Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell Tissue Res ; 375(1): 217-225, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30377783

RESUMEN

Peptides released from trigeminal fibers fulfill well-understood functions in neuroinflammatory processes and in the modulation of nociceptive signal processing. In particular, calcitonin gene-related peptide (CGRP) and substance P (SP), released from afferent nerve terminals, exert paracrine effects on the surrounding tissue and this has been recently highlighted by the prominent parcrine role of CGRP in the development of headache and migraine. Some recent communications suggest that these sensory neuropeptides may also modulate the workings of sensory organs and influence afferent signals from nose, tongue, eyes and ears. Here, we briefly review the evidence for modulatory effects of CGRP and SP in the sensory periphery.


Asunto(s)
Neuropéptidos/metabolismo , Sensación/fisiología , Animales , Humanos , Modelos Biológicos , Percepción
2.
Pflugers Arch ; 470(9): 1335-1348, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29860639

RESUMEN

Physiological processes of vital importance are often safeguarded by compensatory systems that substitute for primary processes in case these are damaged by gene mutation. Ca2+-dependent Cl- secretion in airway epithelial cells may provide such a compensatory mechanism for impaired Cl- secretion via cystic fibrosis transmembrane conductance regulator (CFTR) channels in cystic fibrosis (CF). Anoctamin 1 (ANO1) Ca2+-gated Cl- channels are known to contribute to calcium-dependent Cl- secretion in tracheal and bronchial epithelia. In the present study, two mouse models of CF were examined to assess a potential protective function of Ca2+-dependent Cl- secretion, a CFTR deletion model (cftr-/-), and a CF pathology model that overexpresses the epithelial Na+ channel ß-subunit (ßENaC), which is encoded by the Scnn1b gene, specifically in airway epithelia (Scnn1b-Tg). The expression levels of ANO1 were examined by mRNA and protein content, and the channel protein distribution between ciliated and non-ciliated epithelial cells was analyzed. Moreover, Ussing chamber experiments were conducted to compare Ca2+-dependent Cl- secretion between wild-type animals and the two mouse models. Our results demonstrate that CFTR and ANO1 channels were co-expressed with ENaC in non-ciliated cells of mouse tracheal and bronchial epithelia. Ciliated cells did not express these proteins. Despite co-localization of CFTR and ANO1 in the same cell type, cells in cftr-/- mice displayed no altered expression of ANO1. Similarly, ANO1 expression was unaffected by ßENaC overexpression in the Scnn1b-Tg line. These results suggest that the CF-related environment in the two mouse models did not induce ANO1 overexpression as a compensatory system.


Asunto(s)
Anoctamina-1/metabolismo , Calcio/metabolismo , Canales de Cloruro/metabolismo , Fibrosis Quística/metabolismo , Animales , Bronquios/metabolismo , Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Epitelio/metabolismo , Femenino , Transporte Iónico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mucosa Respiratoria/metabolismo , Transducción de Señal/fisiología , Tráquea/metabolismo
3.
Eur J Neurosci ; 45(4): 587-600, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27891688

RESUMEN

Chemosensation in the mammalian nose comprises detection of odorants, irritants and pheromones. While the traditional view assigned one distinct sub-system to each stimulus type, recent research has produced a more complex picture. Odorants are not only detected by olfactory sensory neurons but also by the trigeminal system. Irritants, in turn, may have a distinct odor, and some pheromones are detected by the olfactory epithelium. Moreover, it is well established that irritants change odor perception and vice versa. A wealth of psychophysical evidence on olfactory-trigeminal interactions in humans contrasts with a paucity of structural insight. In particular, it is unclear whether the two systems communicate just by sharing stimuli, or whether neuronal connections mediate cross-modal signaling. One connection could exist in the olfactory bulb that performs the primary processing of olfactory signals and receives trigeminal innervation. In the present study, neuroanatomical tracing of the mouse ethmoid system illustrates how peptidergic fibers enter the glomerular layer of the olfactory bulb, where local microcircuits process and filter the afferent signal. Biochemical assays reveal release of calcitonin gene-related peptide from olfactory bulb slices and attenuation of cAMP signaling by the neuropeptide. In the non-stimulated tissue, the neuropeptide specifically inhibited the basal activity of calbindin-expressing periglomerular interneurons, but did not affect the basal activity of neurons expressing calretinin, parvalbumin, or tyrosine hydroxylase, nor the activity of astrocytes. This study represents a first step towards understanding trigeminal neuromodulation of olfactory-bulb microcircuits and provides a working hypothesis for trigeminal inhibition of olfactory signal processing. This article is protected by copyright. All rights reserved.

4.
Cerebellum ; 16(5-6): 929-937, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28536821

RESUMEN

Neurons communicate through excitatory and inhibitory synapses. Both lines of communication are adjustable and allow the fine tuning of signal exchange required for learning processes in neural networks. Several distinct modes of plasticity modulate glutamatergic and GABAergic synapses in Purkinje cells of the cerebellar cortex to promote motor control and learning. In the present paper, we present evidence for a role of short-term ionic plasticity in the cerebellar circuit activity. This type of plasticity results from altered chloride driving forces at the synapses that molecular layer interneurons form on Purkinje cell dendrites. Previous studies have provided evidence for transiently diminished chloride gradients at these GABAergic synapses following climbing fiber activity. Electrical stimulation of climbing fibers in acute slices caused a decline of inhibitory postsynaptic currents recorded from Purkinje cells. Dendritic calcium-gated chloride channels of the type anoctamin 2 (ANO2) were proposed to mediate this short-term modulation of inhibition, but the significance of this process for motor control has not been established yet. Here, we report results of behavioral studies obtained from Ano2 -/- mice, a mouse line that was previously shown to lack this particular mode of ionic plasticity. The animals display motor coordination deficits that constitute a condition of mild ataxia. Moreover, motor learning is severely impaired in Ano2 -/- mice, suggesting cerebellar dysfunction. This reduced motor performance of Ano2 -/- mice highlights the significance of inhibitory control for cerebellar function and introduces calcium-dependent short-term ionic plasticity as an efficient control mechanism for neural inhibition.


Asunto(s)
Anoctaminas/deficiencia , Aprendizaje/fisiología , Actividad Motora/fisiología , Trastornos del Movimiento/metabolismo , Animales , Anoctaminas/genética , Cerebelo/metabolismo , Cerebelo/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos del Movimiento/patología , Fuerza Muscular/fisiología
5.
J Physiol ; 594(13): 3501-16, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26857095

RESUMEN

KEY POINTS: In olfactory research it is difficult to deliver stimuli with defined intensity and duration to olfactory sensory neurons. Expression of channelrhodopsin 2 (ChR2) in olfactory sensory neurons provides a means to activate these neurons with light flashes. Appropriate mouse models are available. The present study explores the suitability of an established olfactory marker protein (OMP)/ChR2-yellow fluorescent protein (YFP) mouse model for ex vivo experimentation. Expression of ChR2 in sensory neurons of the main olfactory epithelium, the septal organ and vomeronasal organ is characterized. Expression pattern of ChR2 in olfactory receptor neurons and the properties of light responses indicate that light stimulation does not impact on signal transduction in the chemosensory cilia. Light-induced electro-olfactograms are characterized with light flashes of different intensities, durations and frequencies. The impact of light-induced afferent stimulation on the olfactory bulb is examined with respect to response amplitude, polarity and low-pass filtering. ABSTRACT: For the examination of sensory processing, it is helpful to deliver stimuli in precisely defined temporal and spatial patterns with accurate control of stimulus intensity. This is challenging in experiments with the mammalian olfactory system because airborne odorants have to be transported into the intricate sensory structures of the nose and must dissolve in mucus to be detected by sensory neurons. Defined and reproducible activity can be generated in olfactory sensory neurons that express the light-gated ion channel channelrhodopsin 2 (ChR2). The neurons can be stimulated by light flashes in a controlled fashion by this optogenetic approach. Here we examined the application of an olfactory marker protein (OMP)/ChR2-yellow fluorescent protein (YFP) model for ex vivo exploration of the olfactory epithelium and the olfactory bulb of the mouse. We studied the expression patterns of ChR2 in the main olfactory system, the vomeronasal system, and the septal organ, and we found that ChR2 is absent from the sensory cilia of olfactory sensory neurons. In the olfactory epithelium, we characterized light-induced electro-olfactograms with respect to peripheral encoding of stimulus intensity, stimulus duration and stimulus frequency. In acute slices of the olfactory bulb, we identified specific aspects of the ChR2-induced input signal, concerning its dynamic range, its low-pass filter property and its response to prolonged stimulation. Our study describes the performance of the OMP/ChR2-YFP model for ex vivo experimentation on the peripheral olfactory system and documents its versatility and its limitations for olfactory research.


Asunto(s)
Bulbo Olfatorio/fisiología , Mucosa Olfatoria/fisiología , Animales , Proteínas Bacterianas/fisiología , Channelrhodopsins , Luz , Proteínas Luminiscentes/fisiología , Masculino , Ratones , Modelos Animales , Optogenética , Estimulación Luminosa
6.
Proc Natl Acad Sci U S A ; 113(40): 11063-11065, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27660238

Asunto(s)
Cloruros , Olfato , Humanos , Odorantes
7.
Eur J Neurosci ; 37(4): 572-82, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23205840

RESUMEN

The mammalian olfactory epithelium contains olfactory receptor neurons and trigeminal sensory endings. The former mediate odor detection, the latter the detection of irritants. The two apparently parallel chemosensory systems are in reality interdependent in various well-documented ways. Psychophysical studies have shown that virtually all odorants can act as irritants, and that most irritants have an odor. Thus, the sensory perception of odorants and irritants is based on simultaneous input from the two systems. Moreover, functional interactions between the olfactory system and the trigeminal system exist on both peripheral and central levels. Here we examine the impact of trigeminal stimulation on the odor response of olfactory receptor neurons. Using an odorant with low trigeminal potency (phenylethyl alcohol) and a non-odorous irritant (CO(2) ), we have explored this interaction in psychophysical experiments with human subjects and in electroolfactogram (EOG) recordings from rats. We have demonstrated that simultaneous activation of the trigeminal system attenuates the perception of odor intensity and distorts the EOG response. On the molecular level, we have identified a route for this cross-modal interaction. The neuropeptide calcitonin-gene related peptide (CGRP), which is released from trigeminal sensory fibres upon irritant stimulation, inhibits the odor response of olfactory receptor neurons. CGRP receptors expressed by these neurons mediate this neuromodulatory effect. This study demonstrates a site of trigeminal-olfactory interaction in the periphery. It reveals a pathway for trigeminal impact on olfactory signal processing that influences odor perception.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Percepción Olfatoria/fisiología , Neuronas Receptoras Olfatorias/metabolismo , Transducción de Señal/fisiología , Adulto , Animales , Electrooculografía , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Hibridación in Situ , Irritantes/farmacología , Masculino , Odorantes , Mucosa Olfatoria/metabolismo , Ratas , Receptores Odorantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Trigémino/fisiología , Adulto Joven
8.
Proc Natl Acad Sci U S A ; 107(13): 6052-7, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20231443

RESUMEN

The mammalian olfactory system detects an unlimited variety of odorants with a limited set of odorant receptors. To cope with the complexity of the odor world, each odorant receptor must detect many different odorants. The demand for low odor selectivity creates problems for the transduction process: the initial transduction step, the synthesis of the second messenger cAMP, operates with low efficiency, mainly because odorants bind only briefly to their receptors. Sensory cilia of olfactory receptor neurons have developed an unusual solution to this problem. They accumulate chloride ions at rest and discharge a chloride current upon odor detection. This chloride current amplifies the receptor potential and promotes electrical excitation. We have studied this amplification process by examining identity, subcellular localization, and regulation of its molecular components. We found that the Na(+)/K(+)/2Cl(-) cotransporter NKCC1 is expressed in the ciliary membrane, where it mediates chloride accumulation into the ciliary lumen. Gene silencing experiments revealed that the activity of this transporter depends on the kinases SPAK and OSR1, which are enriched in the cilia together with their own activating kinases, WNK1 and WNK4. A second Cl(-) transporter, the Cl(-)/HCO(3)(-) exchanger SLC4A1, is expressed in the cilia and may support Cl(-) accumulation. The calcium-dependent chloride channel TMEM16B (ANO2) provides a ciliary pathway for the excitatory chloride current. These findings describe a specific set of ciliary proteins involved in anion-based signal amplification. They provide a molecular concept for the unique strategy that allows olfactory sensory neurons to operate as efficient transducers of weak sensory stimuli.


Asunto(s)
Neuronas Receptoras Olfatorias/fisiología , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Secuencia de Bases , Cloruros/metabolismo , Cilios/fisiología , Cartilla de ADN/genética , Retroalimentación Fisiológica , Silenciador del Gen , Transporte Iónico , Ratones , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor , Modelos Neurológicos , Fosforilación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Receptores Odorantes/fisiología , Transducción de Señal/fisiología , Olfato/fisiología , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12 , Proteína Quinasa Deficiente en Lisina WNK 1
9.
Cell Tissue Res ; 347(2): 327-41, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22314846

RESUMEN

Calcium-activated chloride channels are expressed in chemosensory neurons of the nose and contribute to secretory processes and sensory signal transduction. These channels are thought to be members of the family of anoctamins (alternative name: TMEM16 proteins), which are opened by micromolar concentrations of intracellular Ca(2+). Two family members,ANO 1 (TMEM16A) and ANO 2 (TMEM16B), are expressed in the various sensory and respiratory tissues of the nose.We have examined the tissue specificity and sub-cellular localization of these channels in the nasal respiratory epithelium and in the five chemosensory organs of the nose: the main olfactory epithelium, the septal organ of Masera, the vomeronasal organ, the Grueneberg ganglion and the trigeminal system. We have found that the two channels show mutually exclusive expression patterns. ANO 1 is present in the apical membranes of various secretory epithelia in which it is co-localized with the water channel aquaporin 5. It has also been detected in acinar cells and duct cells of subepithelial glands and in the supporting cells of sensory epithelia. In contrast, ANO 2 expression is restricted to chemosensory neurons in which it has been detected in microvillar and ciliary surface structures. The different expression patterns of ANO 1 and ANO 2 have been observed in the olfactory, vomeronasal and respiratory epithelia. No expression has been detected in the Grueneberg ganglion or trigeminal sensory fibers. On the basis of this differential expression, we derive the main functional features of ANO 1 and ANO 2 chloride channels in the nose and suggest their significance for nasal physiology.


Asunto(s)
Canales de Cloruro/metabolismo , Mucosa Nasal/metabolismo , Animales , Anoctamina-1 , Anoctaminas , Ganglios Sensoriales/metabolismo , Ratones , Mucosa Olfatoria/citología , Mucosa Olfatoria/metabolismo , Ratas
10.
Adv Exp Med Biol ; 739: 32-58, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22399394

RESUMEN

In the course of evolution, the strong and unremitting selective pressure on sensory performance has driven the acuity of sensory organs to its physical limits. As a consequence, the study of primary sensory processes illustrates impressively how far a physiological function can be improved, if the survival of a species depends on it. Sensory cells that detect single-photons, single molecules, mechanical motions on a nanometer scale, or incredibly small fluctuations of electromagnetic fields have fascinated physiologists for a long time. It is a great challenge to understand the primary sensory processes on a molecular level. This chapter points out some important recent developments in the search for primary processes in sensory cells that mediate touch perception, hearing, vision, taste, olfaction, as well as the analysis of light polarization and the orientation in the Earth's magnetic field. The data are screened for common transduction strategies and common transduction molecules, an aspect that may be helpful for researchers in the field.


Asunto(s)
Células Receptoras Sensoriales , Animales , Campos Electromagnéticos , Humanos , Fototransducción , Mecanorreceptores , Percepción Olfatoria , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Percepción del Gusto
11.
Biochemistry ; 50(15): 3221-8, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21413724

RESUMEN

Cyclic nucleotide-gated (CNG) channels operate as transduction channels in photoreceptors and olfactory receptor neurons. Direct binding of cGMP or cAMP opens these channels which conduct a mixture of monovalent cations and Ca(2+). Upon activation, CNG channels generate intracellular Ca(2+) signals that play pivotal roles in the transduction cascades of the visual and olfactory systems. Channel activity is controlled by negative feedback mechanisms that involve Ca(2+)-calmodulin, for which all CNG channels possess binding sites. Here we compare the binding properties of the two LQ-type calmodulin binding sites, both of which are thought to be involved in channel regulation. They reside on the isoforms CNGB1 and CNGA4. The CNGB1 subunit is present in rod photoreceptors and olfactory receptor neurons. The CNGA4 subunit is only expressed in olfactory receptor neurons, and there are conflicting results as to its role in calmodulin-mediated feedback inhibition. We examined the interaction of Ca(2+)-calmodulin with two recombinant proteins that encompass either of the two LQ sites. Comparing binding properties, we found that the LQ site of CNGB1 binds Ca(2+)-calmodulin at 10-fold lower Ca(2+) levels than the LQ site of CNGA4. Our data provide biochemical evidence against a contribution of CNGA4 to feedback inhibition. In accordance with previous work on photoreceptor CNG channels, our results indicate that feedback control is the exclusive role of the B-subunits in photoreceptors and olfactory receptor neurons.


Asunto(s)
Calmodulina/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/química , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Calcio/metabolismo , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína
12.
Neuroscience ; 479: 35-47, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34695536

RESUMEN

The spinal trigeminal nucleus caudalis (SpVc) in the mammalian brainstem serves a pivotal function in pain processing. As the main relay center for nociceptive signals, SpVc conducts pain-related signals from various regions of the head toward higher levels of central processing such as the thalamus. SpVc also receives modulatory signals from other brain areas, which can alleviate the perception of headache. We studied the impact of olfactory co-stimulation on pain-related behavior and SpVc neural activity in mice. Using the TRPA1 agonist allyl isothiocyanate (AITC) as noxious stimulus, we quantified the aversive response and the perceived pain intensity by evaluating explorative running and the mouse grimace scale, respectively. We found that the floral odorants phenylethyl alcohol (PEA) and lavender oil mitigated the aversive response to AITC. Consistent with this finding, a newly developed, automated quantification of c-Fos expression in SpVc revealed that co-stimulation with PEA or lavender profoundly reduced network activity in the presence of AITC. These results demonstrated a substantial analgesic potential of odor stimulation in the trigeminal system and provide an explanation for the palliative effect of odors in the treatment of headache.


Asunto(s)
Nocicepción , Olfato , Animales , Encéfalo , Ratones , Odorantes , Núcleo Espinal del Trigémino
13.
PLoS One ; 16(3): e0247801, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33651839

RESUMEN

Modulation of neuronal excitability is a prominent way of shaping the activity of neuronal networks. Recent studies highlight the role of calcium-activated chloride currents in this context, as they can both increase or decrease excitability. The calcium-activated chloride channel Anoctamin 2 (ANO2 alias TMEM16B) has been described in several regions of the mouse brain, including the olivo-cerebellar system. In inferior olivary neurons, ANO2 was proposed to increase excitability by facilitating the generation of high-threshold calcium spikes. An expression of ANO2 in cerebellar Purkinje cells was suggested, but its role in these neurons remains unclear. In the present study, we confirmed the expression of Ano2 mRNA in Purkinje cells and performed electrophysiological recordings to examine the influence of ANO2-chloride channels on the excitability of Purkinje cells by comparing wildtype mice to mice lacking ANO2. Recordings were performed in acute cerebellar slices of adult mice, which provided the possibility to study the role of ANO2 within the cerebellar cortex. Purkinje cells were uncoupled from climbing fiber input to assess specifically the effect of ANO2 channels on Purkinje cell activity. We identified an attenuating effect of ANO2-mediated chloride currents on the instantaneous simple spike activity both during strong current injections and during current injections close to the simple spike threshold. Moreover, we report a reduction of inhibitory currents from GABAergic interneurons upon depolarization, lasting for several seconds. Together with the role of ANO2-chloride channels in inferior olivary neurons, our data extend the evidence for a role of chloride-dependent modulation in the olivo-cerebellar system that might be important for proper cerebellum-dependent motor coordination and learning.


Asunto(s)
Anoctaminas/fisiología , Calcio/metabolismo , Potenciales de la Membrana , Células de Purkinje/fisiología , Animales , Anoctaminas/genética , Calcio/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células de Purkinje/química
14.
Proteomics ; 9(2): 322-34, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19086097

RESUMEN

Olfactory sensory neurons expose to the inhaled air chemosensory cilia which bind odorants and operate as transduction organelles. Odorant receptors in the ciliary membrane activate a transduction cascade which uses cAMP and Ca(2+) for sensory signaling in the ciliary lumen. Although the canonical transduction pathway is well established, molecular components for more complex aspects of sensory transduction, like adaptation, regulation, and termination of the receptor response have not been systematically identified. Moreover, open questions in olfactory physiology include how the cilia exchange solutes with the surrounding mucus, assemble their highly polarized set of proteins, and cope with noxious substances in the ambient air. A specific ciliary proteome would promote research efforts in all of these fields. We have improved a method to detach cilia from rat olfactory sensory neurons and have isolated a preparation specifically enriched in ciliary membrane proteins. Using LC-ESI-MS/MS analysis, we identified 377 proteins which constitute the olfactory cilia proteome. These proteins represent a comprehensive data set for olfactory research since more than 80% can be attributed to the characteristic functions of olfactory sensory neurons and their cilia: signal processing, protein targeting, neurogenesis, solute transport, and cytoprotection. Organellar proteomics thus yielded decisive information about the diverse physiological functions of a sensory organelle.


Asunto(s)
Cilios/química , Neuronas Receptoras Olfatorias/química , Proteínas/química , Proteoma , Receptores Odorantes/química , Adenilil Ciclasas/química , Adenilil Ciclasas/metabolismo , Animales , Cromatografía Liquida , Cilios/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/química , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Bases de Datos de Proteínas , Isoenzimas/química , Isoenzimas/metabolismo , Microscopía Electrónica de Rastreo , Mucosa Olfatoria/anatomía & histología , Neuronas Receptoras Olfatorias/metabolismo , Proteínas/metabolismo , Ratas , Ratas Wistar , Receptores Odorantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
15.
Artículo en Inglés | MEDLINE | ID: mdl-19011871

RESUMEN

In the course of evolution, the strong and unremitting selective pressure on sensory performance has driven the acuity of sensory organs to its physical limits. As a consequence, the study of primary sensory processes illustrates impressively how far a physiological function can be improved if the survival of a species depends on it. Sensory cells that detect single-photons, single molecules, mechanical motions on a nanometer scale, or incredibly small fluctuations of electromagnetic fields have fascinated physiologists for a long time. It is a great challenge to understand the primary sensory processes on a molecular level. This review points out some important recent developments in the search for primary processes in sensory cells that mediate touch perception, hearing, vision, taste, olfaction, as well as the analysis of light polarization and the orientation in the Earth's magnetic field. The data are screened for common transduction strategies and common transduction molecules, an aspect that may be helpful for researchers in the field.


Asunto(s)
Sensación/fisiología , Células Receptoras Sensoriales/fisiología , Transducción de Señal/fisiología , Animales , Evolución Biológica , Audición/fisiología , Humanos , Modelos Biológicos , Fenómenos Físicos , Olfato/fisiología , Gusto/fisiología , Visión Ocular/fisiología
16.
PLoS One ; 14(8): e0211175, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31412038

RESUMEN

Olfactory and trigeminal chemosensory systems reside in parallel within the mammalian nose. Psychophysical studies in people indicate that these two systems interact at a perceptual level. Trigeminal sensations of pungency mask odour perception, while olfactory stimuli can influence trigeminal signal processing tasks such as odour localization. While imaging studies indicate overlap in limbic and cortical somatosensory areas activated by nasal trigeminal and olfactory stimuli, there is also potential cross-talk at the level of the olfactory epithelium, the olfactory bulb and trigeminal brainstem. Here we explored the influence of olfactory and trigeminal signaling in the nasal cavity. A forced choice water consumption paradigm was used to ascertain whether trigeminal and olfactory stimuli could influence behaviour in mice. Mice avoided water sources surrounded by both volatile TRPV1 (cyclohexanone) and TRPA1 (allyl isothiocyanate) irritants and the aversion to cyclohexanone was mitigated when combined with a pure odorant (rose fragrance, phenylethyl alcohol, PEA). To determine whether olfactory-trigeminal interactions within the nose could potentially account for this behavioural effect we recorded from single trigeminal sensory axons innervating the nasal respiratory and olfactory epithelium using an isolated in vitro preparation. To circumvent non-specific effects of chemical stimuli, optical stimulation was used to excite olfactory sensory neurons in mice expressing channel-rhodopsin (ChR2) under the olfactory marker protein (OMP) promoter. Photoactivation of olfactory sensory neurons produced no modulation of axonal action potential conduction in individual trigeminal axons. Similarly, no evidence was found for collateral branching of trigeminal axon that might serve as a conduit for cross-talk between the olfactory and respiratory epithelium and olfactory dura mater. Using direct assessment of action potential activity in trigeminal axons we observed neither paracrine nor axon reflex mediated cross-talk between olfactory and trigeminal sensory systems in the rodent nasal cavity. Our current results suggest that olfactory sensory neurons exert minimal influence on trigeminal signals within the nasal cavity.


Asunto(s)
Cavidad Nasal/inervación , Odorantes/análisis , Vías Olfatorias/efectos de los fármacos , Neuronas Receptoras Olfatorias/fisiología , Nervio Trigémino/fisiología , Potenciales de Acción , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Receptoras Olfatorias/efectos de la radiación , Nervio Trigémino/efectos de los fármacos
17.
Mol Pain ; 4: 32, 2008 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-18700020

RESUMEN

BACKGROUND: Chloride currents in peripheral nociceptive neurons have been implicated in the generation of afferent nociceptive signals, as Cl- accumulation in sensory endings establishes the driving force for depolarizing, and even excitatory, Cl- currents. The intracellular Cl- concentration can, however, vary considerably between individual DRG neurons. This raises the question, whether the contribution of Cl- currents to signal generation differs between individual afferent neurons, and whether the specific Cl- levels in these neurons are subject to modulation. Based on the hypothesis that modulation of the peripheral Cl- homeostasis is involved in the generation of inflammatory hyperalgesia, we examined the effects of inflammatory mediators on intracellular Cl- concentrations and on the expression levels of Cl- transporters in rat DRG neurons. RESULTS: We developed an in vitro assay for testing how inflammatory mediators influence Cl- concentration and the expression of Cl- transporters. Intact DRGs were treated with 100 ng/ml NGF, 1.8 microM ATP, 0.9 microM bradykinin, and 1.4 microM PGE2 for 1-3 hours. Two-photon fluorescence lifetime imaging with the Cl--sensitive dye MQAE revealed an increase of the intracellular Cl- concentration within 2 hours of treatment. This effect coincided with enhanced phosphorylation of the Na+-K+-2Cl- cotransporter NKCC1, suggesting that an increased activity of that transporter caused the early rise of intracellular Cl- levels. Immunohistochemistry of NKCC1 and KCC2, the main neuronal Cl- importer and exporter, respectively, exposed an inverse regulation by the inflammatory mediators. While the NKCC1 immunosignal increased, that of KCC2 declined after 3 hours of treatment. In contrast, the mRNA levels of the two transporters did not change markedly during this time. These data demonstrate a fundamental transition in Cl- homeostasis toward a state of augmented Cl- accumulation, which is induced by a 1-3 hour treatment with inflammatory mediators. CONCLUSION: Our findings indicate that inflammatory mediators impact on Cl- homeostasis in DRG neurons. Inflammatory mediators raise intracellular Cl- levels and, hence, the driving force for depolarizing Cl- efflux. These findings corroborate current concepts for the role of Cl- regulation in the generation of inflammatory hyperalgesia and allodynia. As the intracellular Cl- concentration rises in DRG neurons, afferent signals can be boosted by excitatory Cl- currents in the presynaptic terminals. Moreover, excitatory Cl- currents in peripheral sensory endings may also contribute to the generation or modulation of afferent signals, especially in inflamed tissue.


Asunto(s)
Cloruros/metabolismo , Ganglios Espinales/metabolismo , Homeostasis/fisiología , Mediadores de Inflamación/farmacología , Neuronas/metabolismo , Adenosina Trifosfato/farmacología , Animales , Bradiquinina/farmacología , Dinoprostona/farmacología , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Homeostasis/efectos de los fármacos , Factores de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas
19.
Nat Neurosci ; 7(7): 705-10, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15195096

RESUMEN

An important mechanism by which vertebrate olfactory sensory neurons rapidly adapt to odorants is feedback modulation of the Ca(2+)-permeable cyclic nucleotide-gated (CNG) transduction channels. Extensive heterologous studies of homomeric CNGA2 channels have led to a molecular model of channel modulation based on the binding of calcium-calmodulin to a site on the cytoplasmic amino terminus of CNGA2. Native rat olfactory CNG channels, however, are heteromeric complexes of three homologous but distinct subunits. Notably, in heteromeric channels, we found no role for CNGA2 in feedback modulation. Instead, an IQ-type calmodulin-binding site on CNGB1b and a similar but previously unidentified site on CNGA4 are necessary and sufficient. These sites seem to confer binding of Ca(2+)-free calmodulin (apocalmodulin), which is then poised to trigger inhibition of native channels in the presence of Ca(2+).


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Canales Iónicos/metabolismo , Bulbo Olfatorio/citología , Neuronas Receptoras Olfatorias/fisiología , Secuencias de Aminoácidos/fisiología , Animales , Señalización del Calcio , Células Cultivadas , AMP Cíclico/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Embrión de Mamíferos , Retroalimentación Fisiológica , Humanos , Activación del Canal Iónico/fisiología , Canales Iónicos/química , Riñón , Potenciales de la Membrana/fisiología , Mutagénesis Sitio-Dirigida , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Transfección/métodos
20.
J Gen Physiol ; 127(6): 737-48, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16735757

RESUMEN

In sensory neurons of the peripheral nervous system, receptor potentials can be amplified by depolarizing Cl currents. In mammalian olfactory sensory neurons (OSNs), this anion-based signal amplification results from the sequential activation of two distinct types of transduction channels: cAMP-gated Ca channels and Ca-activated Cl channels. The Cl current increases the initial receptor current about 10-fold and leads to the excitation of the neuron. Here we examine the activation mechanism of the Ca-dependent Cl channel. We focus on calmodulin, which is known to mediate Ca effects on various ion channels. We show that the cell line Odora, which is derived from OSN precursor cells in the rat olfactory epithelium, expresses Ca-activated Cl channels. Single-channel conductance, ion selectivity, voltage dependence, sensitivity to niflumic acid, and Ca sensitivity match between Odora channels and OSN channels. Transfection of Odora cells with CaM mutants reduces the Ca sensitivity of the Cl channels. This result points to the participation of calmodulin in the gating process of Ca-ativated Cl channels, and helps to understand how signal amplification works in the olfactory sensory cilia. Calmodulin was previously shown to mediate feedback inhibition of cAMP-synthesis and of the cAMP-gated Ca channels in OSNs. Our results suggest that calmodulin may also be instrumental in the generation of the excitatory Cl current. It appears to play a pivotal role in the peripheral signal processing of olfactory sensory information. Moreover, recent results from other peripheral neurons, as well as from smooth muscle cells, indicate that the calmodulin-controlled, anion-based signal amplification operates in various cell types where it converts Ca signals into membrane depolarization.


Asunto(s)
Calcio/fisiología , Calmodulina/fisiología , Canales de Cloruro/fisiología , Activación del Canal Iónico/fisiología , Neuronas Receptoras Olfatorias/fisiología , Animales , Calcio/química , Calmodulina/química , Línea Celular , Activación del Canal Iónico/efectos de los fármacos , Neuronas Receptoras Olfatorias/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA