Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 183(1): 284-284.e1, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33007264

RESUMO

Ophthalmic, maxillary, and mandibular branches of the trigeminal nerve provide sensory innervation to orofacial tissues. Trigeminal sensory neurons respond to a diverse array of sensory stimuli to generate distinct sensations, including thermosensation, mechanosensation, itching, and pain. These sensory neurons also detect the distinct sharpness or pungency of many foods and beverages. This SnapShot highlights the transduction ion channels critical to orofacial sensation.


Assuntos
Sensação/fisiologia , Nervo Trigêmeo/anatomia & histologia , Nervo Trigêmeo/fisiologia , Nervos Cranianos/anatomia & histologia , Nervos Cranianos/fisiologia , Humanos , Neurônios Aferentes/fisiologia , Dor/fisiopatologia
2.
Cell ; 155(2): 278-84, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24120130

RESUMO

Living organisms sense their physical environment through cellular mechanotransduction, which converts mechanical forces into electrical and biochemical signals. In turn, signal transduction serves a wide variety of functions, from basic cellular processes as diverse as proliferation, differentiation, migration, and apoptosis up to some of the most sophisticated senses, including touch and hearing. Accordingly, defects in mechanosensing potentially lead to diverse diseases and disorders such as hearing loss, cardiomyopathies, muscular dystrophies, chronic pain, and cancer. Here, we review the status of mechanically activated ion channel discovery and discuss current challenges to define their properties and physiological functions.


Assuntos
Canais Iônicos/metabolismo , Mecanorreceptores/fisiologia , Sensação , Animais , Humanos , Células Receptoras Sensoriais/fisiologia
3.
Circ Res ; 134(5): 572-591, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38422173

RESUMO

The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.


Assuntos
Anemia Hemolítica Congênita , Hipertensão Pulmonar , Animais , Feminino , Humanos , Pressão Sanguínea , Biofísica , Hidropisia Fetal , Mamíferos
4.
Nature ; 516(7529): 121-5, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25471886

RESUMO

The sense of touch provides critical information about our physical environment by transforming mechanical energy into electrical signals. It is postulated that mechanically activated cation channels initiate touch sensation, but the identity of these molecules in mammals has been elusive. Piezo2 is a rapidly adapting, mechanically activated ion channel expressed in a subset of sensory neurons of the dorsal root ganglion and in cutaneous mechanoreceptors known as Merkel-cell-neurite complexes. It has been demonstrated that Merkel cells have a role in vertebrate mechanosensation using Piezo2, particularly in shaping the type of current sent by the innervating sensory neuron; however, major aspects of touch sensation remain intact without Merkel cell activity. Here we show that mice lacking Piezo2 in both adult sensory neurons and Merkel cells exhibit a profound loss of touch sensation. We precisely localize Piezo2 to the peripheral endings of a broad range of low-threshold mechanoreceptors that innervate both hairy and glabrous skin. Most rapidly adapting, mechanically activated currents in dorsal root ganglion neuronal cultures are absent in Piezo2 conditional knockout mice, and ex vivo skin nerve preparation studies show that the mechanosensitivity of low-threshold mechanoreceptors strongly depends on Piezo2. This cellular phenotype correlates with an unprecedented behavioural phenotype: an almost complete deficit in light-touch sensation in multiple behavioural assays, without affecting other somatosensory functions. Our results highlight that a single ion channel that displays rapidly adapting, mechanically activated currents in vitro is responsible for the mechanosensitivity of most low-threshold mechanoreceptor subtypes involved in innocuous touch sensation. Notably, we find that touch and pain sensation are separable, suggesting that as-yet-unknown mechanically activated ion channel(s) must account for noxious (painful) mechanosensation.


Assuntos
Canais Iônicos/metabolismo , Mecanotransdução Celular/fisiologia , Pele/inervação , Tato/fisiologia , Animais , Canais Iônicos/genética , Mecanorreceptores/metabolismo , Mecanotransdução Celular/genética , Células de Merkel/fisiologia , Camundongos , Camundongos Knockout , Células Receptoras Sensoriais/fisiologia , Tato/genética
5.
Nature ; 483(7388): 209-12, 2012 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-22343891

RESUMO

Transduction of mechanical stimuli by receptor cells is essential for senses such as hearing, touch and pain. Ion channels have a role in neuronal mechanotransduction in invertebrates; however, functional conservation of these ion channels in mammalian mechanotransduction is not observed. For example, no mechanoreceptor potential C (NOMPC), a member of transient receptor potential (TRP) ion channel family, acts as a mechanotransducer in Drosophila melanogaster and Caenorhabditis elegans; however, it has no orthologues in mammals. Degenerin/epithelial sodium channel (DEG/ENaC) family members are mechanotransducers in C. elegans and potentially in D. melanogaster; however, a direct role of its mammalian homologues in sensing mechanical force has not been shown. Recently, Piezo1 (also known as Fam38a) and Piezo2 (also known as Fam38b) were identified as components of mechanically activated channels in mammals. The Piezo family are evolutionarily conserved transmembrane proteins. It is unknown whether they function in mechanical sensing in vivo and, if they do, which mechanosensory modalities they mediate. Here we study the physiological role of the single Piezo member in D. melanogaster (Dmpiezo; also known as CG8486). Dmpiezo expression in human cells induces mechanically activated currents, similar to its mammalian counterparts. Behavioural responses to noxious mechanical stimuli were severely reduced in Dmpiezo knockout larvae, whereas responses to another noxious stimulus or touch were not affected. Knocking down Dmpiezo in sensory neurons that mediate nociception and express the DEG/ENaC ion channel pickpocket (ppk) was sufficient to impair responses to noxious mechanical stimuli. Furthermore, expression of Dmpiezo in these same neurons rescued the phenotype of the constitutive Dmpiezo knockout larvae. Accordingly, electrophysiological recordings from ppk-positive neurons revealed a Dmpiezo-dependent, mechanically activated current. Finally, we found that Dmpiezo and ppk function in parallel pathways in ppk-positive cells, and that mechanical nociception is abolished in the absence of both channels. These data demonstrate the physiological relevance of the Piezo family in mechanotransduction in vivo, supporting a role of Piezo proteins in mechanosensory nociception.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Canais Iônicos/metabolismo , Mecanotransdução Celular/fisiologia , Nociceptividade/fisiologia , Células Receptoras Sensoriais/metabolismo , Animais , Sequência Conservada , Proteínas de Drosophila/química , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Deleção de Genes , Células HEK293 , Humanos , Canais Iônicos/química , Canais Iônicos/deficiência , Canais Iônicos/genética , Larva/genética , Larva/metabolismo , Mecanorreceptores/metabolismo , Mecanotransdução Celular/genética , Canais de Sódio/deficiência , Canais de Sódio/genética , Canais de Sódio/metabolismo
6.
Nature ; 483(7388): 176-81, 2012 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-22343900

RESUMO

Mechanotransduction has an important role in physiology. Biological processes including sensing touch and sound waves require as-yet-unidentified cation channels that detect pressure. Mouse Piezo1 (MmPiezo1) and MmPiezo2 (also called Fam38a and Fam38b, respectively) induce mechanically activated cationic currents in cells; however, it is unknown whether Piezo proteins are pore-forming ion channels or modulate ion channels. Here we show that Drosophila melanogaster Piezo (DmPiezo, also called CG8486) also induces mechanically activated currents in cells, but through channels with remarkably distinct pore properties including sensitivity to the pore blocker ruthenium red and single channel conductances. MmPiezo1 assembles as a ∼1.2-million-dalton homo-oligomer, with no evidence of other proteins in this complex. Purified MmPiezo1 reconstituted into asymmetric lipid bilayers and liposomes forms ruthenium-red-sensitive ion channels. These data demonstrate that Piezo proteins are an evolutionarily conserved ion channel family involved in mechanotransduction.


Assuntos
Ativação do Canal Iônico , Canais Iônicos/química , Canais Iônicos/metabolismo , Mecanotransdução Celular/fisiologia , Animais , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Condutividade Elétrica , Células HEK293 , Células HeLa , Humanos , Canais Iônicos/genética , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Porosidade , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(28): 10347-52, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24958852

RESUMO

Mechanosensation is perhaps the last sensory modality not understood at the molecular level. Ion channels that sense mechanical force are postulated to play critical roles in a variety of biological processes including sensing touch/pain (somatosensation), sound (hearing), and shear stress (cardiovascular physiology); however, the identity of these ion channels has remained elusive. We previously identified Piezo1 and Piezo2 as mechanically activated cation channels that are expressed in many mechanosensitive cell types. Here, we show that Piezo1 is expressed in endothelial cells of developing blood vessels in mice. Piezo1-deficient embryos die at midgestation with defects in vascular remodeling, a process critically influenced by blood flow. We demonstrate that Piezo1 is activated by shear stress, the major type of mechanical force experienced by endothelial cells in response to blood flow. Furthermore, loss of Piezo1 in endothelial cells leads to deficits in stress fiber and cellular orientation in response to shear stress, linking Piezo1 mechanotransduction to regulation of cell morphology. These findings highlight an essential role of mammalian Piezo1 in vascular development during embryonic development.


Assuntos
Sistema Cardiovascular/embriologia , Desenvolvimento Embrionário/fisiologia , Células Endoteliais/metabolismo , Canais Iônicos/metabolismo , Mecanotransdução Celular/fisiologia , Animais , Sistema Cardiovascular/citologia , Células Endoteliais/citologia , Canais Iônicos/genética , Camundongos , Camundongos Transgênicos
8.
Proc Natl Acad Sci U S A ; 110(12): 4667-72, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23487782

RESUMO

Mechanotransduction, the pathway by which mechanical forces are translated to biological signals, plays important but poorly characterized roles in physiology. PIEZOs are recently identified, widely expressed, mechanically activated ion channels that are hypothesized to play a role in mechanotransduction in mammals. Here, we describe two distinct PIEZO2 mutations in patients with a subtype of Distal Arthrogryposis Type 5 characterized by generalized autosomal dominant contractures with limited eye movements, restrictive lung disease, and variable absence of cruciate knee ligaments. Electrophysiological studies reveal that the two PIEZO2 mutations affect biophysical properties related to channel inactivation: both E2727del and I802F mutations cause the PIEZO2-dependent, mechanically activated currents to recover faster from inactivation, while E2727del also causes a slowing of inactivation. Both types of changes in kinetics result in increased channel activity in response to a given mechanical stimulus, suggesting that Distal Arthrogryposis Type 5 can be caused by gain-of-function mutations in PIEZO2. We further show that overexpression of mutated PIEZO2 cDNAs does not cause constitutive activity or toxicity to cells, indicating that the observed phenotype is likely due to a mechanotransduction defect. Our studies identify a type of channelopathy and link the dysfunction of mechanically activated ion channels to developmental malformations and joint contractures.


Assuntos
Artrogripose , Doenças Genéticas Inatas , Canais Iônicos/genética , Canais Iônicos/metabolismo , Mecanotransdução Celular/genética , Mutação , Adulto , Artrogripose/genética , Artrogripose/metabolismo , Artrogripose/patologia , Artrogripose/fisiopatologia , Linhagem Celular , Feminino , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Doenças Genéticas Inatas/patologia , Doenças Genéticas Inatas/fisiopatologia , Humanos , Lactente , Recém-Nascido , Masculino
9.
Pflugers Arch ; 467(1): 95-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25037583

RESUMO

Mechanotransduction is the conversion of mechanical stimuli into biological signals. It is involved in the modulation of diverse cellular functions such as migration, proliferation, differentiation, and apoptosis as well as in the detection of sensory stimuli such as air vibration and mechanical contact. Therefore, mechanotransduction is crucial for organ development and homeostasis and plays a direct role in hearing, touch, proprioception, and pain. Multiple molecular players involved in mechanotransduction have been identified in the past, among them ion channels directly activated by cell membrane deformation. Most of these channels have well-established roles in lower organisms but are not conserved in mammals or fail to encode mechanically activated channels in mammals due to non-conservation of mechanotransduction property. A family of mechanically activated channels that counts only two members in human, piezo1 and 2, has emerged recently. Given the lack of valid mechanically activated channel candidates in mammals in the past decades, particular attention is given to piezo channels and their potential roles in various biological functions. This review summarizes our current knowledge on these ion channels.


Assuntos
Canais Iônicos/química , Canais Iônicos/metabolismo , Mecanotransdução Celular/fisiologia , Sensação/fisiologia , Animais , Humanos , Ativação do Canal Iônico/fisiologia , Modelos Biológicos , Estresse Mecânico , Relação Estrutura-Atividade
11.
Nat Chem Biol ; 7(6): 351-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21499266

RESUMO

Intracellular Ca(2+) is essential for diverse cellular functions. Ca(2+) entry into many cell types including immune cells is triggered by depleting endoplasmic reticulum (ER) Ca(2+), a process termed store-operated Ca(2+) entry (SOCE). STIM1 is an ER Ca(2+) sensor. Upon Ca(2+) store depletion, STIM1 clusters at ER-plasma membrane junctions where it interacts with and gates Ca(2+)-permeable Orai1 ion channels. Here we show that STIM1 is also activated by temperature. Heating cells caused clustering of STIM1 at temperatures above 35 °C without depleting Ca(2+) stores and led to Orai1-mediated Ca(2+) influx as a heat off-response (response after cooling). Notably, the functional coupling of STIM1 and Orai1 is prevented at high temperatures, potentially explaining the heat off-response. Additionally, physiologically relevant temperature shifts modulate STIM1-dependent gene expression in Jurkat T cells. Therefore, temperature is an important regulator of STIM1 function.


Assuntos
Cálcio/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Temperatura , Canais de Cálcio/metabolismo , Retículo Endoplasmático , Temperatura Alta , Humanos , Células Jurkat , Proteína ORAI1 , Molécula 1 de Interação Estromal
12.
Neuron ; 110(17): 2713-2727, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35907398

RESUMO

Many ion channels have been described as mechanosensitive according to various criteria. Most broadly defined, an ion channel is called mechanosensitive if its activity is controlled by application of a physical force. The last decade has witnessed a revolution in mechanosensory physiology at the molecular, cellular, and system levels, both in health and in diseases. Since the discovery of the PIEZO proteins as prototypical mechanosensitive channel, many proteins have been proposed to transduce mechanosensory information in mammals. However, few of these newly identified candidates have all the attributes of bona fide, pore-forming mechanosensitive ion channels. In this perspective, we will cover and discuss new data that have advanced our understanding of mechanosensation at the molecular level.


Assuntos
Canais Iônicos , Mecanotransdução Celular , Animais , Canais Iônicos/metabolismo , Mamíferos/metabolismo , Mecanotransdução Celular/fisiologia
13.
J Gen Physiol ; 153(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34694360

RESUMO

Cholangiocytes actively contribute to the final composition of secreted bile. These cells are exposed to abnormal mechanical stimuli during obstructive cholestasis, which has a deep impact on their function. However, the effects of mechanical insults on cholangiocyte function are not understood. Combining gene silencing and pharmacological assays with live calcium imaging, we probed molecular candidates essential for coupling mechanical force to ATP secretion in mouse cholangiocytes. We show that Piezo1 and Pannexin1 are necessary for eliciting the downstream effects of mechanical stress. By mediating a rise in intracellular Ca2+, Piezo1 acts as a mechanosensor responsible for translating cell swelling into activation of Panx1, which triggers ATP release and subsequent signal amplification through P2X4R. Co-immunoprecipitation and pull-down assays indicated physical interaction between Piezo1 and Panx1, which leads to stable plasma membrane complexes. Piezo1-Panx1-P2X4R ATP release pathway could be reconstituted in HEK Piezo1 KO cells. Thus, our data suggest that Piezo1 and Panx1 can form a functional signaling complex that controls force-induced ATP secretion in cholangiocytes. These findings may foster the development of novel therapeutic strategies for biliary diseases.


Assuntos
Trifosfato de Adenosina , Conexinas , Células Epiteliais , Canais Iônicos , Proteínas do Tecido Nervoso , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Conexinas/genética , Células Epiteliais/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Transdução de Sinais
14.
Cell Rep ; 37(5): 109914, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731626

RESUMO

A variety of mechanosensory neurons are involved in touch, proprioception, and pain. Many molecular components of the mechanotransduction machinery subserving these sensory modalities remain to be discovered. Here, we combine recordings of mechanosensitive (MS) currents in mechanosensory neurons with single-cell RNA sequencing. Transcriptional profiles are mapped onto previously identified sensory neuron types to identify cell-type correlates between datasets. Correlation of current signatures with single-cell transcriptomes provides a one-to-one correspondence between mechanoelectric properties and transcriptomically defined neuronal populations. Moreover, a gene-expression differential comparison provides a set of candidate genes for mechanotransduction complexes. Piezo2 is expectedly found to be enriched in rapidly adapting MS current-expressing neurons, whereas Tmem120a and Tmem150c, thought to mediate slow-type MS currents, are uniformly expressed in all mechanosensory neuron subtypes. Further knockdown experiments disqualify them as mediating MS currents in sensory neurons. This dataset constitutes an open resource to explore further the cell-type-specific determinants of mechanosensory properties.


Assuntos
Gânglios Espinais/metabolismo , Perfilação da Expressão Gênica , Mecanotransdução Celular/genética , Neurônios/metabolismo , Transcriptoma , Animais , Gânglios Espinais/citologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Potenciais da Membrana , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Técnicas de Patch-Clamp , RNA-Seq , Análise de Célula Única
15.
PLoS One ; 16(7): e0253562, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34324503

RESUMO

Multiple malformation syndromes (MMS) belong to a group of genetic disorders characterised by neurodevelopmental anomalies and congenital malformations. Here we explore for the first time the genetic aetiology of MMS using whole-exome sequencing (WES) in undiagnosed patients from the Greek-Cypriot population after prior extensive diagnostics workup including karyotype and array-CGH. A total of 100 individuals (37 affected), from 32 families were recruited and family-based WES was applied to detect causative single-nucleotide variants (SNVs) and indels. A genetic diagnosis was reported for 16 MMS patients (43.2%), with 10/17 (58.8%) of the findings being novel. All autosomal dominant findings occurred de novo. Functional studies were also performed to elucidate the molecular mechanism relevant to the abnormal phenotypes, in cases where the clinical significance of the findings was unclear. The 17 variants identified in our cohort were located in 14 genes (PCNT, UBE3A, KAT6A, SPR, POMGNT1, PIEZO2, PXDN, KDM6A, PHIP, HECW2, TFAP2A, CNOT3, AGTPBP1 and GAMT). This study has highlighted the efficacy of WES through the high detection rate (43.2%) achieved for a challenging category of undiagnosed patients with MMS compared to other conventional diagnostic testing methods (10-20% for array-CGH and ~3% for G-banding karyotype analysis). As a result, family-based WES could potentially be considered as a first-tier cost effective diagnostic test for patients with MMS that facilitates better patient management, prognosis and offer accurate recurrence risks to the families.


Assuntos
Anormalidades Múltiplas , Sequenciamento do Exoma , Estudos de Coortes , Humanos , Cariotipagem
17.
Neuron ; 47(2): 179-82, 2005 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16039560

RESUMO

Neuronal Ca2+ channels are key transducers coupling excitability to cellular function. As such, they are tightly regulated by multiple G protein-signaling pathways that finely tune their activity. In addition to fast, direct G(beta)gamma modulation of Ca2+ channels, a slower Galpha(q/11)-mediated mechanism has remained enigmatic despite intensive study. Recent work suggests that membrane phosphoinositides are crucial determinants of Ca2+ channel activity. Here, we discuss their role in Ca2+ channel modulation and the leading theories that seek to elucidate the underlying molecular details of the so-called "mysterious" G(q/11)-mediated signal.


Assuntos
Canais de Cálcio/fisiologia , Fosfatidilinositóis/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Animais , Canais de Cálcio/classificação , Modelos Moleculares , Modelos Neurológicos
18.
Neuron ; 102(2): 373-389.e6, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30819546

RESUMO

Neurons exhibit a limited ability of repair. Given that mechanical forces affect neuronal outgrowth, it is important to investigate whether mechanosensitive ion channels may regulate axon regeneration. Here, we show that DmPiezo, a Ca2+-permeable non-selective cation channel, functions as an intrinsic inhibitor for axon regeneration in Drosophila. DmPiezo activation during axon regeneration induces local Ca2+ transients at the growth cone, leading to activation of nitric oxide synthase and the downstream cGMP kinase Foraging or PKG to restrict axon regrowth. Loss of DmPiezo enhances axon regeneration of sensory neurons in the peripheral and CNS. Conditional knockout of its mammalian homolog Piezo1 in vivo accelerates regeneration, while its pharmacological activation in vitro modestly reduces regeneration, suggesting the role of Piezo in inhibiting regeneration may be evolutionarily conserved. These findings provide a precedent for the involvement of mechanosensitive channels in axon regeneration and add a potential target for modulating nervous system repair.


Assuntos
Axônios/fisiologia , Proteínas de Drosophila/genética , Canais Iônicos/genética , Regeneração/genética , Animais , Cálcio/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Cones de Crescimento/metabolismo , Canais Iônicos/metabolismo , Mecanotransdução Celular/genética , Camundongos , Camundongos Knockout , Regeneração Nervosa/genética , Óxido Nítrico Sintase/metabolismo , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/fisiologia
19.
J Gen Physiol ; 129(1): 57-77, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17190903

RESUMO

Low voltage-activated (LVA) T-type Ca(2+) (I(Ca)T) and NaN/Nav1.9 currents regulate DRG neurons by setting the threshold for the action potential. Although alterations in these channels have been implicated in a variety of pathological pain states, their roles in processing sensory information remain poorly understood. Here, we carried out a detailed characterization of LVA currents in DRG neurons by using a method for better separation of NaN/Nav1.9 and I(Ca)T currents. NaN/Nav1.9 was inhibited by inorganic I(Ca) blockers as follows (IC(50), microM): La(3+) (46) > Cd(2+) (233) > Ni(2+) (892) and by mibefradil, a non-dihydropyridine I(Ca)T antagonist. Amiloride, however, a preferential Cav3.2 channel blocker, had no effects on NaN/Nav1.9 current. Using these discriminative tools, we showed that NaN/Nav1.9, Cav3.2, and amiloride- and Ni(2+)-resistant I(Ca)T (AR-I(Ca)T) contribute differentially to LVA currents in distinct sensory cell populations. NaN/Nav1.9 carried LVA currents into type-I (CI) and type-II (CII) small nociceptors and medium-Adelta-like nociceptive cells but not in low-threshold mechanoreceptors, including putative Down-hair (D-hair) and Aalpha/beta cells. Cav3.2 predominated in CII-nociceptors and in putative D-hair cells. AR-I(Ca)T was restricted to CII-nociceptors, putative D-hair cells, and Aalpha/beta-like cells. These cell types distinguished by their current-signature displayed different types of mechanosensitive channels. CI- and CII-nociceptors displayed amiloride-sensitive high-threshold mechanical currents with slow or no adaptation, respectively. Putative D-hair and Aalpha/beta-like cells had low-threshold mechanical currents, which were distinguished by their adapting kinetics and sensitivity to amiloride. Thus, subspecialized DRG cells express specific combinations of LVA and mechanosensitive channels, which are likely to play a key role in shaping responses of DRG neurons transmitting different sensory modalities.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Cátions/metabolismo , Gânglios Espinais/metabolismo , Mecanorreceptores/metabolismo , Neuropeptídeos/metabolismo , Nociceptores/metabolismo , Canais de Sódio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Amilorida/farmacologia , Animais , Cádmio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Células Cultivadas , Eletrofisiologia , Gânglios Espinais/citologia , Lantânio/farmacologia , Masculino , Mibefradil/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.9 , Neuropeptídeos/efeitos dos fármacos , Níquel/farmacologia , Ratos , Ratos Wistar , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos
20.
Sensors (Basel) ; 7(9): 1667-1682, 2007 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-28903189

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA