Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 410
Filtrar
1.
Anal Chem ; 92(19): 13281-13289, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32880432

RESUMEN

Cell-type-specific metabolic profiling in tissue with heterogeneous composition has been of great interest across all mass spectrometry imaging (MSI) technologies. We report here a powerful new chemical imaging capability in desorption electrospray ionization (DESI) MSI, which enables cell-type-specific and in situ metabolic profiling in complex tissue samples. We accomplish this by combining DESI-MSI with immunofluorescence staining using specific cell-type markers. We take advantage of the variable frequency of each distinct cell type in the lateral septal nucleus (LSN) region of mouse forebrain. This allows computational deconvolution of the cell-type-specific metabolic profile in neurons and astrocytes by convex optimization-a machine learning method. Based on our approach, we observed 107 metabolites that show different distributions and intensities between astrocytes and neurons. We subsequently identified 23 metabolites using high-resolution mass spectrometry (MS) and tandem MS, which include small metabolites such as adenosine and N-acetylaspartate previously associated with astrocytes and neurons, respectively, as well as accumulation of several phospholipid species in neurons which have not been studied before. Overall, this method overcomes the relatively low spatial resolution of DESI-MSI and provides a new platform for in situ metabolic investigation at the cell-type level in complex tissue samples with heterogeneous cell-type composition.


Asunto(s)
Astrocitos/metabolismo , Técnica del Anticuerpo Fluorescente , Prosencéfalo/metabolismo , Animales , Astrocitos/química , Astrocitos/citología , Aprendizaje Automático , Ratones , Neuronas/química , Neuronas/citología , Neuronas/metabolismo , Prosencéfalo/química , Prosencéfalo/citología , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Coloración y Etiquetado
2.
Cereb Cortex ; 29(2): 505-516, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29293918

RESUMEN

SEC14 and Spectrin domain-1 (Sestd1) is a synapse protein that exhibits a striking shift from the presynaptic to postsynaptic space as neurons mature postnatally in the mouse hippocampus. Hippocampal pyramidal neurons from mice with global genetic deletion of Sestd1 have reduced dendrite arbors, spines, and excitatory synapses. Electrophysiologically this correlates with cell-autonomous reductions in both AMPA- and NMDA-excitatory postsynaptic currents in individual hippocampal neurons from which Sestd1 has been deleted in vivo. These neurodevelopmental and functional deficits are associated with increased activation of the Rho family GTPases Rac1 and RhoA. Co-immunoprecipitation and mass spectrometry reveal that the Breakpoint Cluster Region protein, a Rho GTPase activating protein (GAP), forms complexes with Sestd1 in brain tissue. This complements earlier findings that Sestd1 can also partner with other Rho family GAPs and guanine nucleotide exchange factors. Our findings demonstrate that Sestd1 is a developmentally dynamic synaptic regulator of Rho GTPases that contributes to dendrite and excitatory synapse formation within differentiating pyramidal neurons of the forebrain.


Asunto(s)
Proteínas Portadoras/metabolismo , Espinas Dendríticas/metabolismo , Neuropéptidos/metabolismo , Prosencéfalo/metabolismo , Proteínas Proto-Oncogénicas c-bcr/metabolismo , Sinapsis/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Proteínas Portadoras/análisis , Dendritas/química , Dendritas/metabolismo , Espinas Dendríticas/química , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neurogénesis/fisiología , Neuropéptidos/análisis , Técnicas de Cultivo de Órganos , Prosencéfalo/química , Prosencéfalo/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-bcr/análisis , Sinapsis/química , Proteína de Unión al GTP rac1/análisis
3.
Yi Chuan ; 40(5): 390-401, 2018 May 20.
Artículo en Zh | MEDLINE | ID: mdl-29785947

RESUMEN

Cortical neuron migration in the developing mouse forebrain is a complex process, which contains several steps related to cytoskeleton dynamics and remodeling. Neural Wiskott-Aldrich syndrome protein (N-WASP), a member of the WASP-WAVE family, regulates actin cytoskeleton reorganization through the binding of its VCA domain to the Arp2/3 complex. Here we report expression patterns of N-WASP gene in the mouse developing embryonic cortex (E12.5~ E18.5) and find its expression levels are decreased during embryonic development. By using in utero electroporation (IUE) method, we find that either N-WASP overexpression or knockdown impairs cortical neuron migration, and the defects of cortical neuron migration caused by N-WASP overexpression are much more severe than that by its knockdown. N-WASP protein contains four domains: WH1, GBD, polyPro, and VCA. We generated a series of dominant negative N-WASP mutants by modifying these domains. Overexpression of N-WASP mutant lacking domain polyPro, VCA, or WH1, impairs cortical neuron migration. However, overexpression of N-WASP with the H208D point mutation, which abolishes the Cdc42 binding to N-WASP, causes only a marginal defect of cortical neuron migration. Finally, overexpression of the individual domain polyPro or VCA, but not WH1, can recapitulate the defects by N-WASP overexpression. However, overexpression of WH1-GBD fragment has no apparent effect on cortical neuron migration. In conclusion, our data demonstrate that N-WASP regulates cortical neuron migration mainly through its polyPro and VCA domains.


Asunto(s)
Corteza Cerebral/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/química , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Movimiento Celular , Corteza Cerebral/embriología , Ratones , Ratones Endogámicos C57BL , Neuronas/química , Prosencéfalo/química , Prosencéfalo/metabolismo , Dominios Proteicos , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética
4.
RNA Biol ; 14(8): 1064-1074, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27982734

RESUMEN

Circular RNAs (circRNAs) are broadly identified from precursor mRNA (pre-mRNA) back-splicing across various species. Recent studies have suggested a cell-/tissue- specific manner of circRNA expression. However, the distinct expression pattern of circRNAs among species and its underlying mechanism still remain to be explored. Here, we systematically compared circRNA expression from human and mouse, and found that only a small portion of human circRNAs could be determined in parallel mouse samples. The conserved circRNA expression between human and mouse is correlated with the existence of orientation-opposite complementary sequences in introns that flank back-spliced exons in both species, but not the circRNA sequences themselves. Quantification of RNA pairing capacity of orientation-opposite complementary sequences across circRNA-flanking introns by Complementary Sequence Index (CSI) identifies that among all types of complementary sequences, SINEs, especially Alu elements in human, contribute the most for circRNA formation and that their diverse distribution across species leads to the increased complexity of circRNA expression during species evolution. Together, our integrated and comparative reference catalog of circRNAs in different species reveals a species-specific pattern of circRNA expression and suggests a previously under-appreciated impact of fast-evolved SINEs on the regulation of (circRNA) gene expression.


Asunto(s)
Empalme Alternativo , Evolución Biológica , ARN/genética , Elementos de Nucleótido Esparcido Corto , Animales , Emparejamiento Base , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Cerebelo/química , Cerebelo/metabolismo , Corteza Cerebral/química , Corteza Cerebral/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Exones , Perfilación de la Expresión Génica , Humanos , Intrones , Hígado/química , Hígado/metabolismo , Ratones , Miocardio/química , Miocardio/metabolismo , Especificidad de Órganos , Prosencéfalo/química , Prosencéfalo/metabolismo , ARN/química , ARN/metabolismo , ARN Circular , Rombencéfalo/química , Rombencéfalo/metabolismo , Especificidad de la Especie
5.
Bull Exp Biol Med ; 162(3): 293-294, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28091924

RESUMEN

Zinc level in the blood plasma and brain of rats was studied by inductively coupled plasma mass spectrometry. Maximum amount of zinc was observed in the cerebellum (15.0±5.5 µg/mg wet tissue). Single intraperitoneal administration of a zinc donor acyzol (24 mg/kg) did not change the content of this element in the tissues. Repeated injections of acyzol (7 injections over 14 days) significantly increased zinc level in rat plasma and brain. This elevation was most pronounced in the forebrain (cortex and subcortical structures). The rise in zinc concentration in blood plasma correlated with its level in the brain.


Asunto(s)
Cerebelo/metabolismo , Prosencéfalo/metabolismo , Acetato de Zinc/administración & dosificación , Zinc/administración & dosificación , Animales , Cerebelo/química , Complejos de Coordinación/administración & dosificación , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Esquema de Medicación , Imidazoles/química , Inyecciones Intraperitoneales , Masculino , Prosencéfalo/química , Ratas , Ratas Wistar , Zinc/metabolismo , Acetato de Zinc/química , Acetato de Zinc/metabolismo
6.
J Neurosci ; 35(18): 7041-55, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25948256

RESUMEN

Sensory nerves innervating the mucosa of the airways monitor the local environment for the presence of irritant stimuli and, when activated, provide input to the nucleus of the solitary tract (Sol) and paratrigeminal nucleus (Pa5) in the medulla to drive a variety of protective behaviors. Accompanying these behaviors are perceivable sensations that, particularly for stimuli in the proximal end of the airways, can be discrete and localizable. Airway sensations likely reflect the ascending airway sensory circuitry relayed via the Sol and Pa5, which terminates broadly throughout the CNS. However, the relative contribution of the Sol and Pa5 to these ascending pathways is not known. In the present study, we developed and characterized a novel conditional anterograde transneuronal viral tracing system based on the H129 strain of herpes simplex virus 1 and used this system in rats along with conventional neuroanatomical tracing with cholera toxin B to identify subcircuits in the brainstem and forebrain that are in receipt of relayed airway sensory inputs via the Sol and Pa5. We show that both the Pa5 and proximal airways disproportionately receive afferent terminals arising from the jugular (rather than nodose) vagal ganglia and the output of the Pa5 is predominately directed toward the ventrobasal thalamus. We propose the existence of a somatosensory-like pathway from the proximal airways involving jugular ganglia afferents, the Pa5, and the somatosensory thalamus and suggest that this pathway forms the anatomical framework for sensations arising from the proximal airway mucosa.


Asunto(s)
Tronco Encefálico/fisiología , Red Nerviosa/fisiología , Técnicas de Trazados de Vías Neuroanatómicas/métodos , Prosencéfalo/fisiología , Células Receptoras Sensoriales/fisiología , Tráquea/fisiología , Animales , Tronco Encefálico/química , Herpesvirus Humano 1 , Masculino , Red Nerviosa/química , Prosencéfalo/química , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/química , Sinapsis/química , Sinapsis/fisiología , Tráquea/química , Tráquea/inervación
7.
J Neurochem ; 131(2): 147-62, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24985044

RESUMEN

We systematically investigated the purification process of post-synaptic density (PSD) and post-synaptic membrane rafts (PSRs) from the rat forebrain synaptic plasma membranes by examining the components and the structures of the materials obtained after the treatment of synaptic plasma membranes with TX-100, n-octyl ß-d-glucoside (OG) or 3-([3-cholamidopropyl]dimethylammonio)-2-hydroxy-1-propanesulfonate (CHAPSO). These three detergents exhibited distinct separation profiles for the synaptic subdomains. Type I and type II PSD proteins displayed mutually exclusive distribution. After TX-100 treatment, type I PSD was recovered in two fractions: a pellet and an insoluble fraction 8, which contained partially broken PSD-PSR complexes. Conventional PSD was suggested to be a mixture of these two PSD pools and did not contain type II PSD. An association of type I PSD with PSRs was identified in the TX-100 treatment, and those with type II PSD in the OG and CHAPSO treatments. An association of GABA receptors with gephyrin was easily dissociated. OG at a high concentration solubilized the type I PSD proteins. CHAPSO treatment resulted in a variety of distinct fractions, which contained certain novel structures. Two different pools of GluA, either PSD or possibly raft-associated, were identified in the OG and CHAPSO treatments. These results are useful in advancing our understanding of the structural organization of synapses at the molecular level. We systematically investigated the purification process of post-synaptic density (PSD) and synaptic membrane rafts by examining the structures obtained after treatment of the SPMs with TX-100, n-octyl ß-d-glucoside or CHAPSO. Differential distribution of type I and type II PSD, synaptic membrane rafts, and other novel subdomains in the SPM give clues to understand the structural organization of synapses at the molecular level.


Asunto(s)
Detergentes/farmacología , Microdominios de Membrana/ultraestructura , Densidad Postsináptica/ultraestructura , Prosencéfalo/ultraestructura , Membranas Sinápticas/ultraestructura , Animales , Masculino , Microdominios de Membrana/química , Microdominios de Membrana/efectos de los fármacos , Datos de Secuencia Molecular , Densidad Postsináptica/química , Densidad Postsináptica/efectos de los fármacos , Prosencéfalo/química , Prosencéfalo/efectos de los fármacos , Ratas , Ratas Wistar , Membranas Sinápticas/química , Membranas Sinápticas/efectos de los fármacos
8.
Cell Mol Neurobiol ; 34(3): 419-35, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24436034

RESUMEN

D-Serine, a co-agonist at the NMDA receptor (NMDAR), is synthesized from L-serine by the enzyme serine racemase (SR), which is heavily expressed in the forebrain. Although SR was originally reported to be localized exclusively to astrocytes, recent conditional knock out results demonstrate that little SR is expressed in forebrain astrocytes. As a consequence, the cellular location of its product, D-serine, in the brain is also uncertain. Immunocytochemistry now indicates that SR is expressed primarily in forebrain glutamatergic neurons with the remainder in GABAergic interneurons. We utilized SR deficient (SR-/-) mice, which have <15 % of normal D-serine levels, to validate and optimize a D-serine immunohistochemical method. Nearly all of the D-serine in neocortex and hippocampus (HP) is found in neurons, with virtually no D-serine co-localizing with two astrocyte markers. Interestingly, only a subset of the D-serine positive neurons contained SR in the neocortex and HP. Greater than half of the D-serine positive neurons were GABAergic interneurons, with a majority of these neurons containing parvalbumin and/or somatostatin. Only ~25-40 % of interneurons expressed SR in the neocortex and HP. Finally, we demonstrate in human post-mortem neocortex that SR is found in both excitatory and inhibitory neurons, but not in S100ß-containing astrocytes. In sum, these findings conclusively demonstrate that the majority of D-serine is both synthesized and stored in neurons. It will be important to determine the functional significance for the separation of synthesis and storage of D-serine in neurons, as well as the presence of this NMDAR co-agonist in GABAergic interneurons.


Asunto(s)
Neuronas/química , Prosencéfalo/química , Racemasas y Epimerasas/análisis , Serina/análisis , Factores de Edad , Anciano , Animales , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neuronas/enzimología , Neuronas/patología , Prosencéfalo/enzimología , Prosencéfalo/patología , Racemasas y Epimerasas/biosíntesis , Serina/biosíntesis
9.
Eur J Neurosci ; 36(6): 2789-800, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22731249

RESUMEN

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


Asunto(s)
Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Prosencéfalo/patología , Ratas , Animales , Peso Corporal , Factor Neurotrófico Derivado del Encéfalo/análisis , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Glutamina/genética , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Hipotálamo/química , Cuerpos de Inclusión/patología , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Actividad Motora , Mutación , Proteínas del Tejido Nervioso/análisis , Neuropéptidos/análisis , Neuropéptidos/genética , Proteínas Nucleares/análisis , Orexinas , Prosencéfalo/química , Prosencéfalo/fisiopatología , Ratas Sprague-Dawley
10.
Eur J Neurosci ; 35(5): 661-72, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22390178

RESUMEN

Neuronal progenitors are continuously generated in the postnatal rodent subventricular zone and migrate along the rostral migratory stream to supply interneurons in the olfactory bulb. Nonsynaptic GABAergic signaling affects the postnatal neurogenesis by depolarizing neuronal progenitors, which depends on an elevated intracellular Cl(-) concentration. However, the molecular mechanism responsible for Cl(-) accumulation in these cells still remains elusive. Using confocal Ca(2+) imaging, we found that GABA depolarization-induced Ca(2+) increase was either abolished by bumetanide, a specific inhibitor of the Na(+) -K(+) -2Cl(-) cotransporter, or reduced by partial replacement of extracellular Na(+) with Li(+) , in the HEPES buffer but not in the CO(2)/HCO3⁻ buffer. GABA depolarization-induced Ca(2+) increase in CO(2)/HCO3⁻ buffer was abolished by a combination of bumetanide with the anion exchanger inhibitor DIDS or with the carbonic anhydrase inhibitor acetozalimide. Using gramicidin-perforated patch-clamp recording, we further confirmed that bumetanide, together with DIDS or acetozalimide, reduced the intracellular chloride concentration in the neuronal progenitors. In addition, with BrdU labeling, we demonstrated that blocking of the Na(+) -K(+) -2Cl(-) cotransporter, but not anion exchangers, reduced the proliferation of neuronal progenitors. Our results indicate that both the Na(+) -K(+) -2Cl(-) cotransporter and anion exchangers contribute to the elevated intracellular chloride responsible for the depolarizing action of GABA in the postnatal forebrain neuronal progenitors. However, the Na(+) -K(+) -2Cl(-) cotransporter displays an additional effect on neuronal progenitor proliferation.


Asunto(s)
Antiportadores/metabolismo , Cloruros/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Prosencéfalo/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Animales , Animales Recién Nacidos , Homeostasis/fisiología , Ratones , Células-Madre Neurales/química , Células-Madre Neurales/fisiología , Neuronas/química , Neuronas/fisiología , Prosencéfalo/química , Prosencéfalo/citología , Miembro 2 de la Familia de Transportadores de Soluto 12
11.
Neuroinformatics ; 20(4): 1121-1136, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35792992

RESUMEN

Neuronal networks are regulated by three-dimensional spatial and structural properties. Despite robust evidence of functional implications in the modulation of cognition, little is known about the three-dimensional internal organization of cholinergic networks in the forebrain. Cholinergic networks in the forebrain primarily occur in subcortical nuclei, specifically the septum, nucleus basalis, globus pallidus, nucleus accumbens, and the caudate-putamen. Therefore, the present investigation analyzed the three-dimensional spatial organization of 14,000 cholinergic neurons that expressed choline acetyltransferase (ChAT) in these subcortical nuclei of the mouse forebrain. Point process theory and graph signal processing techniques identified three topological principles of organization. First, cholinergic interneuronal distance is not uniform across brain regions. Specifically, in the septum, globus pallidus, nucleus accumbens, and the caudate-putamen, the cholinergic neurons were clustered compared with a uniform random distribution. In contrast, in the nucleus basalis, the cholinergic neurons had a spatial distribution of greater regularity than a uniform random distribution. Second, a quarter of the caudate-putamen is composed of axonal bundles, yet the spatial distribution of cholinergic neurons remained clustered when axonal bundles were accounted for. However, comparison with an inhomogeneous Poisson distribution showed that the nucleus basalis and caudate-putamen findings could be explained by density gradients in those structures. Third, the number of cholinergic neurons varies as a function of the volume of a specific brain region but cell body volume is constant across regions. The results of the present investigation provide topographic descriptions of cholinergic somata distribution and axonal conduits, and demonstrate spatial differences in cognitive control networks. The study provides a comprehensive digital database of the total population of ChAT-positive neurons in the reported structures, with the x,y,z coordinates of each neuron at micrometer resolution. This information is important for future digital cellular atlases and computational models of the forebrain cholinergic system enabling models based on actual spatial geometry.


Asunto(s)
Colina O-Acetiltransferasa , Globo Pálido , Animales , Ratones , Colina O-Acetiltransferasa/análisis , Colina O-Acetiltransferasa/metabolismo , Globo Pálido/química , Globo Pálido/metabolismo , Núcleo Accumbens/química , Núcleo Accumbens/metabolismo , Putamen/química , Putamen/metabolismo , Prosencéfalo/química , Prosencéfalo/metabolismo , Neuronas Colinérgicas/química , Neuronas Colinérgicas/metabolismo , Colinérgicos/análisis , Análisis Espacial
12.
Cell Biochem Funct ; 29(2): 114-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21321971

RESUMEN

IgLONs are a family of four GPI-anchored cell adhesion molecules that regulate neurite outgrowth and synaptogenesis and may act as tumour suppressor genes. Recently we have proposed that two members of the IgLON family act as a heterodimeric complex termed DIgLON. Neurons isolated from chick forebrain co-express all six combinations of IgLONs and the intensity of fluorescence for each pair of IgLONs was highly correlated. Antibody-patching experiments on forebrain neurons show complex formation for IgLON pairs but not between unrelated GPI-anchored glycoproteins. Thus IgLONs are the first GPI-anchored family of glycoproteins shown to form heterodimeric complexes in the plane of the membrane.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Familia de Multigenes , Neuronas/metabolismo , Prosencéfalo/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/química , Pollos , Dimerización , Regulación del Desarrollo de la Expresión Génica , Neuronas/química , Prosencéfalo/química , Prosencéfalo/embriología
13.
Proc Natl Acad Sci U S A ; 105(4): 1249-54, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18212120

RESUMEN

Although prairie voles (Microtus ochrogaster) are socially monogamous, males vary in both sexual and spatial fidelity. Most males form pairbonds, cohabit with one female, and defend territories. Wandering males, in contrast, have expansive home ranges that overlap many males and females. In the laboratory, pairing is regulated by arginine vasopressin and its predominant CNS receptor, vasopressin 1a receptor (V1aR). We investigated individual differences in forebrain V1aR expression of male prairie voles in mixed-sex seminatural enclosures. Individual differences in V1aR were compared with space use measured by radio telemetry and paternity determined with microsatellite markers. Animals engaging in extra-pair fertilizations (EPFs) as either wanderers or paired residents overlapped significantly more in same- and opposite-sex home ranges. Surprisingly, neither social fidelity measured by space use nor sexual fidelity measured by paternity was associated with V1aR expression in the ventral pallidum (VPall) or lateral septum, areas causally related to pairbond formation. In contrast, V1aR expression in the posterior cingulate/retrosplenial cortex (PCing) and laterodorsal thalamus (LDThal), areas implicated in spatial memory, strongly covaried with space use and paternity. Animals engaging in EPFs either as wanderers or paired residents exhibited low levels of LDThal and PCing V1aR expression. Individual differences in brain and behavior parallel differences between prairie voles and promiscuous congeners. The concordance among space use, paternity, and V1aR in spatial circuits suggests a common link between the mechanisms of spatial behaviors and success at EPF. The combined data demonstrate how organismal biology can inform our understanding of individual and species differences in behavioral mechanisms.


Asunto(s)
Arvicolinae/fisiología , Ecosistema , Variación Genética , Neuronas/metabolismo , Receptores de Vasopresinas/fisiología , Conducta Sexual Animal , Territorialidad , Animales , Arvicolinae/genética , Química Encefálica/genética , Química Encefálica/fisiología , Femenino , Fenómenos de Retorno al Lugar Habitual/fisiología , Masculino , Neuronas/química , Neuronas/fisiología , Paternidad , Fenotipo , Valor Predictivo de las Pruebas , Prosencéfalo/química , Prosencéfalo/metabolismo , Prosencéfalo/fisiología , Receptores de Vasopresinas/genética
14.
J Comp Neurol ; 529(9): 2243-2264, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33340092

RESUMEN

Eupnea is generated by neural circuits located in the ponto-medullary brainstem, but can be modulated by higher brain inputs which contribute to volitional control of breathing and the expression of orofacial behaviors, such as vocalization, sniffing, coughing, and swallowing. Surprisingly, the anatomical organization of descending inputs that connect the forebrain with the brainstem respiratory network remains poorly defined. We hypothesized that descending forebrain projections target multiple distributed respiratory control nuclei across the neuroaxis. To test our hypothesis, we made discrete unilateral microinjections of the retrograde tracer cholera toxin subunit B in the midbrain periaqueductal gray (PAG), the pontine Kölliker-Fuse nucleus (KFn), the medullary Bötzinger complex (BötC), pre-BötC, or caudal midline raphé nuclei. We quantified the regional distribution of retrogradely labeled neurons in the forebrain 12-14 days postinjection. Overall, our data reveal that descending inputs from cortical areas predominantly target the PAG and KFn. Differential forebrain regions innervating the PAG (prefrontal, cingulate cortices, and lateral septum) and KFn (rhinal, piriform, and somatosensory cortices) imply that volitional motor commands for vocalization are specifically relayed via the PAG, while the KFn may receive commands to coordinate breathing with other orofacial behaviors (e.g., sniffing, swallowing). Additionally, we observed that the limbic or autonomic (interoceptive) systems are connected to broadly distributed downstream bulbar respiratory networks. Collectively, these data provide a neural substrate to explain how volitional, state-dependent, and emotional modulation of breathing is regulated by the forebrain.


Asunto(s)
Bulbo Raquídeo/fisiología , Mesencéfalo/fisiología , Neuronas/fisiología , Puente/fisiología , Prosencéfalo/fisiología , Mecánica Respiratoria/fisiología , Animales , Femenino , Masculino , Bulbo Raquídeo/química , Mesencéfalo/química , Microinyecciones/métodos , Vías Nerviosas/química , Vías Nerviosas/fisiología , Neuronas/química , Puente/química , Prosencéfalo/química , Trazadores Radiactivos , Ratas , Ratas Sprague-Dawley
15.
Horm Behav ; 58(3): 544-53, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20399213

RESUMEN

Male songbirds often establish territories and attract mates by singing, and some song features can reflect the singer's condition or quality. The quality of the song environment can change, so male songbirds should benefit from assessing the competitiveness of the song environment and appropriately adjusting their own singing behavior and the neural substrates by which song is controlled. In a wide range of taxa, social modulation of behavior is partly mediated by the arginine vasopressin or vasotocin (AVP/AVT) systems. To examine the modulation of singing behavior in response to the quality of the song environment, we compared the song output of laboratory-housed male Lincoln's sparrows (Melospiza lincolnii) exposed to 1 week of chronic playback of songs categorized as either high or low quality, based on song length, complexity, and trill performance. To explore the neural basis of any facultative shifts in behavior, we also quantified the subjects' AVT immunoreactivity (AVT-IR) in three forebrain regions that regulate sociosexual behavior: the medial bed nucleus of the stria terminalis (BSTm), the lateral septum (LS), and the preoptic area. We found that high-quality songs increased singing effort and reduced AVT-IR in the BSTm and LS, relative to low-quality songs. The effect of the quality of the song environment on both singing effort and forebrain AVT-IR raises the hypothesis that AVT within these brain regions plays a role in the modulation of behavior in response to competition that individual males may assess from the prevailing song environment.


Asunto(s)
Prosencéfalo/fisiología , Conducta Sexual Animal/fisiología , Gorriones/fisiología , Vasotocina/fisiología , Vocalización Animal/fisiología , Animales , Masculino , Prosencéfalo/química , Medio Social , Vasotocina/análisis
16.
J Comp Neurol ; 528(8): 1321-1348, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31760659

RESUMEN

Ventralization, a major patterning process in the developing vertebrate neural tube (central nervous system, CNS), depends on Sonic hedgehog (SHH) as a main signaling morphogen. We studied the CNS of late larval and young adult zebrafish in a transgenic shh-GFP line revealing increased neuroanatomical detail due to the progressed differentiation state compared to earlier stages. Some major findings emerge from the present study. (a) shh -GFP is still expressed along the adult zebrafish CNS neuraxis in most locations seen in larvae. (b) We newly identify a ventroposterior shh pallidal domain representing the basal telencephalic signaling center important for basal ganglia development known in other vertebrates (i.e., the anterior entopeduncular area-basal medial ganglionic eminence of mammals). (c) We further show late-emerging shh-GFP positive radial glia cells in the medial zone of the dorsal telencephalon (i.e., the teleostan pallial amygdala). (d) Immunostains for tyrosine hydroxylase demonstrate that there is selective colocalization in adult dopamine cells with shh-GFP in the posterior tuberculum, including in projection cells to striatum, which represents a striking parallel to amniote mesodiencephalic dopamine cell origin from shh expressing floor plate cells. (e) There is no colocalization of shh and islet1 as shown by respective shh-GFP and islet1-GFP lines. (f) The only radially far migrated shh-GFP cells are located in the preglomerular area. (g) There are no adult cerebellar and tectal shh-GFP cells confirming their exclusive role during early development as previously reported by our laboratory.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Globo Pálido/metabolismo , Proteínas Hedgehog/biosíntesis , Prosencéfalo/metabolismo , Telencéfalo/metabolismo , Proteínas de Pez Cebra/biosíntesis , Animales , Animales Modificados Genéticamente , Neuronas Dopaminérgicas/química , Expresión Génica , Globo Pálido/química , Proteínas Hedgehog/análisis , Proteínas Hedgehog/genética , Prosencéfalo/química , Transducción de Señal/fisiología , Telencéfalo/química , Pez Cebra , Proteínas de Pez Cebra/análisis , Proteínas de Pez Cebra/genética
17.
J Comp Neurol ; 528(15): 2551-2568, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32220012

RESUMEN

GABAA receptors are composed of five subunits arranged around a central chloride channel. Their subunits originate from different genes or gene families. The majority of GABAA receptors in the mammalian brain consist of two α-, two ß- and one γ- or δ-subunit. This subunit organization crucially determines the physiological and pharmacological properties of the GABAA receptors. Using immunohistochemistry, we investigated the distribution of 10 GABAA receptor subunits (α1, α2, α3, α4, α5, ß1, ß2, ß3, γ2, and δ) in the fore brain of three female rhesus monkeys (Macaca mulatta). Within the cerebral cortex, subunits α1, α5, ß2, ß3, and γ2 were found in all layers, α2, α3, and ß1 were more concentrated in the inner and outer layers. The caudate/putamen was rich in α1, α2, α5, all three ß-subunits, γ2, and δ. Subunits α3 and α5 were more concentrated in the caudate than in the putamen. In contrast, α1, α2, ß1, ß2, γ2, and δ were highest in the pallidum. Most dorsal thalamic nuclei contained subunits α1, α2, α4, ß2, ß3, and γ2, whereas α1, α3, ß1, and γ2 were most abundant in the reticular nucleus. Within the amygdala, subunits α1, α2, α5, ß1, ß3, γ2, and δ were concentrated in the cortical nucleus, whereas in the lateral and basolateral amygdala α1, α2, α5, ß1, ß3, and δ, and in the central amygdala α1, α2, ß3, and γ2 were most abundant. Interestingly, subunit α3-IR outlined the intercalated nuclei of the amygdala. In the hippocampus, subunits α1, α2, α5, ß2, ß3, γ2, and δ were highly expressed in the dentate molecular layer, whereas α1, α2, α3, α5, ß1, ß2, ß3, and γ2 were concentrated in sector CA1 and the subiculum. The distribution of GABAA receptor subunits in the rhesus monkey was highly heterogeneous indicating a high number of differently assembled receptors. In most areas investigated, notably in the striatum/pallidum, amygdaloid nuclei and in the hippocampus it was more diverse than in the rat and mouse indicating a more heterogeneous and less defined receptor assembly in the monkey than in rodent brain.


Asunto(s)
Prosencéfalo/química , Prosencéfalo/metabolismo , Subunidades de Proteína/biosíntesis , Receptores de GABA-A/biosíntesis , Factores de Edad , Secuencia de Aminoácidos , Animales , Femenino , Inmunohistoquímica , Macaca mulatta , Subunidades de Proteína/análisis , Subunidades de Proteína/genética , Receptores de GABA-A/análisis , Receptores de GABA-A/genética
18.
Neuropharmacology ; 177: 108237, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32710978

RESUMEN

Major depressive disorder (MDD) is a severe mental disorder with a high disability rate worldwide. Selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs) are the most common agents for antidepressant use. SSRIs and SNRIs are believed to achieve antidepressant effects through the activation of serotonergic or noradrenergic systems. However, whether the dopaminergic system is involved remains unclear. In our study, a genetically encoded dopamine sensor and in vivo fiber photometry recordings were used to measure the dopamine concentrations in the medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) after acute intraperitoneal injection of SSRIs or SNRIs. Combined with the behavioral tests, we found that SNRIs increased dopamine concentrations in both the mPFC and the NAc and showed faster antidepressant effects than SSRIs. To verify the enhanced dopamine levels induce the faster antidepressant effects of SNRIs, we employed dopamine receptor antagonists to specifically block the dopaminergic function. The results showed that the faster antidepressant effects of SNRIs were weakened by the dopamine receptor antagonists. Altogether, our study reveals that SNRIs achieve faster antidepressant effects than SSRIs by elevating the dopamine concentrations in the mPFC and the NAc. Our work proposes further mechanisms for the first-line antidepressants, which provides more basis for clinical treatments. This article is part of the special issue on Stress, Addiction and Plasticity.


Asunto(s)
Antidepresivos/administración & dosificación , Depresión/metabolismo , Dopamina/metabolismo , Prosencéfalo/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores de Captación de Serotonina y Norepinefrina/administración & dosificación , Animales , Depresión/tratamiento farmacológico , Depresión/psicología , Dopamina/análisis , Femenino , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Fotometría/métodos , Prosencéfalo/química , Prosencéfalo/efectos de los fármacos , Restricción Física/efectos adversos , Restricción Física/psicología , Resultado del Tratamiento
19.
J Neurosci ; 28(44): 11354-9, 2008 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-18971477

RESUMEN

The serotonergic system, including the serotonin 1A (5-HT(1A)) receptor, has been implicated in the pathophysiology of a number of neuropsychiatric disorders. Current data show substantial interindividual variation in the regional concentration of this receptor site, the source of which is unclear. Monoamine oxidase A (MAO-A) is a key regulator of serotonin metabolism, and polymorphic variation in the X-linked MAO-A gene influences its expression. We hypothesized that polymorphism in the MAO-A gene would be associated with sex-specific variation in 5-HT(1A) receptor expression. We used positron emission tomography and [(11)C]WAY-100635 to quantify 5-HT(1A) receptors in a group of 31 healthy and unmedicated depressed individuals. The same individuals were genotyped for an upstream variable number tandem repeat polymorphism in the promoter of the MAO-A gene. ANOVA of 5-HT(1A) receptor availability demonstrated a significant effect of MAO-A genotype in the raphe nuclei, medial and inferior temporal cortex, insula, medial prefrontal cortex, and anterior cingulate (p < 0.05). The effect persisted when age, race, body mass index, and diagnosis were included in the model. Genotypes with greater putative MAO-A activity were associated with greater 5-HT(1A) receptor availability in women, but not in men. Genotype predicted a substantial 42-74% of the variance in receptor availability in women, depending on the brain region (p < 0.05). Depression diagnosis was not associated with MAO-A genotype or 5-HT(1A) receptor availability in these regions. These results demonstrate a sex-specific interaction between two key molecules of the human serotonergic system, and suggest a neurobiological basis for sexual dimorphism in serotonin-modulated phenotypes.


Asunto(s)
Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Receptor de Serotonina 5-HT1A/genética , Receptor de Serotonina 5-HT1A/metabolismo , Adulto , Tronco Encefálico/química , Tronco Encefálico/metabolismo , Trastorno Depresivo/enzimología , Trastorno Depresivo/genética , Trastorno Depresivo/metabolismo , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo Genético/fisiología , Valor Predictivo de las Pruebas , Prosencéfalo/química , Prosencéfalo/metabolismo , Unión Proteica/fisiología , Caracteres Sexuales
20.
Neurosci Lett ; 453(1): 16-20, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19429007

RESUMEN

Krüppel-like factor 6 (KLF6) is a transcriptional regulator that shows widespread distribution in the peripheral organs of the body. However, it remains uncertain where KLF6 is expressed in the adult forebrain under physiological conditions. Therefore, the present study investigated the spatial patterns of KLF6 expression and identified cell types expressing KLF6 in the forebrain. KLF6 immunoreactivity was widely seen throughout the forebrain including the olfactory bulb, cerebral cortex, hippocampus, septum, amygdala, basal ganglia, thalamus, and hypothalamus. Moreover, KLF6-positive cells were also detected in the radial migratory stream (RMS) and subventricular zone. Immunofluorescent double-labeling revealed that KLF6-immunoreactive cells were co-localized with neuronal nuclei or platelet endothelial cell adhesion molecule-1, a mature neuronal and endothelial marker, respectively, in most forebrain regions. In the RMS, KLF6 was co-expressed with polysialic neural cell adhesion molecule, a marker of neuronal progenitor cells. This is the first report showing that KLF6 protein is expressed in various regions of the adult forebrain and KLF6-positive cells manifest neuronal or endothelial phenotypes under physiological conditions.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/análisis , Prosencéfalo/química , Proteínas Proto-Oncogénicas/análisis , Animales , Núcleo Celular/química , Proteínas de Unión al ADN , Expresión Génica , Inmunohistoquímica , Factor 6 Similar a Kruppel , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/análisis , Molécula L1 de Adhesión de Célula Nerviosa/análisis , Moléculas de Adhesión de Célula Nerviosa/análisis , Neuronas/química , Neuronas/ultraestructura , Proteínas Nucleares/análisis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Ácidos Siálicos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA