Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Neurosci ; 41(11): 2457-2474, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33478988

RESUMO

Transient receptor potential melastatin 3 (TRPM3) is a heat-activated ion channel in primary sensory neurons of the dorsal root ganglia (DRGs). Pharmacological and genetic studies implicated TRPM3 in various pain modalities, but TRPM3 inhibitors were not validated in TRPM3-/- mice. Here we tested two inhibitors of TRPM3 in male and female wild-type and TRPM3-/- mice in nerve injury-induced neuropathic pain. We found that intraperitoneal injection of either isosakuranetin or primidone reduced heat hypersensitivity induced by chronic constriction injury (CCI) of the sciatic nerve in wild-type, but not in TRPM3-/- mice. Primidone was also effective when injected locally in the hindpaw or intrathecally. Consistently, intrathecal injection of the TRPM3 agonist CIM0216 reduced paw withdrawal latency to radiant heat in wild-type, but not in TRPM3-/- mice. Intraperitoneal injection of 2 mg/kg, but not 0.5 mg/kg isosakuranetin, inhibited cold and mechanical hypersensitivity in CCI, both in wild-type and TRPM3-/- mice, indicating a dose-dependent off-target effect. Primidone had no effect on cold sensitivity, and only a marginal effect on mechanical hypersensitivity. Genetic deletion or inhibitors of TRPM3 reduced the increase in the levels of the early genes c-Fos and pERK in the spinal cord and DRGs in CCI mice, suggesting spontaneous activity of the channel. Intraperitoneal isosakuranetin also inhibited spontaneous pain related behavior in CCI in the conditioned place preference assay, and this effect was eliminated in TRPM3-/- mice. Overall, our data indicate a role of TRPM3 in heat hypersensitivity and in spontaneous pain after nerve injury.SIGNIFICANCE STATEMENT Neuropathic pain is a major unsolved medical problem. The heat-activated TRPM3 ion channel is a potential target for novel pain medications, but the pain modalities in which it plays a role are not clear. Here we used a combination of genetic and pharmacological tools to assess the role of this channel in spontaneous pain, heat, cold, and mechanical hypersensitivity in a nerve injury model of neuropathic pain in mice. Our findings indicate a role for TRPM3 in heat hyperalgesia, and spontaneous pain, but not in cold and mechanical hypersensitivity. We also find that not only TRPM3 located in the peripheral nerve termini, but also TRPM3 in the spinal cord or proximal segments of DRG neurons are important for heat hypersensitivity.


Assuntos
Hiperalgesia/metabolismo , Neuralgia/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Feminino , Temperatura Alta , Hiperalgesia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/etiologia , Traumatismos dos Nervos Periféricos/complicações
2.
EMBO Rep ; 21(5): e49124, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32227462

RESUMO

Mechanically activated Piezo2 channels are key players in somatosensory touch, but their regulation by cellular signaling pathways is poorly understood. Dorsal root ganglion (DRG) neurons express a variety of G-protein-coupled receptors that modulate the function of sensory ion channels. Gi-coupled receptors are generally considered inhibitory, as they usually decrease excitability. Paradoxically, activation of Gi-coupled receptors in DRG neurons sometimes induces mechanical hypersensitivity, the mechanism of which is not well understood. Here, we find that activation of Gi-coupled receptors potentiates mechanically activated currents in DRG neurons and heterologously expressed Piezo2 channels, but inhibits Piezo1 currents in heterologous systems in a Gßγ-dependent manner. Pharmacological inhibition of kinases downstream of Gßγ, phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) also abolishes the potentiation of Piezo2 currents. Local injection of sumatriptan, an agonist of the Gi-coupled serotonin 1B/1D receptors, increases mechanical sensitivity in mice, and the effect is abolished by inhibiting PI3K and MAPK. Hence, our studies illustrate an indirect mechanism of action of Gßγ to sensitize Piezo2 currents and alter mechanosensitivity after activation of Gi-coupled receptors.


Assuntos
Gânglios Espinais , Fosfatidilinositol 3-Quinases , Animais , Gânglios Espinais/metabolismo , Canais Iônicos/genética , Camundongos , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
3.
J Biol Chem ; 295(24): 8174-8185, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32345612

RESUMO

The transient receptor potential vanilloid 1 (TRPV1) channel is activated by heat and by capsaicin, the pungent compound in chili peppers. Calcium influx through TRPV1 has been shown to activate a calcium-sensitive phospholipase C (PLC) enzyme and to lead to a robust decrease in phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] levels, which is a major contributor to channel desensitization. Diacylglycerol (DAG), the product of the PLC-catalyzed PI(4,5)P2 hydrolysis, activates protein kinase C (PKC). PKC is known to potentiate TRPV1 activity during activation of G protein-coupled receptors, but it is not known whether DAG modulates TRPV1 during desensitization. We found here that inhibition of diacylglycerol kinase (DAGK) enzymes reduces desensitization of native TRPV1 in dorsal root ganglion neurons as well as of recombinant TRPV1 expressed in HEK293 cells. The effect of DAGK inhibition was eliminated by mutating two PKC-targeted phosphorylation sites, Ser-502 and Ser-800, indicating involvement of PKC. TRPV1 activation induced only a small and transient increase in DAG levels, unlike the robust and more sustained increase induced by muscarinic receptor activation. DAGK inhibition substantially increased the DAG signal evoked by TRPV1 activation but not that evoked by M1 muscarinic receptor activation. Our results show that Ca2+ influx through TRPV1 activates PLC and DAGK enzymes and that the latter limits formation of DAG and negatively regulates TRPV1 channel activity. Our findings uncover a role of DAGK in ion channel regulation.


Assuntos
Diacilglicerol Quinase/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Capsaicina/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citoplasma/metabolismo , Diacilglicerol Quinase/antagonistas & inibidores , Diglicerídeos/metabolismo , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ritanserina/farmacologia
4.
J Neurosci ; 39(31): 6067-6080, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31127000

RESUMO

The cold- and menthol-sensitive transient receptor potential melastatin 8 (TRPM8) channel is important for both physiological temperature detection and cold allodynia. Activation of G-protein-coupled receptors (GPCRs) by proinflammatory mediators inhibits these channels. It was proposed that this inhibition proceeds via direct binding of Gαq to the channel. TRPM8 requires the plasma membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2 or PIP2] for activity. However, it was claimed that a decrease in cellular levels of this lipid upon receptor activation does not contribute to channel inhibition. Here, we show that supplementing the whole-cell patch pipette with PI(4,5)P2 reduced inhibition of TRPM8 by activation of Gαq-coupled receptors in mouse dorsal root ganglion (DRG) neurons isolated from both sexes. Stimulating the same receptors activated phospholipase C (PLC) and decreased plasma membrane PI(4,5)P2 levels in these neurons. PI(4,5)P2 also reduced inhibition of TRPM8 by activation of heterologously expressed muscarinic M1 receptors. Coexpression of a constitutively active Gαq protein that does not couple to PLC inhibited TRPM8 activity, and in cells expressing this protein, decreasing PI(4,5)P2 levels using a voltage-sensitive 5'-phosphatase induced a stronger inhibition of TRPM8 activity than in control cells. Our data indicate that, upon GPCR activation, Gαq binding reduces the apparent affinity of TRPM8 for PI(4,5)P2 and thus sensitizes the channel to inhibition induced by decreasing PI(4,5)P2 levels.SIGNIFICANCE STATEMENT Increased sensitivity to heat in inflammation is partially mediated by inhibition of the cold- and menthol-sensitive transient receptor potential melastatin 8 (TRPM8) ion channels. Most inflammatory mediators act via G-protein-coupled receptors that activate the phospholipase C pathway, leading to the hydrolysis of phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. How receptor activation by inflammatory mediators leads to TRPM8 inhibition is not well understood. Here, we propose that direct binding of Gαq both reduces TRPM8 activity and sensitizes the channel to inhibition by decreased levels of its cofactor, PI(4,5)P2 Our data demonstrate the convergence of two downstream effectors of receptor activation, Gαq and PI(4,5)P2 hydrolysis, in the regulation of TRPM8.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Feminino , Gânglios Espinais/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Mol Pain ; 14: 1744806918763646, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29580154

RESUMO

Primary sensory neurons in the dorsal root ganglia and trigeminal ganglia are responsible for sensing mechanical and thermal stimuli, as well as detecting tissue damage. These neurons express ion channels that respond to thermal, mechanical, or chemical cues, conduct action potentials, and mediate transmitter release. These neurons also express a large number of G-protein coupled receptors, which are major transducers for extracellular signaling molecules, and their activation usually modulates the primary transduction pathways. Receptors that couple to phospholipase C via heterotrimeric Gq/11 proteins and those that activate adenylate cyclase via Gs are considered excitatory; they positively regulate somatosensory transduction and they play roles in inflammatory sensitization and pain, and in some cases also in inducing itch. On the other hand, receptors that couple to Gi/o proteins, such as opioid or GABAB receptors, are generally inhibitory. Their activation counteracts the effect of Gs-stimulation by inhibiting adenylate cyclase, as well as exerts effects on ion channels, usually resulting in decreased excitability. This review will summarize knowledge on Gi-coupled receptors in sensory neurons, focusing on their roles in ion channel regulation and discuss their potential as targets for analgesic and antipruritic medications.


Assuntos
Analgésicos/uso terapêutico , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Receptoras Sensoriais/metabolismo , Córtex Somatossensorial/citologia , Animais , Humanos , Transdução de Sinais
6.
J Physiol ; 594(13): 3609-28, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27062607

RESUMO

KEY POINTS: The cold- and menthol-activated transient receptor potential melastatin 8 (TRPM8) channels are thought to be regulated by phospholipase C (PLC), but neither the specific PLC isoform nor the in vivo relevance of this regulation has been established. Here we identify PLCδ4 as the key PLC isoform involved in regulation of TRPM8 channels in vivo. We show that in small PLCδ4(-/-) TRPM8-positive dorsal root ganglion neurons cold, menthol and WS-12, a selective TRPM8 agonist, evoked significantly larger currents than in wild-type neurons, and action potential frequencies induced by menthol or by current injections were also higher in PLCδ4(-/-) neurons. PLCδ4(-/-) mice showed increased behavioural responses to evaporative cooling, and this effect was inhibited by a TRPM8 antagonist; behavioural responses to heat and mechanical stimuli were not altered. We provide evidence for the involvement of a specific PLC isoform in the regulation of cold sensitivity in mice by regulating TRPM8 activity. ABSTRACT: The transient receptor potential melastatin 8 (TRPM8) ion channel is a major sensor of environmental low temperatures. Ca(2+) -induced activation of phospholipase C (PLC) has been implied in the regulation of TRPM8 channels during menthol- and cold-induced desensitization in vitro. Here we identify PLCδ4 as the key PLC isoform involved in regulation of TRPM8 in sensory dorsal root ganglion (DRG) neurons. We identified two TRPM8-positive neuronal subpopulations, based on their cell body size. Most TRPM8-positive small neurons also responded to capsaicin, and had significantly larger menthol-induced inward current densities than medium-large cells, most of which did not respond to capsaicin. Small, but not medium-large, PLCδ4(-/-) neurons showed significantly larger currents induced by cold, menthol or WS-12, a specific TRPM8 agonist, compared to wild-type (WT) neurons, but TRPM8 protein levels were not different between the two groups. In current-clamp experiments small neurons had more depolarized resting membrane potentials, and required smaller current injections to generate action potentials (APs) than medium-large cells. In small PLCδ4(-/-) neurons, menthol application induced larger depolarizations and generation of APs with frequencies significantly higher compared to WT neurons. In behavioural experiments PLCδ4(-/-) mice showed greater sensitivity to evaporative cooling by acetone than control animals. Pretreatment with the TRPM8 antagonist PBMC reduced cold-induced responses, and the effect was more pronounced in the PLCδ4(-/-) group. Heat and mechanical sensitivity of the PLCδ4(-/-) mice was not different from WT animals. Our data support the involvement of PLCδ4 in the regulation of TRPM8 channel activity in vivo.


Assuntos
Temperatura Baixa , Fosfolipase C delta/fisiologia , Canais de Cátion TRPM/fisiologia , Animais , Comportamento Animal , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Masculino , Camundongos , Camundongos Knockout , Oócitos , Fosfolipase C delta/genética , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Xenopus laevis
7.
J Neurosci ; 33(28): 11451-63, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23843517

RESUMO

Transient Receptor Potential Vanilloid 1 (TRPV1) is a polymodal, Ca(2+)-permeable cation channel crucial to regulation of nociceptor responsiveness. Sensitization of TRPV1 by G-protein coupled receptor (GPCR) agonists to its endogenous activators, such as low pH and noxious heat, is a key factor in hyperalgesia during tissue injury as well as pathological pain syndromes. Conversely, chronic pharmacological activation of TRPV1 by capsaicin leads to calcium influx-induced adaptation of the channel. Paradoxically, both conditions entail activation of phospholipase C (PLC) enzymes, which hydrolyze phosphoinositides. We found that in sensory neurons PLCß activation by bradykinin led to a moderate decrease in phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2), but no sustained change in the levels of its precursor PI(4)P. Preventing this selective decrease in PI(4,5)P2 inhibited TRPV1 sensitization, while selectively decreasing PI(4,5)P2 independently of PLC potentiated the sensitizing effect of protein kinase C (PKC) on the channel, thereby inducing increased TRPV1 responsiveness. Maximal pharmacological TRPV1 stimulation led to a robust decrease of both PI(4,5)P2 and its precursor PI(4)P in sensory neurons. Attenuating the decrease of either lipid significantly reduced desensitization, and simultaneous reduction of PI(4,5)P2 and PI(4)P independently of PLC inhibited TRPV1. We found that, on the mRNA level, the dominant highly Ca(2+)-sensitive PLC isoform in dorsal root ganglia is PLCδ4. Capsaicin-induced desensitization of TRPV1 currents was significantly reduced, whereas capsaicin-induced nerve impulses in the skin-nerve preparation increased in mice lacking this isoform. We propose a comprehensive model in which differential changes in phosphoinositide levels mediated by distinct PLC isoforms result in opposing changes in TRPV1 activity.


Assuntos
Membrana Celular/metabolismo , Nociceptores/metabolismo , Fosfatidilinositóis/antagonistas & inibidores , Fosfatidilinositóis/metabolismo , Canais de Cátion TRPV/fisiologia , Animais , Capsaicina/farmacologia , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptores/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Xenopus laevis
8.
bioRxiv ; 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38464030

RESUMO

Mechanosensitive PIEZO2 ion channels play roles in touch, proprioception, and inflammatory pain. Currently, there are no small molecule inhibitors that selectively inhibit PIEZO2 over PIEZO1. The TMEM120A protein was shown to inhibit PIEZO2 while leaving PIEZO1 unaffected. Here we find that TMEM120A expression elevates cellular levels of phosphatidic acid and lysophosphatidic acid (LPA), aligning with its structural resemblance to lipid-modifying enzymes. Intracellular application of phosphatidic acid or LPA inhibited PIEZO2, but not PIEZO1 activity. Extended extracellular exposure to the non-hydrolyzable phosphatidic acid and LPA analogue carbocyclic phosphatidic acid (ccPA) also inhibited PIEZO2. Optogenetic activation of phospholipase D (PLD), a signaling enzyme that generates phosphatidic acid, inhibited PIEZO2, but not PIEZO1. Conversely, inhibiting PLD led to increased PIEZO2 activity and increased mechanical sensitivity in mice in behavioral experiments. These findings unveil lipid regulators that selectively target PIEZO2 over PIEZO1, and identify the PLD pathway as a regulator of PIEZO2 activity.

9.
J Gen Physiol ; 154(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35819364

RESUMO

PIEZO2 channels mediate rapidly adapting mechanically activated currents in peripheral sensory neurons of the dorsal root ganglia (DRG), and they are indispensable for light touch and proprioception. Relatively little is known about what other proteins regulate PIEZO2 activity in a cellular context. TMEM120A (TACAN) was proposed to act as a high threshold mechanically activated ion channel in nociceptive DRG neurons. Here, we find that Tmem120a coexpression decreased the amplitudes of mechanically activated PIEZO2 currents and increased their threshold of activation. TMEM120A did not inhibit mechanically activated PIEZO1 and TREK1 channels and TMEM120A alone did not result in the appearance of mechanically activated currents above background. Tmem120a and Piezo2 expression in mouse DRG neurons overlapped, and siRNA-mediated knockdown of Tmem120a increased the amplitudes of rapidly adapting mechanically activated currents and decreased their thresholds to mechanical activation. Our data identify TMEM120A as a negative modulator of PIEZO2 channel activity, and do not support TMEM120A being a mechanically activated ion channel.


Assuntos
Canais Iônicos/metabolismo , Mecanotransdução Celular , Animais , Gânglios Espinais/metabolismo , Mecanotransdução Celular/fisiologia , Camundongos , Nociceptores/metabolismo , Células Receptoras Sensoriais/fisiologia
10.
J Physiol ; 589(Pt 24): 6007-27, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22005680

RESUMO

The activity of the cold- and menthol-activated transient receptor potential melastatin 8 (TRPM8) channels diminishes over time in the presence of extracellular Ca(2+), a phenomenon referred to as desensitization or adaptation. Here we show that activation of TRPM8 by cold or menthol evokes a decrease in cellular phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] levels. The decrease in PtdIns(4,5)P(2) levels was accompanied by increased inositol 1,4,5 trisphosphate (InsP(3)) production, and was inhibited by loading the cells with the Ca(2+) chelator BAPTA-AM, showing that it was the consequence of the activation of phospholipase C (PLC) by increased intracellular Ca(2+) concentrations. PtdIns(4,5)P(2) hydrolysis showed excellent temporal correlation with current desensitization in simultaneous patch clamp and fluorescence-based PtdIns(4,5)P(2) level measurements. Intracellular dialysis of PtdIns(4,5)P(2) inhibited desensitization both in native neuronal and recombinant TRPM8 channels. PtdIns(4)P, the precursor of PtdIns(4,5)P(2), did not inhibit desensitization, consistent with its minimal effect in excised patches. Omission of MgATP from the intracellular solution accelerated desensitization, and MgATP reactivated TRPM8 channels in excised patches in a phosphatidylinositol 4-kinase (PI4K)-dependent manner. PLC-independent depletion of PtdIns(4,5)P(2) using a voltage-sensitive phosphatase (ci-VSP) inhibited TRPM8 currents, and omission of ATP from the intracellular solution inhibited recovery from this inhibition. Inhibitors of PKC had no effect on the kinetics of desensitization. We conclude that Ca(2+) influx through TRPM8 activates a Ca(2+)-sensitive PLC isoform, and the resulting depletion of PtdIns(4,5)P(2) plays a major role in desensitization of both cold and menthol responses.


Assuntos
Adaptação Fisiológica/fisiologia , Temperatura Baixa , Mentol/farmacologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Canais de Cátion TRPM/fisiologia , Trifosfato de Adenosina/fisiologia , Animais , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Células HEK293 , Humanos , Camundongos , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína Quinase C/fisiologia
11.
Methods Enzymol ; 652: 49-79, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34059290

RESUMO

Ion channel are embedded in the lipid bilayers of biological membranes. Membrane phospholipids constitute a barrier to ion movement, and they have been considered for a long time as a passive environment for channel proteins. Membrane phospholipids, however, do not only serve as a passive amphipathic environment, but they also modulate channel activity by direct specific lipid-protein interactions. Phosphoinositides are quantitatively minor components of biological membranes, and they play roles in many cellular functions, including membrane traffic, cellular signaling and cytoskeletal organization. Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is mainly found in the inner leaflet of the plasma membrane. Its role as a potential ion channel regulator was first appreciated over two decades ago and by now this lipid is a well-established cofactor or regulator of many different ion channels. The past two decades witnessed the steady development of techniques to study ion channel regulation by phosphoinositides with progress culminating in recent cryoEM structures that allowed visualization of how PI(4,5)P2 opens some ion channels. This chapter will provide an overview of the methods to study regulation by phosphoinositides, focusing on plasma membrane ion channels and PI(4,5)P2.


Assuntos
Fosfatidilinositol 4,5-Difosfato , Fosfatidilinositóis , Membrana Celular , Canais Iônicos , Bicamadas Lipídicas
12.
Elife ; 92020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32343227

RESUMO

Transient Receptor Potential Melastatin 3 (TRPM3) is a Ca2+ permeable non-selective cation channel activated by heat and chemical agonists such as pregnenolone sulfate and CIM0216. TRPM3 mutations in humans were recently reported to be associated with intellectual disability and epilepsy; the functional effects of those mutations, however, were not reported. Here, we show that both disease-associated mutations in the human TRPM3 render the channel overactive, but likely via different mechanisms. The Val to Met substitution in the S4-S5 loop induced a larger increase in basal activity and agonist sensitivity at room temperature than the Pro to Gln substitution in the extracellular segment of S6. In contrast, heat activation was increased more by the S6 mutant than by the S4-S5 segment mutant. Both mutants were inhibited by the TRPM3 antagonist primidone, suggesting a potential therapeutic intervention to treat this disease.


Inherited brain disorders often cause severe problems for those affected by them. One example is a group of diseases, collectively termed "developmental and epileptic encephalopathies", or DEE for short. People with these diseases usually have both epilepsy and intellectual disabilities, and in some patients these conditions are associated with two mutations that change a gene called TRPM3. The TRPM3 gene encodes a protein called an ion channel. Ion channels form pores on the surfaces of cells. When channels are active, the pores open, allowing charged particles ­ which, in the case of TRPM3, are sodium and calcium ions ­ to pass through, carrying tiny electrical currents. In the nervous system, ion channels help nerve cells communicate and also allow them to sense changes in the environment. The TRPM3 channel is known to open in response to heat and certain chemical "activators". In mice, TRPM3 is found in sensory nerve cells, where it acts as a heat sensor. Although altering TRPM3 in mice affects their ability to sense intense or painful heat stimuli, they are otherwise completely normal and have no symptoms resembling human DEE disorders. Although TRPM3 is found in the human brain, little is known about its role there or what effects the DEE-associated mutations have on its activity. Zhao et al. therefore set out to determine, whether each of the mutation was a 'loss of function', meaning that it stopped the channel from opening, or a 'gain of function', meaning it made the channel open more often. Frog egg cells and mammalian cells grown in the laboratory were engineered to produce the TRPM3 ion channel. Measurements of electrical activity on these cells revealed that the two mutations seen in people with DEE were both 'gain of function'. Both mutants were more sensitive to heat and chemical activators than the normal protein. They were also more active overall, even without any stimuli. However, one mutation had a greater effect on heat sensitivity, while the other caused a larger increase in chemical-induced activity. Imaging experiments revealed that both mutant channels also increased the amount of calcium inside the cells. This could explain why the mutations cause disease, since abnormally high calcium levels can damage nerve cells. In addition, the epilepsy drug primidone switched off the mutant channels, pointing to potential treatment of this disease using primidone.


Assuntos
Pregnenolona/farmacologia , Canais de Cátion TRPM/efeitos dos fármacos , Canais de Cátion TRPM/genética , Cálcio/metabolismo , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Mutação/efeitos dos fármacos , Mutação/genética
13.
Br J Pharmacol ; 176(17): 3110-3125, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31074038

RESUMO

BACKGROUND AND PURPOSE: Opioids remain the most efficient medications against severe pain; they act on receptors that couple to heterotrimeric G-proteins in the Gαi/o family. Opioids exert many of their acute effects through modulating ion channels via Gßγ subunits. Many of their side effects are attributed to ß-arrestin recruitment. Several biased agonists that do not recruit ß-arrestins, but activate G-protein-dependent pathways, have recently been developed. While these compounds have been proposed to be full agonists of G-protein signalling in several high throughput pharmacological assays, their effects were not studied on ion channel targets. EXPERIMENTAL APPROACH: Here, we used patch-clamp electrophysiology and Ca2+ imaging to test the effects of TRV130, PZM21, and herkinorin, three G-protein-biased agonists of µ-opioid receptors, on ion channel targets of Gαi/o /Gßγ signalling. We also studied G-protein dissociation using a FRET-based assay. KEY RESULTS: All three biased agonists induced smaller activation of G-protein-coupled inwardly rectifying K+ channels (Kir 3.2) and smaller inhibition of transient receptor potential melastatin (TRPM3) channels than the full µ receptor agonist DAMGO. Co-application of TRV130 or PZM21, but not herkinorin, alleviated the effects of DAMGO on both channels. PZM21 and TRV130 also decreased the effect of morphine on Kir 3.2 channels. The CaV 2.2 channel was also inhibited less by PZM21 and TRV130 than by DAMGO. We also found that TRV130, PZM21, and herkinorin were less effective than DAMGO at inducing dissociation of the Gαi /Gßγ complex. CONCLUSION AND IMPLICATIONS: TRV130, PZM21, and potentially herkinorin are partial agonists of µ receptors.


Assuntos
Canais Iônicos/antagonistas & inibidores , Receptores Opioides mu/agonistas , Transdução de Sinais/efeitos dos fármacos , Compostos de Espiro/farmacologia , Tiofenos/farmacologia , Ureia/análogos & derivados , Animais , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Canais Iônicos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores Opioides mu/metabolismo , Compostos de Espiro/química , Relação Estrutura-Atividade , Tiofenos/química , Ureia/química , Ureia/farmacologia
14.
Elife ; 82019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31647410

RESUMO

Transient receptor potential vanilloid 5 (TRPV5) is a highly calcium selective ion channel that acts as the rate-limiting step of calcium reabsorption in the kidney. The lack of potent, specific modulators of TRPV5 has limited the ability to probe the contribution of TRPV5 in disease phenotypes such as hypercalcemia and nephrolithiasis. Here, we performed structure-based virtual screening (SBVS) at a previously identified TRPV5 inhibitor binding site coupled with electrophysiology screening and identified three novel inhibitors of TRPV5, one of which exhibits high affinity, and specificity for TRPV5 over other TRP channels, including its close homologue TRPV6. Cryo-electron microscopy of TRPV5 in the presence of the specific inhibitor and its parent compound revealed novel binding sites for this channel. Structural and functional analysis have allowed us to suggest a mechanism of action for the selective inhibition of TRPV5 and lay the groundwork for rational design of new classes of TRPV5 modulators.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/química , Sítios de Ligação , Microscopia Crioeletrônica , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Conformação Proteica
15.
Elife ; 82019 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-31566564

RESUMO

Transient receptor potential vanilloid 2 (TRPV2) plays a critical role in neuronal development, cardiac function, immunity, and cancer. Cannabidiol (CBD), the non-psychotropic therapeutically active ingredient of Cannabis sativa, is an activator of TRPV2 and also modulates other transient receptor potential (TRP) channels. Here, we determined structures of the full-length rat TRPV2 channel in apo and CBD-bound states in nanodiscs by cryo-electron microscopy. We show that CBD interacts with TRPV2 through a hydrophobic pocket located between S5 and S6 helices of adjacent subunits, which differs from known ligand and lipid binding sites in other TRP channels. CBD-bound TRPV2 structures revealed that the S4-S5 linker plays a critical role in channel gating upon CBD binding. Additionally, nanodiscs permitted us to visualize two distinct TRPV2 apo states in a lipid environment. Together these results provide a foundation to further understand TRPV channel gating, their divergent physiological functions, and to accelerate structure-based drug design.


Assuntos
Canabidiol/farmacologia , Lipídeos/química , Canais de Cátion TRPV/genética , Animais , Sítios de Ligação/efeitos dos fármacos , Cannabis/química , Microscopia Crioeletrônica , Humanos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Ligantes , Mutação/genética , Conformação Proteica em alfa-Hélice/efeitos dos fármacos , Estrutura Secundária de Proteína , Ratos , Canais de Cátion TRPV/ultraestrutura
16.
J Gen Physiol ; 150(11): 1554-1566, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30333107

RESUMO

The transient receptor potential channel vanilloid type 1 (TRPV1) is activated by a variety of endogenous and exogenous stimuli and is involved in nociception and body temperature regulation. Although the structure of TRPV1 has been experimentally determined in both the closed and open states, very little is known about its activation mechanism. In particular, the conformational changes that occur in the pore domain and result in ionic conduction have not yet been identified. Here we suggest a hypothetical molecular mechanism for TRPV1 activation, which involves rotation of a conserved asparagine in S6 from a position facing the S4-S5 linker toward the pore. This rotation is associated with hydration of the pore and dehydration of the four peripheral cavities located between each S6 and S4-S5 linker. In light of our hypothesis, we perform bioinformatics analyses of TRP and other evolutionary related ion channels, evaluate newly available structures, and reexamine previously reported water accessibility and mutagenesis experiments. These analyses provide several independent lines of evidence to support our hypothesis. Finally, we show that our proposed molecular mechanism is compatible with the prevailing theory that the selectivity filter acts as a secondary gate in TRPV1.


Assuntos
Canais de Cátion TRPV/metabolismo , Asparagina , Simulação de Dinâmica Molecular , Conformação Proteica , Rotação
17.
J Phys Chem Lett ; 9(6): 1260-1264, 2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29439562

RESUMO

The nonselective cation channel TRPV1 is responsible for transducing noxious stimuli into action potentials propagating through peripheral nerves. It is activated by temperatures greater than 43 °C, while remaining completely nonconductive at temperatures lower than this threshold. The origin of this sharp response, which makes TRPV1 a biological temperature sensor, is not understood. Here we used molecular dynamics simulations and free energy calculations to characterize the molecular determinants of the transition between nonconductive and conductive states. We found that hydration of the pore and thus ion permeation depends critically on the polar character of its molecular surface: in this narrow hydrophobic enclosure, the motion of a polar side-chain is sufficient to stabilize either the dry or wet state. The conformation of this side-chain is in turn coupled to the hydration state of four peripheral cavities, which undergo a dewetting transition at the activation temperature.


Assuntos
Simulação de Dinâmica Molecular , Canais de Cátion TRPV/química , Canais de Cátion TRPV/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Movimento , Porosidade , Conformação Proteica , Termodinâmica
18.
Elife ; 62017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28829742

RESUMO

Transient receptor potential melastatin 3 (TRPM3) channels are activated by heat, and chemical ligands such as pregnenolone sulphate (PregS) and CIM0216. Here, we show that activation of receptors coupled to heterotrimeric Gi/o proteins inhibits TRPM3 channels. This inhibition was alleviated by co-expression of proteins that bind the ßγ subunits of heterotrimeric G-proteins (Gßγ). Co-expression of Gßγ, but not constitutively active Gαi or Gαo, inhibited TRPM3 currents. TRPM3 co-immunoprecipitated with Gß, and purified Gßγ proteins applied to excised inside-out patches inhibited TRPM3 currents, indicating a direct effect. Baclofen and somatostatin, agonists of Gi-coupled receptors, inhibited Ca2+ signals induced by PregS and CIM0216 in mouse dorsal root ganglion (DRG) neurons. The GABAB receptor agonist baclofen also inhibited inward currents induced by CIM0216 in DRG neurons, and nocifensive responses elicited by this TRPM3 agonist in mice. Our data uncover a novel signaling mechanism regulating TRPM3 channels.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/farmacologia , Subunidades gama da Proteína de Ligação ao GTP/farmacologia , Canais de Cátion TRPM/efeitos dos fármacos , Animais , Baclofeno/antagonistas & inibidores , Escala de Avaliação Comportamental , Cálcio/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/farmacologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pregnenolona/farmacologia , Somatostatina/antagonistas & inibidores
19.
Neurosci Res ; 55(4): 421-5, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16730827

RESUMO

RFa-related peptides play a significant role in the processing of pain in the CNS of mammals. Recently it has been found that, when applied subcutaneously, these peptides elicit a powerful algogenic effect. The question arises whether this peripheral effect can be connected with the ability of RFa-related peptides to decrease the rate of desensitization of acid sensing ionic channels (ASICs) expressed in primary sensory neurons. We have addressed this question by comparing the effects of neuropeptide SF (NPSF), mammalian RFa peptide, in ASIC3-/- and wild-type C57BL/6J mice. Knockout of ASIC3 gene results in the changes in some of the behavioral parameters. However, subcutaneous injections of the NPSF into the n.saphenous innervation area result in a clearly nociceptive behavior in both strains of mice. There is no significant difference in the total time of licking of injected paw in the ASIC3-/- (194+/-22s) and C57BL/6J (227+/-25s) animals. Thus peripheral algogenic effects of NPSF cannot be explained only in terms of their action on the ASIC3 channels and involves some other, still unidentified mechanism.


Assuntos
Neuropeptídeos/farmacologia , Nociceptores/fisiologia , Dor/metabolismo , Células Receptoras Sensoriais/fisiologia , Canais de Sódio/genética , Canais Iônicos Sensíveis a Ácido , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Pé/inervação , Pé/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptores/efeitos dos fármacos , Dor/induzido quimicamente , Dor/genética , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Especificidade da Espécie
20.
Cell Rep ; 4(2): 302-315, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23850286

RESUMO

The TRPM8 ion channel is expressed in sensory neurons and is responsible for sensing environmental cues, such as cold temperatures and chemical compounds, including menthol and icilin. The channel functional activity is regulated by various physical and chemical factors and is likely to be preconditioned by its molecular composition. Our studies indicate that the TRPM8 channel forms a structural-functional complex with the polyester poly-(R)-3-hydroxybutyrate (PHB). We identified by mass spectrometry a number of PHB-modified peptides in the N terminus of the TRPM8 protein and in its extracellular S3-S4 linker. Removal of PHB by enzymatic hydrolysis and site-directed mutagenesis of both the serine residues that serve as covalent anchors for PHB and adjacent hydrophobic residues that interact with the methyl groups of the polymer resulted in significant inhibition of TRPM8 channel activity. We conclude that the TRPM8 channel undergoes posttranslational modification by PHB and that this modification is required for its normal function.


Assuntos
Hidroxibutiratos/metabolismo , Poliésteres/metabolismo , Canais de Cátion TRPM/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Técnicas de Cultura de Células , Células HEK293 , Humanos , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Proibitinas , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Relação Estrutura-Atividade , Canais de Cátion TRPM/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA