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










Base de dados
Intervalo de ano de publicação
1.
Glia ; 71(4): 945-956, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36495059

RESUMO

Signal propagation is the essential function of nerves. Lysophosphatidic acid 18:1 (LPA) allows the selective stimulation of calcium signaling in Schwann cells but not neurons. Here, the time course of slowing and amplitude reduction on compound action potentials due to LPA exposure was observed in myelinated and unmyelinated fibers of the mouse, indicating a clear change of axonal function. Teased nerve fiber imaging showed that Schwann cell activation is also present in axon-attached Schwann cells in freshly isolated peripheral rat nerves. The LPA receptor 1 was primarily localized at the cell extensions in isolated rat Schwann cells, suggesting a role in cell migration. Structural investigation of rat C-fibers demonstrated that LPA leads to an evagination of the axons from their Schwann cells. In A-fibers, the nodes of Ranvier appeared unchanged, but the Schmidt-Lanterman incisures were shortened and myelination reduced. The latter might increase leak current, reducing the potential spread to the next node of Ranvier and explain the changes in conduction velocity. The observed structural changes provide a plausible explanation for the functional changes in myelinated and unmyelinated axons of peripheral nerves and the reported sensory sensations such as itch and pain.


Assuntos
Nervos Periféricos , Células de Schwann , Camundongos , Ratos , Animais , Nervos Periféricos/fisiologia , Células de Schwann/fisiologia , Bainha de Mielina , Fibras Nervosas Mielinizadas/fisiologia , Axônios/fisiologia
2.
Physiol Rep ; 10(6): e15194, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35340127

RESUMO

The most widely used formalin test to screen antinociceptive drug candidates is still apostrophized as targeting inflammatory pain, in spite of strong opposing evidence published. In our rat skin-nerve preparation ex vivo, recording from all classes of sensory single-fibers (n = 32), 30 units were transiently excited by formaldehyde concentrations 1-100 mM applied to receptive fields (RFs) for 3 min, C and Aδ-fibers being more sensitive (1-30 mM) than Aß-fibers. From 30 mM on, ~1% of the concentration usually injected in vivo, all RFs were defunctionalized and conduction in an isolated sciatic nerve preparation was irreversibly blocked. Thus, formaldehyde, generated a state of 'anesthesia dolorosa' in the RFs in so far as after a quiescent interphase all fibers with unmyelinated terminals developed a second phase of vigorous discharge activity which correlated well in time course and magnitude with published pain-related behaviors. Sural nerve filament recordings in vivo confirmed that higher formalin concentrations (> 42 mM) have to be injected to the skin to induce this second phase of discharge. Patch-clamp and calcium-imaging confirmed TRPA1 as the primary transducer of formaldehyde (10 mM) effects on mouse sensory neurons. However, stimulated CGRP release from isolated skin of TRPA1+/+ and TRPA1-/- mice showed a convergence of the saturating concentration-response curves at 100 mM formaldehyde, which did not occur with nerve and trachea preparations. Finally, skin-nerve recordings from C and Aδ-fibers of TRPA1-/- mice revealed a massive reduction in formaldehyde (30 mM)-evoked discharge. However, the remaining activity was still biphasic, thus confirming additional unspecific excitotoxic actions of the fixative that diffuses along still excitable axons as previously published. The multiplicity of formaldehyde's actions requires extensive discussion and literature review, leading to a fundamental reevaluation of the formalin test.


Assuntos
Dor , Roedores , Animais , Camundongos , Dor/induzido quimicamente , Medição da Dor , Ratos , Células Receptoras Sensoriais , Pele/inervação
3.
Exp Neurol ; 346: 113838, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34450183

RESUMO

Painful diabetic neuropathy occurs in approximately 20% of diabetic patients with underlying pathomechanisms not fully understood. We evaluated the contribution of the CaV3.2 isoform of T-type calcium channel to hyperglycemia-induced changes in cutaneous sensory C-fiber functions and neuropeptide release employing the streptozotocin (STZ) diabetes model in congenic mouse strains including global knockouts (KOs). Hyperglycemia established for 3-5 weeks in male C57BL/6J mice led to major reorganizations in peripheral C-fiber functions. Unbiased electrophysiological screening of mechanosensitive single-fibers in isolated hairy hindpaw skin revealed a relative loss of (polymodal) heat sensing in favor of cold sensing. In healthy CaV3.2 KO mice both heat and cold sensitivity among the C-fibers seemed underrepresented in favor of exclusive mechanosensitivity, low-threshold in particular, which deficit became significant in the diabetic KOs. Diabetes also led to a marked increase in the incidence of spontaneous discharge activity among the C-fibers of wildtype mice, which was reduced by the specific CaV3.2 blocker TTA-P2 and largely absent in the KOs. Evaluation restricted to the peptidergic class of nerve fibers - measuring KCl-stimulated CGRP release - revealed a marked reduction in the sciatic nerve by TTA-P2 in healthy but not diabetic wildtypes, the latter showing CGRP release that was as much reduced as in healthy and, to the same extent, in diabetic CaV3.2 KOs. These data suggest that diabetes abrogates all CaV3.2 functionality in the peripheral nerve axons. In striking contrast, diabetes markedly increased the KCl-stimulated CGRP release from isolated hairy skin of wildtypes but not KO mice, and TTA-P2 reversed this increase, strongly suggesting a de novo expression of CaV3.2 in peptidergic cutaneous nerve endings which may contribute to the enhanced spontaneous activity. De-glycosylation by neuraminidase showed clear desensitizing effects, both in regard to spontaneous activity and stimulated CGRP release, but included actions independent of CaV3.2. However, as diabetes-enhanced glycosylation is decisive for intra-axonal trafficking, it may account for the substantial reorganizations of the CaV3.2 distribution. The results may strengthen the validation of CaV3.2 channel as a therapeutic target of treating painful diabetic neuropathy.


Assuntos
Canais de Cálcio Tipo T/biossíntese , Neuropatias Diabéticas/metabolismo , Neuralgia/metabolismo , Nociceptores/metabolismo , Pele/metabolismo , Animais , Canais de Cálcio Tipo T/genética , Diabetes Mellitus Experimental , Neuropatias Diabéticas/genética , Neuropatias Diabéticas/patologia , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/genética , Neuralgia/patologia , Nociceptores/patologia , Técnicas de Cultura de Órgãos , Pele/inervação , Pele/patologia
4.
Mol Pain ; 14: 1744806918811699, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30345869

RESUMO

BACKGROUND: Etomidate is a preferred drug for the induction of general anesthesia in cardiovascular risk patients. As with propofol and other perioperatively used anesthetics, the application of aqueous etomidate formulations causes an intensive burning pain upon injection. Such algogenic properties of etomidate have been attributed to the solubilizer propylene glycol which represents 35% of the solution administered clinically. The aim of this study was to investigate the underlying molecular mechanisms which lead to injection pain of aqueous etomidate formulations. RESULTS: Activation of the nociceptive transient receptor potential (TRP) ion channels TRPA1 and TRPV1 was studied in a transfected HEK293t cell line by whole-cell voltage clamp recordings of induced inward ion currents. Calcium influx in sensory neurons of wild-type and trp knockout mice was ratiometrically measured by Fura2-AM staining. Stimulated calcitonin gene-related peptide release from mouse sciatic nerves was detected by enzyme immunoassay. Painfulness of different etomidate formulations was tested in a translational human pain model. Etomidate as well as propylene glycol proved to be effective agonists of TRPA1 and TRPV1 ion channels at clinically relevant concentrations. Etomidate consistently activated TRPA1, but there was also evidence for a contribution of TRPV1 in dependence of drug concentration ranges and species specificities. Distinct N-terminal cysteine and lysine residues seemed to mediate gating of TRPA1, although the electrophile scavenger N-acetyl-L-cysteine did not prevent its activation by etomidate. Propylene glycol-induced activation of TRPA1 and TRPV1 appeared independent of the concomitant high osmolarity. Intradermal injections of etomidate as well as propylene glycol evoked severe burning pain in the human pain model that was absent with emulsification of etomidate. CONCLUSIONS: Data in our study provided evidence that pain upon injection of clinical aqueous etomidate formulations is not an unspecific effect of hyperosmolarity but rather due to a specific action mediated by activated nociceptive TRPA1 and TRPV1 ion channels in sensory neurons.


Assuntos
Etomidato/farmacologia , Dor/fisiopatologia , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cálcio/metabolismo , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dor/induzido quimicamente , Dor/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo
5.
PLoS One ; 12(11): e0188008, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29141003

RESUMO

BACKGROUND: Local anaesthetics (LA) reduce neuronal excitability by inhibiting voltage-gated Na+ channels. When applied at high concentrations in the direct vicinity of nerves, LAs can also induce relevant irritation and neurotoxicity via mechanisms involving an increase of intracellular Ca2+. In the present study we explored the role of the Ca2+-permeable ion channels TRPA1 and TRPV1 for lidocaine-induced Ca2+-influx, neuropeptide release and neurotoxicity in mouse sensory neurons. METHODS: Cultured dorsal root ganglion (DRG) neurons from wildtype and mutant mice lacking TRPV1, TRPA1 or both channels were explored by means of calcium imaging, whole-cell patch clamp recordings and trypan blue staining for cell death. Release of calcitonin gene-related peptide (CGRP) from isolated mouse peripheral nerves was determined with ELISA. RESULTS: Lidocaine up to 10 mM induced a concentration-dependent reversible increase in intracellular Ca2+ in DRG neurons from wildtype and mutant mice lacking one of the two receptors, but not in neurons lacking both TRPA1 and TRPV1. 30 mM lidocaine also released Ca2+ from intracellular stores, presumably from the endoplasmic reticulum. While 10 mM lidocaine evoked an axonal CGRP release requiring expression of either TRPA1 or TRPV1, CGRP release induced by 30 mM lidocaine again mobilized internal Ca2+ stores. Lidocaine-evoked cell death required neither TRPV1 nor TRPA1. SUMMARY: Depending on the concentration, lidocaine employs TRPV1, TRPA1 and intracellular Ca2+ stores to induce a Ca2+-dependent release of the neuropeptide CGRP. Lidocaine-evoked cell death does not seem to require Ca2+ influx through TRPV1 or TRPV1.


Assuntos
Cálcio/metabolismo , Lidocaína/farmacologia , Células Receptoras Sensoriais/efeitos dos fármacos , Canal de Cátion TRPA1/fisiologia , Canais de Cátion TRPV/fisiologia , Animais , Apoptose/efeitos dos fármacos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Células Cultivadas , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Transporte de Íons , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Células Receptoras Sensoriais/metabolismo
6.
Sci Rep ; 7(1): 5447, 2017 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-28710476

RESUMO

Phospholipids occurring in cell membranes and lipoproteins are converted into oxidized phospholipids (OxPL) by oxidative stress promoting atherosclerotic plaque formation. Here, OxPL were characterized as novel targets in acute and chronic inflammatory pain. Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (OxPAPC) and its derivatives were identified in inflamed tissue by mass spectrometry and binding assays. They elicited calcium influx, hyperalgesia and induced pro-nociceptive peptide release. Genetic, pharmacological and mass spectrometric evidence in vivo as well as in vitro confirmed the role of transient receptor potential channels (TRPA1 and TRPV1) as OxPAPC targets. Treatment with the monoclonal antibody E06 or with apolipoprotein A-I mimetic peptide D-4F, capturing OxPAPC in atherosclerosis, prevented inflammatory hyperalgesia, and in vitro TRPA1 activation. Administration of D-4F or E06 to rats profoundly ameliorated mechanical hyperalgesia and inflammation in collagen-induced arthritis. These data reveal a clinically relevant role for OxPAPC in inflammation offering therapy for acute and chronic inflammatory pain treatment by scavenging OxPAPC.


Assuntos
Anticorpos Monoclonais/farmacologia , Apolipoproteína A-I/farmacologia , Artrite Experimental/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Dor/tratamento farmacológico , Fosfatidilcolinas/antagonistas & inibidores , Canal de Cátion TRPA1/genética , Canais de Cátion TRPV/genética , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Colágeno Tipo II/administração & dosagem , Feminino , Expressão Gênica , Células HEK293 , Membro Posterior , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Masculino , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Dor/induzido quimicamente , Dor/metabolismo , Dor/patologia , Técnicas de Patch-Clamp , Fosfatidilcolinas/metabolismo , Fosfatidilcolinas/farmacologia , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Canal de Cátion TRPA1/antagonistas & inibidores , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
7.
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
8.
Pain ; 157(11): 2504-2516, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27434506

RESUMO

Crotalphine is a structural analogue to a novel analgesic peptide that was first identified in the crude venom from the South American rattlesnake Crotalus durissus terrificus. Although crotalphine's analgesic effect is well established, its direct mechanism of action remains unresolved. The aim of the present study was to investigate the effect of crotalphine on ion channels in peripheral pain pathways. We found that picomolar concentrations of crotalphine selectively activate heterologously expressed and native TRPA1 ion channels. TRPA1 activation by crotalphine required intact N-terminal cysteine residues and was followed by strong and long-lasting desensitization of the channel. Homologous desensitization of recombinant TRPA1 and heterologous desensitization in cultured dorsal root ganglia neurons was observed. Likewise, crotalphine acted on peptidergic TRPA1-expressing nerve endings ex vivo as demonstrated by suppression of calcitonin gene-related peptide release from the trachea and in vivo by inhibition of chemically induced and inflammatory hypersensitivity in mice. The crotalphine-mediated desensitizing effect was abolished by the TRPA1 blocker HC030031 and absent in TRPA1-deficient mice. Taken together, these results suggest that crotalphine is the first peptide to mediate antinociception selectively and at subnanomolar concentrations by targeting TRPA1 ion channels.


Assuntos
Analgésicos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Peptídeos/uso terapêutico , Canais de Potencial de Receptor Transitório/metabolismo , Potenciais de Ação/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Bradicinina/toxicidade , Cálcio/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Gânglios Espinais/citologia , Células HEK293 , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/etiologia , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Peptídeos/farmacologia , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/genética , Zimosan/toxicidade
9.
Sci Rep ; 6: 28621, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27356469

RESUMO

We demonstrate a novel dual strategy against inflammation and pain through body-wide desensitization of nociceptors via TRPA1. Attenuation of experimental colitis by capsazepine (CPZ) has long been attributed to its antagonistic action on TRPV1 and associated inhibition of neurogenic inflammation. In contrast, we found that CPZ exerts its anti-inflammatory effects via profound desensitization of TRPA1. Micromolar CPZ induced calcium influx in isolated dorsal root ganglion (DRG) neurons from wild-type (WT) but not TRPA1-deficient mice. CPZ-induced calcium transients in human TRPA1-expressing HEK293t cells were blocked by the selective TRPA1 antagonists HC 030031 and A967079 and involved three cysteine residues in the N-terminal domain. Intriguingly, both colonic enemas and drinking water with CPZ led to profound systemic hypoalgesia in WT and TRPV1(-/-) but not TRPA1(-/-) mice. These findings may guide the development of a novel class of disease-modifying drugs with anti-inflammatory and anti-nociceptive effects.


Assuntos
Analgésicos/farmacologia , Anti-Inflamatórios/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Capsaicina/análogos & derivados , Dor/tratamento farmacológico , Óleos de Plantas/farmacologia , Canal de Cátion TRPA1/metabolismo , Acetanilidas/farmacologia , Animais , Capsaicina/farmacologia , Células HEK293 , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Mostardeira , Oximas/farmacologia , Dor/genética , Dor/metabolismo , Purinas/farmacologia , Canal de Cátion TRPA1/antagonistas & inibidores , Canal de Cátion TRPA1/genética
10.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27030709

RESUMO

BACKGROUND: Opioids are the gold standard for the treatment of acute pain despite serious side effects in the central and enteric nervous system. µ-opioid receptors (MOPs) are expressed and functional at the terminals of sensory axons, when activated by exogenous or endogenous ligands. However, the presence and function of MOP along nociceptive axons remains controversial particularly in naïve animals. Here, we characterized axonal MOPs by immunofluorescence, ultrastructural, and functional analyses. Furthermore, we evaluated hypertonic saline as a possible enhancer of opioid receptor function. RESULTS: Comparative immunolabeling showed that, among several tested antibodies, which all provided specific MOP detection in the rat central nervous system (CNS), only one monoclonal MOP-antibody yielded specificity and reproducibility for MOP detection in the rat peripheral nervous system including the sciatic nerve. Double immunolabeling documented that MOP immunoreactivity was confined to calcitonin gene-related peptide (CGRP) positive fibers and fiber bundles. Almost identical labeling and double labeling patterns were found using mcherry-immunolabeling on sciatic nerves of mice producing a MOP-mcherry fusion protein (MOP-mcherry knock-in mice). Preembedding immunogold electron microscopy on MOP-mcherry knock-in sciatic nerves indicated presence of MOP in cytoplasm and at membranes of unmyelinated axons. Application of [D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin (DAMGO) or fentanyl dose-dependently inhibited depolarization-induced CGRP release from rat sciatic nerve axons ex vivo, which was blocked by naloxone. When the lipophilic opioid fentanyl was applied perisciatically in naïve Wistar rats, mechanical nociceptive thresholds increased. Subthreshold doses of fentanyl or the hydrophilic opioid DAMGO were only effective if injected together with hypertonic saline. In vitro, using ß-arrestin-2/MOP double-transfected human embryonic kidney cells, DAMGO as well as fentanyl lead to a recruitment of ß-arrestin-2 to the membrane followed by a ß-arrestin-2 reappearance in the cytosol and MOP internalization. Pretreatment with hypertonic saline prevented MOP internalization. CONCLUSION: MOPs are present and functional in the axonal membrane from naïve animals. Hypertonic saline acutely decreases ligand-induced internalization of MOP and thereby might improve MOP function. Further studies should explore potential clinical applications of opioids together with enhancers for regional analgesia.


Assuntos
Analgesia , Axônios/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Axônios/efeitos dos fármacos , Axônios/ultraestrutura , Comportamento Animal/efeitos dos fármacos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Endocitose/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Fentanila/farmacologia , Técnicas de Introdução de Genes , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Nociceptividade/efeitos dos fármacos , Potássio/farmacologia , Ratos Wistar , Reprodutibilidade dos Testes , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , beta-Arrestinas/metabolismo
11.
J Physiol ; 594(19): 5529-41, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27105013

RESUMO

KEY POINTS: This study examines conduction in peripheral nerves and its use dependence in tetrodotoxin-resistant (TTXr) sodium channel (Nav 1.8, Nav 1.9) knockout and wildtype animals. We observed use-dependent decreases of single fibre and compound action potential amplitude in peripheral mouse C-fibres (wildtype). This matches the previously published hypothesis that increased Na/K-pump activity is not the underlying mechanism for use-dependent changes of neural conduction. Knocking out TTXr sodium channels influences use-dependent changes of conductive properties (action potential amplitude, latency, conduction safety) in the order Nav 1.8 KO > Nav 1.9KO > wildtype. This is most likely explained by different subsets of presumably (relatively) Nav 1.7-rich conducting fibres in knockout animals as compared to wildtypes, in combination with reduced per-pulse sodium influx. ABSTRACT: Use dependency of peripheral nerves, especially of nociceptors, correlates with receptive properties. Slow inactivation of voltage-gated sodium channels has been discussed to be the underlying mechanism - pointing to a receptive class-related difference of sodium channel equipment. Using electrophysiological recordings of single unmyelinated cutaneous fibres and their compound action potential (AP), we evaluated use-dependent changes in mouse peripheral nerves, and the contribution of the tetrodotoxin-resistant (TTXr) sodium channels Nav 1.8 and Nav 1.9 to these changes. Nerve fibres were electrically stimulated using single or double pulses at 2 Hz. Use-dependent changes of latency, AP amplitude, and duration as well as the fibres' ability to follow the stimulus were evaluated. AP amplitudes substantially diminished in used fibres from C57BL/6 but increased in Nav 1.8 knockout (KO) mice, with Nav 1.9 KO in between. Activity-induced latency slowing was in contrast the most pronounced in Nav 1.8 KOs and the least in wildtype mice. The genotype was also predictive of how long fibres could follow the double pulsed stimulus with wildtype fibres blocking first and Nav 1.8 KO fibres enduring the longest. In contrast, changes in spike duration were less pronounced and displayed no significant tendency. Thus, all major measures of peripheral nerve accommodation (amplitude, latency and durability) depended on genotype. All use-dependent changes appeared in the order NaV 1.8 KO > NaV 1.9 KO > wildtype, which is most likely explained by the relative contribution of Nav 1.7 varying in the same order and the amounts of per-pulse sodium influx expected in the opposite order.


Assuntos
Fibras Nervosas/fisiologia , Nociceptores/fisiologia , Canais de Sódio/fisiologia , Potenciais de Ação , Animais , Resistência a Medicamentos , Estimulação Elétrica , Feminino , Pé/inervação , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Condução Nervosa , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/genética , Tetrodotoxina/farmacologia
12.
Anesthesiology ; 124(5): 1153-65, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26859646

RESUMO

BACKGROUND: The relatively membrane-impermeable lidocaine derivative QX-314 has been reported to permeate the ion channels transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential cation channel, subfamily A, member 1 (TRPA1) to induce a selective inhibition of sensory neurons. This approach is effective in rodents, but it also seems to be associated with neurotoxicity. The authors examined whether the human isoforms of TRPV1 and TRPA1 allow intracellular entry of QX-314 to mediate sodium channel inhibition and cytotoxicity. METHODS: Human embryonic kidney 293 (HEK-293) cells expressing wild-type or mutant human (h) TRPV1 or TRPA1 constructs as well as the sodium channel Nav1.7 were investigated by means of patch clamp and ratiometric calcium imaging. Cytotoxicity was examined by flow cytometry. RESULTS: Activation of hTRPA1 by carvacrol and hTRPV1 by capsaicin produced a QX-314-independent reduction of sodium current amplitudes. However, permeation of QX-314 through hTRPV1 or hTRPA1 was evident by a concentration-dependent, use-dependent inhibition of Nav1.7 activated at 10 Hz. Five and 30 mM QX-314 activated hTRPV1 via mechanisms involving the intracellular vanilloid-binding domain and hTRPA1 via unknown mechanisms independent of intracellular cysteins. Expression of hTRPV1, but not hTRPA1, was associated with a QX-314-induced cytotoxicity (viable cells 48 ± 5% after 30 mM QX-314) that was ameliorated by the TRPV1 antagonist 4-(3-chloro-2-pyridinyl)-N-[4-(1,1-dimethylethyl)phenyl]-1-piperazinecarboxamide (viable cells 81 ± 5%). CONCLUSIONS: The study data demonstrate that QX-314 directly activates and permeates the human isoforms of TRPV1 and TRPA1 to induce inhibition of sodium channels, but also a TRPV1-dependent cytotoxicity. These results warrant further validation of this approach in more intact preparations and may be valuable for the development of this concept into clinical practice.


Assuntos
Anestésicos Locais/farmacologia , Canais de Cálcio/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Lidocaína/análogos & derivados , Proteínas do Tecido Nervoso/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Lidocaína/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Proteínas do Tecido Nervoso/agonistas , Canal de Cátion TRPA1 , Canais de Cátion TRPV/agonistas , Canais de Potencial de Receptor Transitório/agonistas
13.
Pain ; 155(6): 1079-1090, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24525276

RESUMO

There is emerging evidence that hyperpolarization-activated cation (HCN) channels are involved in the development of pathological pain, including allodynia and hyperalgesia. Mice lacking the HCN isoform 2 display reduced heat but unchanged mechanical pain behavior, as recently shown in preclinical models of acute inflammatory pain. However, the impact of HCN2 to chronic pain conditions is less clear and has not been examined so far. In this report, we study the role of HCN2 in the complete Freund's adjuvant inflammation model reflecting chronic pain conditions. We used sensory neuron-specific as well as inducible global HCN2 mutants analyzing pain behavior in persistent inflammation and complemented this by region-specific administration of an HCN channel blocker. Our results demonstrate that the absence of HCN2 in primary sensory neurons reduces tactile hypersensitivity in chronic inflammatory conditions but leaves heat hypersensitivity unaffected. This result is in remarkable contrast to the recently described role of HCN2 in acute inflammatory conditions. We show that chronic inflammation results in an increased expression of HCN2 and causes sensitization in peripheral and spinal terminals of the pain transduction pathway. The contribution of HCN2 to peripheral sensitization mechanisms was further supported by single-fiber recordings from isolated skin-nerve preparations and by conduction velocity measurements of saphenous nerve preparations. Global HCN2 mutants revealed that heat hypersensitivity-unaffected in peripheral HCN2 mutants-was diminished by the additional disruption of central HCN2 channels, suggesting that thermal hyperalgesia under chronic inflammatory conditions is mediated by HCN2 channels beyond primary sensory afferents.


Assuntos
Temperatura Alta/efeitos adversos , Hiperalgesia/genética , Hiperalgesia/patologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/fisiologia , Canais de Potássio/fisiologia , Tato/genética , Animais , Células Cultivadas , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Estimulação Física/efeitos adversos
14.
J Neurosci ; 33(1): 315-26, 2013 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-23283344

RESUMO

Specific somatosensations may be processed by different subsets of primary afferents. C-fibers expressing heat-sensitive TRPV1 channels are proposed, for example, to be heat but not mechanical pain detectors. To phenotype in rats the sensory function of TRPV1(+) afferents, we rapidly and selectively silenced only their activity, by introducing the membrane-impermeant sodium channel blocker QX-314 into these axons via the TRPV1 channel pore. Using tandem mass spectrometry we show that upon activation with capsaicin, QX-314 selectively accumulates in the cytosol only of TRPV1-expressing cells, and not in control cells. Exposure to QX-314 and capsaicin induces in small DRG neurons a robust sodium current block within 30 s. In sciatic nerves, application of extracellular QX-314 with capsaicin persistently reduces C-fiber but not A-fiber compound action potentials and this effect does not occur in TRPV1(-/-) mice. Behavioral phenotyping after selectively silencing TRPV1(+) sciatic nerve axons by perineural injections of QX-314 and capsaicin reveals deficits in heat and mechanical pressure but not pinprick or light touch perception. The response to intraplantar capsaicin is substantially reduced, as expected. During inflammation, silencing TRPV1(+) axons abolishes heat, mechanical, and cold hyperalgesia but tactile and cold allodynia remain following peripheral nerve injury. These results indicate that TRPV1-expressing sensory neurons process particular thermal and mechanical somatosensations, and that the sensory channels activated by mechanical and cold stimuli to produce pain in naive/inflamed rats differ from those in animals after peripheral nerve injury.


Assuntos
Axônios/fisiologia , Comportamento Animal/fisiologia , Dor Crônica/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPV/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Anestésicos Locais/farmacologia , Animais , Axônios/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Capsaicina/farmacologia , Modelos Animais de Doenças , Lidocaína/análogos & derivados , Lidocaína/farmacologia , Masculino , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/fisiopatologia , Células Receptoras Sensoriais/efeitos dos fármacos
15.
J Biol Chem ; 287(34): 28291-306, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22740698

RESUMO

Neuropathic pain can develop as an agonizing sequela of diabetes mellitus and chronic uremia. A chemical link between both conditions of altered metabolism is the highly reactive compound methylglyoxal (MG), which accumulates in all cells, in particular neurons, and leaks into plasma as an index of the severity of the disorder. The electrophilic structure of this cytotoxic ketoaldehyde suggests TRPA1, a receptor channel deeply involved in inflammatory and neuropathic pain, as a molecular target. We demonstrate that extracellularly applied MG accesses specific intracellular binding sites of TRPA1, activating inward currents and calcium influx in transfected cells and sensory neurons, slowing conduction velocity in unmyelinated peripheral nerve fibers, and stimulating release of proinflammatory neuropeptides from and action potential firing in cutaneous nociceptors. Using a model peptide of the N terminus of human TRPA1, we demonstrate the formation of disulfide bonds based on MG-induced modification of cysteines as a novel mechanism. In conclusion, MG is proposed to be a candidate metabolite that causes neuropathic pain in metabolic disorders and thus is a promising target for medicinal chemistry.


Assuntos
Canais de Cálcio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/metabolismo , Nociceptores/metabolismo , Aldeído Pirúvico/metabolismo , Canais de Cátion TRPC/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Potenciais de Ação , Animais , Sítios de Ligação , Canais de Cálcio/genética , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Neuralgia/dietoterapia , Neuralgia/genética , Neuralgia/patologia , Neurônios/metabolismo , Neurônios/patologia , Neuropeptídeos/metabolismo , Nociceptores/patologia , Ratos , Canal de Cátion TRPA1 , Canais de Cátion TRPC/genética , Canais de Potencial de Receptor Transitório/genética
16.
Proc Natl Acad Sci U S A ; 109(29): E2018-27, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22733753

RESUMO

Selective targeting of sensory or nociceptive neurons in peripheral nerves remains a clinically desirable goal. Delivery of promising analgesic drugs is often impeded by the perineurium, which functions as a diffusion barrier attributable to tight junctions. We used perineurial injection of hypertonic saline as a tool to open the perineurial barrier transiently in rats and elucidated the molecular action principle in mechanistic detail: Hypertonic saline acts via metalloproteinase 9 (MMP9). The noncatalytic hemopexin domain of MMP9 binds to the low-density lipoprotein receptor-related protein-1, triggers phosphorylation of extracellular signal-regulated kinase 1/2, and induces down-regulation of the barrier-forming tight junction protein claudin-1. Perisciatic injection of any component of this pathway, including MMP9 hemopexin domain or claudin-1 siRNA, enables an opioid peptide ([D-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin) and a selective sodium channel (NaV1.7)-blocking toxin (ProToxin-II) to exert antinociceptive effects without motor impairment. The latter, as well as the classic TTX, blocked compound action potentials in isolated nerves only after disruption of the perineurial barrier, which, in return, allowed endoneurally released calcitonin gene-related peptide to pass through the nerve sheaths. Our data establish the function and regulation of claudin-1 in the perineurium as the major sealing component, which could be modulated to facilitate drug delivery or, potentially, reseal the barrier under pathological conditions.


Assuntos
Analgésicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Nervos Periféricos/metabolismo , Solução Salina Hipertônica/administração & dosagem , Analgésicos/metabolismo , Animais , Western Blotting , Claudina-1 , Espectroscopia Dielétrica , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imunofluorescência , Metaloproteinase 9 da Matriz/farmacologia , Proteínas de Membrana/metabolismo , Limiar da Dor/efeitos dos fármacos , Fosforilação , RNA Interferente Pequeno/genética , Ratos , Solução Salina Hipertônica/metabolismo
17.
Anesthesiology ; 116(6): 1335-46, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22504149

RESUMO

BACKGROUND: Opioids induce analgesia mainly by inhibiting synaptic transmission via G protein-coupled opioid receptors. In addition to analgesia, buprenorphine induces a pronounced antihyperalgesia and is an effective adjuvant to local anesthetics. These properties only partially apply to other opioids, and thus targets other than opioid receptors are likely to be employed. Here we asked if buprenorphine inhibits voltage-gated Na(+) channels. METHODS: Na(+) currents were examined by whole cell patch clamp recordings on different recombinant Na(+) channel α-subunits. The effect of buprenorphine on unmyelinated mouse C-fibers was examined with the skin-nerve preparation. Data are presented as mean ± SEM. RESULTS: Buprenorphine induced a concentration-dependent tonic (IC(50) 33 ± 2 µM) and use-dependent block of endogenous Na(+) channels in ND7/23 cells. This block was state-dependent and displayed slow on and off characteristics. The effect of buprenorphine was reduced on local anesthetic insensitive Nav1.4-mutant constructs and was more pronounced on the inactivation-deficient Nav1.4-WCW mutant. Neuronal (Nav1.3, Nav1.7, and Nav1.8), cardiac (Nav1.5), and skeletal muscle (Nav1.4) α-subunits displayed small differences in tonic block, but similar degrees of use-dependent block. According to our patch clamp data, buprenorphine blocked electrically evoked action potentials in C-fiber nerve terminals. Buprenorphine was more potent than other opioids, including morphine (IC(50) 378 ± 20 µM), fentanyl (IC(50) 95 ± 5 µM), sufentanil (IC(50) 111 ± 6 µM), remifenatil (IC(50) 612 ± 17 µM), and tramadol (IC(50) 194 ± 9 µM). CONCLUSIONS: Buprenorphine is a potent local anesthetic and blocks voltage-gated Na(+) channels via the local anesthetic binding site. This property is likely to be relevant when buprenorphine is used for pain treatment and for local anesthesia.


Assuntos
Anestésicos Locais/farmacologia , Buprenorfina/farmacologia , Entorpecentes/farmacologia , Receptores Opioides mu/agonistas , Bloqueadores dos Canais de Sódio , Canais de Sódio/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Linhagem Celular , Interpretação Estatística de Dados , Gânglios Espinais/citologia , Humanos , Lidocaína/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.4 , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas Amielínicas/efeitos dos fármacos , Técnicas de Patch-Clamp , Pele/inervação , Canais de Sódio/genética , Canais de Sódio/fisiologia , Tetrodotoxina/farmacologia
18.
J Biol Chem ; 285(45): 34781-92, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20826794

RESUMO

Anesthetic agents can induce a paradox activation and sensitization of nociceptive sensory neurons and, thus, potentially facilitate pain processing. Here we identify distinct molecular mechanisms that mediate an activation of sensory neurons by 2,6-diisopropylphenol (propofol), a commonly used intravenous anesthetic known to elicit intense pain upon injection. Clinically relevant concentrations of propofol activated the recombinant transient receptor potential (TRP) receptors TRPA1 and TRPV1 heterologously expressed in HEK293t cells. In dorsal root ganglion (DRG) neurons, propofol-induced activation correlated better to expression of TRPA1 than of TRPV1. However, pretreatment with the protein kinase C activator 4ß-phorbol 12-myristate 13-acetate (PMA) resulted in a significantly sensitized propofol-induced activation of TRPV1 in DRG neurons as well as in HEK293t cells. Pharmacological and genetic silencing of both TRPA1 and TRPV1 only partially abrogated propofol-induced responses in DRG neurons. The remaining propofol-induced activation was abolished by the selective γ-aminobutyric acid, type A (GABA(A)) receptor antagonist picrotoxin. Propofol but not GABA evokes a release of calcitonin gene-related peptide, a key component of neurogenic inflammation, from isolated peripheral nerves of wild-type but not TRPV1 and TRPA1-deficient mice. Moreover, propofol but not GABA induced an intense pain upon intracutaneous injection. As both the release of calcitonin gene-related peptide and injection pain by propofol seem to be independent of GABA(A) receptors, our data identify TRPV1 and TRPA1 as key molecules for propofol-induced excitation of sensory neurons. This study warrants further investigations into the role of anesthetics to induce nociceptor sensitization and to foster postoperative pain.


Assuntos
Anestésicos Intravenosos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Nociceptores/metabolismo , Propofol/farmacologia , Receptores de GABA-A , Canais de Cátion TRPV/biossíntese , Canais de Potencial de Receptor Transitório/biossíntese , Animais , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Antagonistas GABAérgicos/farmacologia , Gânglios Espinais , Células HEK293 , Humanos , Inflamação/genética , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Picrotoxina/farmacologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Canal de Cátion TRPA1 , Canais de Cátion TRPV/genética , Canais de Potencial de Receptor Transitório/genética
19.
Pflugers Arch ; 459(5): 751-63, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20174994

RESUMO

The generation of action potentials in nociceptive neurons is accomplished by the tetrodotoxin-resistant (TTXr) Na+ channel Na(v)1.8. Following nerve injury, a redistribution of Na(v)1.8 from dorsal root ganglion (DRG) neurons into peripheral axons contributes to hyperexcitability and possibly to neuropathic pain. Na(v)1.8 has been reported to display a lower sensitivity to block by Na+ channel blockers as compared to TTX-sensitive (TTXs) Na(v) subunits. Furthermore, the antinociceptive efficacy of lidocaine is increased in Na(v)1.8-knockout mice. Here, we asked if Na(v)1.8 expression can reduce the susceptibility of sensory neurons to block by lidocaine. Employing wild-type and Na(v)1.8-knockout mice, we examined C-fibers in the skin-nerve preparation and Na+ currents in DRG neurons by patch-clamp recordings. Deletion of Na(v)1.8 resulted in an enhanced tonic block of Na+ currents in DRG neurons held at -80 mV but not at -140 mV. Accordingly, lower concentrations of lidocaine were required for a conduction block of C-fibers from Na(v)1.8-knockout as compared to wild-type mice. The efficacy of lidocaine on neurons lacking Na(v)1.8 was further increased by cold temperatures, due to a synergistic hyperpolarizing shift of the slow inactivation of TTXs Na+ channels by lidocaine and cooling. Finally, the approximately 90% reduction of TTXr Na+ currents in injured neurons from mice with a peripheral nerve injury was accompanied with an enhanced tonic block by lidocaine. In conclusion, our data demonstrate that the expression of Na(v)1.8 in sensory neurons can confine the antinociceptive efficacy of lidocaine and other Na+ channel blockers employed for pain treatment.


Assuntos
Anestésicos Locais/farmacologia , Lidocaína/farmacologia , Nociceptores/efeitos dos fármacos , Canais de Sódio/metabolismo , Tetrodotoxina/farmacologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.8 , Nociceptores/fisiologia , Canais de Sódio/genética
20.
J Clin Invest ; 118(2): 763-76, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18172555

RESUMO

Local anesthetics (LAs) block the generation and propagation of action potentials by interacting with specific sites of voltage-gated Na(+) channels. LAs can also excite sensory neurons and be neurotoxic through mechanisms that are as yet undefined. Nonspecific cation channels of the transient receptor potential (TRP) channel family that are predominantly expressed by nociceptive sensory neurons render these neurons sensitive to a variety of insults. Here we demonstrated that the LA lidocaine activated TRP channel family receptors TRPV1 and, to a lesser extent, TRPA1 in rodent dorsal root ganglion sensory neurons as well as in HEK293t cells expressing TRPV1 or TRPA1. Lidocaine also induced a TRPV1-dependent release of calcitonin gene-related peptide (CGRP) from isolated skin and peripheral nerve. Lidocaine sensitivity of TRPV1 required segments of the putative vanilloid-binding domain within and adjacent to transmembrane domain 3, was diminished under phosphatidylinositol 4,5-bisphosphate depletion, and was abrogated by a point mutation at residue R701 in the proximal C-terminal TRP domain. These data identify TRPV1 and TRPA1 as putative key elements of LA-induced nociceptor excitation. This effect is sufficient to release CGRP, a key component of neurogenic inflammation, and warrants investigation into the role of TRPV1 and TRPA1 in LA-induced neurotoxicity.


Assuntos
Anestésicos Locais/toxicidade , Lidocaína/toxicidade , Neurônios Aferentes/efeitos dos fármacos , Canais de Cátion TRPV/agonistas , Animais , Anquirinas , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Canais de Cálcio/efeitos dos fármacos , Capsaicina/farmacologia , Linhagem Celular , Potenciais Evocados/efeitos dos fármacos , Humanos , Neurônios Aferentes/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/antagonistas & inibidores , Fármacos do Sistema Sensorial/farmacologia , Canal de Cátion TRPA1 , Canais de Cátion TRPC , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...