Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 35(12): e22025, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34758144

RESUMEN

Mepyramine, a first-generation antihistamine targeting the histamine H(1) receptor, was extensively prescribed to patients suffering from allergic reactions and urticaria. Serious adverse effects, especially in case of overdose, were frequently reported, including drowsiness, impaired thinking, convulsion, and coma. Many of these side effects were associated with the blockade of histaminergic or cholinergic receptors. Here we show that mepyramine directly inhibits a variety of voltage-gated sodium channels, including the Tetrodotoxin-sensitive isoforms and the main isoforms (Nav1.7, Nav1.8, and Nav1.9) of nociceptors. Estimated IC50 were within the range of drug concentrations detected in poisoned patients. Mepyramine inhibited sodium channels through fast- or slow-inactivated state preference depending on the isoform. Moreover, mepyramine inhibited the firing responses of C- and Aß-type nerve fibers in ex vivo skin-nerve preparations. Locally applied mepyramine had analgesic effects on the scorpion toxin-induced excruciating pain and produced pain relief in acute, inflammatory, and chronic pain models. Collectively, these data provide evidence that mepyramine has the potential to be developed as a topical analgesic agent.


Asunto(s)
Artritis Experimental/complicaciones , Ganglios Espinales/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/fisiología , Nociceptores/efectos de los fármacos , Dolor/tratamiento farmacológico , Pirilamina/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Potenciales de Acción , Animales , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Antagonistas de los Receptores Histamínicos H1/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.8/química , Nociceptores/metabolismo , Nociceptores/patología , Dolor/etiología , Dolor/metabolismo , Dolor/patología
3.
Nat Commun ; 10(1): 4253, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31534133

RESUMEN

Medication-overuse headaches (MOH) occur with both over-the-counter and pain-relief medicines, including paracetamol, opioids and combination analgesics. The mechanisms that lead to MOH are still uncertain. Here, we show that abnormal activation of Nav1.9 channels by Nitric Oxide (NO) is responsible for MOH induced by triptan migraine medicine. Deletion of the Scn11a gene in MOH mice abrogates NO-mediated symptoms, including cephalic and extracephalic allodynia, photophobia and phonophobia. NO strongly activates Nav1.9 in dural afferent neurons from MOH but not normal mice. Abnormal activation of Nav1.9 triggers CGRP secretion, causing artery dilatation and degranulation of mast cells. In turn, released mast cell mediators potentiates Nav1.9 in meningeal nociceptors, exacerbating inflammation and pain signal. Analysis of signaling networks indicates that PKA is downregulated in trigeminal neurons from MOH mice, relieving its inhibitory action on NO-Nav1.9 coupling. Thus, anomalous activation of Nav1.9 channels by NO, as a result of chronic medication, promotes MOH.


Asunto(s)
Cefaleas Secundarias/patología , Trastornos Migrañosos/patología , Canal de Sodio Activado por Voltaje NAV1.9/metabolismo , Neuronas Aferentes/metabolismo , Óxido Nítrico/metabolismo , Triptaminas/efectos adversos , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Degranulación de la Célula/fisiología , Células Cultivadas , Femenino , Cefaleas Secundarias/inducido químicamente , Hiperalgesia/fisiopatología , Masculino , Mastocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.9/genética , Neuronas Aferentes/efectos de los fármacos , Nociceptores/fisiología , Dolor/fisiopatología , Uso Excesivo de Medicamentos Recetados/efectos adversos
4.
Pflugers Arch ; 467(1): 109-19, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25416542

RESUMEN

Physical contact with the external world occurs through specialized neural structures called mechanoreceptors. Cutaneous mechanoreceptors provide information to the central nervous system (CNS) about touch, pressure, vibration, and skin stretch. The physiological function of these mechanoreceptors is to convert physical forces into neuronal signals. Key questions concern the molecular identity of the mechanoelectric transducer channels and the mechanisms by which the physical parameters of the mechanical stimulus are encoded into patterns of action potentials (APs). Compelling data indicate that the biophysical traits of mechanosensitive channels combined with the collection of voltage-gated channels are essential to describe the nature of the stimulus. Recent research also points to a critical role of the auxiliary cell-nerve ending communication in encoding stimulus properties. This review describes the characteristics of ion channels responsible for translating mechanical stimuli into the neural codes that underlie touch perception and pain.


Asunto(s)
Ganglios Espinales/fisiología , Mecanorreceptores/fisiología , Mecanotransducción Celular/fisiología , Fenómenos Fisiológicos de la Piel , Piel/inervación , Tacto/fisiología , Potenciales de Acción/fisiología , Vías Aferentes/fisiología , Animales , Humanos , Modelos Biológicos , Percepción del Dolor/fisiología
5.
Cell Rep ; 5(2): 378-88, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24139797

RESUMEN

C-low-threshold mechanoreceptors (C-LTMRs) are unique among C-unmyelinated primary sensory neurons. These neurons convey two opposite aspects of touch sensation: a sensation of pleasantness, and a sensation of injury-induced mechanical pain. Here, we show that TAFA4 is a specific marker of C-LTMRs. Genetic labeling in combination with electrophysiological recordings show that TAFA4+ neurons have intrinsic properties of mechano-nociceptors. TAFA4-null mice exhibit enhanced mechanical and chemical hypersensitivity following inflammation and nerve injury as well as increased excitability of spinal cord lamina IIi neurons, which could be reversed by intrathecal or bath application of recombinant TAFA4 protein. In wild-type C57/Bl6 mice, intrathecal administration of TAFA4 strongly reversed carrageenan-induced mechanical hypersensitivity, suggesting a potent analgesic role of TAFA4 in pain relief. Our data provide insights into how C-LTMR-derived TAFA4 modulates neuronal excitability and controls the threshold of somatic sensation.


Asunto(s)
Citocinas/metabolismo , Nociceptores/metabolismo , Dolor/fisiopatología , Estrés Mecánico , Animales , Carragenina/toxicidad , Citocinas/genética , Citocinas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Dolor/metabolismo , Umbral del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
6.
Neuron ; 77(5): 899-914, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23473320

RESUMEN

Molecular determinants of threshold sensitivity of mammalian mechanoreceptors are unknown. Here, we identify a mechanosensitive (MS) K(+) current (IKmech) that governs mechanical threshold and adaptation of distinct populations of mechanoreceptors. Toxin profiling and transgenic mouse studies indicate that IKmech is carried by Kv1.1-Kv1.2 heteromers. Mechanosensitivity is attributed to Kv1.1 subunits, through facilitation of voltage-dependent open probability. IKmech is expressed in high-threshold C-mechano-nociceptors (C-HTMRs) and Aß-mechanoreceptors, but not in low-threshold C-mechanoreceptors. IKmech opposes depolarization induced by slow/ultraslow MS cation currents in C-HTMRs, thereby shifting mechanical threshold for firing to higher values. However, due to kinetics mismatch with rapidly-adapting MS cation currents, IKmech tunes firing adaptation but not mechanical threshold in Aß-mechanoreceptors. Expression of Kv1.1 dominant negative or inhibition of Kv1.1/IKmech caused severe mechanical allodynia but not heat hyperalgesia. By balancing the activity of excitatory mechanotransducers, Kv1.1 acts as a mechanosensitive brake that regulates mechanical sensitivity of fibers associated with mechanical perception.


Asunto(s)
Canal de Potasio Kv.1.1/fisiología , Dolor/fisiopatología , Tacto/fisiología , Animales , Interpretación Estadística de Datos , Canales de Potasio Éter-A-Go-Go/fisiología , Hiperalgesia/fisiopatología , Inmunohistoquímica , Canales de Potasio KCNQ/fisiología , Canal de Potasio Kv.1.1/genética , Mecanorreceptores/fisiología , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Fibras Nerviosas/fisiología , Nociceptores/fisiología , Umbral del Dolor/fisiología , Técnicas de Placa-Clamp , Estimulación Física , Canales de Potasio Calcio-Activados/fisiología , Células Receptoras Sensoriales/fisiología , Umbral Sensorial/fisiología , Transducción de Señal/fisiología
7.
Cell Calcium ; 53(5-6): 327-37, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23528238

RESUMEN

Primary cilium has emerged as mechanosensor to subtle flow variations in epithelial cells, but its role in shear stress detection remains controversial. To probe the function of this non-motile organelle in shear stress detection by cells, we compared calcium signalling responses induced by shear stress in ciliated and unciliated MDCK cells. Cytosolic free Ca²âº ([Ca²âº]i) was measured using Fura-PE3 video imaging fluorescence microscopy in response to shear stress due to laminar flow (385 µl s⁻¹). Our results show that both unciliated and ciliated MDCK cells are shear stress sensitive via ATP release and autocrine feedback through purinergic receptors. However, purinergic calcium signals differed in response intensity and receptor subtypes. In unciliated cells, shear stress-induced elevation in [Ca²âº]i was predominantly mediated through P2X receptors (P2XR). In contrast, calcium mobilization in ciliated MDCK cells resulted from P2YRs and store-operated Ca²âº-permeable channels besides P2XRs. These findings lend support to the hypothesis that ATP release in response to shear stress is independent of the primary cilium and that transduction of mechanical strain into a specific biochemical responses stems on the mobilization of different sets of purinergic receptors.


Asunto(s)
Adenosina Trifosfato/metabolismo , Cilios/fisiología , Mecanotransducción Celular , Receptores Purinérgicos P2X/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Resistencia al Corte , Estrés Mecánico , Animales , Comunicación Autocrina , Señalización del Calcio , Perros , Retroalimentación Fisiológica , Células de Riñón Canino Madin Darby , Microscopía Fluorescente , Especificidad de Órganos , Cultivo Primario de Células
8.
Methods Mol Biol ; 998: 159-70, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23529428

RESUMEN

Mechanotransduction, the conversion of a mechanical stimulus into a biological response, constitutes the basis of a variety of physiological functions such as the senses of touch, balance, proprioception, blood pressure, and hearing. In vertebrates, mechanosensation is mediated by mechanosensory neurons, whose cell bodies are located in trigeminal and dorsal root ganglia. Here, we describe an in vitro model of mechanotransduction that provides an opportunity to explore the properties of mechanosensitive channels in mammalian sensory neurons. The mechano-clamp method allows applying local force on plasma membrane of whole-cell patch-clamped sensory neurons. This technique uses a mechanical probe driven by a computer-assisted piezoelectric microstage to repeatedly stimulate sensory neurons with accurate control of stimulus strength, duration, and speed.


Asunto(s)
Electricidad , Fenómenos Mecánicos , Técnicas de Placa-Clamp/métodos , Células Receptoras Sensoriales/citología , Animales , Fenómenos Electrofisiológicos , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Masculino , Mecanotransducción Celular , Técnicas de Placa-Clamp/instrumentación , Ratas , Ratas Wistar
9.
Channels (Austin) ; 6(4): 234-45, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23146937

RESUMEN

Cutaneous mechanoreceptors are localized in the various layers of the skin where they detect a wide range of mechanical stimuli, including light brush, stretch, vibration and noxious pressure. This variety of stimuli is matched by a diverse array of specialized mechanoreceptors that respond to cutaneous deformation in a specific way and relay these stimuli to higher brain structures. Studies across mechanoreceptors and genetically tractable sensory nerve endings are beginning to uncover touch sensation mechanisms. Work in this field has provided researchers with a more thorough understanding of the circuit organization underlying the perception of touch. Novel ion channels have emerged as candidates for transduction molecules and properties of mechanically gated currents improved our understanding of the mechanisms of adaptation to tactile stimuli. This review highlights the progress made in characterizing functional properties of mechanoreceptors in hairy and glabrous skin and ion channels that detect mechanical inputs and shape mechanoreceptor adaptation.


Asunto(s)
Mecanorreceptores/metabolismo , Tacto/fisiología , Animales , Humanos , Canales Iónicos/metabolismo , Mecanorreceptores/química , Neuronas/fisiología , Piel/metabolismo , Médula Espinal/metabolismo
10.
Pain ; 153(2): 473-484, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22172548

RESUMEN

Menthol is a natural compound of plant origin known to produce cool sensation via the activation of the TRPM8 channel. It is also frequently part of topical analgesic drugs available in a pharmacy, although its mechanism of action is still unknown. Compelling evidence indicates that voltage-gated Na(+) channels are critical for experiencing pain sensation. We tested the hypothesis that menthol may block voltage-gated Na(+) channels in dorsal root ganglion (DRG) neurons. By use of a patch clamp, we evaluated the effects of menthol application on tetrodotoxin (TTX)-resistant Nav1.8 and Nav1.9 channel subtypes in DRG neurons, and on TTX-sensitive Na(+) channels in immortalized DRG neuron-derived F11 cells. The results indicate that menthol inhibits Na(+) channels in a concentration-, voltage-, and frequency-dependent manner. Menthol promoted fast and slow inactivation states, causing use-dependent depression of Na(+) channel activity. In current clamp recordings, menthol inhibited firing at high-frequency stimulation with minimal effects on normal neuronal activity. We found that low concentrations of menthol cause analgesia in mice, relieving pain produced by a Na(+) channel-targeting toxin. We conclude that menthol is a state-selective blocker of Nav1.8, Nav1.9, and TTX-sensitive Na(+) channels, indicating a role for Na(+) channel blockade in the efficacy of menthol as topical analgesic compound.


Asunto(s)
Analgésicos/farmacología , Mentol/farmacología , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Canales de Sodio/metabolismo , Animales , Línea Celular Tumoral , Células Híbridas , Masculino , Ratones , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.8 , Canal de Sodio Activado por Voltaje NAV1.9 , Inhibición Neural/fisiología , Neuroblastoma/metabolismo , Neuroblastoma/fisiopatología , Dolor/metabolismo , Cultivo Primario de Células , Ratas , Ratas Wistar , Canales de Sodio/fisiología
11.
Nat Protoc ; 6(7): 979-90, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21720312

RESUMEN

Mechanotransduction constitutes the basis of a variety of physiological processes, such as the senses of touch, balance, proprioception and hearing. In vertebrates, mechanosensation is mediated by mechanosensory receptors. The aptitude of these mechanoreceptors for detecting mechanical information relies on the presence of mechanosensitive channels that transform mechanical forces into electrical signals. However, advances in understanding mechanical transduction processes have proven difficult because sensory nerve endings have historically been inaccessible to patch-clamp recording. We report here an in vitro model of mechanotransduction that allows the application of focal force on sensory neuron membrane during whole-cell patch clamping. This technique, called mechano-clamp, provides an opportunity to explore the properties and identities of mechanotransducer channels in mammalian sensory neurons. The protocol-from tissue dissociation to patch-clamp recording-can be completed in 7 h.


Asunto(s)
Ganglios Espinales/fisiología , Mecanorreceptores/fisiología , Mecanotransducción Celular/fisiología , Técnicas de Placa-Clamp , Animales , Masculino , Estimulación Física , Ratas , Ratas Wistar
12.
Nat Rev Neurosci ; 12(3): 139-53, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21304548

RESUMEN

The somatosensory system mediates fundamental physiological functions, including the senses of touch, pain and proprioception. This variety of functions is matched by a diverse array of mechanosensory neurons that respond to force in a specific fashion. Mechanotransduction begins at the sensory nerve endings, which rapidly transform mechanical forces into electrical signals. Progress has been made in establishing the functional properties of mechanoreceptors, but it has been remarkably difficult to characterize mechanotranducer channels at the molecular level. However, in the past few years, new functional assays have provided insights into the basic properties and molecular identity of mechanotransducer channels in mammalian sensory neurons. The recent identification of novel families of proteins as mechanosensing molecules will undoubtedly accelerate our understanding of mechanotransduction mechanisms in mammalian somatosensation.


Asunto(s)
Potenciales Evocados Somatosensoriales/fisiología , Mecanorreceptores/fisiología , Mecanotransducción Celular/fisiología , Células Receptoras Sensoriales/fisiología , Animales , Humanos , Tacto/fisiología
13.
J Neurosci ; 30(40): 13384-95, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20926665

RESUMEN

How desensitization of mechanotransducer currents regulates afferent signal generation in mammalian sensory neurons is essentially unknown. Here, we dissected desensitization mechanisms of mechanotransducer channels in rat sensory neurons that mediate the sense of touch and pain. We identified four types of mechanotransducer currents that distribute differentially in cutaneous nociceptors and mechanoreceptors and that differ in desensitization rates. Desensitization of mechanotransducer channels in mechanoreceptors was fast and mediated by channel inactivation and adaptation, which reduces the mechanical force sensed by the transduction channel. Both processes were promoted by negative voltage. These properties of mechanotransducer channels suited them to encode the dynamic parameters of the stimulus. In contrast, inactivation and adaptation of mechanotransducer channels in nociceptors had slow time courses and were suited to encode duration of the stimulus. Thus, desensitization properties of mechanotransducer currents relate to their functions as sensors of phasic and tonic stimuli and enable sensory neurons to achieve efficient stimulus representation.


Asunto(s)
Activación del Canal Iónico/fisiología , Mecanorreceptores/fisiología , Mecanotransducción Celular/fisiología , Inhibición Neural/fisiología , Células Receptoras Sensoriales/fisiología , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Animales , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Activación del Canal Iónico/genética , Masculino , Mecanotransducción Celular/genética , Inhibición Neural/genética , Nociceptores/fisiología , Dolor/genética , Dolor/fisiopatología , Técnicas de Placa-Clamp/métodos , Fenotipo , Ratas , Ratas Wistar , Factores de Tiempo , Tacto/genética , Tacto/fisiología
14.
EMBO J ; 29(7): 1176-91, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20168298

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in two genes, PKD1 and PKD2, which encode polycystin-1 (PC1) and polycystin-2 (PC2), respectively. Earlier work has shown that PC1 and PC2 assemble into a polycystin complex implicated in kidney morphogenesis. PC2 also assembles into homomers of uncertain functional significance. However, little is known about the molecular mechanisms that direct polycystin complex assembly and specify its functions. We have identified a coiled coil in the C-terminus of PC2 that functions as a homodimerization domain essential for PC1 binding but not for its self-oligomerization. Dimerization-defective PC2 mutants were unable to reconstitute PC1/PC2 complexes either at the plasma membrane (PM) or at PM-endoplasmic reticulum (ER) junctions but could still function as ER Ca(2+)-release channels. Expression of dimerization-defective PC2 mutants in zebrafish resulted in a cystic phenotype but had lesser effects on organ laterality. We conclude that C-terminal dimerization of PC2 specifies the formation of polycystin complexes but not formation of ER-localized PC2 channels. Mutations that affect PC2 C-terminal homo- and heteromerization are the likely molecular basis of cyst formation in ADPKD.


Asunto(s)
Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Línea Celular , Dimerización , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Expresión Génica , Humanos , Riñón/patología , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Canales Catiónicos TRPP/genética , Técnicas del Sistema de Dos Híbridos , Pez Cebra/genética
15.
Neuron ; 59(3): 439-49, 2008 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-18701069

RESUMEN

Small-conductance Ca2+-activated K+ (SK) channels are widely expressed in neuronal tissues where they underlie post-spike hyperpolarizations, regulate spike-frequency adaptation, and shape synaptic responses. SK channels constitutively interact with calmodulin (CaM), which serves as Ca2+ sensor, and with protein kinase CK2 and protein phosphatase 2A, which modulate their Ca2+ gating. By recording coupled activities of Ca2+ and SK2 channels, we showed that SK2 channels can be inhibited by neurotransmitters independently of changes in the activity of the priming Ca2+ channels. This inhibition involvesSK2-associated CK2 and results from a 3-fold reduction in the Ca2+ sensitivity of channel gating. CK2phosphorylated SK2-bound CaM but not KCNQ2-bound CaM, thereby selectively regulating SK2 channels. We extended these observations to sensory neurons by showing that noradrenaline inhibits SK current and increases neuronal excitability in aCK2-dependent fashion. Hence, neurotransmitter-initiated signaling cascades can dynamically regulate Ca2+ sensitivity of SK channels and directly influence somatic excitability.


Asunto(s)
Calcio/metabolismo , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Potenciales de Acción , Animales , Apamina/farmacología , Autorradiografía/métodos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/fisiología , Calmodulina , Células Cultivadas , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos/farmacología , Ganglios Espinales/citología , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/efectos de la radiación , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/efectos de la radiación , Microinyecciones/métodos , Neuronas/fisiología , Técnicas de Placa-Clamp , Ratas , Transducción de Señal , Ganglio Cervical Superior/citología
16.
Sensors (Basel) ; 7(9): 1667-1682, 2007 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-28903189

RESUMEN

Mechanoreceptive sensory neurons innervating the skin, skeletal muscles andviscera signal both innocuous and noxious information necessary for proprioception, touchand pain. These neurons are responsible for the transduction of mechanical stimuli intoaction potentials that propagate to the central nervous system. The ability of these cells todetect mechanical stimuli impinging on them relies on the presence of mechanosensitivechannels that transduce the external mechanical forces into electrical and chemical signals.Although a great deal of information regarding the molecular and biophysical properties ofmechanosensitive channels in prokaryotes has been accumulated over the past two decades,less is known about the mechanosensitive channels necessary for proprioception and thesenses of touch and pain. This review summarizes the most pertinent data onmechanosensitive channels of mammalian somatosensory neurons, focusing on theirproperties, pharmacology and putative identity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...