Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pflugers Arch ; 472(7): 865-880, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32601768

RESUMO

Chronic pain is a global problem affecting up to 20% of the world's population and has a significant economic, social and personal cost to society. Sensory neurons of the dorsal root ganglia (DRG) detect noxious stimuli and transmit this sensory information to regions of the central nervous system (CNS) where activity is perceived as pain. DRG neurons express multiple voltage-gated sodium channels that underlie their excitability. Research over the last 20 years has provided valuable insights into the critical roles that two channels, NaV1.7 and NaV1.9, play in pain signalling in man. Gain of function mutations in NaV1.7 cause painful conditions while loss of function mutations cause complete insensitivity to pain. Only gain of function mutations have been reported for NaV1.9. However, while most NaV1.9 mutations lead to painful conditions, a few are reported to cause insensitivity to pain. The critical roles these channels play in pain along with their low expression in the CNS and heart muscle suggest they are valid targets for novel analgesic drugs.


Assuntos
Dor Crônica/genética , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.9/genética , Animais , Sistema Nervoso Central/patologia , Dor Crônica/patologia , Gânglios Espinais/patologia , Humanos
2.
Pain ; 161(5): 1100-1108, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31929383

RESUMO

Alleviating chronic pain is challenging, due to lack of drugs that effectively inhibit nociceptors without off-target effects on motor or central neurons. Dorsal root ganglia (DRG) contain nociceptive and non-nociceptive neurons. Drug screening on cultured DRG neurons, rather than cell lines, allows for the identification of drugs most potent on nociceptors with no effects on non-nociceptors (as a proxy for unwanted side effects on central nervous system and motor neurons). However, screening using DRG neurons is currently a low-throughput process, and there is a need for assays to speed this process for analgesic drug discovery. We previously showed that veratridine elicits distinct response profiles in sensory neurons. Here, we show evidence that a veratridine-based calcium assay allows for an unbiased and efficient assessment of a drug effect on nociceptors (targeted neurons) and non-nociceptors (nontargeted neurons). We confirmed the link between the oscillatory profile and nociceptors, and the slow-decay profile and non-nociceptors using 3 transgenic mouse lines of known pain phenotypes. We used the assay to show that blockers for Nav1.7 and Nav1.8 channels, which are validated targets for analgesics, affect non-nociceptors at concentrations needed to effectively inhibit nociceptors. However, a combination of low doses of both blockers had an additive effect on nociceptors without a significant effect on non-nociceptors, indicating that the assay can also be used to screen for combinations of existing or novel drugs for the greatest selective inhibition of nociceptors.


Assuntos
Células Receptoras Sensoriais , Analgésicos/farmacologia , Animais , Gânglios Espinais , Camundongos , Camundongos Endogâmicos C57BL , Nociceptores
3.
Pain ; 159(8): 1641-1651, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29697531

RESUMO

Voltage-gated potassium (Kv) channels are increasingly recognised as key regulators of nociceptive excitability. Kcns1 is one of the first potassium channels to be associated with neuronal hyperexcitability and mechanical sensitivity in the rat, as well as pain intensity and risk of developing chronic pain in humans. Here, we show that in mice, Kcns1 is predominantly expressed in the cell body and axons of myelinated sensory neurons positive for neurofilament-200, including Aδ-fiber nociceptors and low-threshold Aß mechanoreceptors. In the spinal cord, Kcns1 was detected in laminae III to V of the dorsal horn where most sensory A fibers terminate, as well as large motoneurons of the ventral horn. To investigate Kcns1 function specifically in the periphery, we generated transgenic mice in which the gene is deleted in all sensory neurons but retained in the central nervous system. Kcns1 ablation resulted in a modest increase in basal mechanical pain, with no change in thermal pain processing. After neuropathic injury, Kcns1 KO mice exhibited exaggerated mechanical pain responses and hypersensitivity to both noxious and innocuous cold, consistent with increased A-fiber activity. Interestingly, Kcns1 deletion also improved locomotor performance in the rotarod test, indicative of augmented proprioceptive signalling. Our results suggest that restoring Kcns1 function in the periphery may be of some use in ameliorating mechanical and cold pain in chronic states.


Assuntos
Neuralgia/metabolismo , Limiar da Dor/fisiologia , Células do Corno Posterior/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Camundongos , Camundongos Knockout , Destreza Motora/fisiologia , Neuralgia/genética , Estimulação Física , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Propriocepção/fisiologia
4.
Pain ; 159(3): 496-506, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29194125

RESUMO

The sodium channel NaV1.7 contributes to action potential (AP) generation and propagation. Loss-of-function mutations in patients lead to congenital indifference to pain, though it remains unclear where on the way from sensory terminals to central nervous system the signalling is disrupted. We confirm that conditional deletion of NaV1.7 in advillin-expressing sensory neurons leads to impaired heat and mechanical nociception in behavioural tests. With single-fiber recordings from isolated skin, we found (1) a significantly lower prevalence of heat responsiveness to normally mechanosensitive C-fibers, although (2) the rare heat responses seemed quite vigorous, and (3) heat-induced calcitonin gene-related peptide release was normal. In biophysical respects, although electrical excitability, rheobase, and chronaxy were normal, (4) axonal conduction velocity was 20% slower than in congenic wild-type mice (5) and when challenged with double pulses (<100 milliseconds interval), the second AP showed more pronounced latency increase (6). On prolonged electrical stimulation at 2 Hz, (7) activity-dependent slowing of nerve fiber conduction was markedly less, and (8) was less likely to result in conduction failure of the mutant single fibers. Finally, recording of compound APs from the whole saphenous nerve confirmed slower conduction and less activity-dependent slowing as well as the functional absence of a large subpopulation of C-fibers (9) in conditional NaV1.7 knockouts. In conclusion, the clear deficits in somatic primary afferent functions shown in our study may be complemented by previously reported synaptic dysfunction and opioidergic inhibition, together accounting for the complete insensitivity to pain in the human mutants lacking NaV1.7.


Assuntos
Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Dor/genética , Potenciais de Ação/genética , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibras Nervosas Amielínicas/fisiologia , Dor/fisiopatologia , Medição da Dor/métodos , Limiar da Dor/fisiologia , Estimulação Física/efeitos adversos , Células Receptoras Sensoriais/fisiologia
5.
Sci Rep ; 7: 45221, 2017 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-28338073

RESUMO

Nociceptors are a subpopulation of dorsal root ganglia (DRG) neurons that detect noxious stimuli and signal pain. Veratridine (VTD) is a voltage-gated sodium channel (VGSC) modifier that is used as an "agonist" in functional screens for VGSC blockers. However, there is very little information on VTD response profiles in DRG neurons and how they relate to neuronal subtypes. Here we characterised VTD-induced calcium responses in cultured mouse DRG neurons. Our data shows that the heterogeneity of VTD responses reflects distinct subpopulations of sensory neurons. About 70% of DRG neurons respond to 30-100 µM VTD. We classified VTD responses into four profiles based upon their response shape. VTD response profiles differed in their frequency of occurrence and correlated with neuronal size. Furthermore, VTD response profiles correlated with responses to the algesic markers capsaicin, AITC and α, ß-methylene ATP. Since VTD response profiles integrate the action of several classes of ion channels and exchangers, they could act as functional "reporters" for the constellation of ion channels/exchangers expressed in each sensory neuron. Therefore our findings are relevant to studies and screens using VTD to activate DRG neurons.


Assuntos
Sinalização do Cálcio , Gânglios Espinais/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Veratridina/farmacologia , Animais , Capsaicina/farmacologia , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Masculino , Moduladores de Transporte de Membrana/farmacologia , Camundongos , Nociceptividade , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/fisiologia
6.
Pain ; 158(1): 58-67, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27780178

RESUMO

The upregulation of the tetrodotoxin-resistant voltage-gated sodium channel NaV1.9 has previously been associated with inflammatory hyperalgesia. Na1.9 knockout (KO) mice, however, did not seem insensitive in conventional tests of acute nociception. Using electrophysiological, neurochemical, and behavioral techniques, we now show NaV1.9-null mice exhibit impaired mechanical and thermal sensory capacities and reduced electrical excitability of nociceptors. In single-fiber recordings from isolated skin, the electrical threshold of NaV1.9 KO C fibers was elevated by 55% and the median von Frey threshold was 32 mN in contrast to 8 mN in wild types (WTs). The prevalence of C mechano-heat-sensitive (CMH) fibers was only 25.6% in NaV1.9 KO animals compared to 75.8% in the WT group, and the heat threshold of these CMH fibers was 40.4°C in the control vs 44°C in the KO group. Compound action potential recordings from isolated sciatic nerve segments of NaV1.9 KO mice revealed lower activity-induced slowing of conduction velocity upon noxious heat stimulation: 8% vs 30% in WTs. Heat-induced calcitonin gene-related peptide release from the skin was less in the KO than in the WT group. The reduced noxious heat sensitivity was finally confirmed with the Hargreaves test using 2 rates of radiant heating of the plantar hind paws. In conclusion, NaV1.9 presumably contributes to acute thermal and mechanical nociception in mice, most likely through increasing the excitability but probably also by amplifying receptor potentials irrespective of the stimulus modality.


Assuntos
Hiperalgesia , Canal de Sódio Disparado por Voltagem NAV1.9/deficiência , Fibras Nervosas Amielínicas/fisiologia , Nociceptores/fisiologia , Potenciais de Ação/genética , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Feminino , Temperatura Alta/efeitos adversos , Hiperalgesia/genética , Hiperalgesia/patologia , Hiperalgesia/fisiopatologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.9/genética , Condução Nervosa/genética , Limiar da Dor/fisiologia , Estimulação Física/efeitos adversos , Pele/inervação
7.
Toxins (Basel) ; 8(3)2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26999206

RESUMO

Loss-of-function mutations of Na(V)1.7 lead to congenital insensitivity to pain, a rare condition resulting in individuals who are otherwise normal except for the inability to sense pain, making pharmacological inhibition of Na(V)1.7 a promising therapeutic strategy for the treatment of pain. We characterized a novel mouse model of Na(V)1.7-mediated pain based on intraplantar injection of the scorpion toxin OD1, which is suitable for rapid in vivo profiling of Na(V)1.7 inhibitors. Intraplantar injection of OD1 caused spontaneous pain behaviors, which were reversed by co-injection with Na(V)1.7 inhibitors and significantly reduced in Na(V)1.7(-/-) mice. To validate the use of the model for profiling Na(V)1.7 inhibitors, we determined the Na(V) selectivity and tested the efficacy of the reported Na(V)1.7 inhibitors GpTx-1, PF-04856264 and CNV1014802 (raxatrigine). GpTx-1 selectively inhibited Na(V)1.7 and was effective when co-administered with OD1, but lacked efficacy when delivered systemically. PF-04856264 state-dependently and selectively inhibited Na(V)1.7 and significantly reduced OD1-induced spontaneous pain when delivered locally and systemically. CNV1014802 state-dependently, but non-selectively, inhibited Na(V) channels and was only effective in the OD1 model when delivered systemically. Our novel model of Na(V)1.7-mediated pain based on intraplantar injection of OD1 is thus suitable for the rapid in vivo characterization of the analgesic efficacy of Na(V)1.7 inhibitors.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/fisiologia , Dor/tratamento farmacológico , Peptídeos/uso terapêutico , Éteres Fenílicos/uso terapêutico , Prolina/análogos & derivados , Venenos de Escorpião/uso terapêutico , Bloqueadores dos Canais de Sódio/uso terapêutico , Venenos de Aranha/uso terapêutico , Analgésicos , Animais , Comportamento Animal/efeitos dos fármacos , Células CHO , Cricetulus , Modelos Animais de Doenças , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/fisiologia , Dor/induzido quimicamente , Prolina/uso terapêutico , Veia Safena/inervação , Sulfonamidas/uso terapêutico
8.
Acta Neuropathol Commun ; 3: 74, 2015 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-26610346

RESUMO

BACKGROUND: Tamoxifen (TAM) is an important cancer therapeutic and an experimental tool for effecting genetic recombination using the inducible Cre-Lox technique. Despite its widespread use in the clinic and laboratory, we know little about its effects on the nervous system. This is of significant concern because TAM, via unknown mechanisms, induces cognitive impairment in humans. A hallmark of cellular stress is induction of Activating Transcription Factor 3 (Atf3), and so to determine whether TAM induces cellular stress in the adult nervous system, we generated a knock-in mouse in which Atf3 promoter activity drives transcription of TAM-dependent Cre recombinase (Cre-ERT2); when crossed with tdtomato reporter mice, Atf3 induction results in robust and permanent genetic labeling of cells in which it is up-regulated even transiently. RESULTS: We found that granular neurons of the olfactory bulb and dentate gyrus, vascular cells and ependymal cells throughout the brain, and peripheral sensory neurons expressed tdtomato in response to TAM treatment. We also show that TAM induced Atf3 up-regulation through inhibition of cholesterol epoxide hydrolase (ChEH): reporter expression was mitigated by delivery in vitamin E-rich wheat germ oil (vitamin E depletes ChEH substrates), and was partially mimicked by a ChEH-specific inhibitor. CONCLUSIONS: This work demonstrates that TAM stresses cells of the adult central and peripheral nervous systems and highlights concerns about clinical and experimental use of TAM. We propose TAM administration in vitamin E-rich vehicles such as wheat germ oil as a simple remedy.


Assuntos
Colesterol/metabolismo , Sistema Nervoso/citologia , Neurônios/fisiologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Regulação para Cima/efeitos dos fármacos , Fator 3 Ativador da Transcrição/genética , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Relação Dose-Resposta a Droga , Epóxido Hidrolases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Lectinas de Plantas/genética , Lectinas de Plantas/metabolismo , Óleos de Plantas/farmacologia , Regiões Promotoras Genéticas , Vitamina E/farmacologia
9.
PLoS One ; 10(6): e0128670, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26053673

RESUMO

In vitro cell lines from DRG neurons aid drug discovery because they can be used for early stage, high-throughput screens for drugs targeting pain pathways, with minimal dependence on animals. We have established a conditionally immortal DRG cell line from the Immortomouse. Using immunocytochemistry, RT-PCR and calcium microfluorimetry, we demonstrate that the cell line MED17.11 expresses markers of cells committed to the sensory neuron lineage. Within a few hours under differentiating conditions, MED17.11 cells extend processes and following seven days of differentiation, express markers of more mature DRG neurons, such as NaV1.7 and Piezo2. However, at least at this time-point, the nociceptive marker NaV1.8 is not expressed, but the cells respond to compounds known to excite nociceptors, including the TRPV1 agonist capsaicin, the purinergic receptor agonist ATP and the voltage gated sodium channel agonist, veratridine. Robust calcium transients are observed in the presence of the inflammatory mediators bradykinin, histamine and norepinephrine. MED17.11 cells have the potential to replace or reduce the use of primary DRG culture in sensory, pain and developmental research by providing a simple model to study acute nociception, neurite outgrowth and the developmental specification of DRG neurons.


Assuntos
Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Nociceptores/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Clonais , Gânglios Espinais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Canais de Sódio/metabolismo , Transfecção , Veratridina/farmacologia
10.
J Physiol ; 592(21): 4677-96, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25172946

RESUMO

Mouse pancreatic ß- and α-cells are equipped with voltage-gated Na(+) currents that inactivate over widely different membrane potentials (half-maximal inactivation (V0.5) at -100 mV and -50 mV in ß- and α-cells, respectively). Single-cell PCR analyses show that both α- and ß-cells have Nav1.3 (Scn3) and Nav1.7 (Scn9a) α subunits, but their relative proportions differ: ß-cells principally express Nav1.7 and α-cells Nav1.3. In α-cells, genetically ablating Scn3a reduces the Na(+) current by 80%. In ß-cells, knockout of Scn9a lowers the Na(+) current by >85%, unveiling a small Scn3a-dependent component. Glucagon and insulin secretion are inhibited in Scn3a(-/-) islets but unaffected in Scn9a-deficient islets. Thus, Nav1.3 is the functionally important Na(+) channel α subunit in both α- and ß-cells because Nav1.7 is largely inactive at physiological membrane potentials due to its unusually negative voltage dependence of inactivation. Interestingly, the Nav1.7 sequence in brain and islets is identical and yet the V0.5 for inactivation is >30 mV more negative in ß-cells. This may indicate the presence of an intracellular factor that modulates the voltage dependence of inactivation.


Assuntos
Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.3/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Sódio/fisiologia , Animais , Regulação da Expressão Gênica , Células Secretoras de Glucagon/efeitos dos fármacos , Glucose , Células HEK293 , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.3/genética , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Neurotoxinas/farmacologia , Isoformas de Proteínas , Subunidades Proteicas
11.
Pain ; 155(10): 1962-75, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24972070

RESUMO

Chronic visceral pain affects millions of individuals worldwide and remains poorly understood, with current therapeutic options constrained by gastrointestinal adverse effects. Visceral pain is strongly associated with inflammation and distension of the gut. Here we report that the voltage-gated sodium channel subtype NaV1.9 is expressed in half of gut-projecting rodent dorsal root ganglia sensory neurons. We show that NaV1.9 is required for normal mechanosensation, for direct excitation and for sensitization of mouse colonic afferents by mediators from inflammatory bowel disease tissues, and by noxious inflammatory mediators individually. Excitatory responses to ATP or PGE2 were substantially reduced in NaV1.9(-/-) mice. Deletion of NaV1.9 substantially attenuates excitation and subsequent mechanical hypersensitivity after application of inflammatory soup (IS) (bradykinin, ATP, histamine, PGE2, and 5HT) to visceral nociceptors located in the serosa and mesentery. Responses to mechanical stimulation of mesenteric afferents were also reduced by loss of NaV1.9, and there was a rightward shift in stimulus-response function to ramp colonic distension. By contrast, responses to rapid, high-intensity phasic distension of the colon are initially unaffected; however, run-down of responses to repeat phasic distension were exacerbated in NaV1.9(-/-) afferents. Finally colonic afferent activation by supernatants derived from inflamed human tissue was greatly reduced in NaV1.9(-/-) mice. These results demonstrate that NaV1.9 is required for persistence of responses to intense mechanical stimulation, contributes to inflammatory mechanical hypersensitivity, and is essential for activation by noxious inflammatory mediators, including those from diseased human bowel. These observations indicate that NaV1.9 represents a high-value target for development of visceral analgesics.


Assuntos
Colo/inervação , Hiperalgesia/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.9/metabolismo , Fibras Aferentes Viscerais/metabolismo , Potenciais de Ação/efeitos dos fármacos , Trifosfato de Adenosina/farmacologia , Adolescente , Adulto , Idoso , Animais , Colo/metabolismo , Colo/fisiopatologia , Dinoprostona/farmacologia , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Humanos , Hiperalgesia/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.9/genética , Estimulação Física , Fibras Aferentes Viscerais/efeitos dos fármacos , Fibras Aferentes Viscerais/fisiopatologia , Adulto Jovem
12.
Cell Rep ; 6(2): 301-12, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24440715

RESUMO

Nav1.7, a peripheral neuron voltage-gated sodium channel, is essential for pain and olfaction in mice and humans. We examined the role of Nav1.7 as well as Nav1.3, Nav1.8, and Nav1.9 in different mouse models of chronic pain. Constriction-injury-dependent neuropathic pain is abolished when Nav1.7 is deleted in sensory neurons, unlike nerve-transection-related pain, which requires the deletion of Nav1.7 in sensory and sympathetic neurons for pain relief. Sympathetic sprouting that develops in parallel with nerve-transection pain depends on the presence of Nav1.7 in sympathetic neurons. Mechanical and cold allodynia required distinct sets of neurons and different repertoires of sodium channels depending on the nerve injury model. Surprisingly, pain induced by the chemotherapeutic agent oxaliplatin and cancer-induced bone pain do not require the presence of Nav1.7 sodium channels or Nav1.8-positive nociceptors. Thus, similar pain phenotypes arise through distinct cellular and molecular mechanisms. Therefore, rational analgesic drug therapy requires patient stratification in terms of mechanisms and not just phenotype.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Neuralgia/metabolismo , Dor Nociceptiva/metabolismo , Nociceptores/metabolismo , Animais , Hiperalgesia/metabolismo , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Neuralgia/genética , Nociceptividade , Dor Nociceptiva/genética , Nociceptores/fisiologia
13.
Hum Mol Genet ; 21(16): 3655-67, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22641814

RESUMO

Spontaneous neural activity promotes axon growth in many types of developing neurons, including motoneurons. In motoneurons from a mouse model of spinal muscular atrophy (SMA), defects in axonal growth and presynaptic function correlate with a reduced frequency of spontaneous Ca(2+) transients in axons which are mediated by N-type Ca(2+) channels. To characterize the mechanisms that initiate spontaneous Ca(2+) transients, we investigated the role of voltage-gated sodium channels (VGSCs). We found that low concentrations of the VGSC inhibitors tetrodotoxin (TTX) and saxitoxin (STX) reduce the rate of axon growth in cultured embryonic mouse motoneurons without affecting their survival. STX was 5- to 10-fold more potent than TTX and Ca(2+) imaging confirmed that low concentrations of STX strongly reduce the frequency of spontaneous Ca(2+) transients in somatic and axonal regions. These findings suggest that the Na(V)1.9, a VGSC that opens at low thresholds, could act upstream of spontaneous Ca(2+) transients. qPCR from cultured and laser-microdissected spinal cord motoneurons revealed abundant expression of Na(V)1.9. Na(V)1.9 protein is preferentially localized in axons and growth cones. Suppression of Na(V)1.9 expression reduced axon elongation. Motoneurons from Na(V)1.9(-/-) mice showed the reduced axon growth in combination with reduced spontaneous Ca(2+) transients in the soma and axon terminals. Thus, Na(V)1.9 function appears to be essential for activity-dependent axon growth, acting upstream of spontaneous Ca(2+) elevation through voltage-gated calcium channels (VGCCs). Na(V)1.9 activation could therefore serve as a target for modulating axonal regeneration in motoneuron diseases such as SMA in which presynaptic activity of VGCCs is reduced.


Assuntos
Axônios/metabolismo , Cálcio/metabolismo , Neurônios Motores/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.9/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Cones de Crescimento/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/efeitos dos fármacos , Atrofia Muscular Espinal/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.9/genética , Coelhos , Saxitoxina/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Medula Espinal/citologia , Medula Espinal/metabolismo , Tetrodotoxina/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo
14.
Nat Commun ; 3: 791, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22531176

RESUMO

Human acute and inflammatory pain requires the expression of voltage-gated sodium channel Nav1.7 but its significance for neuropathic pain is unknown. Here we show that Nav1.7 expression in different sets of mouse sensory and sympathetic neurons underlies distinct types of pain sensation. Ablating Nav1.7 gene (SCN9A) expression in all sensory neurons using Advillin-Cre abolishes mechanical pain, inflammatory pain and reflex withdrawal responses to heat. In contrast, heat-evoked pain is retained when SCN9A is deleted only in Nav1.8-positive nociceptors. Surprisingly, responses to the hotplate test, as well as neuropathic pain, are unaffected when SCN9A is deleted in all sensory neurons. However, deleting SCN9A in both sensory and sympathetic neurons abolishes these pain sensations and recapitulates the pain-free phenotype seen in humans with SCN9A loss-of-function mutations. These observations demonstrate an important role for Nav1.7 in sympathetic neurons in neuropathic pain, and provide possible insights into the mechanisms that underlie gain-of-function Nav1.7-dependent pain conditions.


Assuntos
Neuralgia/metabolismo , Percepção da Dor , Células Receptoras Sensoriais/metabolismo , Canais de Sódio/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Humanos , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.7 , Neuralgia/genética , Neuralgia/psicologia , Canais de Sódio/genética , Sistema Nervoso Simpático/citologia
15.
Mol Pain ; 8: 21, 2012 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-22449024

RESUMO

BACKGROUND: Members of the degenerin/epithelial (DEG/ENaC) sodium channel family are mechanosensors in C elegans, and Nav1.7 and Nav1.8 voltage-gated sodium channel knockout mice have major deficits in mechanosensation. ß and γENaC sodium channel subunits are present with acid sensing ion channels (ASICs) in mammalian sensory neurons of the dorsal root ganglia (DRG). The extent to which epithelial or voltage-gated sodium channels are involved in transduction of mechanical stimuli is unclear. RESULTS: Here we show that deleting ß and γENaC sodium channels in sensory neurons does not result in mechanosensory behavioural deficits. We had shown previously that Nav1.7/Nav1.8 double knockout mice have major deficits in behavioural responses to noxious mechanical pressure. However, all classes of mechanically activated currents in DRG neurons are unaffected by deletion of the two sodium channels. In contrast, the ability of Nav1.7/Nav1.8 knockout DRG neurons to generate action potentials is compromised with 50% of the small diameter sensory neurons unable to respond to electrical stimulation in vitro. CONCLUSION: Behavioural deficits in Nav1.7/Nav1.8 knockout mice reflects a failure of action potential propagation in a mechanosensitive set of sensory neurons rather than a loss of primary transduction currents. DEG/ENaC sodium channels are not mechanosensors in mouse sensory neurons.


Assuntos
Mecanotransdução Celular/fisiologia , Células Receptoras Sensoriais/metabolismo , Canais de Sódio/metabolismo , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Animais , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Mecanotransdução Celular/genética , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.7 , Canal de Sódio Disparado por Voltagem NAV1.8 , Canais de Sódio/genética
16.
Pain ; 152(6): 1238-1248, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21295407

RESUMO

Opioid receptors are major actors in pain control and are broadly distributed throughout the nervous system. A major challenge in pain research is the identification of key opioid receptor populations within nociceptive pathways, which control physiological and pathological pain. In particular, the respective contribution of peripheral vs. central receptors remains unclear, and it has not been addressed by genetic approaches. To investigate the contribution of peripheral delta opioid receptors in pain control, we created conditional knockout mice where delta receptors are deleted specifically in peripheral Na(V)1.8-positive primary nociceptive neurons. Mutant mice showed normal pain responses to acute heat and to mechanical and formalin stimuli. In contrast, mutant animals showed a remarkable increase of mechanical allodynia under both inflammatory pain induced by complete Freund adjuvant and neuropathic pain induced by partial sciatic nerve ligation. In these 2 models, heat hyperalgesia was virtually unchanged. SNC80, a delta agonist administered either systemically (complete Freund adjuvant and sciatic nerve ligation) or into a paw (sciatic nerve ligation), reduced thermal hyperalgesia and mechanical allodynia in control mice. However, these analgesic effects were absent in conditional mutant mice. In conclusion, this study reveals the existence of delta opioid receptor-mediated mechanisms, which operate at the level of Na(V)1.8-positive nociceptive neurons. Delta receptors in these neurons tonically inhibit mechanical hypersensitivity in both inflammatory and neuropathic pain, and they are essential to mediate delta opioid analgesia under conditions of persistent pain. This delta receptor population represents a feasible therapeutic target to alleviate chronic pain while avoiding adverse central effects. The conditional knockout of delta-opioid receptor in primary afferent Na(V)1.8 neurons augmented mechanical allodynia in persistent pain models and abolished delta opioid analgesia in these models.


Assuntos
Gânglios Espinais/patologia , Nociceptores/fisiologia , Dor/genética , Dor/patologia , Receptores Opioides delta/deficiência , Analgésicos Opioides/uso terapêutico , Análise de Variância , Animais , Benzamidas/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Adjuvante de Freund/efeitos adversos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Inflamação/induzido quimicamente , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Canal de Sódio Disparado por Voltagem NAV1.8 , Nociceptores/efeitos dos fármacos , Dor/etiologia , Medição da Dor/métodos , Piperazinas/uso terapêutico , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Canais de Sódio/genética , Canais de Sódio/metabolismo , Isótopos de Enxofre/farmacocinética
17.
Mol Pain ; 6: 77, 2010 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-21059214

RESUMO

BACKGROUND: EphB receptors and their ephrin-B ligands play an important role in nervous system development, as well as synapse formation and plasticity in the adult brain. Recent studies show that intrathecal treatment with EphB-receptor activator ephrinB2-Fc induced thermal hyperalgesia and mechanical allodynia in rat, indicating that ephrin-B2 in small dorsal root ganglia (DRG) neurons and EphB receptors in the spinal cord modulate pain processing. To examine the role of ephrin-B2 in peripheral pain pathways, we deleted ephrin-B2 in Nav1.8+ nociceptive sensory neurons with the Cre-loxP system. Sensory neuron numbers and terminals were examined using neuronal makers. Pain behavior in acute, inflammatory and neuropathic pain models was assessed in the ephrin-B2 conditional knockout (CKO) mice. We also investigated the c-Fos expression and NMDA receptor NR2B phosphorylation in ephrin-B2 CKO mice and littermate controls. RESULTS: The ephrin-B2 CKO mice were healthy with no sensory neuron loss. However, pain-related behavior was substantially altered. Although acute pain behavior and motor co-ordination were normal, inflammatory pain was attenuated in ephrin-B2 mutant mice. Complete Freund's adjuvant (CFA)-induced mechanical hyperalgesia was halved. Formalin-induced pain behavior was attenuated in the second phase, and this correlated with diminished tyrosine phosphorylation of N-methyl-D-aspartic acid (NMDA) receptor subunit NR2B in the dorsal horn. Thermal hyperalgesia and mechanical allodynia were significantly reduced in the Seltzer model of neuropathic pain. CONCLUSIONS: Presynaptic ephrin-B2 expression thus plays an important role in regulating inflammatory pain through the regulation of synaptic plasticity in the dorsal horn and is also involved in the pathogenesis of some types of neuropathic pain.


Assuntos
Efrina-B2/metabolismo , Inflamação/complicações , Inflamação/metabolismo , Neuralgia/complicações , Neuralgia/metabolismo , Nociceptores/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Doença Aguda , Animais , Comportamento Animal , Sobrevivência Celular , Modelos Animais de Doenças , Éxons/genética , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Deleção de Genes , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/patologia , Integrases/metabolismo , Camundongos , Microglia/metabolismo , Microglia/patologia , Canal de Sódio Disparado por Voltagem NAV1.8 , Neuralgia/patologia , Neurônios Aferentes/metabolismo , Neurônios Aferentes/patologia , Nociceptores/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Sódio/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia
18.
J Neurosci ; 30(32): 10860-71, 2010 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-20702715

RESUMO

To examine the role of small RNAs in peripheral pain pathways, we deleted the enzyme Dicer in mouse postmitotic damage-sensing neurons. We used a Nav1.8-Cre mouse to target those nociceptors important for inflammatory pain. The conditional null mice were healthy with a normal number of sensory neurons and normal acute pain thresholds. Behavioral studies showed that inflammatory pain was attenuated or abolished. Inflammatory mediators failed to enhance excitability of Nav1.8+ sensory neurons from null mutant mice. Acute noxious input into the dorsal horn of the spinal cord was apparently normal, but the increased input associated with inflammatory pain measured using c-Fos staining was diminished. Microarray and quantitative real-time reverse-transcription PCR (qRT-PCR) analysis showed that Dicer deletion lead to the upregulation of many broadly expressed mRNA transcripts in dorsal root ganglia. By contrast, nociceptor-associated mRNA transcripts (e.g., Nav1.8, P2xr3, and Runx-1) were downregulated, resulting in lower levels of protein and functional expression. qRT-PCR analysis also showed lowered levels of expression of nociceptor-specific pre-mRNA transcripts. MicroRNA microarray and deep sequencing identified known and novel nociceptor microRNAs in mouse Nav1.8+ sensory neurons that may regulate nociceptor gene expression.


Assuntos
Regulação da Expressão Gênica/genética , Nociceptores/metabolismo , Limiar da Dor/fisiologia , Dor/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Canais de Sódio/metabolismo , Análise de Variância , Animais , Cerebelo/citologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , RNA Helicases DEAD-box/deficiência , Modelos Animais de Doenças , Endorribonucleases/deficiência , Feminino , Adjuvante de Freund/efeitos adversos , Gânglios Espinais/metabolismo , Perfilação da Expressão Gênica/métodos , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.8 , Proteínas do Tecido Nervoso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Dor/induzido quimicamente , Dor/genética , Medição da Dor , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X3 , Ribonuclease III , Canais de Sódio/deficiência , Canais de Sódio/genética , Medula Espinal/fisiopatologia , Fatores de Tempo
19.
J Neurosci ; 30(11): 3983-94, 2010 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-20237269

RESUMO

Small unmyelinated sensory neurons classified as nociceptors are divided into two subpopulations based on phenotypic differences, including expression of neurotrophic factor receptors. Approximately half of unmyelinated nociceptors express the NGF receptor TrkA, and half express the GDNF family ligand (GFL) receptor Ret. The function of NGF/TrkA signaling in the TrkA population of nociceptors has been extensively studied, and NGF/TrkA signaling is a well established mediator of pain. The GFLs are analgesic in models of neuropathic pain emphasizing the importance of understanding the physiological function of GFL/Ret signaling in nociceptors. However, perinatal lethality of Ret-null mice has precluded the study of the physiological role of GFL/Ret signaling in the survival, maintenance, and function of nociceptors in viable mice. We deleted Ret exclusively in nociceptors by crossing nociceptor-specific Na(v)1.8 Cre and Ret conditional mice to produce Ret-Na(v)1.8 conditional knock-out (CKO) mice. Loss of Ret exclusively in nociceptors results in a reduction in nociceptor number and size, indicating that Ret signaling is important for the survival and trophic support of these cells. Ret-Na(v)1.8 CKO mice exhibit reduced epidermal innervation but normal central projections. In addition, Ret-Na(v)1.8 CKO mice have increased sensitivity to cold and increased formalin-induced pain, demonstrating that Ret signaling modulates the function of nociceptors in vivo. Enhanced inflammation-induced pain may be mediated by decreased prostatic acid phosphatase (PAP), as PAP levels are markedly reduced in Ret-Na(v)1.8 CKO mice. The results of this study identify the physiological role of endogenous Ret signaling in the survival and function of nociceptors.


Assuntos
Nociceptores/fisiologia , Proteínas Proto-Oncogênicas c-ret/fisiologia , Transdução de Sinais/fisiologia , Animais , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Feminino , Formaldeído/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.8 , Nociceptores/citologia , Medição da Dor/métodos , Proteínas Proto-Oncogênicas c-ret/deficiência , Proteínas Proto-Oncogênicas c-ret/genética , Transdução de Sinais/genética , Canais de Sódio/deficiência , Canais de Sódio/genética
20.
J Neurosci ; 30(6): 2138-49, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-20147541

RESUMO

Chronic pain associated with inflammation is a common clinical problem, and the underlying mechanisms have only begun to be unraveled. GRK2 regulates cellular signaling by promoting G-protein-coupled receptor (GPCR) desensitization and direct interaction with downstream kinases including p38. The aim of this study was to determine the contribution of GRK2 to regulation of inflammatory pain and to unravel the underlying mechanism. GRK2(+/-) mice with an approximately 50% reduction in GRK2 developed increased and markedly prolonged thermal hyperalgesia and mechanical allodynia after carrageenan-induced paw inflammation or after intraplantar injection of the GPCR-binding chemokine CCL3. The effect of reduced GRK2 in specific cells was investigated using Cre-Lox technology. Carrageenan- or CCL3-induced hyperalgesia was increased but not prolonged in mice with decreased GRK2 only in Na(v)1.8 nociceptors. In vitro, reduced neuronal GRK2 enhanced CCL3-induced TRPV1 sensitization. In vivo, CCL3-induced acute hyperalgesia in GRK2(+/-) mice was mediated via TRPV1. Reduced GRK2 in microglia/monocytes only was required and sufficient to transform acute carrageenan- or CCL3-induced hyperalgesia into chronic hyperalgesia. Chronic hyperalgesia in GRK2(+/-) mice was associated with ongoing microglial activation and increased phospho-p38 and tumor necrosis factor alpha (TNF-alpha) in the spinal cord. Inhibition of spinal cord microglial, p38, or TNF-alpha activity by intrathecal administration of specific inhibitors reversed ongoing hyperalgesia in GRK2(+/-) mice. Microglia/macrophage GRK2 expression was reduced in the lumbar ipsilateral spinal cord during neuropathic pain, underlining the pathophysiological relevance of microglial GRK2. Thus, we identified completely novel cell-specific roles of GRK2 in regulating acute and chronic inflammatory hyperalgesia.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Dor/enzimologia , Dor/fisiopatologia , Animais , Astrócitos/metabolismo , Células Cultivadas , Quimiocina CCL3/farmacologia , Quimiocina CCL3/fisiologia , Feminino , Quinase 2 de Receptor Acoplado a Proteína G/genética , Hiperalgesia/enzimologia , Hiperalgesia/fisiopatologia , Inflamação/enzimologia , Inflamação/fisiopatologia , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/enzimologia , Dor/imunologia , Doenças do Sistema Nervoso Periférico/enzimologia , Doenças do Sistema Nervoso Periférico/imunologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/enzimologia , Medula Espinal/enzimologia , Canais de Cátion TRPV/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...