is critical for maintaining energy homeostasis. However, the functional implications of TrkB-fexpressing neurons in the DMH (DMHTrkB) on energy expenditure are unclear. Additionally, the neurocircuitry underlying the effect of DMHTrkB neurons on energy homeostasis has not been explored. In this study, we show that activation of DMHTrkB neurons leads to a robust increase in adaptive thermogenesis and energy expenditure without altering heart rate or blood pressure, while silencing DMHTrkB neurons impairs thermogenesis. Furthermore, we reveal neuroanatomically and functionally distinct populations of DMHTrkB neurons that regulate food intake or thermogenesis. Activation of DMHTrkB neurons projecting to the raphe pallidus (RPa) stimulates thermogenesis and increased energy expenditure, whereas DMHTrkB neurons that send collaterals to the paraventricular hypothalamus (PVH) and preoptic area (POA) inhibit feeding. Together, our findings provide evidence that DMHTrkB neuronal activity plays an important role in regulating energy expenditure and delineate distinct neurocircuits that underly the separate effects of DMHTrkB neuronal activity on food intake and thermogenesis.


Asunto(s)
Regulación del Apetito/genética , Metabolismo Energético/genética , Glicoproteínas de Membrana/genética , Núcleo Hipotalámico Paraventricular/metabolismo , Área Preóptica/metabolismo , Proteínas Tirosina Quinasas/genética , Termogénesis/genética , Animales , Ingestión de Alimentos/genética , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis/genética , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Núcleo Pálido del Rafe/citología , Núcleo Pálido del Rafe/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Área Preóptica/citología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Proteína Fluorescente Roja
8.
J Mol Neurosci ; 70(11): 1858-1870, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32430868

RESUMEN

There are two major isoforms of NTRK2 (neurotrophic receptor tyrosine kinase 2, or TrkB), full-length isoform with tyrosine kinase (TK) domain intact (+) and spliced isoform without tyrosine kinase domain (TK(-)). Within each isoform, there exist subtypes with minor modifications of the protein sequences. In human, the NTRK2 mRNA transcripts encoding TK(+) have same 3'UTRs, while the transcripts encoding subtypes of NTRK2 TK(-) have two completely different 3'UTRs. In mouse, the mRNA transcripts encoding same NTRK2 protein sequence for either TK(+) or TK(-) have long or short 3'UTRs, respectively. The physiological functions of these different 3'UTRs are still unknown. Pilocarpine stimulation increased Ntrk2 mRNA levels in soma, while the increase in synaptosome was smaller. FISH results further showed that mouse Ntrk2 transcripts with different 3'UTRs were distributed differently in cultured cortical neurons. The transcripts with long 3'UTR were distributed more in apical dendrites compared with transcripts with short 3'UTR. Our results provide evidence of non-coding 3'UTR function in regulating mRNA distribution in neurons.


Asunto(s)
Glicoproteínas de Membrana/genética , Neuronas/metabolismo , Proteínas Tirosina Quinasas/genética , Transporte de ARN , ARN Mensajero/metabolismo , Regiones no Traducidas 3' , Animales , Células Cultivadas , Corteza Cerebral/citología , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , Sinaptosomas/metabolismo
9.
Nat Commun ; 11(1): 1729, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32265438

RESUMEN

The TrkB receptor is critical for the control of energy balance, as mutations in its gene (NTRK2) lead to hyperphagia and severe obesity. The main neural substrate mediating the appetite-suppressing activity of TrkB, however, remains unknown. Here, we demonstrate that selective Ntrk2 deletion within paraventricular hypothalamus (PVH) leads to severe hyperphagic obesity. Furthermore, chemogenetic activation or inhibition of TrkB-expressing PVH (PVHTrkB) neurons suppresses or increases food intake, respectively. PVHTrkB neurons project to multiple brain regions, including ventromedial hypothalamus (VMH) and lateral parabrachial nucleus (LPBN). We find that PVHTrkB neurons projecting to LPBN are distinct from those to VMH, yet Ntrk2 deletion in PVH neurons projecting to either VMH or LPBN results in hyperphagia and obesity. Additionally, TrkB activation with BDNF increases firing of these PVH neurons. Therefore, TrkB signaling is a key regulator of a previously uncharacterized neuronal population within the PVH that impinges upon multiple circuits to govern appetite.


Asunto(s)
Hiperfagia/metabolismo , Glicoproteínas de Membrana/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Apetito/genética , Conducta Alimentaria/fisiología , Femenino , Hiperfagia/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiopatología , Proteínas Tirosina Quinasas/genética , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/metabolismo
10.
Nat Commun ; 11(1): 1797, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286273

RESUMEN

Mutations that inactivate negative translation regulators cause autism spectrum disorders (ASD), which predominantly affect males and exhibit social interaction and communication deficits and repetitive behaviors. However, the cells that cause ASD through elevated protein synthesis resulting from these mutations remain unknown. Here we employ conditional overexpression of translation initiation factor eIF4E to increase protein synthesis in specific brain cells. We show that exaggerated translation in microglia, but not neurons or astrocytes, leads to autism-like behaviors in male mice. Although microglial eIF4E overexpression elevates translation in both sexes, it only increases microglial density and size in males, accompanied by microglial shift from homeostatic to a functional state with enhanced phagocytic capacity but reduced motility and synapse engulfment. Consequently, cortical neurons in the mice have higher synapse density, neuroligins, and excitation-to-inhibition ratio compared to control mice. We propose that functional perturbation of male microglia is an important cause for sex-biased ASD.


Asunto(s)
Trastorno Autístico/metabolismo , Conducta Animal , Microglía/metabolismo , Biosíntesis de Proteínas , Animales , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular , Femenino , Perfilación de la Expresión Génica , Genotipo , Homeostasis , Masculino , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Neuronas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fagocitosis , Corteza Prefrontal/metabolismo , Corteza Prefrontal/ultraestructura , Conducta Social , Sinapsis/metabolismo
11.
Nat Commun ; 10(1): 3622, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31399584

RESUMEN

Caspase-2 is the most evolutionarily conserved member in the caspase family of proteases and is constitutively expressed in most cell types including neurons; however, its physiological function remains largely unknown. Here we report that caspase-2 plays a critical role in synaptic plasticity and cognitive flexibility. We found that caspase-2 deficiency led to deficits in dendritic spine pruning, internalization of AMPA receptors and long-term depression. Our results indicate that caspase-2 degrades Rictor, a key mTOR complex 2 (mTORC2) component, to inhibit Akt activation, which leads to enhancement of the GSK3ß activity and thereby long-term depression. Furthermore, we found that mice lacking caspase-2 displayed elevated levels of anxiety, impairment in reversal water maze learning, and little memory loss over time. These results not only uncover a caspase-2-mTORC2-Akt-GSK3ß signaling pathway, but also suggest that caspase-2 is important for memory erasing and normal behaviors by regulating synaptic number and transmission.


Asunto(s)
Caspasa 2/metabolismo , Cognición/fisiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores AMPA/metabolismo , Transducción de Señal/fisiología , Animales , Ansiedad , Conducta Animal , Proteínas Portadoras/metabolismo , Caspasa 2/genética , Espinas Dendríticas/genética , Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Depresión Sináptica a Largo Plazo/genética , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/metabolismo , Ratones , Ratones Noqueados , Plasticidad Neuronal , Neuronas/metabolismo , Receptores de Glutamato/metabolismo
12.
Proc Natl Acad Sci U S A ; 116(8): 3256-3261, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30718415

RESUMEN

Genetic evidence indicates that brain-derived neurotrophic factor (BDNF) signaling through the TrkB receptor plays a critical role in the control of energy balance. Mutations in the BDNF or the TrkB-encoding NTRK2 gene have been found to cause severe obesity in humans and mice. However, it remains unknown which brain neurons express TrkB to control body weight. Here, we report that TrkB-expressing neurons in the dorsomedial hypothalamus (DMH) regulate food intake. We found that the DMH contains both glutamatergic and GABAergic TrkB-expressing neurons, some of which also express the leptin receptor (LepR). As revealed by Fos immunohistochemistry, a significant number of TrkB-expressing DMH (DMHTrkB) neurons were activated upon either overnight fasting or after refeeding. Chemogenetic activation of DMHTrkB neurons strongly suppressed feeding in the dark cycle when mice are physiologically hungry, whereas chemogenetic inhibition of DMHTrkB neurons greatly promoted feeding in the light cycle when mice are physiologically satiated, without affecting feeding in the dark cycle. Neuronal tracing revealed that DMHTrkB neurons do not innervate neurons expressing agouti-related protein in the arcuate nucleus, indicating that DMHTrkB neurons are distinct from previously identified LepR-expressing GABAergic DMH neurons that suppress feeding. Furthermore, selective Ntrk2 deletion in the DMH of adult mice led to hyperphagia, reduced energy expenditure, and obesity. Thus, our data show that DMHTrkB neurons are a population of neurons that are necessary and sufficient to suppress appetite and maintain physiological satiation. Pharmacological activation of these neurons could be a therapeutic intervention for the treatment of obesity.


Asunto(s)
Ingestión de Alimentos/genética , Metabolismo Energético/genética , Glicoproteínas de Membrana/genética , Obesidad/genética , Proteínas Tirosina Quinasas/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Neuronas GABAérgicas/metabolismo , Regulación de la Expresión Génica/genética , Homeostasis/genética , Humanos , Hipotálamo/metabolismo , Ratones , Obesidad/tratamiento farmacológico , Obesidad/patología , Fotoperiodo , Receptores de Leptina
13.
Cell Metab ; 29(4): 917-931.e4, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30661931

RESUMEN

Anxiety disorders are associated with body weight changes in humans. However, the mechanisms underlying anxiety-induced weight changes remain poorly understood. Using Emx1Cre/+ mice, we deleted the gene for brain-derived neurotrophic factor (BDNF) in the cortex, hippocampus, and some amygdalar subregions. The resulting mutant mice displayed impaired GABAergic transmission and elevated anxiety. They were leaner when fed either a chow diet or a high-fat diet, owing to higher sympathetic activity, basal metabolic rate, brown adipocyte thermogenesis, and beige adipocyte formation, compared to control mice. BDNF re-expression in the amygdala rescued the anxiety and metabolic phenotypes in mutant mice. Conversely, anxiety induced by amygdala-specific Bdnf deletion or administration of an inverse GABAA receptor agonist increased energy expenditure. These results reveal that increased activities in anxiogenic circuits can reduce body weight by promoting adaptive thermogenesis and basal metabolism via the sympathetic nervous system and suggest that amygdalar GABAergic neurons are a link between anxiety and metabolic dysfunction.


Asunto(s)
Ansiolíticos/farmacología , Bromazepam/farmacología , Carbolinas/farmacología , Metabolismo Energético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Ansiolíticos/administración & dosificación , Peso Corporal/efectos de los fármacos , Bromazepam/administración & dosificación , Carbolinas/administración & dosificación , Dieta , Ratones , Ratones Endogámicos , Obesidad/inducido químicamente , Obesidad/metabolismo
14.
Elife ; 72018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29419376

RESUMEN

Stress can be a motivational force for decisive action and adapting to novel environment; whereas, exposure to chronic stress contributes to the development of depression and anxiety. However, the molecular mechanisms underlying stress-responsive behaviors are not fully understood. Here, we identified the orphan receptor GPR158 as a novel regulator operating in the prefrontal cortex (PFC) that links chronic stress to depression. GPR158 is highly upregulated in the PFC of human subjects with major depressive disorder. Exposure of mice to chronic stress also increased GPR158 protein levels in the PFC in a glucocorticoid-dependent manner. Viral overexpression of GPR158 in the PFC induced depressive-like behaviors. In contrast GPR158 ablation, led to a prominent antidepressant-like phenotype and stress resiliency. We found that GPR158 exerts its effects via modulating synaptic strength altering AMPA receptor activity. Taken together, our findings identify a new player in mood regulation and introduce a pharmacological target for managing depression.


Asunto(s)
Depresión/fisiopatología , Regulación de la Expresión Génica , Corteza Prefrontal/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Estrés Psicológico , Animales , Humanos , Ratones
15.
PLoS One ; 12(5): e0177610, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28494017

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by CAG repeat expansion within exon1 of the HTT gene. The gene generates two mRNA variants that carry either a short or long 3' untranslated region (3'UTR) while encoding the same protein. It remains unknown whether the two mRNA variants play distinct roles in HD pathogenesis. We found that the long HTT 3'UTR was capable of guiding mRNA to neuronal dendrites, suggesting that some long-form HTT mRNA is transported to dendrites for local protein synthesis. To assay roles of two HTT mRNA variants in cell bodies, we expressed mRNA harboring HTT exon1 containing 23x or 145x CAGs with the short or long 3'UTR. We found that mutant mRNA containing the short 3'UTR produced more protein aggregates and caused more apoptosis in both cultured neurons and HEK293 cells, compared with mutant mRNA containing the long 3'UTR. Although the two 3'UTRs did not affect mRNA stability, we detected higher levels of protein synthesis from mRNA containing the short 3'UTR than from mRNA containing the long 3'UTR. These results indicate that the long HTT 3'UTR suppresses translation. Thus, short-form mutant HTT mRNA will be more efficient in producing toxic protein than its long-form counterpart.


Asunto(s)
Regulación de la Expresión Génica , Proteína Huntingtina/genética , Proteína Huntingtina/toxicidad , Proteínas Mutantes/genética , Proteínas Mutantes/toxicidad , Biosíntesis de Proteínas , Regiones no Traducidas 3'/genética , Animales , Células HEK293 , Humanos , Proteína Huntingtina/química , Proteínas Mutantes/química , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Agregado de Proteínas/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Multimerización de Proteína/efectos de los fármacos , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Solubilidad , Fracciones Subcelulares/metabolismo
16.
Proc Natl Acad Sci U S A ; 114(4): E619-E628, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28069946

RESUMEN

Stability of neuronal connectivity is critical for brain functions, and morphological perturbations are associated with neurodegenerative disorders. However, how neuronal morphology is maintained in the adult brain remains poorly understood. Here, we identify Wnt5a, a member of the Wnt family of secreted morphogens, as an essential factor in maintaining dendritic architecture in the adult hippocampus and for related cognitive functions in mice. Wnt5a expression in hippocampal neurons begins postnatally, and its deletion attenuated CaMKII and Rac1 activity, reduced GluN1 glutamate receptor expression, and impaired synaptic plasticity and spatial learning and memory in 3-mo-old mice. With increased age, Wnt5a loss caused progressive attrition of dendrite arbors and spines in Cornu Ammonis (CA)1 pyramidal neurons and exacerbated behavioral defects. Wnt5a functions cell-autonomously to maintain CA1 dendrites, and exogenous Wnt5a expression corrected structural anomalies even at late-adult stages. These findings reveal a maintenance factor in the adult brain, and highlight a trophic pathway that can be targeted to ameliorate dendrite loss in pathological conditions.


Asunto(s)
Dendritas/fisiología , Hipocampo/fisiología , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Proteína Wnt-5a/fisiología , Animales , Células Cultivadas , Masculino , Ratones Mutantes , Ratones Transgénicos , Neuronas/fisiología , Ratas Sprague-Dawley , Agudeza Visual , Proteína Wnt-5a/genética
17.
Curr Biol ; 26(22): 2992-3003, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27773571

RESUMEN

It is well recognized that G-protein-coupled receptors (GPCRs) can activate Ras-regulated kinase pathways to produce lasting changes in neuronal function. Mechanisms by which GPCRs transduce these signals and their relevance to brain disorders are not well understood. Here, we identify a major Ras regulator, neurofibromin 1 (NF1), as a direct effector of GPCR signaling via Gßγ subunits in the striatum. We find that binding of Gßγ to NF1 inhibits its ability to inactivate Ras. Deletion of NF1 in striatal neurons prevents the opioid-receptor-induced activation of Ras and eliminates its coupling to Akt-mTOR-signaling pathway. By acting in the striatal medium spiny neurons of the direct pathway, NF1 regulates opioid-induced changes in Ras activity, thereby sensitizing mice to psychomotor and rewarding effects of morphine. These results delineate a novel mechanism of GPCR signaling to Ras pathways and establish a critical role of NF1 in opioid addiction.


Asunto(s)
Analgésicos Opioides/metabolismo , Neurofibromina 1/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Proteínas ras/metabolismo , Animales , Femenino , Masculino , Ratones , Neostriado/metabolismo , Neurofibromina 1/metabolismo , Neuronas/metabolismo , Unión Proteica
18.
Mol Metab ; 5(10): 903-917, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27689003

RESUMEN

OBJECTIVE: Obesity and type 2 diabetes (T2D) lead to various life-threatening diseases such as coronary heart disease, stroke, osteoarthritis, asthma, and neurodegeneration. Therefore, extensive research is ongoing to identify novel pathways that can be targeted in obesity/T2D. Deletion of the inositol pyrophosphate (5-IP7) biosynthetic enzyme, inositol hexakisphosphate kinase-1 (IP6K1), protects mice from high fat diet (HFD) induced obesity (DIO) and insulin resistance. Yet, whether this pathway is a valid pharmacologic target in obesity/T2D is not known. Here, we demonstrate that TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl)purine], a pan-IP6K inhibitor, has strong anti-obesity and anti-diabetic effects in DIO mice. METHODS: Q-NMR, GTT, ITT, food intake, energy expenditure, QRT-PCR, ELISA, histology, and immunoblot studies were conducted in short (2.5-week)- and long (10-week)-term TNP treated DIO C57/BL6 WT and IP6K1-KO mice, under various diet and temperature conditions. RESULTS: TNP, when injected at the onset of HFD-feeding, decelerates initiation of DIO and insulin resistance. Moreover, TNP facilitates weight loss and restores metabolic parameters, when given to DIO mice. However, TNP does not reduce weight gain in HFD-fed IP6K1-KO mice. TNP specifically enhances insulin sensitivity in DIO mice via Akt activation. TNP decelerates weight gain primarily by enhancing thermogenic energy expenditure in the adipose tissue. Accordingly, TNP's effect on body weight is partly abolished whereas its impact on glucose homeostasis is preserved at thermoneutral temperature. CONCLUSION: Pharmacologic inhibition of the inositol pyrophosphate pathway has strong therapeutic potential in obesity, T2D, and other metabolic diseases.

19.
Nat Rev Neurosci ; 17(5): 282-92, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27052383

RESUMEN

Energy balance--that is, the relationship between energy intake and energy expenditure--is regulated by a complex interplay of hormones, brain circuits and peripheral tissues. Leptin is an adipocyte-derived cytokine that suppresses appetite and increases energy expenditure. Ironically, obese individuals have high levels of plasma leptin and are resistant to leptin treatment. Neurotrophic factors, particularly ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF), are also important for the control of body weight. CNTF can overcome leptin resistance in order to reduce body weight, although CNTF and leptin activate similar signalling cascades. Mutations in the gene encoding BDNF lead to insatiable appetite and severe obesity.


Asunto(s)
Peso Corporal/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Respuesta de Saciedad/fisiología , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Factor Neurotrófico Ciliar/farmacología , Factor Neurotrófico Ciliar/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Humanos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Respuesta de Saciedad/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
20.
Mol Endocrinol ; 30(5): 494-503, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27003443

RESUMEN

Brain-derived neurotrophic factor (BDNF) expressed in the paraventricular hypothalamus (PVH) has been shown to play a key role in regulating energy intake and energy expenditure. BDNF is also expressed in other hypothalamic nuclei; however, the role in the control of energy balance for BDNF produced in these structures remains largely unknown. We found that deleting the Bdnf gene in the ventromedial hypothalamus (VMH) during embryogenesis using the Sf1-Cre transgene had no effect on body weight in mice. In contrast, deleting the Bdnf gene in the adult VMH using Cre-expressing virus led to significant hyperphagia and obesity. These observations indicate that the lack of a hyperphagia phenotype in the Sf1-Cre/Bdnf mutant mice is likely due to developmental compensation. To investigate the role of BDNF expressed in other hypothalamic areas, we employed the hypothalamus-specific Nkx2.1-Cre transgene to delete the Bdnf gene. We found that the Nkx2.1-Cre transgene could abolish BDNF expression in many hypothalamic nuclei, but not in the PVH, and that the resulting mutant mice developed modest obesity due to reduced energy expenditure. Thus, BDNF produced in the VMH plays a role in regulating energy intake. Furthermore, BDNF expressed in hypothalamic areas other than PVH and VMH is also involved in the control of energy expenditure.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Peso Corporal/fisiología , Ingestión de Energía/fisiología , Hiperfagia/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Factores de Empalme de ARN/metabolismo , Factor Nuclear Tiroideo 1/metabolismo , Transgenes/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...