Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
eNeuro ; 11(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38834299

RESUMEN

Viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), use respiratory epithelial cells as an entry point for infection. Within the nasal cavity, the olfactory epithelium (OE) is particularly sensitive to infections which may lead to olfactory dysfunction. In patients suffering from coronavirus disease 2019, deficits in olfaction have been characterized as a distinctive symptom. Here, we used the K18hACE2 mice to study the spread of SARS-CoV-2 infection and inflammation in the olfactory system (OS) after 7 d of infection. In the OE, we found that SARS-CoV-2 selectively targeted the supporting/sustentacular cells (SCs) and macrophages from the lamina propria. In the brain, SARS-CoV-2 infected some microglial cells in the olfactory bulb (OB), and there was a widespread infection of projection neurons in the OB, piriform cortex (PC), and tubular striatum (TuS). Inflammation, indicated by both elevated numbers and morphologically activated IBA1+ cells (monocyte/macrophage lineages), was preferentially increased in the OE septum, while it was homogeneously distributed throughout the layers of the OB, PC, and TuS. Myelinated OS axonal tracts, the lateral olfactory tract, and the anterior commissure, exhibited decreased levels of 2',3'-cyclic-nucleotide 3'-phosphodiesterase, indicative of myelin defects. Collectively, our work supports the hypothesis that SARS-CoV-2 infected SC and macrophages in the OE and, centrally, microglia and subpopulations of OS neurons. The observed inflammation throughout the OS areas and central myelin defects may account for the long-lasting olfactory deficit.


Asunto(s)
COVID-19 , Vaina de Mielina , Bulbo Olfatorio , Mucosa Olfatoria , SARS-CoV-2 , Animales , COVID-19/patología , COVID-19/complicaciones , Ratones , Mucosa Olfatoria/patología , Mucosa Olfatoria/virología , Bulbo Olfatorio/patología , Bulbo Olfatorio/virología , Vaina de Mielina/patología , Vaina de Mielina/metabolismo , Microglía/patología , Microglía/metabolismo , Microglía/virología , Ratones Transgénicos , Enzima Convertidora de Angiotensina 2/metabolismo , Trastornos del Olfato/patología , Trastornos del Olfato/virología , Modelos Animales de Enfermedad , Masculino , Inflamación/patología , Inflamación/virología , Macrófagos/patología , Femenino
2.
J Neurosci ; 43(6): 1051-1071, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36596700

RESUMEN

Parkinson's disease (PD) is characterized by multiple symptoms including olfactory dysfunction, whose underlying mechanisms remain unclear. Here, we explored pathologic changes in the olfactory pathway of transgenic (Tg) mice of both sexes expressing the human A30P mutant α-synuclein (α-syn; α-syn-Tg mice) at 6-7 and 12-14 months of age, representing early and late-stages of motor progression, respectively. α-Syn-Tg mice at late stages exhibited olfactory behavioral deficits, which correlated with severe α-syn pathology in projection neurons (PNs) of the olfactory pathway. In parallel, olfactory bulb (OB) neurogenesis in α-syn-Tg mice was reduced in the OB granule cells at six to seven months and OB periglomerular cells at 12-14 months, respectively, both of which could contribute to olfactory dysfunction. Proteomic analyses showed a disruption in endocytic and exocytic pathways in the OB during the early stages which appeared exacerbated at the synaptic terminals when the mice developed olfactory deficits at 12-14 months. Our data suggest that (1) the α-syn-Tg mice recapitulate the olfactory functional deficits seen in PD; (2) olfactory structures exhibit spatiotemporal disparities for vulnerability to α-syn pathology; (3) α-syn pathology is restricted to projection neurons in the olfactory pathway; (4) neurogenesis in adult α-syn-Tg mice is reduced in the OB; and (5) synaptic endocytosis and exocytosis defects in the OB may further explain olfactory deficits.SIGNIFICANCE STATEMENT Olfactory dysfunction is a characteristic symptom of Parkinson's disease (PD). Using the human A30P mutant α-synuclein (α-syn)-expressing mouse model, we demonstrated the appearance of olfactory deficits at late stages of the disease, which was accompanied by the accumulation of α-syn pathology in projection neurons (PNs) of the olfactory system. This dysfunction included a reduction in olfactory bulb (OB) neurogenesis as well as changes in synaptic vesicular transport affecting synaptic function, both of which are likely contributing to olfactory behavioral deficits.


Asunto(s)
Trastornos del Olfato , Enfermedad de Parkinson , Masculino , Femenino , Ratones , Humanos , Animales , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Olfato , Proteómica , Ratones Transgénicos , Neurogénesis , Trastornos del Olfato/genética , Modelos Animales de Enfermedad
3.
eNeuro ; 10(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36697258

RESUMEN

Microglia invade the neuroblast migratory corridor of the rostral migratory stream (RMS) early in development. The early postnatal RMS does not yet have the dense astrocyte and vascular scaffold that helps propel forward migrating neuroblasts, which led us to consider whether microglia help regulate conditions permissive to neuroblast migration in the RMS. GFP-labeled microglia in CX3CR-1GFP/+ mice assemble primarily along the outer borders of the RMS during the first postnatal week, where they exhibit predominantly an ameboid morphology and associate with migrating neuroblasts. Microglia ablation for 3 d postnatally does not impact the density of pulse labeled BrdU+ neuroblasts nor the distance migrated by tdTomato electroporated neuroblasts in the RMS. However, microglia wrap DsRed-labeled neuroblasts in the RMS of P7 CX3CR-1GFP/+;DCXDsRed/+ mice and express the markers CD68, CLEC7A, MERTK, and IGF-1, suggesting active regulation in the developing RMS. Microglia depletion for 14 d postnatally further induced an accumulation of CC3+ DCX+ apoptotic neuroblasts in the RMS, a wider RMS and extended patency of the lateral ventricle extension in the olfactory bulb. These findings illustrate the importance of microglia in maintaining a healthy neuroblast population and an environment permissive to neuroblast migration in the early postnatal RMS.


Asunto(s)
Microglía , Células-Madre Neurales , Ratones , Animales , Células-Madre Neurales/fisiología , Ventrículos Laterales , Movimiento Celular/fisiología , Bulbo Olfatorio/fisiología
4.
Nat Neurosci ; 25(9): 1119, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35902651
5.
Front Cell Neurosci ; 15: 658480, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33994949

RESUMEN

Odor stimuli consist of thousands of possible molecules, each molecule with many different properties, each property a dimension of the stimulus. Processing these high dimensional stimuli would appear to require many stages in the brain to reach odor perception, yet, in mammals, after the sensory receptors this is accomplished through only two regions, the olfactory bulb and olfactory cortex. We take a first step toward a fundamental understanding by identifying the sequence of local operations carried out by microcircuits in the pathway. Parallel research provided strong evidence that processed odor information is spatial representations of odor molecules that constitute odor images in the olfactory bulb and odor objects in olfactory cortex. Paleontology provides a unique advantage with evolutionary insights providing evidence that the basic architecture of the olfactory pathway almost from the start ∼330 million years ago (mya) has included an overwhelming input from olfactory sensory neurons combined with a large olfactory bulb and olfactory cortex to process that input, driven by olfactory receptor gene duplications. We identify a sequence of over 20 microcircuits that are involved, and expand on results of research on several microcircuits that give the best insights thus far into the nature of the high dimensional processing.

6.
J Neurophysiol ; 124(2): 375-387, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32639901

RESUMEN

The first compartmental computer models of brain neurons using the Rall method predicted novel and unexpected dendrodendritic interactions between mitral and granule cells in the olfactory bulb. We review the models from a 50-year perspective on the work that has challenged, supported, and extended the original proposal that these interactions mediate both lateral inhibition and oscillatory activity, essential steps in the neural basis of olfactory processing and perception. We highlight strategies behind the neurophysiological experiments and the Rall methods that enhance the ability of detailed compartmental modeling to give counterintuitive predictions that lead to deeper insights into neural organization at the synaptic and circuit level. The application of these methods to mechanisms of neurogenesis and plasticity are exciting challenges for the future.


Asunto(s)
Ondas Encefálicas/fisiología , Dendritas/fisiología , Modelos Teóricos , Inhibición Neural/fisiología , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Bulbo Olfatorio/fisiología , Percepción Olfatoria/fisiología , Sinapsis/fisiología , Animales
7.
eNeuro ; 6(5)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31554664

RESUMEN

The formation of the olfactory nerve and olfactory bulb (OB) glomeruli begins embryonically in mice. However, the development of the olfactory system continues throughout life with the addition of new olfactory sensory neurons (OSNs) in the olfactory epithelium (OE). Much attention has been given to the perinatal innervation of the OB by OSN axons, but in the young adult the process of OSN maturation and axon targeting to the OB remains controversial. To address this gap in understanding, we used BrdU to label late-born OSNs in young adult mice at postnatal day 25 (P25-born OSNs) and timed their molecular maturation following basal cell division. We show that OSNs in young adults undergo a sequential molecular development with the expression of GAP 43 (growth-associated protein 43) > AC3 (adenylyl cyclase 3) > OMP (olfactory marker protein), consecutively, in a time frame of ∼8 d. To assess OSN axon development, we implemented an in vivo fate-mapping strategy to label P25-born OSNs with ZsGreen. Using sampling intervals of 24 h, we demonstrate the progressive extension of OSN axons in the OE, through the foramen of the cribriform plate, and onto the surface of the OB. OSN axons reached the OB and began to target and robustly innervate specific glomeruli ∼10 d following basal cell division, a time point at which OMP expression becomes evident. Our data demonstrate a sequential process of correlated axon extension and molecular maturation that is similar to that seen in the neonate, but on a slightly longer timescale and with regional differences in the OE.


Asunto(s)
Bulbo Olfatorio/citología , Bulbo Olfatorio/crecimiento & desarrollo , Mucosa Olfatoria/citología , Mucosa Olfatoria/crecimiento & desarrollo , Neuronas Receptoras Olfatorias/citología , Animales , Ratones , Neurogénesis/fisiología
8.
Mol Cell Neurosci ; 98: 82-96, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31200100

RESUMEN

The olfactory tubercle (OT) is located in the ventral-medial region of the brain where it receives primary input from olfactory bulb (OB) projection neurons and processes olfactory behaviors related to motivation, hedonics of smell and sexual encounters. The OT is part of the dopamine reward system that shares characteristics with the striatum. Together with the nucleus accumbens, the OT has been referred to as the "ventral striatum". However, despite its functional importance little is known about the embryonic development of the OT and the phenotypic properties of the OT cells. Here, using thymidine analogs, we establish that mouse OT neurogenesis occurs predominantly between E11-E15 in a lateral-to-medial gradient. Then, using a piggyBac multicolor technique we characterized the migratory route of OT neuroblasts from their embryonic point of origin. Following neurogenesis in the ventral lateral ganglionic eminence (vLGE), neuroblasts destined for the OT followed a dorsal-ventral pathway we named "ventral migratory course" (VMC). Upon reaching the nascent OT, neurons established a prototypical laminar distribution that was determined, in part, by the progenitor cell of origin. A phenotypic analysis of OT neuroblasts using a single-color piggyBac technique, showed that OT shared the molecular specification of striatal neurons. In addition to primary afferent input from the OB, the OT also receives a robust dopaminergic input from ventral tegmentum (Ikemoto, 2007). We used tyrosine hydroxylase (TH) expression as a proxy for dopaminergic innervation and showed that TH onset occurs at E13 and progressively increased until postnatal stages following an 'inside-out' pattern. Postnatally, we established the myelination in the OT occurring between P7 and P14, as shown with CNPase staining, and we characterized the cellular phenotypes populating the OT by immunohistochemistry. Collectively, this work provides the first detailed analysis of the developmental and maturation processes occurring in mouse OT, and demonstrates the striatal nature of the OT as part of the ventral striatum (vST).


Asunto(s)
Neurogénesis , Tubérculo Olfatorio/embriología , Animales , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Femenino , Masculino , Ratones , Vaina de Mielina/metabolismo , Tubérculo Olfatorio/citología , Tubérculo Olfatorio/crecimiento & desarrollo
9.
Cell Rep ; 26(10): 2805-2817.e9, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30840899

RESUMEN

Heterozygous coding mutations in TRIO are associated with neurodevelopmental disorders, including autism, schizophrenia, bipolar disorder, and epilepsy, and impair TRIO's biochemical activities. To model mutant alleles, we ablated one or both Trio alleles from excitatory neurons in the cortex and hippocampus of mice. Trio haploinsufficiency increases anxiety and impairs social preference and motor coordination. Trio loss reduces forebrain size and dendritic arborization but increases dendritic spine densities. Cortical synapses in Trio haploinsufficient mice are small, exhibit pre- and postsynaptic deficits, and cannot undergo long-term potentiation. Similar phenotypes are observed in Trio knockout mice. Overall, Trio haploinsufficiency causes severe disease-relevant deficits in behavior and neuronal structure and function. Interestingly, phosphodiesterase 4A5 (PDE4A5) levels are reduced and protein kinase A (PKA) signaling is increased when TRIO levels are reduced. Elevation of PDE4A5 and drug-based attenuation of PKA signaling rescue Trio haploinsufficiency-related dendritic spine defects, suggesting an avenue for therapeutic intervention for TRIO-related neurodevelopmental disorders.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Trastornos del Neurodesarrollo/genética , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Sinapsis/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Noqueados
10.
J Comp Neurol ; 527(7): 1278-1289, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30628061

RESUMEN

Many functions of glial cells depend on the formation of selective glial networks mediated by gap junctions formed by members of the connexin family. Olfactory ensheathing cells (OECs) are specialized glia associated with olfactory sensory neuron axons. Like other glia, they form selective networks, however, the connexins that support OEC connectivity in vivo have not been identified. We used an in vivo mouse model to selectively delete candidate connexin genes with temporal control from OECs and address the physiological consequences. Using this model, we effectively abolished the expression of connexin 43 (Cx43) in OECs in both juvenile and adult mice. Cx43-deleted OECs exhibited features consistent with the loss of gap junctions including reduced membrane conductance, largely reduced sensitivity to the gap junction blocker meclofenamic acid and loss of dye coupling. This indicates that Cx43, a typically astrocytic connexin, is the main connexin forming functional channels in OECs. Despite these changes in functional properties, the deletion of Cx43 deletion did not alter the density of OECs. The strategy used here may prove useful to delete other candidate genes to better understand the functional roles of OECs in vivo.


Asunto(s)
Conexina 43/fisiología , Uniones Comunicantes/fisiología , Técnicas de Inactivación de Genes , Neuroglía/fisiología , Bulbo Olfatorio/citología , Envejecimiento/metabolismo , Animales , Conexina 43/deficiencia , Conexina 43/genética , Cruzamientos Genéticos , Femenino , Uniones Comunicantes/efectos de los fármacos , Genes Reporteros , Genes Sintéticos , Integrasas/genética , Masculino , Ácido Meclofenámico/farmacología , Ratones , Ratones Noqueados , Proteína Proteolipídica de la Mielina/genética , Bulbo Olfatorio/metabolismo , Técnicas de Placa-Clamp , Tamoxifeno/farmacología
11.
Cereb Cortex ; 29(1): 1-16, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29136113

RESUMEN

Piriform cortex (PC) is a 3-layer paleocortex receiving primary afferent input from the olfactory bulb. The past decade has seen significant progress in understanding the synaptic, cellular and functional organization of PC, but PC embryogenesis continues to be enigmatic. Here, using birthdating strategies and clonal analyses, we probed the early development and laminar specificity of neurogenesis/gliogenesis as it relates to the organization of the PC. Our data demonstrate a temporal sequence of laminar-specific neurogenesis following the canonical "inside-out" pattern, with the notable exception of PC Layer II which exhibited an inverse "outside-in" temporal neurogenic pattern. Of interest, we found no evidence of a neurogenic gradient along the anterior to posterior axis, although the timing of neuronal migration and laminar development was delayed rostrally by approximately 24 h. To begin probing if lineage affected cell fate in the PC, we labeled PC neuroblasts using a multicolor technique and analyzed their laminar organization. Our results suggested that PC progenitors were phenotypically committed to reach specific layers early in the development. Collectively, these studies shed new light on the determinants of the laminar specificity of neuronal/glial organization in PC and the likely role of subpopulations of committed progenitors in regulating PC embryogenesis.


Asunto(s)
Linaje de la Célula/fisiología , Movimiento Celular/fisiología , Neurogénesis/fisiología , Neuroglía/fisiología , Corteza Piriforme/citología , Corteza Piriforme/crecimiento & desarrollo , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Embarazo
12.
Front Cell Neurosci ; 12: 295, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233329

RESUMEN

Voltage-gated calcium (Cav) channels are a prerequisite for signal transmission at the first olfactory sensory neuron (OSN) synapse within the glomeruli of the main olfactory bulb (MOB). We showed previously that the N-type Cav channel subunit Cav2.2 is present in the vast majority of glomeruli and plays a central role in presynaptic transmitter release. Here, we identify a distinct subset of glomeruli in the MOB of adult mice that is characterized by expression of the P/Q-type channel subunit Cav2.1. Immunolocalization shows that Cav2.1+ glomeruli reside predominantly in the medial and dorsal MOB, and in the vicinity of the necklace glomerular region close to the accessory olfactory bulb. Few glomeruli are detected on the ventral and lateral MOB. Cav2.1 labeling in glomeruli colocalizes with the presynaptic marker vGlut2 in the axon terminals of OSNs. Electron microscopy shows that Cav2.1+ presynaptic boutons establish characteristic asymmetrical synapses with the dendrites of second-order neurons in the glomerular neuropil. Cav2.1+ glomeruli receive axonal input from OSNs that express molecules of canonical OSNs: olfactory marker protein, the ion channel Cnga2, and the phosphodiesterase Pde4a. In the main olfactory epithelium, Cav2.1 labels a distinct subpopulation of OSNs whose distribution mirrors the topography of the MOB glomeruli, that shows the same molecular signature, and is already present at birth. Together, these experiments identify a unique Cav2.1+ multiglomerular domain in the MOB that may form a previously unrecognized olfactory subsystem distinct from other groups of necklace glomeruli that rely on cGMP signaling mechanisms.

13.
Brain Struct Funct ; 223(9): 4067-4085, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30141078

RESUMEN

The anterior commissure (AC) is a phylogenetically conserved inter-hemispheric connection found among vertebrates with bilateral symmetry. The AC connects predominantly olfactory areas but many aspects of its development and structure are unknown. To fill this gap, we investigated the embryonic and postnatal development of the AC by tracing axons with DiI and the piggyback transposon multicolor system. With this strategy, we show that axon growth during establishment of the AC follows a strictly regulated timeline of events that include waiting periods ("regressive strategies") as well as periods of active axon outgrowth ("progressive strategies"). We also provide evidence that these processes may be regulated in the midline via overexpression of chondroitin sulfate proteoglycans. Additionally, we demonstrate that the ipsi- and contralateral innervation of piriform cortex occurs simultaneously. Morphologically, we found that 20% of axons were myelinated by postnatal day (P) 22, in a process that occurred fundamentally around P14. By immunohistochemistry, we described the presence of glial cells and two new subtypes of neurons: one expressing a calretinin (CR)-/MAP2+ phenotype, distributed homogeneously inside the AC; and the other expressing a CR+/MAP2+ phenotype that lies beneath the bed nucleus of the stria terminalis. Our results are consistent with the notion that the AC follows a strictly regulated program during the embryonic and postnatal development similarly to other distal targeting axonal tracts.


Asunto(s)
Comisura Anterior Cerebral/embriología , Corteza Piriforme/embriología , Animales , Comisura Anterior Cerebral/ultraestructura , Axones/ultraestructura , Femenino , Masculino , Ratones , Vaina de Mielina/ultraestructura , Neuroglía/citología , Neuronas/citología , Corteza Piriforme/citología
14.
Cell Rep ; 24(6): 1523-1535, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30089263

RESUMEN

Hyperactivating mutations in the non-receptor tyrosine phosphatase SHP2 cause Noonan syndrome (NS). NS is associated with cognitive deficits, but how hyperactivation of SHP2 in NS changes neuron function is not well understood. We find that mice bearing an NS-associated SHP2 allele (NS mice) have selectively impaired Schaffer collateral-CA1 NMDA (N-methyl-D-aspartate) receptor (NMDAR)-mediated neurotransmission and that residual NMDAR-mediated currents decay faster in NS mice because of reduced contribution of GluN1:GluN2B diheteromers. Consistent with altered GluN2B function, we identify GluN2B Y1252 as an NS-associated SHP2 substrate both in vitro and in vivo. Mutation of Y1252 does not alter recombinant GluN1:GluN2B receptor kinetics. Instead, phospho-Y1252 binds the actin-regulatory adaptor protein Nck2, and this interaction is required for proper NMDAR function. These results establish SHP2 and Nck2 as NMDAR regulatory proteins and strongly suggest that NMDAR dysfunction contributes to NS cognitive deficits.


Asunto(s)
Síndrome de Noonan/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Síndrome de Noonan/metabolismo , Transducción de Señal
15.
Cell Rep ; 21(5): 1281-1292, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29091766

RESUMEN

Synapses in the developing brain are structurally dynamic but become stable by early adulthood. We demonstrate here that an α5-subunit-containing laminin stabilizes synapses during this developmental transition. Hippocampal neurons deposit laminin α5 at synapses during adolescence as connections stabilize. Disruption of laminin α5 in neurons causes dramatic fluctuations in dendritic spine head size that can be rescued by exogenous α5-containing laminin. Conditional deletion of laminin α5 in vivo increases dendritic spine size and leads to an age-dependent loss of synapses accompanied by behavioral defects. Remaining synapses have larger postsynaptic densities and enhanced neurotransmission. Finally, we provide evidence that laminin α5 acts through an integrin α3ß1-Abl2 kinase-p190RhoGAP signaling cascade and partners with laminin ß2 to regulate dendritic spine density and behavior. Together, our results identify laminin α5 as a stabilizer of dendritic spines and synapses in the brain and elucidate key cellular and molecular mechanisms by which it acts.


Asunto(s)
Laminina/metabolismo , Neuronas/metabolismo , Sinapsis/fisiología , Animales , Conducta Animal , Espinas Dendríticas/fisiología , Potenciales Evocados/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hibridación Fluorescente in Situ , Integrinas/metabolismo , Laminina/deficiencia , Laminina/genética , Ratones , Ratones Noqueados , Microscopía Electrónica , Técnicas de Placa-Clamp , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
16.
J Comp Neurol ; 525(7): 1743-1755, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28152579

RESUMEN

The ionotropic serotonin receptor, 5-HT3 , is expressed by many developing neurons within the central nervous system. Since the olfactory epithelium continues to generate new olfactory sensory neurons (OSNs) throughout life, we investigated the possibility that 5-HT3 is expressed in the adult epithelium. Using a transgenic mouse in which the promoter for the 5-HT3a subunit drives expression of green fluorescent protein (GFP), we assessed the expression of this marker in the olfactory epithelium of adult mice. Both the native 5-HT3a mRNA and GFP are expressed within globose basal cells of the olfactory and vomeronasal epithelium in adult mice. Whereas the 5-HT3a mRNA disappears relatively quickly after final cell division, the GFP label persists for about 5 days, thereby labeling immature OSNs in both the main olfactory system and vomeronasal organ. The GFP-labeled cells include both proliferative globose basal cells as well as immature OSNs exhibiting the hallmarks of ongoing differentiation including GAP43, PGP9.5, but the absence of olfactory marker protein. Some of the GFP-labeled OSNs show characteristics of more mature yet still developing OSNs including the presence of cilia extending from the apical knob and expression of NaV1.5, a component of the transduction cascade. These findings suggest that 5-HT3a is indicative of a proliferative or developmental state, regardless of age, and that the 5-HT3A GFP mice may prove useful for future studies of neurogenesis in the olfactory epithelium. J. Comp. Neurol. 525:1743-1755, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas Receptoras Olfatorias/citología , Receptores de Serotonina 5-HT3/biosíntesis , Células Madre Adultas/citología , Animales , Proteínas Fluorescentes Verdes , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Células-Madre Neurales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Mol Cell Neurosci ; 74: 78-86, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27073125

RESUMEN

Transmission of olfactory information to higher brain regions is mediated by olfactory bulb (OB) projection neurons, the mitral and tufted cells. Although mitral/tufted cells are often characterized as the OB counterpart of cortical projection neurons (also known as pyramidal neurons), they possess several unique morphological characteristics and project specifically to the olfactory cortices. Moreover, the molecular networks contributing to the generation of mitral/tufted cells during development are largely unknown. To understand the developmental patterns of gene expression in mitral/tufted cells in the OB, we performed transcriptome analyses targeting purified OB projection neurons at different developmental time points with next-generation RNA sequencing (RNA-seq). Through these analyses, we found 1202 protein-coding genes that are temporally differentially-regulated in developing projection neurons. Among them, 388 genes temporally changed their expression level only in projection neurons. The data provide useful resource to study the molecular mechanisms regulating development of mitral/tufted cells. We further compared the gene expression profiles of developing mitral/tufted cells with those of three cortical projection neuron subtypes, subcerebral projection neurons, corticothalamic projection neurons, and callosal projection neurons, and found that the molecular signature of developing olfactory projection neuron bears resemblance to that of subcerebral neurons. We also identified 3422 events that change the ratio of splicing isoforms in mitral/tufted cells during maturation. Interestingly, several genes expressed a novel isoform not previously reported. These results provide us with a broad perspective of the molecular networks underlying the development of OB projection neurons.


Asunto(s)
Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Transcriptoma , Animales , Regulación del Desarrollo de la Expresión Génica , Ratones , Bulbo Olfatorio/citología , Bulbo Olfatorio/embriología , Sistemas de Lectura Abierta
18.
Glia ; 63(9): 1646-59, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25856239

RESUMEN

The olfactory nerve is permissive for axon growth throughout life. This has been attributed in part to the olfactory ensheathing glial cells that encompass the olfactory sensory neuron fascicles. Olfactory ensheathing cells (OECs) also promote axon growth in vitro and when transplanted in vivo to sites of injury. The mechanisms involved remain largely unidentified owing in part to the limited knowledge of the physiological properties of ensheathing cells. Glial cells rely for many functions on the properties of the potassium channels expressed; however, those expressed in ensheathing cells are unknown. Here we show that OECs express voltage-dependent potassium currents compatible with inward rectifier (Kir ) and delayed rectifier (KDR ) channels. Together with gap junction coupling, these contribute to the heterogeneity of membrane properties observed in OECs. The relevance of K(+) currents expressed by ensheathing cells is discussed in relation to plasticity of the olfactory nerve.


Asunto(s)
Vaina de Mielina/fisiología , Nervio Olfatorio/citología , Nervio Olfatorio/fisiología , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Conexina 43/metabolismo , Femenino , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Inmunohistoquímica , Masculino , Ratones , Vaina de Mielina/efectos de los fármacos , Nervio Olfatorio/efectos de los fármacos , Técnicas de Placa-Clamp , Potasio/metabolismo , Técnicas de Cultivo de Tejidos
19.
Proc Natl Acad Sci U S A ; 112(18): 5821-6, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25902488

RESUMEN

Odorant receptors (OR) are strongly implicated in coalescence of olfactory sensory neuron (OSN) axons and the formation of olfactory bulb (OB) glomeruli. However, when ORs are first expressed relative to basal cell division and OSN axon extension is unknown. We developed an in vivo fate-mapping strategy that enabled us to follow OSN maturation and axon extension beginning at basal cell division. In parallel, we mapped the molecular development of OSNs beginning at basal cell division, including the onset of OR expression. Our data show that ORs are first expressed around 4 d following basal cell division, 24 h after OSN axons have reached the OB. Over the next 6+ days the OSN axons navigate the OB nerve layer and ultimately coalesce in glomeruli. These data provide a previously unidentified perspective on the role of ORs in homophilic OSN axon adhesion and lead us to propose a new model dividing axon extension into two phases. Phase I is OR-independent and accounts for up to 50% of the time during which axons approach the OB and begin navigating the olfactory nerve layer. Phase II is OR-dependent and concludes as OSN axons coalesce in glomeruli.


Asunto(s)
Axones/metabolismo , Bulbo Olfatorio/fisiología , Receptores Odorantes/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Electroporación , Proteína GAP-43/metabolismo , Inmunohistoquímica , Hibridación in Situ , Riñón/metabolismo , Ratones , Mitosis , Neurogénesis , Neuronas/metabolismo , Neuronas Aferentes/citología , Odorantes , Bulbo Olfatorio/citología , Nervio Olfatorio/citología , Neuronas Receptoras Olfatorias/metabolismo , Olfato/genética , Células Madre/citología , Tamoxifeno/química
20.
J Comp Neurol ; 523(8): 1145-61, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25420934

RESUMEN

Odor information relayed by olfactory bulb projection neurons, mitral and tufted cells (M/T), is modulated by pairs of reciprocal dendrodendritic synaptic circuits in the external plexiform layer (EPL). Interneurons, which are accounted for largely by granule cells, receive depolarizing input from M/T dendrites and in turn inhibit current spread in M/T dendrites via hyperpolarizing reciprocal dendrodendritic synapses. Because the location of dendrodendritic synapses may significantly affect the cascade of odor information, we assessed synaptic properties and density within sublaminae of the EPL and along the length of M/T secondary dendrites. In electron micrographs the M/T to granule cell synapse appeared to predominate and was equivalent in both the outer and inner EPL. However, the dendrodendritic synapses from granule cell spines onto M/T dendrites were more prevalent in the outer EPL. In contrast, individual gephyrin-immunoreactive (IR) puncta, a postsynaptic scaffolding protein at inhibitory synapses used here as a proxy for the granule to M/T dendritic synapse was equally distributed throughout the EPL. Of significance to the organization of intrabulbar circuits, gephyrin-IR synapses are not uniformly distributed along M/T secondary dendrites. Synaptic density, expressed as a function of surface area, increases distal to the cell body. Furthermore, the distributions of gephyrin-IR puncta are heterogeneous and appear as clusters along the length of the M/T dendrites. Consistent with computational models, our data suggest that temporal coding in M/T cells is achieved by precisely located inhibitory input and that distance from the soma is compensated for by an increase in synaptic density.


Asunto(s)
Dendritas/ultraestructura , Bulbo Olfatorio/ultraestructura , Sinapsis/ultraestructura , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Dendritas/metabolismo , Femenino , Inmunohistoquímica , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica , Bulbo Olfatorio/anatomía & histología , Bulbo Olfatorio/metabolismo , Fotomicrografía , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...