Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(16): e2309211121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593081

RESUMEN

Vesicular release of neurotransmitters and hormones relies on the dynamic assembly of the exocytosis/trans-SNARE complex through sequential interactions of synaptobrevins, syntaxins, and SNAP-25. Despite SNARE-mediated release being fundamental for intercellular communication in all excitable tissues, the role of auxiliary proteins modulating the import of reserve vesicles to the active zone, and thus, scaling repetitive exocytosis remains less explored. Secretagogin is a Ca2+-sensor protein with SNAP-25 being its only known interacting partner. SNAP-25 anchors readily releasable vesicles within the active zone, thus being instrumental for 1st phase release. However, genetic deletion of secretagogin impedes 2nd phase release instead, calling for the existence of alternative protein-protein interactions. Here, we screened the secretagogin interactome in the brain and pancreas, and found syntaxin-4 grossly overrepresented. Ca2+-loaded secretagogin interacted with syntaxin-4 at nanomolar affinity and 1:1 stoichiometry. Crystal structures of the protein complexes revealed a hydrophobic groove in secretagogin for the binding of syntaxin-4. This groove was also used to bind SNAP-25. In mixtures of equimolar recombinant proteins, SNAP-25 was sequestered by secretagogin in competition with syntaxin-4. Kd differences suggested that secretagogin could shape unidirectional vesicle movement by sequential interactions, a hypothesis supported by in vitro biological data. This mechanism could facilitate the movement of transport vesicles toward release sites, particularly in the endocrine pancreas where secretagogin, SNAP-25, and syntaxin-4 coexist in both α- and ß-cells. Thus, secretagogin could modulate the pace and fidelity of vesicular hormone release by differential protein interactions.


Asunto(s)
Fusión de Membrana , Secretagoginas , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Secretagoginas/metabolismo , Membrana Celular/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Exocitosis , Comunicación Celular , Sintaxina 1/metabolismo , Unión Proteica
2.
Proc Natl Acad Sci U S A ; 120(31): e2222095120, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37487094

RESUMEN

The locus coeruleus (LC) is a small nucleus in the pons from which ascending and descending projections innervate major parts of the central nervous system. Its major transmitter is norepinephrine (NE). This system is evolutionarily conserved, including in humans, and its functions are associated with wakefulness and related to disorders, such as depression. Here, we performed single-cell ribonucleic acid-sequencing (RNA-seq) to subdivide neurons in the LC (24 clusters in total) into 3 NE, 17 glutamate, and 5 γ-aminobutyric acid (GABA) subtypes, and to chart their neuropeptide, cotransmitter, and receptor profiles. We found that NE neurons expressed at least 19 neuropeptide transcripts, notably galanin (Gal) but not Npy, and >30 neuropeptide receptors. Among the galanin receptors, Galr1 was expressed in ~19% of NE neurons, as was also confirmed by in situ hybridization. Unexpectedly, Galr1 was highly expressed in GABA neurons surrounding the NE ensemble. Patch-clamp electrophysiology and cell-type-specific Ca2+-imaging using GCaMP6s revealed that a GalR1 agonist inhibits up to ~35% of NE neurons. This effect is direct and does not rely on feed-forward GABA inhibition. Our results define a role for the galanin system in NE functions, and a conceptual framework for the action of many other peptides and their receptors.


Asunto(s)
Galanina , Hormonas Peptídicas , Humanos , Animales , Ratones , Locus Coeruleus , Neuronas , Ácido Glutámico , Norepinefrina
3.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34108238

RESUMEN

Galanin receptor1 (GalR1) transcript levels are elevated in the rat ventral periaqueductal gray (vPAG) after chronic mild stress (CMS) and are related to depression-like behavior. To explore the mechanisms underlying the elevated GalR1 expression, we carried out molecular biological experiments in vitro and in animal behavioral experiments in vivo. It was found that a restricted upstream region of the GalR1 gene, from -250 to -220, harbors an E-box and plays a negative role in the GalR1 promoter activity. The transcription factor Scratch2 bound to the E-box to down-regulate GalR1 promoter activity and lower expression levels of the GalR1 gene. The expression of Scratch2 was significantly decreased in the vPAG of CMS rats. Importantly, local knockdown of Scratch2 in the vPAG caused elevated expression of GalR1 in the same region, as well as depression-like behaviors. RNAscope analysis revealed that GalR1 mRNA is expressed together with Scratch2 in both GABA and glutamate neurons. Taking these data together, our study further supports the involvement of GalR1 in mood control and suggests a role for Scratch2 as a regulator of depression-like behavior by repressing the GalR1 gene in the vPAG.


Asunto(s)
Conducta Animal , Depresión/patología , Sustancia Gris Periacueductal/patología , Receptor de Galanina Tipo 1/metabolismo , Factores de Transcripción/metabolismo , Animales , Elementos E-Box/genética , Neuronas GABAérgicas/metabolismo , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Células PC12 , Regiones Promotoras Genéticas/genética , Unión Proteica , Ratas , Receptor de Galanina Tipo 1/genética , Estrés Psicológico/complicaciones , Factores de Transcripción/genética , Sitio de Iniciación de la Transcripción
4.
J Clin Med ; 10(8)2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917176

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disorder as yet without effective therapy. Symptoms of this disorder typically reflect cortical malfunction with local neurohistopathology, which biased investigators to search for focal triggers and molecular mechanisms. Cortex, however, receives massive afferents from caudal brain structures, which do not only convey specific information but powerfully tune ensemble activity. Moreover, there is evidence that the start of AD is subcortical. The brainstem harbors monoamine systems, which establish a dense innervation in both allo- and neocortex. Monoaminergic synapses can co-release neuropeptides either by precisely terminating on cortical neurons or, when being "en passant", can instigate local volume transmission. Especially due to its early damage, malfunction of the ascending monoaminergic system emerges as an early sign and possible trigger of AD. This review summarizes the involvement and cascaded impairment of brainstem monoaminergic neurons in AD and discusses cellular mechanisms that lead to their dysfunction. We highlight the significance and therapeutic challenges of transmitter co-release in ascending activating system, describe the role and changes of local connections and distant afferents of brainstem nuclei in AD, and summon the rapidly increasing diagnostic window during the last few years.

5.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33558223

RESUMEN

The perception of and response to danger is critical for an individual's survival and is encoded by subcortical neurocircuits. The amygdaloid complex is the primary neuronal site that initiates bodily reactions upon external threat with local-circuit interneurons scaling output to effector pathways. Here, we categorize central amygdala neurons that express secretagogin (Scgn), a Ca2+-sensor protein, as a subset of protein kinase Cδ (PKCδ)+ interneurons, likely "off cells." Chemogenetic inactivation of Scgn+/PKCδ+ cells augmented conditioned response to perceived danger in vivo. While Ca2+-sensor proteins are typically implicated in shaping neurotransmitter release presynaptically, Scgn instead localized to postsynaptic compartments. Characterizing its role in the postsynapse, we found that Scgn regulates the cell-surface availability of NMDA receptor 2B subunits (GluN2B) with its genetic deletion leading to reduced cell membrane delivery of GluN2B, at least in vitro. Conclusively, we describe a select cell population, which gates danger avoidance behavior with secretagogin being both a selective marker and regulatory protein in their excitatory postsynaptic machinery.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Interneuronas/metabolismo , Proteína Quinasa C-delta/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Secretagoginas/metabolismo , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/fisiología , Animales , Reacción de Prevención , Línea Celular Tumoral , Células Cultivadas , Miedo , Femenino , Humanos , Interneuronas/fisiología , Masculino , Transporte de Proteínas , Ratas , Ratas Wistar , Secretagoginas/genética , Potenciales Sinápticos
6.
Proc Natl Acad Sci U S A ; 116(51): 25958-25967, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31796600

RESUMEN

Psychostimulant use is an ever-increasing socioeconomic burden, including a dramatic rise during pregnancy. Nevertheless, brain-wide effects of psychostimulant exposure are incompletely understood. Here, we performed Fos-CreERT2-based activity mapping, correlated for pregnant mouse dams and their fetuses with amphetamine, nicotine, and caffeine applied acutely during midgestation. While light-sheet microscopy-assisted intact tissue imaging revealed drug- and age-specific neuronal activation, the indusium griseum (IG) appeared indiscriminately affected. By using GAD67gfp/+ mice we subdivided the IG into a dorsolateral domain populated by γ-aminobutyric acidergic interneurons and a ventromedial segment containing glutamatergic neurons, many showing drug-induced activation and sequentially expressing Pou3f3/Brn1 and secretagogin (Scgn) during differentiation. We then combined Patch-seq and circuit mapping to show that the ventromedial IG is a quasi-continuum of glutamatergic neurons (IG-Vglut1+) reminiscent of dentate granule cells in both rodents and humans, whose dendrites emanate perpendicularly toward while their axons course parallel with the superior longitudinal fissure. IG-Vglut1+ neurons receive VGLUT1+ and VGLUT2+ excitatory afferents that topologically segregate along their somatodendritic axis. In turn, their efferents terminate in the olfactory bulb, thus being integral to a multisynaptic circuit that could feed information antiparallel to the olfactory-cortical pathway. In IG-Vglut1+ neurons, prenatal psychostimulant exposure delayed the onset of Scgn expression. Genetic ablation of Scgn was then found to sensitize adult mice toward methamphetamine-induced epilepsy. Overall, our study identifies brain-wide targets of the most common psychostimulants, among which Scgn+/Vglut1+ neurons of the IG link limbic and olfactory circuits.


Asunto(s)
Mapeo Encefálico , Encéfalo/metabolismo , Regulación de la Expresión Génica , Lóbulo Límbico/metabolismo , Animales , Axones/metabolismo , Encéfalo/diagnóstico por imagen , Dendritas/metabolismo , Femenino , Glutamato Descarboxilasa/genética , Humanos , Interneuronas/metabolismo , Lóbulo Límbico/anatomía & histología , Lóbulo Límbico/efectos de los fármacos , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Factores del Dominio POU/genética , Factores del Dominio POU/metabolismo , Secretagoginas/genética , Secretagoginas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/genética , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Ácido gamma-Aminobutírico/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(10): E2006-E2015, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28223495

RESUMEN

The rostral migratory stream (RMS) is viewed as a glia-enriched conduit of forward-migrating neuroblasts in which chemorepulsive signals control the pace of forward migration. Here we demonstrate the existence of a scaffold of neurons that receive synaptic inputs within the rat, mouse, and human fetal RMS equivalents. These neurons express secretagogin, a Ca2+-sensor protein, to execute an annexin V-dependent externalization of matrix metalloprotease-2 (MMP-2) for reconfiguring the extracellular matrix locally. Mouse genetics combined with pharmacological probing in vivo and in vitro demonstrate that MMP-2 externalization occurs on demand and that its loss slows neuroblast migration. Loss of function is particularly remarkable upon injury to the olfactory bulb. Cumulatively, we identify a signaling cascade that provokes structural remodeling of the RMS through recruitment of MMP-2 by a previously unrecognized neuronal constituent. Given the life-long presence of secretagogin-containing neurons in human, this mechanism might be exploited for therapeutic benefit in rescue strategies.


Asunto(s)
Calcio/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Secretagoginas/genética , Animales , Anexina A5/genética , Anexina A5/metabolismo , Movimiento Celular , Feto , Regulación de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Microtomía , Neuroglía/ultraestructura , Neuronas/ultraestructura , Bulbo Olfatorio/citología , Cultivo Primario de Células , Ratas , Ratas Wistar , Secretagoginas/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Técnicas de Cultivo de Tejidos
8.
Proc Natl Acad Sci U S A ; 113(52): E8472-E8481, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27940914

RESUMEN

Major depressive disorder (MDD) is a substantial burden to patients, families, and society, but many patients cannot be treated adequately. Rodent experiments suggest that the neuropeptide galanin (GAL) and its three G protein-coupled receptors, GAL1-3, are involved in mood regulation. To explore the translational potential of these results, we assessed the transcript levels (by quantitative PCR), DNA methylation status (by bisulfite pyrosequencing), and GAL peptide by RIA of the GAL system in postmortem brains from depressed persons who had committed suicide and controls. Transcripts for all four members were detected and showed marked regional variations, GAL and galanin receptor 1 (GALR1) being most abundant. Striking increases in GAL and GALR3 mRNA levels, especially in the noradrenergic locus coeruleus and the dorsal raphe nucleus, in parallel with decreased DNA methylation, were found in both male and female suicide subjects as compared with controls. In contrast, GAL and GALR3 transcript levels were decreased, GALR1 was increased, and DNA methylation was increased in the dorsolateral prefrontal cortex of male suicide subjects, however, there were no changes in the anterior cingulate cortex. Thus, GAL and its receptor GALR3 are differentially methylated and expressed in brains of MDD subjects in a region- and sex-specific manner. Such an epigenetic modification in GALR3, a hyperpolarizing receptor, might contribute to the dysregulation of noradrenergic and serotonergic neurons implicated in the pathogenesis of MDD. Thus, one may speculate that a GAL3 antagonist could have antidepressant properties by disinhibiting the firing of these neurons, resulting in increased release of noradrenaline and serotonin in forebrain areas involved in mood regulation.


Asunto(s)
Trastorno Depresivo Mayor/metabolismo , Galanina/metabolismo , Receptor de Galanina Tipo 1/metabolismo , Receptor de Galanina Tipo 3/metabolismo , Adulto , Afecto , Anciano , Encéfalo/metabolismo , Encéfalo/patología , Mapeo Encefálico , Estudios de Casos y Controles , Metilación de ADN , Trastorno Depresivo Mayor/genética , Núcleo Dorsal del Rafe/metabolismo , Femenino , Galanina/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Locus Coeruleus/metabolismo , Masculino , Persona de Mediana Edad , Receptor de Galanina Tipo 1/genética , Receptor de Galanina Tipo 3/genética , Factores Sexuales , Suicidio
9.
Proc Natl Acad Sci U S A ; 113(43): E6686-E6695, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27791037

RESUMEN

Pain is a critical component hindering recovery and regaining of function after surgery, particularly in the elderly. Understanding the role of pain signaling after surgery may lead to novel interventions for common complications such as delirium and postoperative cognitive dysfunction. Using a model of tibial fracture with intramedullary pinning in male mice, associated with cognitive deficits, we characterized the effects on the primary somatosensory system. Here we show that tibial fracture with pinning triggers cold allodynia and up-regulates nerve injury and inflammatory markers in dorsal root ganglia (DRGs) and spinal cord up to 2 wk after intervention. At 72 h after surgery, there is an increase in activating transcription factor 3 (ATF3), the neuropeptides galanin and neuropeptide Y (NPY), brain-derived neurotrophic factor (BDNF), as well as neuroinflammatory markers including ionized calcium-binding adaptor molecule 1 (Iba1), glial fibrillary acidic protein (GFAP), and the fractalkine receptor CX3CR1 in DRGs. Using an established model of complete transection of the sciatic nerve for comparison, we observed similar but more pronounced changes in these markers. However, protein levels of BDNF remained elevated for a longer period after fracture. In the hippocampus, BDNF protein levels were increased, yet there were no changes in Bdnf mRNA in the parent granule cell bodies. Further, c-Fos was down-regulated in the hippocampus, together with a reduction in neurogenesis in the subgranular zone. Taken together, our results suggest that attenuated BDNF release and signaling in the dentate gyrus may account for cognitive and mental deficits sometimes observed after surgery.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Disfunción Cognitiva/genética , Giro Dentado/metabolismo , Ganglios Espinales/metabolismo , Neuropéptido Y/genética , Dolor/genética , Fracturas de la Tibia/cirugía , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Giro Dentado/fisiopatología , Fijación Intramedular de Fracturas/efectos adversos , Galanina/genética , Galanina/metabolismo , Ganglios Espinales/fisiopatología , Regulación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neuropéptido Y/metabolismo , Dolor/etiología , Dolor/metabolismo , Dolor/patología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Transducción de Señal , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Fracturas de la Tibia/genética , Fracturas de la Tibia/metabolismo , Fracturas de la Tibia/fisiopatología
10.
Proc Natl Acad Sci U S A ; 113(32): E4726-35, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27457954

RESUMEN

The neuropeptide galanin coexists in rat brain with serotonin in the dorsal raphe nucleus and with noradrenaline in the locus coeruleus (LC), and it has been suggested to be involved in depression. We studied rats exposed to chronic mild stress (CMS), a rodent model of depression. As expected, these rats showed several endophenotypes relevant to depression-like behavior compared with controls. All these endophenotypes were normalized after administration of a selective serotonin reuptake inhibitor. The transcripts for galanin and two of its receptors, galanin receptor 1 (GALR1) and GALR2, were analyzed with quantitative real-time PCR using laser capture microdissection in the following brain regions: the hippocampal formation, LC, and ventral periaqueductal gray (vPAG). Only Galr1 mRNA levels were significantly increased, and only in the latter region. After knocking down Galr1 in the vPAG with an siRNA technique, all parameters of the depressive behavioral phenotype were similar to controls. Thus, the depression-like behavior in rats exposed to CMS is likely related to an elevated expression of Galr1 in the vPAG, suggesting that a GALR1 antagonist could have antidepressant effects.


Asunto(s)
Depresión/etiología , Sustancia Gris Periacueductal/fisiología , Receptor de Galanina Tipo 1/fisiología , Animales , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Locus Coeruleus/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Receptor de Galanina Tipo 1/antagonistas & inhibidores , Serotonina/fisiología , Ácido gamma-Aminobutírico/fisiología
11.
Proc Natl Acad Sci U S A ; 113(3): E368-77, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26668381

RESUMEN

An increased incidence in the sleep-disorder narcolepsy has been associated with the 2009-2010 pandemic of H1N1 influenza virus in China and with mass vaccination campaigns against influenza during the pandemic in Finland and Sweden. Pathogenetic mechanisms of narcolepsy have so far mainly focused on autoimmunity. We here tested an alternative working hypothesis involving a direct role of influenza virus infection in the pathogenesis of narcolepsy in susceptible subjects. We show that infection with H1N1 influenza virus in mice that lack B and T cells (Recombinant activating gene 1-deficient mice) can lead to narcoleptic-like sleep-wake fragmentation and sleep structure alterations. Interestingly, the infection targeted brainstem and hypothalamic neurons, including orexin/hypocretin-producing neurons that regulate sleep-wake stability and are affected in narcolepsy. Because changes occurred in the absence of adaptive autoimmune responses, the findings show that brain infections with H1N1 virus have the potential to cause per se narcoleptic-like sleep disruption.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Narcolepsia/fisiopatología , Narcolepsia/virología , Neuronas/fisiología , Sueño , Vigilia , Animales , Antígenos Virales/inmunología , Electroencefalografía , Proteínas de Homeodominio/metabolismo , Hipotálamo/fisiopatología , Hipotálamo/virología , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Bulbo Olfatorio/fisiopatología , Bulbo Olfatorio/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/fisiopatología , Infecciones por Orthomyxoviridae/virología
12.
Proc Natl Acad Sci U S A ; 112(45): E6185-94, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26494286

RESUMEN

Endocannabinoids are implicated in the control of glucose utilization and energy homeostasis by orchestrating pancreatic hormone release. Moreover, in some cell niches, endocannabinoids regulate cell proliferation, fate determination, and migration. Nevertheless, endocannabinoid contributions to the development of the endocrine pancreas remain unknown. Here, we show that α cells produce the endocannabinoid 2-arachidonoylglycerol (2-AG) in mouse fetuses and human pancreatic islets, which primes the recruitment of ß cells by CB1 cannabinoid receptor (CB1R) engagement. Using subtractive pharmacology, we extend these findings to anandamide, a promiscuous endocannabinoid/endovanilloid ligand, which impacts both the determination of islet size by cell proliferation and α/ß cell sorting by differential activation of transient receptor potential cation channel subfamily V member 1 (TRPV1) and CB1Rs. Accordingly, genetic disruption of TRPV1 channels increases islet size whereas CB1R knockout augments cellular heterogeneity and favors insulin over glucagon release. Dietary enrichment in ω-3 fatty acids during pregnancy and lactation in mice, which permanently reduces endocannabinoid levels in the offspring, phenocopies CB1R(-/-) islet microstructure and improves coordinated hormone secretion. Overall, our data mechanistically link endocannabinoids to cell proliferation and sorting during pancreatic islet formation, as well as to life-long programming of hormonal determinants of glucose homeostasis.


Asunto(s)
Endocannabinoides/metabolismo , Islotes Pancreáticos/embriología , Morfogénesis/fisiología , Receptor Cannabinoide CB1/metabolismo , Canales Catiónicos TRPV/metabolismo , Análisis de Varianza , Animales , Ácidos Grasos Omega-3/administración & dosificación , Femenino , Feto/metabolismo , Prueba de Tolerancia a la Glucosa , Procesamiento de Imagen Asistido por Computador , Islotes Pancreáticos/anatomía & histología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Embarazo
13.
Proc Natl Acad Sci U S A ; 112(31): E4326-35, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195742

RESUMEN

Synaptosomal-associated protein of 25 kDa (SNAP-25) is a key molecule in the soluble N-ethylmaleimide-sensitive factor attachment protein (SNARE) complex mediating fast Ca(2+)-triggered release of hormones and neurotransmitters, and both splice variants, SNAP-25a and SNAP-25b, can participate in this process. Here we explore the hypothesis that minor alterations in the machinery mediating regulated membrane fusion can increase the susceptibility for metabolic disease and precede obesity and type 2 diabetes. Thus, we used a mouse mutant engineered to express normal levels of SNAP-25 but only SNAP-25a. These SNAP-25b-deficient mice were exposed to either a control or a high-fat/high-sucrose diet. Monitoring of food intake, body weight, hypothalamic function, and lipid and glucose homeostases showed that SNAP-25b-deficient mice fed with control diet developed hyperglycemia, liver steatosis, and adipocyte hypertrophy, conditions dramatically exacerbated when combined with the high-fat/high-sucrose diet. Thus, modified SNARE function regulating stimulus-dependent exocytosis can increase the vulnerability to and even provoke metabolic disease. When combined with a high-fat/high-sucrose diet, this vulnerability resulted in diabesity. Our SNAP-25b-deficient mouse may represent a diabesity model.


Asunto(s)
Enfermedades Metabólicas/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Adiposidad , Animales , Glucemia/metabolismo , Peso Corporal , Dislipidemias/patología , Ingestión de Energía , Metabolismo Energético , Conducta Alimentaria , Femenino , Homeostasis , Hipertrofia , Hipotálamo/metabolismo , Insulina/metabolismo , Secreción de Insulina , Leptina/sangre , Hígado/metabolismo , Hígado/patología , Masculino , Enfermedades Metabólicas/sangre , Ratones Obesos , Fenotipo , Receptores de Leptina/metabolismo , Proteína 25 Asociada a Sinaptosomas/deficiencia
14.
Proc Natl Acad Sci U S A ; 111(16): E1666-73, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24706871

RESUMEN

Galanin is a stress-inducible neuropeptide and cotransmitter in serotonin and norepinephrine neurons with a possible role in stress-related disorders. Here we report that variants in genes for galanin (GAL) and its receptors (GALR1, GALR2, GALR3), despite their disparate genomic loci, conferred increased risk of depression and anxiety in people who experienced childhood adversity or recent negative life events in a European white population cohort totaling 2,361 from Manchester, United Kingdom and Budapest, Hungary. Bayesian multivariate analysis revealed a greater relevance of galanin system genes in highly stressed subjects compared with subjects with moderate or low life stress. Using the same method, the effect of the galanin system genes was stronger than the effect of the well-studied 5-HTTLPR polymorphism in the serotonin transporter gene (SLC6A4). Conventional multivariate analysis using general linear models demonstrated that interaction of galanin system genes with life stressors explained more variance (1.7%, P = 0.005) than the life stress-only model. This effect replicated in independent analysis of the Manchester and Budapest subpopulations, and in males and females. The results suggest that the galanin pathway plays an important role in the pathogenesis of depression in humans by increasing the vulnerability to early and recent psychosocial stress. Correcting abnormal galanin function in depression could prove to be a novel target for drug development. The findings further emphasize the importance of modeling environmental interaction in finding new genes for depression.


Asunto(s)
Encéfalo/metabolismo , Depresión/genética , Galanina/genética , Interacción Gen-Ambiente , Receptores de Galanina/genética , Estrés Psicológico/genética , Adulto , Teorema de Bayes , Simulación por Computador , Demografía , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Modelos Logísticos , Masculino , Modelos Biológicos , Análisis Multinivel , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Transducción de Señal
15.
Proc Natl Acad Sci U S A ; 111(12): E1149-58, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24616509

RESUMEN

Neuronal calcium (Ca(2+))-binding proteins 1 and 2 (NECAB1/2) are members of the phylogenetically conserved EF-hand Ca(2+)-binding protein superfamily. To date, NECABs have been explored only to a limited extent and, so far, not at all at the spinal level. Here, we describe the distribution, phenotype, and nerve injury-induced regulation of NECAB1/NECAB2 in mouse dorsal root ganglia (DRGs) and spinal cord. In DRGs, NECAB1/2 are expressed in around 70% of mainly small- and medium-sized neurons. Many colocalize with calcitonin gene-related peptide and isolectin B4, and thus represent nociceptors. NECAB1/2 neurons are much more abundant in DRGs than the Ca(2+)-binding proteins (parvalbumin, calbindin, calretinin, and secretagogin) studied to date. In the spinal cord, the NECAB1/2 distribution is mainly complementary. NECAB1 labels interneurons and a plexus of processes in superficial layers of the dorsal horn, commissural neurons in the intermediate area, and motor neurons in the ventral horn. Using CLARITY, a novel, bilaterally connected neuronal system with dendrites that embrace the dorsal columns like palisades is observed. NECAB2 is present in cell bodies and presynaptic boutons across the spinal cord. In the dorsal horn, most NECAB1/2 neurons are glutamatergic. Both NECAB1/2 are transported into dorsal roots and peripheral nerves. Peripheral nerve injury reduces NECAB2, but not NECAB1, expression in DRG neurons. Our study identifies NECAB1/2 as abundant Ca(2+)-binding proteins in pain-related DRG neurons and a variety of spinal systems, providing molecular markers for known and unknown neuron populations of mechanosensory and pain circuits in the spinal cord.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Ganglios Espinales/metabolismo , Neuronas/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Médula Espinal/citología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Dolor/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Médula Espinal/metabolismo
16.
Proc Natl Acad Sci U S A ; 110(6): E536-45, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341594

RESUMEN

Using riboprobe in situ hybridization, we studied the localization of the transcripts for the neuropeptide galanin and its receptors (GalR1-R3), tryptophan hydroxylase 2, tyrosine hydroxylase, and nitric oxide synthase as well as the three vesicular glutamate transporters (VGLUT 1-3) in the locus coeruleus (LC) and the dorsal raphe nucleus (DRN) regions of postmortem human brains. Quantitative real-time PCR (qPCR) was used also. Galanin and GalR3 mRNA were found in many noradrenergic LC neurons, and GalR3 overlapped with serotonin neurons in the DRN. The qPCR analysis at the LC level ranked the transcripts in the following order in the LC: galanin >> GalR3 >> GalR1 > GalR2; in the DRN the ranking was galanin >> GalR3 >> GalR1 = GalR2. In forebrain regions the ranking was GalR1 > galanin > GalR2. VGLUT1 and -2 were strongly expressed in the pontine nuclei but could not be detected in LC or serotonin neurons. VGLUT2 transcripts were found in very small, nonpigmented cells in the LC and in the lateral and dorsal aspects of the periaqueductal central gray. Nitric oxide synthase was not detected in serotonin neurons. These findings show distinct differences between the human brain and rodents, especially rat, in the distribution of the galanin system and some other transmitter systems. For example, GalR3 seems to be the important galanin receptor in both the human LC and DRN versus GalR1 and -2 in the rodent brain. Such knowledge may be important when considering therapeutic principles and drug development.


Asunto(s)
Encéfalo/metabolismo , Galanina/genética , Galanina/metabolismo , Neurotransmisores/metabolismo , Animales , Humanos , Hibridación in Situ , Locus Coeruleus/metabolismo , Neurotransmisores/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Núcleos del Rafe/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Galanina Tipo 1/genética , Receptor de Galanina Tipo 1/metabolismo , Receptor de Galanina Tipo 2/genética , Receptor de Galanina Tipo 2/metabolismo , Receptor de Galanina Tipo 3/genética , Receptor de Galanina Tipo 3/metabolismo , Especificidad de la Especie , Distribución Tisular , Proteína 1 de Transporte Vesicular de Glutamato/genética , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(2): 690-5, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23267110

RESUMEN

Diabetic peripheral neuropathy (DPN) is the most common complication in both type 1 and type 2 diabetes. Here we studied some phenotypic features of a well-established animal model of type 2 diabetes, the leptin receptor-deficient db(-)/db(-) mouse, and also the effect of long-term (6 mo) treatment with coenzyme Q10 (CoQ10), an endogenous antioxidant. Diabetic mice at 8 mo of age exhibited loss of sensation, hypoalgesia (an increase in mechanical threshold), and decreases in mechanical hyperalgesia, cold allodynia, and sciatic nerve conduction velocity. All these changes were virtually completely absent after the 6-mo, daily CoQ10 treatment in db(-)/db(-) mice when started at 7 wk of age. There was a 33% neuronal loss in the lumbar 5 dorsal root ganglia (DRGs) of the db(-)/db(-) mouse versus controls at 8 mo of age, which was significantly attenuated by CoQ10. There was no difference in neuron number in 5/6-wk-old mice between diabetic and control mice. We observed a strong down-regulation of phospholipase C (PLC) ß3 in the DRGs of diabetic mice at 8 mo of age, a key molecule in pain signaling, and this effect was also blocked by the 6-mo CoQ10 treatment. Many of the phenotypic, neurochemical regulations encountered in lumbar DRGs in standard models of peripheral nerve injury were not observed in diabetic mice at 8 mo of age. These results suggest that reactive oxygen species and reduced PLCß3 expression may contribute to the sensory deficits in the late-stage diabetic db(-)/db(-) mouse, and that early long-term administration of the antioxidant CoQ10 may represent a promising therapeutic strategy for type 2 diabetes neuropathy.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Neuronas/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/prevención & control , Receptores de Leptina/deficiencia , Ubiquinona/análogos & derivados , Factores de Edad , Animales , Western Blotting , Diabetes Mellitus Tipo 2/patología , Estimulación Eléctrica , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Hiperalgesia/patología , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Conducción Nerviosa/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/etiología , Fosfolipasa C beta/metabolismo , Receptores de Leptina/genética , Nervio Ciático/lesiones , Nervio Ciático/patología , Estadísticas no Paramétricas , Ubiquinona/farmacología
18.
Proc Natl Acad Sci U S A ; 109(16): 6259-64, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22474393

RESUMEN

Expanding the repertoire of molecularly diverse neurons in the human nervous system is paramount to characterizing the neuronal networks that underpin sensory processing. Defining neuronal identities is particularly timely in the human olfactory system, whose structural differences from nonprimate macrosmatic species have recently gained momentum. Here, we identify clusters of bipolar neurons in a previously unknown outer "shell" domain of the human olfactory tract, which express secretagogin, a cytosolic Ca(2+) binding protein. These "shell" neurons are wired into the olfactory circuitry because they can receive mixed synaptic inputs. Unexpectedly, secretagogin is often coexpressed with polysialylated-neural cell adhesion molecule, ß-III-tubulin, and calretinin, suggesting that these neurons represent a cell pool that might have escaped terminal differentiation into the olfactory circuitry. We hypothesized that secretagogin-containing "shell" cells may be eliminated from the olfactory axis under neurodegenerative conditions. Indeed, the density, but not the morphological or neurochemical integrity, of secretagogin-positive neurons selectively decreases in the olfactory tract in Alzheimer's disease. In conclusion, secretagogin identifies a previously undescribed cell pool whose cytoarchitectonic arrangements and synaptic connectivity are poised to modulate olfactory processing in humans.


Asunto(s)
Proteínas de Unión al Calcio/biosíntesis , Diferenciación Celular , Neuronas/metabolismo , Vías Olfatorias/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Autopsia , Recuento de Células , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Vías Olfatorias/citología , Secretagoginas , Sinaptofisina/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(44): 18108-13, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22025706

RESUMEN

The anorectic anx/anx mouse exhibits disturbed feeding behavior and aberrances, including neurodegeneration, in peptidergic neurons in the appetite regulating hypothalamic arcuate nucleus. Poor feeding in infants, as well as neurodegeneration, are common phenotypes in human disorders caused by dysfunction of the mitochondrial oxidative phosphorylation system (OXPHOS). We therefore hypothesized that the anorexia and degenerative phenotypes in the anx/anx mouse could be related to defects in the OXPHOS. In this study, we found reduced efficiency of hypothalamic OXPHOS complex I assembly and activity in the anx/anx mouse. We also recorded signs of increased oxidative stress in anx/anx hypothalamus, possibly as an effect of the decreased hypothalamic levels of fully assembled complex I, that were demonstrated by native Western blots. Furthermore, the Ndufaf1 gene, encoding a complex I assembly factor, was genetically mapped to the anx interval and found to be down-regulated in anx/anx mice. These results suggest that the anorexia and hypothalamic neurodegeneration of the anx/anx mouse are associated with dysfunction of mitochondrial complex I.


Asunto(s)
Anorexia/fisiopatología , Hipotálamo/fisiopatología , Mitocondrias/fisiología , Alelos , Animales , Anorexia/genética , Hipotálamo/metabolismo , Ratones , Mitocondrias/metabolismo , Fosforilación Oxidativa , Estrés Oxidativo
20.
Proc Natl Acad Sci U S A ; 108(41): 17201-6, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21949401

RESUMEN

Although brain-derived neurotrophic factor (BDNF) is known to regulate circuit development and synaptic plasticity, its exact role in neuronal network activity remains elusive. Using mutant mice (TrkB-PV(-/-)) in which the gene for the BDNF receptor, tyrosine kinase B receptor (trkB), has been specifically deleted in parvalbumin-expressing, fast-spiking GABAergic (PV+) interneurons, we show that TrkB is structurally and functionally important for the integrity of the hippocampal network. The amplitude of glutamatergic inputs to PV+ interneurons and the frequency of GABAergic inputs to excitatory pyramidal cells were reduced in the TrkB-PV(-/-) mice. Functionally, rhythmic network activity in the gamma-frequency band (30-80 Hz) was significantly decreased in hippocampal area CA1. This decrease was caused by a desynchronization and overall reduction in frequency of action potentials generated in PV+ interneurons of TrkB-PV(-/-) mice. Our results show that the integration of PV+ interneurons into the hippocampal microcircuit is impaired in TrkB-PV(-/-) mice, resulting in decreased rhythmic network activity in the gamma-frequency band.


Asunto(s)
Región CA1 Hipocampal/fisiología , Interneuronas/fisiología , Receptor trkB/fisiología , Potenciales de Acción , Animales , Factor Neurotrófico Derivado del Encéfalo/fisiología , Región CA1 Hipocampal/citología , Recuento de Células , Dendritas/fisiología , Neuronas GABAérgicas/fisiología , Ratones , Ratones Noqueados , Red Nerviosa/citología , Red Nerviosa/fisiología , Parvalbúminas/metabolismo , Receptor trkB/deficiencia , Receptor trkB/genética , Transducción de Señal , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...