Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 606(7912): 137-145, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614217

RESUMO

Nerve injury leads to chronic pain and exaggerated sensitivity to gentle touch (allodynia) as well as a loss of sensation in the areas in which injured and non-injured nerves come together1-3. The mechanisms that disambiguate these mixed and paradoxical symptoms are unknown. Here we longitudinally and non-invasively imaged genetically labelled populations of fibres that sense noxious stimuli (nociceptors) and gentle touch (low-threshold afferents) peripherally in the skin for longer than 10 months after nerve injury, while simultaneously tracking pain-related behaviour in the same mice. Fully denervated areas of skin initially lost sensation, gradually recovered normal sensitivity and developed marked allodynia and aversion to gentle touch several months after injury. This reinnervation-induced neuropathic pain involved nociceptors that sprouted into denervated territories precisely reproducing the initial pattern of innervation, were guided by blood vessels and showed irregular terminal connectivity in the skin and lowered activation thresholds mimicking low-threshold afferents. By contrast, low-threshold afferents-which normally mediate touch sensation as well as allodynia in intact nerve territories after injury4-7-did not reinnervate, leading to an aberrant innervation of tactile end organs such as Meissner corpuscles with nociceptors alone. Genetic ablation of nociceptors fully abrogated reinnervation allodynia. Our results thus reveal the emergence of a form of chronic neuropathic pain that is driven by structural plasticity, abnormal terminal connectivity and malfunction of nociceptors during reinnervation, and provide a mechanistic framework for the paradoxical sensory manifestations that are observed clinically and can impose a heavy burden on patients.


Assuntos
Hiperalgesia , Neuralgia , Nociceptores , Pele , Animais , Dor Crônica/fisiopatologia , Hiperalgesia/fisiopatologia , Mecanorreceptores/patologia , Camundongos , Neuralgia/fisiopatologia , Nociceptores/patologia , Pele/inervação , Pele/fisiopatologia
2.
J Biol Chem ; 299(6): 104782, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37146970

RESUMO

PKA is a downstream effector of many inflammatory mediators that induce pain hypersensitivity by increasing the mechanosensitivity of nociceptive sensory afferent. Here, we examine the molecular mechanism underlying PKA-dependent modulation of the mechanically activated ion channel PIEZO2, which confers mechanosensitivity to many nociceptors. Using phosphorylation site prediction algorithms, we identified multiple putative and highly conserved PKA phosphorylation sites located on intracellular intrinsically disordered regions of PIEZO2. Site-directed mutagenesis and patch-clamp recordings showed that substitution of one or multiple putative PKA sites within a single intracellular domain does not alter PKA-induced PIEZO2 sensitization, whereas mutation of a combination of nine putative sites located on four different intracellular regions completely abolishes PKA-dependent PIEZO2 modulation, though it remains unclear whether all or just some of these nine sites are required. By demonstrating that PIEZO1 is not modulated by PKA, our data also reveal a previously unrecognized functional difference between PIEZO1 and PIEZO2. Moreover, by demonstrating that PKA only modulates PIEZO2 currents evoked by focal mechanical indentation of the cell, but not currents evoked by pressure-induced membrane stretch, we provide evidence suggesting that PIEZO2 is a polymodal mechanosensor that engages different protein domains for detecting different types of mechanical stimuli.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico , Canais Iônicos , Mecanotransdução Celular , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Mecanotransdução Celular/genética , Dor/fisiopatologia , Domínios Proteicos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Transporte Proteico/genética
3.
Proc Natl Acad Sci U S A ; 116(28): 14260-14269, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31235572

RESUMO

Piezo channels are mechanically activated ion channels that confer mechanosensitivity to a variety of different cell types. Piezos oligomerize as propeller-shaped homotrimers that are thought to locally curve the membrane into spherical domes that project into the cell. While several studies have identified domains and amino acids that control important properties such as ion permeability and selectivity as well as inactivation kinetics and voltage sensitivity, only little is known about intraprotein interactions that govern mechanosensitivity-the most unique feature of PIEZOs. Here we used site-directed mutagenesis and patch-clamp recordings to investigate the mechanogating mechanism of PIEZO2. We demonstrate that charged amino acids at the interface between the beam domain-i.e., a long α-helix that protrudes from the intracellular side of the "propeller" blade toward the inner vestibule of the channel-and the C-terminal domain (CTD) as well as hydrophobic interactions between the highly conserved Y2807 of the CTD and pore-lining helices are required to ensure normal mechanosensitivity of PIEZO2. Moreover, single-channel recordings indicate that a previously unrecognized intrinsically disordered domain located adjacent to the beam acts as a cytosolic plug that limits ion permeation possibly by clogging the inner vestibule of both PIEZO1 and PIEZO2. Thus, we have identified several intraprotein domain interfaces that control the mechanical activation of PIEZO1 and PIEZO2 and which might thus serve as promising targets for drugs that modulate the mechanosensitivity of Piezo channels.

4.
Mol Pain ; 14: 1744806918814640, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30387376

RESUMO

Nerve growth factor is an inflammatory mediator that induces long-lasting hyperalgesia, which can partially be attributed to nerve growth factor-induced sensitization of primary afferent nociceptors. It was shown that nerve growth factor increases the excitability of polymodal C-fibre nociceptors by modulating tetrodotoxin-sensitive and tetrodotoxin-resistant voltage-gated sodium channels, but hitherto only little is known about the effects of nerve growth factor on sodium currents in other nociceptor subtypes that express the nerve growth factor receptor TrkA. We previously characterized two reporter mouse lines that allow the unequivocal identification of two important subclasses of TrkA-expressing nociceptors - i.e. neuropeptide Y receptor type 2 (NPY2R+ ) Aδ-fibre nociceptors that mediate pinprick pain and nicotinic acetylcholine receptor alpha-3 subunit (CHRNA3+ ) silent nociceptors, which are the most abundant TrkA+ nociceptors in visceral organs and deep somatic tissues. Here, we utilized these mouse lines to investigate the expression patterns and the possible nerve growth factor-dependent modulation of sodium channels in these neurons using whole-cell patch-clamp recordings and quantitative real-time polymerase chain reaction. We demonstrate that NPY2R+ nociceptors, CHRNA3+ 'silent' nociceptors and polymodal C-fibre nociceptors express different combinations of sodium channel α- and ß-subunits and accordingly exhibit functionally different sodium currents. Moreover, we demonstrate that nerve growth factor produces robust hyperpolarizing shifts in the half-activation voltage of tetrodotoxin-resistant currents in NPY2R+ nociceptors and polymodal C-fibre nociceptors and also shifts the half-activation of tetrodotoxin-sensitive currents in polymodal C-fibre nociceptors. In silent nociceptors, however, nerve growth factor solely increases the current density of the tetrodotoxin-resistant current but does not alter other sodium channel properties. Considering the different peripheral target tissues and the previously reported roles in different forms of pain of the nociceptor subpopulations that were examined here, our results suggest that nerve growth factor differentially contributes to the development visceral and cutaneous pain hypersensitivity and highlights the importance of developing different therapeutic strategies for different forms of pain.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Nociceptores/metabolismo , Tetrodotoxina/farmacologia , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Gânglios Espinais/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nociceptores/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/metabolismo
5.
PLoS Biol ; 10(5): e1001318, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563300

RESUMO

In all vertebrates hearing and touch represent two distinct sensory systems that both rely on the transformation of mechanical force into electrical signals. There is an extensive literature describing single gene mutations in humans that cause hearing impairment, but there are essentially none for touch. Here we first asked if touch sensitivity is a heritable trait and second whether there are common genes that influence different mechanosensory senses like hearing and touch in humans. Using a classical twin study design we demonstrate that touch sensitivity and touch acuity are highly heritable traits. Quantitative phenotypic measures of different mechanosensory systems revealed significant correlations between touch and hearing acuity in a healthy human population. Thus mutations in genes causing deafness genes could conceivably negatively influence touch sensitivity. In agreement with this hypothesis we found that a proportion of a cohort of congenitally deaf young adults display significantly impaired measures of touch sensitivity compared to controls. In contrast, blind individuals showed enhanced, not diminished touch acuity. Finally, by examining a cohort of patients with Usher syndrome, a genetically well-characterized deaf-blindness syndrome, we could show that recessive pathogenic mutations in the USH2A gene influence touch acuity. Control Usher syndrome cohorts lacking demonstrable pathogenic USH2A mutations showed no impairment in touch acuity. Our study thus provides comprehensive evidence that there are common genetic elements that contribute to touch and hearing and has identified one of these genes as USH2A.


Assuntos
Perda Auditiva/genética , Mecanotransdução Celular , Tato/genética , Síndromes de Usher/genética , Adolescente , Adulto , Fatores Etários , Barorreflexo , Estudos de Coortes , Proteínas da Matriz Extracelular/genética , Feminino , Testes Genéticos , Genótipo , Perda Auditiva/congênito , Humanos , Padrões de Herança , Masculino , Mutação , Fenótipo , Temperatura , Adulto Jovem
6.
Physiology (Bethesda) ; 28(3): 142-50, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23636260

RESUMO

The hairs of the skin not only function to prevent heat loss but also have important sensory functions. Recent work has now established that each hair of the skin is innervated by one or more of three types of mechanoreceptor ending. Each of these three mechanoreceptor types possesses distinct molecular features and detects distinctive information about skin touch, which is relayed to specific brain locations in a somatotopic fashion.


Assuntos
Cabelo/fisiologia , Mecanorreceptores/fisiologia , Mecanotransdução Celular , Pele/inervação , Tato , Vias Aferentes/fisiologia , Animais , Potenciais Evocados , Folículo Piloso/inervação , Humanos , Neurônios Aferentes/fisiologia
7.
Handb Exp Pharmacol ; 220: 251-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24668476

RESUMO

Nerve growth factor (NGF) is central to the development and functional regulation of sensory neurons that signal the first events that lead to pain. These sensory neurons, called nociceptors, require NGF in the early embryo to survive and also for their functional maturation. The long road from the discovery of NGF and its roles during development to the realization that NGF plays a major role in the pathophysiology of inflammatory pain will be reviewed. In particular, we will discuss the various signaling events initiated by NGF that lead to long-lasting thermal and mechanical hyperalgesia in animals and in man. It has been realized relatively recently that humanized function blocking antibodies directed against NGF show remarkably analgesic potency in human clinical trials for painful conditions as varied as osteoarthritis, lower back pain, and interstitial cystitis. Thus, anti-NGF medication has the potential to make a major impact on day-to-day chronic pain treatment in the near future. It is therefore all the more important to understand the precise pathways and mechanisms that are controlled by NGF to both initiate and sustain mechanical and thermal hyperalgesia. Recent work suggests that NGF-dependent regulation of the mechanosensory properties of sensory neurons that signal mechanical pain may open new mechanistic avenues to refine and exploit relevant molecular targets for novel analgesics.


Assuntos
Hiperalgesia/etiologia , Fator de Crescimento Neural/fisiologia , Nociceptividade/fisiologia , Dor/tratamento farmacológico , Animais , Desenvolvimento Embrionário , Humanos , Fator de Crescimento Neural/antagonistas & inibidores
8.
Channels (Austin) ; 18(1): 2355123, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38754025

RESUMO

PIEZO1 and PIEZO2 are mechanically activated ion channels that confer mechanosensitivity to various cell types. PIEZO channels are commonly examined using the so-called poking technique, where currents are recorded in the whole-cell configuration of the patch-clamp technique, while the cell surface is mechanically stimulated with a small fire-polished patch pipette. Currently, there is no gold standard for mechanical stimulation, and therefore, stimulation protocols differ significantly between laboratories with regard to stimulation velocity, angle, and size of the stimulation probe. Here, we systematically examined the impact of variations in these three stimulation parameters on the outcomes of patch-clamp recordings of PIEZO1 and PIEZO2. We show that the inactivation kinetics of PIEZO1 and, to a lesser extent, of PIEZO2 change with the angle at which the probe that is used for mechanical stimulation is positioned and, even more prominently, with the size of its tip. Moreover, we found that the mechanical activation threshold of PIEZO2, but not PIEZO1, decreased with increasing stimulation speeds. Thus, our data show that two key outcome parameters of PIEZO-related patch-clamp studies are significantly affected by common variations in the mechanical stimulation protocols, which calls for caution when comparing data from different laboratories and highlights the need to establish a gold standard for mechanical stimulation to improve comparability and reproducibility of data obtained with the poking technique.


Assuntos
Canais Iônicos , Técnicas de Patch-Clamp , Canais Iônicos/metabolismo , Humanos , Cinética , Células HEK293 , Mecanotransdução Celular
9.
Pain ; 165(6): 1336-1347, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38739766

RESUMO

ABSTRACT: Evidence from previous studies supports the concept that spinal cord injury (SCI)-induced neuropathic pain (NP) has its neural roots in the peripheral nervous system. There is uncertainty about how and to which degree mechanoreceptors contribute. Sensorimotor activation-based interventions (eg, treadmill training) have been shown to reduce NP after experimental SCI, suggesting transmission of pain-alleviating signals through mechanoreceptors. The aim of the present study was to understand the contribution of mechanoreceptors with respect to mechanical allodynia in a moderate mouse contusion SCI model. After genetic ablation of tropomyosin receptor kinase B expressing mechanoreceptors before SCI, mechanical allodynia was reduced. The identical genetic ablation after SCI did not yield any change in pain behavior. Peptidergic nociceptor sprouting into lamina III/IV below injury level as a consequence of SCI was not altered by either mechanoreceptor ablation. However, skin-nerve preparations of contusion SCI mice 7 days after injury yielded hyperexcitability in nociceptors, not in mechanoreceptors, which makes a substantial direct contribution of mechanoreceptors to NP maintenance unlikely. Complementing animal data, quantitative sensory testing in human SCI subjects indicated reduced mechanical pain thresholds, whereas the mechanical detection threshold was not altered. Taken together, early mechanoreceptor ablation modulates pain behavior, most likely through indirect mechanisms. Hyperexcitable nociceptors seem to be the main drivers of SCI-induced NP. Future studies need to focus on injury-derived factors triggering early-onset nociceptor hyperexcitability, which could serve as targets for more effective therapeutic interventions.


Assuntos
Modelos Animais de Doenças , Hiperalgesia , Mecanorreceptores , Camundongos Endogâmicos C57BL , Traumatismos da Medula Espinal , Animais , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Camundongos , Hiperalgesia/fisiopatologia , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Mecanorreceptores/metabolismo , Mecanorreceptores/fisiologia , Masculino , Humanos , Limiar da Dor/fisiologia , Feminino , Medição da Dor , Camundongos Transgênicos , Neuralgia/etiologia , Neuralgia/metabolismo , Neuralgia/fisiopatologia
10.
J Clin Invest ; 134(7)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38557489

RESUMO

Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.


Assuntos
Canais de Cálcio , Cálcio , Camundongos , Animais , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Pâncreas/metabolismo , Exocitose/fisiologia , Vesículas Secretórias/genética
11.
EMBO J ; 28(10): 1479-91, 2009 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-19322198

RESUMO

Somatic sensation relies on the transduction of physical stimuli into electrical signals by sensory neurons of the dorsal root ganglia. Little is known about how and when during development different types of sensory neurons acquire transduction competence. We directly investigated the emergence of electrical excitability and mechanosensitivity of embryonic and postnatal mouse sensory neurons. We show that sensory neurons acquire mechanotransduction competence coincident with peripheral target innervation. Mechanotransduction competence arises in different sensory lineages in waves, coordinated by distinct developmental mechanisms. Sensory neurons that are mechanoreceptors or proprioceptors acquire mature mechanotransduction indistinguishable from the adult already at E13. This process is independent of neurotrophin-3 and may be driven by a genetic program. In contrast, most nociceptive (pain sensing) sensory neurons acquire mechanosensitive competence as a result of exposure to target-derived nerve growth factor. The highly regulated process of mechanosensory acquisition unveiled here, reveals new strategies to identify molecules required for sensory neuron mechanotransduction.


Assuntos
Desenvolvimento Embrionário , Mecanorreceptores/fisiologia , Mecanotransdução Celular , Sistema Nervoso/crescimento & desenvolvimento , Células Receptoras Sensoriais , Animais , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Neural/fisiologia , Nociceptores/fisiologia , Propriocepção
12.
EMBO Rep ; 12(4): 292-5, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21415857

RESUMO

The 102nd biannual Boehringer Ingelheim Fonds International Titisee Conference took place in October 2010. In the welcoming atmosphere of the small lakeside resort in the Black Forest, southern Germany, scientists from around the world gathered to discuss current topics and challenges in the area of sensory biology. The research presented covered all of the classical Aristotelian senses (and beyond) and provided a glimpse at recent progress and recurring themes in the sensory systems.


Assuntos
Sensação/fisiologia , Animais , Congressos como Assunto , Audição/genética , Audição/fisiologia , Humanos , Mecanotransdução Celular/genética , Mecanotransdução Celular/fisiologia , Modelos Biológicos , Sensação/genética , Olfato/genética , Olfato/fisiologia , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
13.
Nat Commun ; 14(1): 1899, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37019973

RESUMO

Mechanically silent nociceptors are sensory afferents that are insensitive to noxious mechanical stimuli under normal conditions but become sensitized to such stimuli during inflammation. Using RNA-sequencing and quantitative RT-PCR we demonstrate that inflammation upregulates the expression of the transmembrane protein TMEM100 in silent nociceptors and electrophysiology revealed that over-expression of TMEM100 is required and sufficient to un-silence silent nociceptors in mice. Moreover, we show that mice lacking TMEM100 do not develop secondary mechanical hypersensitivity-i.e., pain hypersensitivity that spreads beyond the site of inflammation-during knee joint inflammation and that AAV-mediated overexpression of TMEM100 in articular afferents in the absence of inflammation is sufficient to induce mechanical hypersensitivity in remote skin regions without causing knee joint pain. Thus, our work identifies TMEM100 as a key regulator of silent nociceptor un-silencing and reveals a physiological role for this hitherto enigmatic afferent subclass in triggering spatially remote secondary mechanical hypersensitivity during inflammation.


Assuntos
Nociceptores , Dor , Animais , Camundongos , Inflamação/metabolismo , Articulação do Joelho , Nociceptores/metabolismo , Dor/metabolismo , Pele/metabolismo
14.
Nat Commun ; 13(1): 1365, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35292651

RESUMO

A central question in mechanobiology is how mechanical forces acting in or on cells are transmitted to mechanically-gated PIEZO channels that convert these forces into biochemical signals. Here we examined the role of the intracellular domains of PIEZO2, which account for 25% of the channel, and demonstrate that these domains fine-tune properties such as poking and stretch-sensitivity, velocity coding and single channel conductance. Moreover, we show that the intrinsically disordered linker between the transmembrane helices twelve and thirteen (IDR5) is required for the activation of PIEZO2 by cytoskeleton-transmitted forces. The deletion of IDR5 abolishes PIEZO2-mediated inhibition of neurite outgrowth, while it only partially affected its sensitivity to cell indentation and does not alter its stretch sensitivity. Thus, we propose that PIEZO2 is a polymodal mechanosensor that detects different types of mechanical stimuli via different force transmission pathways, which highlights the importance of utilizing multiple complementary assays when investigating PIEZO function.


Assuntos
Canais Iônicos , Mecanotransdução Celular , Citoesqueleto/metabolismo , Canais Iônicos/metabolismo , Mecanotransdução Celular/fisiologia
15.
Cell Rep ; 35(9): 109191, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34077727

RESUMO

The vasculature is innervated by a network of peripheral afferents that sense and regulate blood flow. Here, we describe a system of non-peptidergic sensory neurons with cell bodies in the spinal ganglia that regulate vascular tone in the distal arteries. We identify a population of mechanosensitive neurons, marked by tropomyosin receptor kinase C (TrkC) and tyrosine hydroxylase in the dorsal root ganglia, which projects to blood vessels. Local stimulation of TrkC neurons decreases vessel diameter and blood flow, whereas systemic activation increases systolic blood pressure and heart rate variability via the sympathetic nervous system. Ablation of the neurons provokes variability in local blood flow, leading to a reduction in systolic blood pressure, increased heart rate variability, and ultimately lethality within 48 h. Thus, a population of TrkC+ sensory neurons forms part of a sensory-feedback mechanism that maintains cardiovascular homeostasis through the autonomic nervous system.


Assuntos
Pressão Sanguínea/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Comportamento Animal , Fluoresceína/metabolismo , Gânglios Espinais/fisiologia , Frequência Cardíaca/fisiologia , Camundongos Transgênicos , Receptor trkC/metabolismo
16.
Nat Neurosci ; 24(1): 74-81, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33288907

RESUMO

Fingertip mechanoreceptors comprise sensory neuron endings together with specialized skin cells that form the end-organ. Exquisitely sensitive, vibration-sensing neurons are associated with Meissner's corpuscles in the skin. In the present study, we found that USH2A, a transmembrane protein with a very large extracellular domain, was found in terminal Schwann cells within Meissner's corpuscles. Pathogenic USH2A mutations cause Usher's syndrome, associated with hearing loss and visual impairment. We show that patients with biallelic pathogenic USH2A mutations also have clear and specific impairments in vibrotactile touch perception, as do mutant mice lacking USH2A. Forepaw rapidly adapting mechanoreceptors innervating Meissner's corpuscles, recorded from Ush2a-/- mice, showed large reductions in vibration sensitivity. However, the USH2A protein was not found in sensory neurons. Thus, loss of USH2A in corpuscular end-organs reduced mechanoreceptor sensitivity as well as vibration perception. Thus, a tether-like protein is required to facilitate detection of small-amplitude vibrations essential for the perception of fine-grained tactile surfaces.


Assuntos
Proteínas da Matriz Extracelular/genética , Mecanorreceptores/metabolismo , Sensação/fisiologia , Vibração , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Knockout , Mutação/genética , Células de Schwann/fisiologia , Pele/inervação , Tato/fisiologia , Síndromes de Usher/genética
17.
Front Cell Neurosci ; 14: 13, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32116559

RESUMO

Pronounced activity-dependent slowing of conduction has been used to characterize mechano-insensitive, "silent" nociceptors and might be due to high expression of NaV1.8 and could, therefore, be characterized by their tetrodotoxin-resistance (TTX-r). Nociceptor-class specific differences in action potential characteristics were studied by: (i) in vitro calcium imaging in single porcine nerve growth factor (NGF)-responsive neurites; (ii) in vivo extracellular recordings in functionally identified porcine silent nociceptors; and (iii) in vitro patch-clamp recordings from murine silent nociceptors, genetically defined by nicotinic acetylcholine receptor subunit alpha-3 (CHRNA3) expression. Porcine TTX-r neurites (n = 26) in vitro had more than twice as high calcium transients per action potential as compared to TTX-s neurites (n = 18). In pig skin, silent nociceptors (n = 14) characterized by pronounced activity-dependent slowing of conduction were found to be TTX-r, whereas polymodal nociceptors were TTX-s (n = 12) and had only moderate slowing. Mechano-insensitive cold nociceptors were also TTX-r but showed less activity-dependent slowing than polymodal nociceptors. Action potentials in murine silent nociceptors differed from putative polymodal nociceptors by longer duration and higher peak amplitudes. Longer duration AP in silent murine nociceptors linked to increased sodium load would be compatible with a pronounced activity-dependent slowing in pig silent nociceptors and longer AP durations could be in line with increased calcium transients per action potential observed in vitro in TTX-resistant NGF responsive porcine neurites. Even though there is no direct link between slowing and TTX-resistant channels, the results indicate that axons of silent nociceptors not only differ in their receptive but also in their axonal properties.

18.
Neuron ; 107(6): 1141-1159.e7, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32735781

RESUMO

Diabetic peripheral neuropathy (DPN) is a highly frequent and debilitating clinical complication of diabetes that lacks therapies. Cellular oxidative stress regulates post-translational modifications, including SUMOylation. Here, using unbiased screens, we identified key enzymes in metabolic pathways and ion channels as novel molecular targets of SUMOylation that critically regulated their activity. Sensory neurons of diabetic patients and diabetic mice demonstrated changes in the SUMOylation status of metabolic enzymes and ion channels. In support of this, profound metabolic dysfunction, accelerated neuropathology, and sensory loss were observed in diabetic gene-targeted mice selectively lacking the ability to SUMOylate proteins in peripheral sensory neurons. TRPV1 function was impaired by diabetes-induced de-SUMOylation as well as by metabolic imbalance elicited by de-SUMOylation of metabolic enzymes, facilitating diabetic sensory loss. Our results unexpectedly uncover an endogenous post-translational mechanism regulating diabetic neuropathy in patients and mouse models that protects against metabolic dysfunction, nerve damage, and altered sensory perception.


Assuntos
Neuropatias Diabéticas/metabolismo , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Nociceptividade , Células Receptoras Sensoriais/metabolismo , Sumoilação , Canais de Cátion TRPV/metabolismo , Animais , Células Cultivadas , Ciclo do Ácido Cítrico , Neuropatias Diabéticas/fisiopatologia , Feminino , Gânglios Espinais/citologia , Glicólise , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
J Physiol ; 587(Pt 14): 3493-503, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19505980

RESUMO

Mechanical stimuli impinging on the skin are converted into electrical signals by mechanically gated ion channels located at the peripheral nerve endings of dorsal root ganglion (DRG) neurons. Under inflammatory conditions sensory neurons are commonly sensitised to mechanical stimuli; a putative mechanism that may contribute to such sensitisation of sensory neurons is enhanced responsiveness of mechanotransduction ion channels. Here we show that the algogens UTP and ATP potentiate mechanosensitive RA currents in peptidergic nociceptive DRG neurons and reduce thresholds for mechanically induced action potential firing in these neurones. Pharmacological characterisation suggests that this effect is mediated by the Gq-coupled P2Y(2) nucleotide receptor. Moreover, using the in vitro skin nerve technique, we show that UTP also increases action potential firing rates in response to mechanical stimuli in a subpopulation of skin C-fibre nociceptors. Together our findings suggest that UTP sensitises a subpopulation of cutaneous C-fibre nociceptors via a previously undescribed G-protein-dependent potentiation of mechanically activated RA-type currents.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Potenciação de Longa Duração/fisiologia , Mecanotransdução Celular/fisiologia , Potenciais da Membrana/fisiologia , Nociceptores/fisiologia , Células Receptoras Sensoriais/fisiologia , Pele/inervação , Trifosfato de Adenosina/administração & dosagem , Animais , Células Cultivadas , Potenciação de Longa Duração/efeitos dos fármacos , Mecanotransdução Celular/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Nociceptores/efeitos dos fármacos , Estimulação Física/métodos , Células Receptoras Sensoriais/efeitos dos fármacos , Pele/efeitos dos fármacos , Fenômenos Fisiológicos da Pele , Uridina Trifosfato/administração & dosagem
20.
Elife ; 72018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29521261

RESUMO

Piezo2 ion channels are critical determinants of the sense of light touch in vertebrates. Yet, their regulation is only incompletely understood. We recently identified myotubularin related protein-2 (Mtmr2), a phosphoinositide (PI) phosphatase, in the native Piezo2 interactome of murine dorsal root ganglia (DRG). Here, we demonstrate that Mtmr2 attenuates Piezo2-mediated rapidly adapting mechanically activated (RA-MA) currents. Interestingly, heterologous Piezo1 and other known MA current subtypes in DRG appeared largely unaffected by Mtmr2. Experiments with catalytically inactive Mtmr2, pharmacological blockers of PI(3,5)P2 synthesis, and osmotic stress suggest that Mtmr2-dependent Piezo2 inhibition involves depletion of PI(3,5)P2. Further, we identified a PI(3,5)P2 binding region in Piezo2, but not Piezo1, that confers sensitivity to Mtmr2 as indicated by functional analysis of a domain-swapped Piezo2 mutant. Altogether, our results propose local PI(3,5)P2 modulation via Mtmr2 in the vicinity of Piezo2 as a novel mechanism to dynamically control Piezo2-dependent mechanotransduction in peripheral sensory neurons.


Assuntos
Canais Iônicos/genética , Mecanotransdução Celular/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Células Receptoras Sensoriais/metabolismo , Animais , Membrana Celular/genética , Membrana Celular/metabolismo , Gânglios Espinais/crescimento & desenvolvimento , Gânglios Espinais/fisiologia , Humanos , Canais Iônicos/química , Camundongos , Pressão Osmótica/fisiologia , Nervos Periféricos/metabolismo , Nervos Periféricos/fisiologia , Fosfoinositídeo Fosfolipase C/genética , Fosfolipídeos/química , Fosfolipídeos/genética , Proteínas Tirosina Fosfatases não Receptoras/antagonistas & inibidores , Células Receptoras Sensoriais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA