Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Pharmacol ; 103(4): 221-229, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36635052

RESUMO

Sodium channel inhibitors used as local anesthetics, antiarrhythmics, or antiepileptics typically have the property of use-dependent inhibition, whereby inhibition is enhanced by repetitive channel activation. For targeting pain, Nav1.8 channels are an attractive target because they are prominent in primary pain-sensing neurons, with little or no expression in most other kinds of neurons, and a number of Nav1.8-targeted compounds have been developed. We examined the characteristics of Nav1.8 inhibition by one of the most potent Nav1.8 inhibitors so far described, A-887826, and found that when studied with physiologic resting potentials and physiologic temperatures, inhibition had strong "reverse use dependence", whereby inhibition was relieved by repetitive short depolarizations. This effect was much stronger with A-887826 than with A-803467, another Nav1.8 inhibitor. The use-dependent relief from inhibition was seen in both human Nav1.8 channels studied in a cell line and in native Nav1.8 channels in mouse dorsal root ganglion (DRG) neurons. In native Nav1.8 channels, substantial relief of inhibition occurred during repetitive stimulation by action potential waveforms at 5 Hz, suggesting that the phenomenon is likely important under physiologic conditions. SIGNIFICANCE STATEMENT: Nav1.8 sodium channels are expressed in primary pain-sensing neurons and are a prime current target for new drugs for pain. This work shows that one of the most potent Nav1.8 inhibitors, A-887826, has the unusual property that inhibition is relieved by repeated short depolarizations. This "reverse use dependence" may reduce inhibition during physiological firing and should be selected against in drug development.


Assuntos
Morfolinas , Canal de Sódio Disparado por Voltagem NAV1.8 , Neurônios , Niacinamida , Dor , Animais , Humanos , Camundongos , Gânglios Espinais , Potenciais da Membrana , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Canal de Sódio Disparado por Voltagem NAV1.8/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Dor/tratamento farmacológico , Dor/metabolismo , Ratos Sprague-Dawley , Ratos
2.
Cell Calcium ; 106: 102635, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35944383

RESUMO

High-voltage-activated calcium channels (HVACCs) are promising targets for developing analgesics given their roles in controlling synaptic transmission, neuronal excitability and neuropeptide release in primary nociceptive neurons. Despite previous efforts in developing HVACCs inhibitors of various drug modalities, it remains undetermined whether targeting HVACCs directly by a gene therapy approach could lead to pain alleviation in vivo. To test this, Sun and colleagues adopted a post-translational ubiquitination-based knockdown method targeting HVACCs in primary sensory neurons. They showed ablation of HVACC currents in a subset of primary sensory neurons, dampened hyperexcitability of sensory neurons after nerve injury and reduced pain behavior with no apparent adverse effects [1]. The results open the possibility of targeting ion channels with ubiquitination-based knockdown as a promising gene therapy candidate for pain treatment in future clinical studies.


Assuntos
Células Receptoras Sensoriais , Transmissão Sináptica , Animais , Canais de Cálcio/metabolismo , Gânglios Espinais/metabolismo , Dor , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo
3.
Elife ; 112022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35179483

RESUMO

Cannabidiol (CBD), a chemical found in the Cannabis sativa plant, is a clinically effective antiepileptic drug whose mechanism of action is unknown. Using a fluorescence-based thallium flux assay, we performed a large-scale screen and found enhancement of flux through heterologously expressed human Kv7.2/7.3 channels by CBD. Patch-clamp recordings showed that CBD acts at submicromolar concentrations to shift the voltage dependence of Kv7.2/7.3 channels in the hyperpolarizing direction, producing a dramatic enhancement of current at voltages near -50 mV. CBD enhanced native M-current in mouse superior cervical ganglion starting at concentrations of 30 nM and also enhanced M-current in rat hippocampal neurons. The potent enhancement of Kv2/7.3 channels by CBD may contribute to its effectiveness as an antiepileptic drug by reducing neuronal hyperexcitability.


Assuntos
Canabidiol/farmacologia , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Neurônios/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/genética , Neurônios/efeitos dos fármacos , Ratos
4.
Elife ; 112022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-35129439

RESUMO

Olfactory information is encoded in lateral entorhinal cortex (LEC) by two classes of layer 2 (L2) principal neurons: fan and pyramidal cells. However, the functional properties of L2 cells and how they contribute to odor coding are unclear. Here, we show in awake mice that L2 cells respond to odors early during single sniffs and that LEC is essential for rapid discrimination of both odor identity and intensity. Population analyses of L2 ensembles reveal that rate coding distinguishes odor identity, but firing rates are only weakly concentration dependent and changes in spike timing can represent odor intensity. L2 principal cells differ in afferent olfactory input and connectivity with inhibitory circuits and the relative timing of pyramidal and fan cell spikes provides a temporal code for odor intensity. Downstream, intensity is encoded purely by spike timing in hippocampal CA1. Together, these results reveal the unique processing of odor information by LEC subcircuits and highlight the importance of temporal coding in higher olfactory areas.


Assuntos
Córtex Entorrinal/fisiologia , Neurônios/fisiologia , Odorantes , Olfato/fisiologia , Humanos
5.
Nat Neurosci ; 25(2): 168-179, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34931070

RESUMO

Bacterial products can act on neurons to alter signaling and function. In the present study, we found that dorsal root ganglion (DRG) sensory neurons are enriched for ANTXR2, the high-affinity receptor for anthrax toxins. Anthrax toxins are composed of protective antigen (PA), which binds to ANTXR2, and the protein cargoes edema factor (EF) and lethal factor (LF). Intrathecal administration of edema toxin (ET (PA + EF)) targeted DRG neurons and induced analgesia in mice. ET inhibited mechanical and thermal sensation, and pain caused by formalin, carrageenan or nerve injury. Analgesia depended on ANTXR2 expressed by Nav1.8+ or Advillin+ neurons. ET modulated protein kinase A signaling in mouse sensory and human induced pluripotent stem cell-derived sensory neurons, and attenuated spinal cord neurotransmission. We further engineered anthrax toxins to introduce exogenous protein cargoes, including botulinum toxin, into DRG neurons to silence pain. Our study highlights interactions between a bacterial toxin and nociceptors, which may lead to the development of new pain therapeutics.


Assuntos
Antraz , Bacillus anthracis , Toxinas Bacterianas , Células-Tronco Pluripotentes Induzidas , Animais , Antraz/microbiologia , Antraz/terapia , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Gânglios Espinais/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Nociceptores/metabolismo , Dor , Receptores de Peptídeos/metabolismo
6.
J Neurosci ; 41(30): 6371-6387, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34131037

RESUMO

The nonpsychoactive phytocannabinoid cannabidiol (CBD) has been shown to have analgesic effects in animal studies but little is known about its mechanism of action. We examined the effects of CBD on intrinsic excitability of primary pain-sensing neurons. Studying acutely dissociated capsaicin-sensitive mouse DRG neurons at 37°C, we found that CBD effectively inhibited repetitive action potential firing, from 15-20 action potentials evoked by 1 s current injections in control to 1-3 action potentials with 2 µm CBD. Reduction of repetitive firing was accompanied by a reduction of action potential height, widening of action potentials, reduction of the afterhyperpolarization, and increased propensity to enter depolarization block. Voltage-clamp experiments showed that CBD inhibited both TTX-sensitive and TTX-resistant (TTX-R) sodium currents in a use-dependent manner. CBD showed strong state-dependent inhibition of TTX-R channels, with fast binding to inactivated channels during depolarizations and slow unbinding on repolarization. CBD alteration of channel availability at various voltages suggested that CBD binds especially tightly [Kd (dissociation constant), ∼150 nm] to the slow inactivated state of TTX-R channels, which can be substantially occupied at voltages as negative as -40 mV. Remarkably, CBD was more potent in inhibiting TTX-R channels and inhibiting action potential firing than the local anesthetic bupivacaine. We conclude that CBD might produce some of its analgesic effects by direct effects on neuronal excitability, with tight binding to the slow inactivated state of Nav1.8 channels contributing to effective inhibition of repetitive firing by modest depolarizations.SIGNIFICANCE STATEMENT Cannabidiol (CBD) has been shown to inhibit pain in various rodent models, but the mechanism of this effect is unknown. We describe the ability of CBD to inhibit repetitive action potential firing in primary nociceptive neurons from mouse dorsal root ganglia and analyze the effects on voltage-dependent sodium channels. We find that CBD interacts with TTX-resistant sodium channels in a state-dependent manner suggesting particularly tight binding to slow inactivated states of Nav1.8 channels, which dominate the overall inactivation of Nav1.8 channels for small maintained depolarizations from the resting potential. The results suggest that CBD can exert analgesic effects in part by directly inhibiting repetitive firing of primary nociceptors and suggest a strategy of identifying compounds that bind selectively to slow inactivated states of Nav1.8 channels for developing effective analgesics.


Assuntos
Analgésicos/farmacologia , Canabidiol/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Nociceptores/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Células Cultivadas , Feminino , Gânglios Espinais , Masculino , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.8/efeitos dos fármacos , Nociceptores/metabolismo
7.
Elife ; 82019 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-31765298

RESUMO

Voltage-dependent sodium and calcium channels in pain-initiating nociceptor neurons are attractive targets for new analgesics. We made a permanently charged cationic derivative of an N-type calcium channel-inhibitor. Unlike cationic derivatives of local anesthetic sodium channel blockers like QX-314, this cationic compound inhibited N-type calcium channels more effectively with extracellular than intracellular application. Surprisingly, the compound is also a highly effective sodium channel inhibitor when applied extracellularly, producing more potent inhibition than lidocaine or bupivacaine. The charged inhibitor produced potent and long-lasting analgesia in mouse models of incisional wound and inflammatory pain, inhibited release of the neuropeptide calcitonin gene-related peptide (CGRP) from dorsal root ganglion neurons, and reduced inflammation in a mouse model of allergic asthma, which has a strong neurogenic component. The results show that some cationic molecules applied extracellularly can powerfully inhibit both sodium channels and calcium channels, thereby blocking both nociceptor excitability and pro-inflammatory peptide release.


Assuntos
Canais de Cálcio Tipo N/genética , Inflamação Neurogênica/tratamento farmacológico , Dor/tratamento farmacológico , Canais de Sódio/genética , Animais , Bupivacaína/farmacologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/patologia , Humanos , Lidocaína/análogos & derivados , Lidocaína/farmacologia , Camundongos , Inflamação Neurogênica/genética , Inflamação Neurogênica/patologia , Nociceptores , Dor/genética , Dor/patologia , Bloqueadores dos Canais de Sódio/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...