Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Cell ; 184(4): 969-982.e13, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33571427

RESUMO

Iron overload causes progressive organ damage and is associated with arthritis, liver damage, and heart failure. Elevated iron levels are present in 1%-5% of individuals; however, iron overload is undermonitored and underdiagnosed. Genetic factors affecting iron homeostasis are emerging. Individuals with hereditary xerocytosis, a rare disorder with gain-of-function (GOF) mutations in mechanosensitive PIEZO1 ion channel, develop age-onset iron overload. We show that constitutive or macrophage expression of a GOF Piezo1 allele in mice disrupts levels of the iron regulator hepcidin and causes iron overload. We further show that PIEZO1 is a key regulator of macrophage phagocytic activity and subsequent erythrocyte turnover. Strikingly, we find that E756del, a mild GOF PIEZO1 allele present in one-third of individuals of African descent, is strongly associated with increased plasma iron. Our study links macrophage mechanotransduction to iron metabolism and identifies a genetic risk factor for increased iron levels in African Americans.


Assuntos
Canais Iônicos/metabolismo , Ferro/metabolismo , Negro ou Afro-Americano , Envelhecimento/metabolismo , Alelos , Animais , Estudos de Coortes , Contagem de Eritrócitos , Eritropoese , Mutação com Ganho de Função/genética , Hepatócitos/metabolismo , Hepcidinas/sangue , Hepcidinas/metabolismo , Humanos , Ferro/sangue , Sobrecarga de Ferro/metabolismo , Macrófagos/metabolismo , Mecanotransdução Celular , Camundongos Endogâmicos C57BL , Fagocitose , Fenótipo , Estresse Fisiológico
2.
Cell ; 164(3): 499-511, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26824658

RESUMO

The volume-regulated anion channel (VRAC) is activated when a cell swells, and it plays a central role in maintaining cell volume in response to osmotic challenges. SWELL1 (LRRC8A) was recently identified as an essential component of VRAC. However, the identity of the pore-forming subunits of VRAC and how the channel is gated by cell swelling are unknown. Here, we show that SWELL1 and up to four other LRRC8 subunits assemble into heterogeneous complexes of ∼800 kDa. When reconstituted into bilayers, LRRC8 complexes are sufficient to form anion channels activated by osmolality gradients. In bilayers, as well as in cells, the single-channel conductance of the complexes depends on the LRRC8 composition. Finally, low ionic strength (Γ) in the absence of an osmotic gradient activates the complexes in bilayers. These data demonstrate that LRRC8 proteins together constitute the VRAC pore and that hypotonic stress can activate VRAC through a decrease in cytoplasmic Γ.


Assuntos
Canais Iônicos/metabolismo , Proteínas de Membrana/metabolismo , Células HeLa , Humanos , Canais Iônicos/química , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Osmose
3.
Nat Rev Mol Cell Biol ; 18(12): 771-783, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28974772

RESUMO

Cellular mechanotransduction, the process of translating mechanical forces into biological signals, is crucial for a wide range of physiological processes. A role for ion channels in sensing mechanical forces has been proposed for decades, but their identity in mammals remained largely elusive until the discovery of Piezos. Recent research on Piezos has underscored their importance in somatosensation (touch perception, proprioception and pulmonary respiration), red blood cell volume regulation, vascular physiology and various human genetic disorders.


Assuntos
Doenças Genéticas Inatas/metabolismo , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Propriocepção , Mecânica Respiratória , Percepção do Tato , Animais , Doenças Genéticas Inatas/genética , Humanos , Canais Iônicos/genética
4.
Cell ; 157(2): 447-458, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24725410

RESUMO

Maintenance of a constant cell volume in response to extracellular or intracellular osmotic changes is critical for cellular homeostasis. Activation of a ubiquitous volume-regulated anion channel (VRAC) plays a key role in this process; however, its molecular identity in vertebrates remains unknown. Here, we used a cell-based fluorescence assay and performed a genome-wide RNAi screen to find components of VRAC. We identified SWELL1 (LRRC8A), a member of a four-transmembrane protein family with unknown function, as essential for hypotonicity-induced iodide influx. SWELL1 is localized to the plasma membrane, and its knockdown dramatically reduces endogenous VRAC currents and regulatory cell volume decrease in various cell types. Furthermore, point mutations in SWELL1 cause a significant change in VRAC anion selectivity, demonstrating that SWELL1 is an essential VRAC component. These findings enable further molecular characterization of the VRAC channel complex and genetic studies for understanding the function of VRAC in normal physiology and disease.


Assuntos
Tamanho Celular , Proteínas de Membrana/metabolismo , Animais , Membrana Celular/química , Membrana Celular/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Estudo de Associação Genômica Ampla , Células HEK293 , Células HeLa , Humanos , Iodetos/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Interferência de RNA
5.
Nature ; 620(7976): 1117-1125, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37587339

RESUMO

PIEZOs are mechanosensitive ion channels that convert force into chemoelectric signals1,2 and have essential roles in diverse physiological settings3. In vitro studies have proposed that PIEZO channels transduce mechanical force through the deformation of extensive blades of transmembrane domains emanating from a central ion-conducting pore4-8. However, little is known about how these channels interact with their native environment and which molecular movements underlie activation. Here we directly observe the conformational dynamics of the blades of individual PIEZO1 molecules in a cell using nanoscopic fluorescence imaging. Compared with previous structural models of PIEZO1, we show that the blades are significantly expanded at rest by the bending stress exerted by the plasma membrane. The degree of expansion varies dramatically along the length of the blade, where decreased binding strength between subdomains can explain increased flexibility of the distal blade. Using chemical and mechanical modulators of PIEZO1, we show that blade expansion and channel activation are correlated. Our findings begin to uncover how PIEZO1 is activated in a native environment. More generally, as we reliably detect conformational shifts of single nanometres from populations of channels, we expect that this approach will serve as a framework for the structural analysis of membrane proteins through nanoscopic imaging.


Assuntos
Canais Iônicos , Membrana Celular/metabolismo , Fluorescência , Canais Iônicos/química , Canais Iônicos/metabolismo , Modelos Moleculares , Movimento , Conformação Proteica , Análise de Célula Única
6.
Nature ; 607(7917): 104-110, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35732741

RESUMO

Itch triggers scratching, a behavioural defence mechanism that aids in the removal of harmful irritants and parasites1. Chemical itch is triggered by many endogenous and exogenous cues, such as pro-inflammatory histamine, which is released during an allergic reaction1. Mechanical itch can be triggered by light sensations such as wool fibres or a crawling insect2. In contrast to chemical itch pathways, which have been extensively studied, the mechanisms that underlie the transduction of mechanical itch are largely unknown. Here we show that the mechanically activated ion channel PIEZO1 (ref. 3) is selectively expressed by itch-specific sensory neurons and is required for their mechanically activated currents. Loss of PIEZO1 function in peripheral neurons greatly reduces mechanically evoked scratching behaviours and both acute and chronic itch-evoked sensitization. Finally, mice expressing a gain-of-function Piezo1 allele4 exhibit enhanced mechanical itch behaviours. Our studies reveal the polymodal nature of itch sensory neurons and identify a role for PIEZO1 in the sensation of itch.


Assuntos
Canais Iônicos , Prurido , Alelos , Animais , Canais Iônicos/deficiência , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Prurido/genética , Prurido/fisiopatologia , Sensação , Células Receptoras Sensoriais/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-33975957

RESUMO

Plant roots adapt to the mechanical constraints of the soil to grow and absorb water and nutrients. As in animal species, mechanosensitive ion channels in plants are proposed to transduce external mechanical forces into biological signals. However, the identity of these plant root ion channels remains unknown. Here, we show that Arabidopsis thaliana PIEZO1 (PZO1) has preserved the function of its animal relatives and acts as an ion channel. We present evidence that plant PIEZO1 is expressed in the columella and lateral root cap cells of the root tip, which are known to experience robust mechanical strain during root growth. Deleting PZO1 from the whole plant significantly reduced the ability of its roots to penetrate denser barriers compared to wild-type plants. pzo1 mutant root tips exhibited diminished calcium transients in response to mechanical stimulation, supporting a role of PZO1 in root mechanotransduction. Finally, a chimeric PZO1 channel that includes the C-terminal half of PZO1 containing the putative pore region was functional and mechanosensitive when expressed in naive mammalian cells. Collectively, our data suggest that Arabidopsis PIEZO1 plays an important role in root mechanotransduction and establish PIEZOs as physiologically relevant mechanosensitive ion channels across animal and plant kingdoms.


Assuntos
Proteínas de Arabidopsis/fisiologia , Arabidopsis/fisiologia , Mecanotransdução Celular/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Raízes de Plantas/fisiologia
8.
Nature ; 509(7502): 622-6, 2014 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-24717433

RESUMO

How we sense touch remains fundamentally unknown. The Merkel cell-neurite complex is a gentle touch receptor in the skin that mediates slowly adapting responses of Aß sensory fibres to encode fine details of objects. This mechanoreceptor complex was recognized to have an essential role in sensing gentle touch nearly 50 years ago. However, whether Merkel cells or afferent fibres themselves sense mechanical force is still debated, and the molecular mechanism of mechanotransduction is unknown. Synapse-like junctions are observed between Merkel cells and associated afferents, and yet it is unclear whether Merkel cells are inherently mechanosensitive or whether they can rapidly transmit such information to the neighbouring nerve. Here we show that Merkel cells produce touch-sensitive currents in vitro. Piezo2, a mechanically activated cation channel, is expressed in Merkel cells. We engineered mice deficient in Piezo2 in the skin, but not in sensory neurons, and show that Merkel-cell mechanosensitivity completely depends on Piezo2. In these mice, slowly adapting responses in vivo mediated by the Merkel cell-neurite complex show reduced static firing rates, and moreover, the mice display moderately decreased behavioural responses to gentle touch. Our results indicate that Piezo2 is the Merkel-cell mechanotransduction channel and provide the first line of evidence that Piezo channels have a physiological role in mechanosensation in mammals. Furthermore, our data present evidence for a two-receptor-site model, in which both Merkel cells and innervating afferents act together as mechanosensors. The two-receptor system could provide this mechanoreceptor complex with a tuning mechanism to achieve highly sophisticated responses to a given mechanical stimulus.


Assuntos
Canais Iônicos/metabolismo , Mecanotransdução Celular , Células de Merkel/metabolismo , Tato/fisiologia , Potenciais de Ação , Animais , Condutividade Elétrica , Feminino , Técnicas In Vitro , Canais Iônicos/deficiência , Canais Iônicos/genética , Masculino , Mecanotransdução Celular/genética , Camundongos , Camundongos Knockout , Neuritos/metabolismo , Neurônios Aferentes/metabolismo , Pele/citologia , Pele/inervação , Tato/genética
9.
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
10.
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
11.
Proc Natl Acad Sci U S A ; 108(48): 19413-8, 2011 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22087007

RESUMO

The voltage-gated sodium channel Na(v)1.8 is known to function in the transmission of pain signals induced by cold, heat, and mechanical stimuli. Sequence variants of human Na(v)1.8 have been linked to altered cardiac conduction. We identified an allele of Scn10a encoding the α-subunit of Na(v)1.8 among mice homozygous for N-ethyl-N-nitrosourea-induced mutations. The allele creates a dominant neurobehavioral phenotype termed Possum, characterized by transient whole-body tonic immobility induced by pinching the skin at the back of the neck ("scruffing"). The Possum mutation enhanced Na(v)1.8 sodium currents and neuronal excitability and heightened sensitivity of mutants to cold stimuli. Striking electroencephalographic changes were observed concomitant with the scruffing-induced behavioral change. In addition, electrocardiography demonstrated that Possum mice exhibited marked sinus bradycardia and R-R variability upon scruffing, abrogated by infusion of atropine. However, atropine failed to prevent or mitigate the tonic immobility response. Hyperactive sodium conduction via Na(v)1.8 thus leads to a complex neurobehavioral phenotype, which resembles catatonia in schizophrenic humans and tonic immobility in other mammals upon application of a discrete stimulus; no other form of mechanosensory stimulus could induce the immobility phenotype. Our data confirm the involvement of Na(v)1.8 in transducing pain initiated by cold and additionally implicate Na(v)1.8 in previously unknown functions in the central nervous system and heart.


Assuntos
Resposta de Imobilidade Tônica/fisiologia , Mutação/genética , Fenótipo , Canais de Sódio/genética , Animais , Atropina/farmacologia , Bradicardia/genética , Eletrocardiografia , Eletroencefalografia , Resposta de Imobilidade Tônica/efeitos dos fármacos , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.8 , Canais de Sódio/fisiologia
12.
bioRxiv ; 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-37873218

RESUMO

The dimeric two-pore OSCA/TMEM63 family has recently been identified as mechanically activated ion channels. Previously, based on the unique features of the structure of OSCA1.2, we postulated the potential involvement of several structural elements in sensing membrane tension1. Interestingly, while OSCA1, 2, and 3 clades are activated by membrane stretch in cell-attached patches (i.e., they are stretch-activated channels), they differ in their ability to transduce membrane deformation induced by a blunt probe (poking). In an effort to understand the domains contributing to mechanical signal transduction, we used cryo-electron microscopy to solve the structure of Arabidopsis thaliana (At) OSCA3.1, which, unlike AtOSCA1.2, only produced stretch- but not poke-activated currents in our initial characterization2. Mutagenesis and electrophysiological assessment of conserved and divergent putative mechanosensitive features of OSCA1.2 reveal a selective disruption of the macroscopic currents elicited by poking without considerable effects on stretch-activated currents (SAC). Our results support the involvement of the amphipathic helix and lipid-interacting residues in the membrane fenestration in the response to poking. Our findings position these two structural elements as potential sources of functional diversity within the family.

13.
Elife ; 122024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38592763

RESUMO

The dimeric two-pore OSCA/TMEM63 family has recently been identified as mechanically activated ion channels. Previously, based on the unique features of the structure of OSCA1.2, we postulated the potential involvement of several structural elements in sensing membrane tension (Jojoa-Cruz et al., 2018). Interestingly, while OSCA1, 2, and 3 clades are activated by membrane stretch in cell-attached patches (i.e. they are stretch-activated channels), they differ in their ability to transduce membrane deformation induced by a blunt probe (poking). Here, in an effort to understand the domains contributing to mechanical signal transduction, we used cryo-electron microscopy to solve the structure of Arabidopsis thaliana (At) OSCA3.1, which, unlike AtOSCA1.2, only produced stretch- but not poke-activated currents in our initial characterization (Murthy et al., 2018). Mutagenesis and electrophysiological assessment of conserved and divergent putative mechanosensitive features of OSCA1.2 reveal a selective disruption of the macroscopic currents elicited by poking without considerable effects on stretch-activated currents (SAC). Our results support the involvement of the amphipathic helix and lipid-interacting residues in the membrane fenestration in the response to poking. Our findings position these two structural elements as potential sources of functional diversity within the family.


Assuntos
Arabidopsis , Microscopia Crioeletrônica , Arabidopsis/genética , Membrana Celular , Mecanotransdução Celular , Mutagênese
14.
Nature ; 445(7127): 541-5, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17237762

RESUMO

The nervous system senses peripheral damage through nociceptive neurons that transmit a pain signal. TRPA1 is a member of the Transient Receptor Potential (TRP) family of ion channels and is expressed in nociceptive neurons. TRPA1 is activated by a variety of noxious stimuli, including cold temperatures, pungent natural compounds, and environmental irritants. How such diverse stimuli activate TRPA1 is not known. We observed that most compounds known to activate TRPA1 are able to covalently bind cysteine residues. Here we use click chemistry to show that derivatives of two such compounds, mustard oil and cinnamaldehyde, covalently bind mouse TRPA1. Structurally unrelated cysteine-modifying agents such as iodoacetamide (IA) and (2-aminoethyl)methanethiosulphonate (MTSEA) also bind and activate TRPA1. We identified by mass spectrometry fourteen cytosolic TRPA1 cysteines labelled by IA, three of which are required for normal channel function. In excised patches, reactive compounds activated TRPA1 currents that were maintained at least 10 min after washout of the compound in calcium-free solutions. Finally, activation of TRPA1 by disulphide-bond-forming MTSEA is blocked by the reducing agent dithiothreitol (DTT). Collectively, our data indicate that covalent modification of reactive cysteines within TRPA1 can cause channel activation, rapidly signalling potential tissue damage through the pain pathway.


Assuntos
Cisteína/metabolismo , Dissulfetos/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Noxas/farmacologia , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/metabolismo , Acroleína/análogos & derivados , Acroleína/química , Acroleína/metabolismo , Acroleína/farmacologia , Animais , Cisteína/química , Dissulfetos/química , Ditiotreitol/farmacologia , Condutividade Elétrica , Metanossulfonato de Etila/análogos & derivados , Metanossulfonato de Etila/química , Metanossulfonato de Etila/metabolismo , Metanossulfonato de Etila/farmacologia , Humanos , Camundongos , Mostardeira/química , Mostardeira/metabolismo , Noxas/química , Noxas/metabolismo , Dor/induzido quimicamente , Dor/fisiopatologia , Óleos de Plantas/química , Óleos de Plantas/metabolismo , Óleos de Plantas/farmacologia , Canais de Potencial de Receptor Transitório/química
15.
Science ; 379(6628): 201-206, 2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36634173

RESUMO

Distal arthrogryposis (DA) is a collection of rare disorders that are characterized by congenital joint contractures. Most DA mutations are in muscle- and joint-related genes, and the anatomical defects originate cell-autonomously within the musculoskeletal system. However, gain-of-function mutations in PIEZO2, a principal mechanosensor in somatosensation, cause DA subtype 5 (DA5) through unknown mechanisms. We show that expression of a gain-of-function PIEZO2 mutation in proprioceptive sensory neurons that mainly innervate muscle spindles and tendons is sufficient to induce DA5-like phenotypes in mice. Overactive PIEZO2 causes anatomical defects through increased activity within the peripheral nervous system during postnatal development. Furthermore, botulinum toxin (Botox) and a dietary fatty acid that modulates PIEZO2 activity reduce DA5-like deficits. This reveals a role for somatosensory neurons: Excessive mechanosensation within these neurons disrupts musculoskeletal development.


Assuntos
Artrogripose , Contratura , Canais Iônicos , Mecanotransdução Celular , Células Receptoras Sensoriais , Animais , Camundongos , Artrogripose/genética , Artrogripose/fisiopatologia , Contratura/genética , Contratura/fisiopatologia , Mecanotransdução Celular/genética , Mutação , Células Receptoras Sensoriais/fisiologia , Canais Iônicos/genética
16.
Neuron ; 111(16): 2488-2501.e8, 2023 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-37321223

RESUMO

Sensory neurons detect mechanical forces from both the environment and internal organs to regulate physiology. PIEZO2 is a mechanosensory ion channel critical for touch, proprioception, and bladder stretch sensation, yet its broad expression in sensory neurons suggests it has undiscovered physiological roles. To fully understand mechanosensory physiology, we must know where and when PIEZO2-expressing neurons detect force. The fluorescent styryl dye FM 1-43 was previously shown to label sensory neurons. Surprisingly, we find that the vast majority of FM 1-43 somatosensory neuron labeling in mice in vivo is dependent on PIEZO2 activity within the peripheral nerve endings. We illustrate the potential of FM 1-43 by using it to identify novel PIEZO2-expressing urethral neurons that are engaged by urination. These data reveal that FM 1-43 is a functional probe for mechanosensitivity via PIEZO2 activation in vivo and will facilitate the characterization of known and novel mechanosensory processes in multiple organ systems.


Assuntos
Canais Iônicos , Mecanotransdução Celular , Camundongos , Animais , Mecanotransdução Celular/fisiologia , Canais Iônicos/metabolismo , Células Receptoras Sensoriais/fisiologia , Compostos de Piridínio/metabolismo
17.
J Neurosci ; 30(21): 7300-9, 2010 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-20505096

RESUMO

Lysophosphatidic acid (LPA) is a membrane-derived lysophospholipid that can induce pleomorphic effects in neural progenitor cells (NPCs) from the cerebral cortex, including alterations in ionic conductance. LPA-induced, calcium-mediated conductance changes have been reported; however, the underlying molecular mechanisms have not been determined. We show here that activation of specific cognate receptors accounts for nearly all intracellular calcium responses evoked by LPA in acutely cultured nestin-positive NPCs from the developing mouse cerebral cortex. Fast-onset changes in intracellular calcium levels required release from thapsigargin-sensitive stores by a pertussis toxin-insensitive mechanism. The influx of extracellular calcium through Cd(2+)/Ni(2+)-insensitive influx pathways, approximately one-half of which were Gd(3+) sensitive, contributed to the temporal diversity of responses. Quantitative reverse transcription-PCR revealed the presence of all five known LPA receptors in primary NPCs, with prominent expression of LPA(1), LPA(2), and LPA(4). Combined genetic and pharmacological studies indicated that NPC responses were mediated by LPA(1) (approximately 30% of the cells), LPA(2) (approximately 30%), a combination of receptors on single cells (approximately 30%), and non-LPA(1,2,3) pathways (approximately 10%). LPA responsivity was significantly reduced in more differentiated TuJ1(+) cells within cultures. Calcium transients in a large proportion of LPA-responsive NPCs were also initiated by the closely related signaling lipid S1P (sphingosine-1-phosphate). These data demonstrate for the first time the involvement of LPA receptors in mediating surprisingly diverse NPC calcium responses involving multiple receptor subtypes that function within a single cell. Compared with other known factors, lysophospholipids represent the major activator of calcium signaling identified within NPCs at this early stage in corticogenesis.


Assuntos
Sinalização do Cálcio/fisiologia , Córtex Cerebral/citologia , Líquido Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Neurogênese/fisiologia , Neurônios/citologia , Animais , Cloreto de Cádmio/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Ácido Glutâmico/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/classificação , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Isoxazóis/farmacologia , Lisofosfolipídeos/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/classificação , Proteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Níquel/farmacologia , Organofosfatos/farmacologia , Toxina Pertussis/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Gravidez , Propionatos/farmacologia , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tapsigargina/farmacologia
18.
J Neurosci ; 29(1): 153-8, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19129393

RESUMO

Maintaining physiological pH is required for survival, and exposure to alkaline chemicals such as ammonia (smelling salts) elicits severe pain and inflammation through unknown mechanisms. TRPV1, the capsaicin receptor, is an integrator of noxious stimuli including heat and extracellular acidic pH. Here, we report that ammonia activates TRPV1, TRPA1 (another polymodal nocisensor), and other unknown receptor(s) expressed in sensory neurons. Ammonia and intracellular alkalization activate TRPV1 through a mechanism that involves a cytoplasmic histidine residue, not used by other TRPV1 agonists such as heat, capsaicin or low pH. Our studies show that TRPV1 detects both acidic and basic deviations from homeostatic pH.


Assuntos
Ácidos/farmacologia , Cloreto de Amônio/farmacologia , Células Receptoras Sensoriais/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Capsaicina/farmacologia , Linhagem Celular Transformada , Gânglios Espinais/citologia , Humanos , Concentração de Íons de Hidrogênio , Proteínas Luminescentes/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Mutagênese Sítio-Dirigida/métodos , Técnicas de Patch-Clamp , Pirazinas/farmacologia , Piridinas/farmacologia , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/deficiência , Transfecção/métodos
19.
Nat Neurosci ; 9(4): 493-500, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16520735

RESUMO

Menthol is a cooling compound derived from mint leaves and is extensively used as a flavoring chemical. Menthol activates transient receptor potential melastatin 8 (TRPM8), an ion channel also activated by cold, voltage and phosphatidylinositol-4,5-bisphosphate (PIP2). Here we investigated the mechanism by which menthol activates mouse TRPM8. Using a new high-throughput approach, we screened a random mutant library consisting of approximately 14,000 individual TRPM8 mutants for clones that are affected in their response to menthol while retaining channel function. We identified determinants of menthol sensitivity in two regions: putative transmembrane segment 2 (S2) and the C-terminal TRP domain. Analysis of these mutants indicated that activation by menthol involves a gating mechanism distinct and separable from gating by cold, voltage or PIP2. Notably, TRP domain mutations mainly attenuated menthol efficacy, suggesting that this domain influences events downstream of initial binding. In contrast, S2 mutations strongly shifted the concentration dependence of menthol activation, raising the possibility that S2 influences menthol binding.


Assuntos
Temperatura Baixa , Mentol/metabolismo , Canais de Cátion TRPM/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Biblioteca Gênica , Mentol/química , Camundongos , Dados de Sequência Molecular , Estrutura Molecular , Mutagênese , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Pirimidinonas/metabolismo , Canais de Cátion TRPM/química , Canais de Cátion TRPM/genética
20.
Brain Res Rev ; 60(1): 2-23, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19203589

RESUMO

Pain perception begins with the activation of primary sensory nociceptors. Over the past decade, flourishing research has revealed that members of the Transient Receptor Potential (TRP) ion channel family are fundamental molecules that detect noxious stimuli and transduce a diverse range of physical and chemical energy into action potentials in somatosensory nociceptors. Here we highlight the roles of TRP vanilloid 1 (TRPV1), TRP melastatin 8 (TRPM8) and TRP ankyrin 1 (TRPA1) in the activation of nociceptors by heat and cold environmental stimuli, mechanical force, and by chemicals including exogenous plant and environmental compounds as well as endogenous inflammatory molecules. The contribution of these channels to pain and somatosensation is discussed at levels ranging from whole animal behavior to molecular modulation by intracellular signaling proteins. An emerging theme is that TRP channels are not simple ion channel transducers of one or two stimuli, but instead serve multidimensional roles in signaling sensory stimuli that are exceptionally diverse in modality and in their environmental milieu.


Assuntos
Sistema Nervoso/fisiopatologia , Nociceptores/fisiologia , Dor/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Anquirinas/fisiologia , Humanos , Sistema Nervoso/metabolismo , Dor/metabolismo , Sensação/fisiologia , Canais de Cátion TRPM/fisiologia , Canais de Cátion TRPV/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA