Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Pain ; 165(2): 470-486, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37733484

ABSTRACT

ABSTRACT: Lipid-rich diet is the major cause of obesity, affecting 13% of the worldwide adult population. Obesity is a major risk factor for metabolic syndrome that includes hyperlipidemia and diabetes mellitus. The early phases of metabolic syndrome are often associated with hyperexcitability of peripheral small diameter sensory fibers and painful diabetic neuropathy. Here, we investigated the effect of high-fat diet-induced obesity on the activity of dorsal root ganglion (DRG) sensory neurons and pain perception. We deciphered the underlying cellular mechanisms involving the acid-sensing ion channel 3 (ASIC3). We show that mice made obese through consuming high-fat diet developed the metabolic syndrome and prediabetes that was associated with heat pain hypersensitivity, whereas mechanical sensitivity was not affected. Concurrently, the slow conducting C fibers in the skin of obese mice showed increased activity on heating, whereas their mechanosensitivity was not altered. Although ASIC3 knockout mice fed with high-fat diet became obese, and showed signs of metabolic syndrome and prediabetes, genetic deletion, and in vivo pharmacological inhibition of ASIC3, protected mice from obesity-induced thermal hypersensitivity. We then deciphered the mechanisms involved in the heat hypersensitivity of mice and found that serum from high-fat diet-fed mice was enriched in lysophosphatidylcholine (LPC16:0, LPC18:0, and LPC18:1). These enriched lipid species directly increased the activity of DRG neurons through activating the lipid sensitive ASIC3 channel. Our results identify ASIC3 channel in DRG neurons and circulating lipid species as a mechanism contributing to the hyperexcitability of nociceptive neurons that can cause pain associated with lipid-rich diet consumption and obesity.


Subject(s)
Metabolic Syndrome , Prediabetic State , Animals , Mice , Acid Sensing Ion Channels/metabolism , Diet, High-Fat/adverse effects , Ganglia, Spinal/metabolism , Lipids , Metabolic Syndrome/metabolism , Obesity , Pain , Prediabetic State/metabolism , Sensory Receptor Cells/metabolism
2.
PLoS Comput Biol ; 19(4): e1010993, 2023 04.
Article in English | MEDLINE | ID: mdl-37068087

ABSTRACT

Dorsal horn of the spinal cord is an important crossroad of pain neuraxis, especially for the neuronal plasticity mechanisms that can lead to chronic pain states. Windup is a well-known spinal pain facilitation process initially described several decades ago, but its exact mechanism is still not fully understood. Here, we combine both ex vivo and in vivo electrophysiological recordings of rat spinal neurons with computational modeling to demonstrate a role for ASIC1a-containing channels in the windup process. Spinal application of the ASIC1a inhibitory venom peptides mambalgin-1 and psalmotoxin-1 (PcTx1) significantly reduces the ability of deep wide dynamic range (WDR) neurons to develop windup in vivo. All deep WDR-like neurons recorded from spinal slices exhibit an ASIC current with biophysical and pharmacological characteristics consistent with functional expression of ASIC1a homomeric channels. A computational model of WDR neuron supplemented with different ASIC1a channel parameters accurately reproduces the experimental data, further supporting a positive contribution of these channels to windup. It also predicts a calcium-dependent windup decrease for elevated ASIC conductances, a phenomenon that was experimentally validated using the Texas coral snake ASIC-activating toxin (MitTx) and calcium-activated potassium channel inhibitory peptides (apamin and iberiotoxin). This study supports a dual contribution to windup of calcium permeable ASIC1a channels in deep laminae projecting neurons, promoting it upon moderate channel activity, but ultimately leading to calcium-dependent windup inhibition associated to potassium channels when activity increases.


Subject(s)
Calcium , Pain , Animals , Rats , Calcium/metabolism , Computer Simulation , Neurons/physiology , Peptides , Apamin/metabolism
3.
Biomolecules ; 13(2)2023 02 09.
Article in English | MEDLINE | ID: mdl-36830710

ABSTRACT

Lipids, especially lysophosphatidylcholine LPC16:0, have been shown to be involved in chronic joint pain through the activation of acid-sensing ion channels (ASIC3). The aim of the present study was to investigate the lipid contents of the synovial fluids from controls and patients suffering from chronic joint pain in order to identify characteristic lipid signatures associated with specific joint diseases. For this purpose, lipids were extracted from the synovial fluids and analyzed by mass spectrometry. Lipidomic analyses identified certain choline-containing lipid classes and molecular species as biomarkers of chronic joint pain, regardless of the pathology, with significantly higher levels detected in the patient samples. Moreover, correlations were observed between certain lipid levels and the type of joint pathologies. Interestingly, LPC16:0 levels appeared to correlate with the metabolic status of patients while other choline-containing lipids were more specifically associated with the inflammatory state. Overall, these data point at selective lipid species in synovial fluid as being strong predictors of specific joint pathologies which could help in the selection of the most adapted treatment.


Subject(s)
Joint Diseases , Humans , Joint Diseases/metabolism , Synovial Fluid/chemistry , Lipids/analysis , Biomarkers/metabolism , Arthralgia/metabolism
4.
Toxins (Basel) ; 14(10)2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36287977

ABSTRACT

Acid-sensing ion channels (ASICs) are voltage-independent H+-gated cation channels largely expressed in the nervous system of rodents and humans. At least six isoforms (ASIC1a, 1b, 2a, 2b, 3 and 4) associate into homotrimers or heterotrimers to form functional channels with highly pH-dependent gating properties. This review provides an update on the pharmacological profiles of animal peptide toxins targeting ASICs, including PcTx1 from tarantula and related spider toxins, APETx2 and APETx-like peptides from sea anemone, and mambalgin from snake, as well as the dimeric protein snake toxin MitTx that have all been instrumental to understanding the structure and the pH-dependent gating of rodent and human cloned ASICs and to study the physiological and pathological roles of native ASICs in vitro and in vivo. ASICs are expressed all along the pain pathways and the pharmacological data clearly support a role for these channels in pain. ASIC-targeting peptide toxins interfere with ASIC gating by complex and pH-dependent mechanisms sometimes leading to opposite effects. However, these dual pH-dependent effects of ASIC-inhibiting toxins (PcTx1, mambalgin and APETx2) are fully compatible with, and even support, their analgesic effects in vivo, both in the central and the peripheral nervous system, as well as potential effects in humans.


Subject(s)
Acid Sensing Ion Channels , Spider Venoms , Animals , Humans , Rodentia/metabolism , Spider Venoms/chemistry , Peptides/chemistry , Analgesics/pharmacology , Pain/drug therapy
5.
Front Mol Neurosci ; 15: 880651, 2022.
Article in English | MEDLINE | ID: mdl-35774865

ABSTRACT

Lysophosphatidyl-choline (LPC), a member of the phospholipid family, is an emerging player in pain. It is known to modulate different pain-related ion channels, including Acid-Sensing Ion Channel 3 (ASIC3), a cationic channel mainly expressed in peripheral sensory neurons. LPC potentiates ASIC3 current evoked by mild acidifications, but can also activate the channel at physiological pH. Very recently, LPC has been associated to chronic pain in patients suffering from fibromyalgia or osteoarthritis. Accordingly, repetitive injections of LPC within mouse muscle or joint generate both persistent pain-like and anxiety-like behaviors in an ASIC3-dependent manner. LPC has also been reported to generate acute pain behaviors when injected intraplantarly in rodents. Here, we explore the mechanism of action of a single cutaneous injection of LPC by studying its effects on spinal dorsal horn neurons. We combine pharmacological, molecular and functional approaches including in vitro patch clamp recordings and in vivo recordings of spinal neuronal activity. We show that a single cutaneous injection of LPC exclusively affects the nociceptive pathway, inducing an ASIC3-dependent sensitization of nociceptive fibers that leads to hyperexcitabilities of both high threshold (HT) and wide dynamic range (WDR) spinal neurons. ASIC3 is involved in LPC-induced increase of WDR neuron's windup as well as in WDR and HT neuron's mechanical hypersensitivity, and it participates, together with TRPV1, to HT neuron's thermal hypersensitivity. The nociceptive input induced by a single LPC cutaneous rather induces short-term sensitization, contrary to previously described injections in muscle and joint. If the effects of peripheral LPC on nociceptive pathways appear to mainly depend on peripheral ASIC3 channels, their consequences on pain may also depend on the tissue injected. Our findings contribute to a better understanding of the nociceptive signaling pathway activated by peripheral LPC via ASIC3 channels, which is an important step regarding the ASIC3-dependent roles of this phospholipid in acute and chronic pain conditions.

6.
Pain ; 163(10): 1999-2013, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35086123

ABSTRACT

ABSTRACT: Rheumatic diseases are often associated to debilitating chronic pain, which remains difficult to treat and requires new therapeutic strategies. We had previously identified lysophosphatidylcholine (LPC) in the synovial fluids from few patients and shown its effect as a positive modulator of acid-sensing ion channel 3 (ASIC3) able to induce acute cutaneous pain in rodents. However, the possible involvement of LPC in chronic joint pain remained completely unknown. Here, we show, from 2 independent cohorts of patients with painful rheumatic diseases, that the synovial fluid levels of LPC are significantly elevated, especially the LPC16:0 species, compared with postmortem control subjects. Moreover, LPC16:0 levels correlated with pain outcomes in a cohort of osteoarthritis patients. However, LPC16:0 do not appear to be the hallmark of a particular joint disease because similar levels are found in the synovial fluids of a second cohort of patients with various rheumatic diseases. The mechanism of action was next explored by developing a pathology-derived rodent model. Intra-articular injections of LPC16:0 is a triggering factor of chronic joint pain in both male and female mice, ultimately leading to persistent pain and anxiety-like behaviors. All these effects are dependent on ASIC3 channels, which drive sufficient peripheral inputs to generate spinal sensitization processes. This study brings evidences from mouse and human supporting a role for LPC16:0 via ASIC3 channels in chronic pain arising from joints, with potential implications for pain management in osteoarthritis and possibly across other rheumatic diseases.


Subject(s)
Acid Sensing Ion Channels , Chronic Pain , Osteoarthritis , Acid Sensing Ion Channels/metabolism , Animals , Arthralgia/etiology , Female , Humans , Lysophosphatidylcholines/toxicity , Male , Mice , Osteoarthritis/complications
7.
Pain ; 163(8): 1542-1559, 2022 08 01.
Article in English | MEDLINE | ID: mdl-34924556

ABSTRACT

ABSTRACT: Several bone conditions, eg, bone cancer, osteoporosis, and rheumatoid arthritis (RA), are associated with a risk of developing persistent pain. Increased osteoclast activity is often the hallmark of these bony pathologies and not only leads to bone remodeling but is also a source of pronociceptive factors that sensitize the bone-innervating nociceptors. Although historically bone loss in RA has been believed to be a consequence of inflammation, both bone erosion and pain can occur years before the symptom onset. Here, we have addressed the disconnection between inflammation, pain, and bone erosion by using a combination of 2 monoclonal antibodies isolated from B cells of patients with RA. We have found that mice injected with B02/B09 monoclonal antibodies (mAbs) developed a long-lasting mechanical hypersensitivity that was accompanied by bone erosion in the absence of joint edema or synovitis. Intriguingly, we have noted a lack of analgesic effect of naproxen and a moderate elevation of few inflammatory factors in the ankle joints suggesting that B02/B09-induced pain-like behavior does not depend on inflammatory processes. By contrast, we found that inhibiting osteoclast activity and acid-sensing ion channel 3 signaling prevented the development of B02/B09-mediated mechanical hypersensitivity. Moreover, we have identified secretory phospholipase A2 and lysophosphatidylcholine 16:0 as critical components of B02/B09-induced pain-like behavior and shown that treatment with a secretory phospholipase A2 inhibitor reversed B02/B09-induced mechanical hypersensitivity and bone erosion. Taken together, our study suggests a potential link between bone erosion and pain in a state of subclinical inflammation and offers a step forward in understanding the mechanisms of bone pain in diseases such as RA.


Subject(s)
Acid Sensing Ion Channels , Arthritis, Rheumatoid , Osteoclasts , Pain , Acid Sensing Ion Channels/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/drug therapy , Inflammation/complications , Mice , Osteoclasts/pathology , Pain/pathology
8.
Nat Commun ; 12(1): 1557, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692361

ABSTRACT

Fragile X syndrome (FXS) is the most frequent form of inherited intellectual disability and the best-described monogenic cause of autism. CGG-repeat expansion in the FMR1 gene leads to FMR1 silencing, loss-of-expression of the Fragile X Mental Retardation Protein (FMRP), and is a common cause of FXS. Missense mutations in the FMR1 gene were also identified in FXS patients, including the recurrent FMRP-R138Q mutation. To investigate the mechanisms underlying FXS caused by this mutation, we generated a knock-in mouse model (Fmr1R138Q) expressing the FMRP-R138Q protein. We demonstrate that, in the hippocampus of the Fmr1R138Q mice, neurons show an increased spine density associated with synaptic ultrastructural defects and increased AMPA receptor-surface expression. Combining biochemical assays, high-resolution imaging, electrophysiological recordings, and behavioural testing, we also show that the R138Q mutation results in impaired hippocampal long-term potentiation and socio-cognitive deficits in mice. These findings reveal the functional impact of the FMRP-R138Q mutation in a mouse model of FXS.


Subject(s)
Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Fragile X Mental Retardation Protein/metabolism , Mutation, Missense/physiology , Receptors, Glutamate/metabolism , Animals , Biotinylation , Brain/metabolism , Brain/physiopathology , Cells, Cultured , Cognitive Dysfunction/metabolism , Female , Fragile X Mental Retardation Protein/genetics , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Immunoblotting , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Male , Mice , Mutation, Missense/genetics , Patch-Clamp Techniques , Receptors, Glutamate/genetics
9.
Nat Commun ; 9(1): 757, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29472612

ABSTRACT

Fragile X syndrome (FXS) is the most frequent inherited cause of intellectual disability and the best-studied monogenic cause of autism. FXS results from the functional absence of the fragile X mental retardation protein (FMRP) leading to abnormal pruning and consequently to synaptic communication defects. Here we show that FMRP is a substrate of the small ubiquitin-like modifier (SUMO) pathway in the brain and identify its active SUMO sites. We unravel the functional consequences of FMRP sumoylation in neurons by combining molecular replacement strategy, biochemical reconstitution assays with advanced live-cell imaging. We first demonstrate that FMRP sumoylation is promoted by activation of metabotropic glutamate receptors. We then show that this increase in sumoylation controls the homomerization of FMRP within dendritic mRNA granules which, in turn, regulates spine elimination and maturation. Altogether, our findings reveal the sumoylation of FMRP as a critical activity-dependent regulatory mechanism of FMRP-mediated neuronal function.


Subject(s)
Dendritic Spines/metabolism , Fragile X Mental Retardation Protein/metabolism , Sumoylation , Amino Acid Sequence , Animals , Cells, Cultured , Dendritic Spines/genetics , Dendritic Spines/pathology , Female , Fragile X Mental Retardation Protein/chemistry , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Models, Neurological , Phenotype , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Secretory Vesicles/metabolism , Sequence Homology, Amino Acid
10.
Front Neurosci ; 10: 542, 2016.
Article in English | MEDLINE | ID: mdl-27932946

ABSTRACT

The neuropeptide neurotensin (NT) elicits numerous pharmacological effects through three different receptors (NTSR1, NTSR2, and NTSR3 also called sortilin). Pharmacological approaches and generation of NTSR1 and NTSR2-deficient mice allowed to determine the NT-induced antipsychotic like behavior, the inhibitory of weak fear memory and the nociceptive signaling in a rat formalin tonic pain model to NTSR1. Conversely, the effects of NT on thermal and tonic nociceptions were mediated by NTSR2. However, the role of NTSR3/sortilin on the neurotensinergic system was not investigated. Here, by using C57Bl/6J mouse model in which the gene coding for NTSR3/sortilin has been inactivated, we observed a modification of the expression of both NTSR2 and NT itself. Quantitative PCR and protein expression using Western blot analyses and AlphaLisa™ technology resulted in the observation that brain NTSR2 as well as brain and blood NT were 2-fold increased in KO mice leading to a resistance of these mice to thermal and chemical pain. These data confirm that NTSR3/sortilin interacts with other NT receptors (i.e., NTSR2) and that its deletion modifies also the affinity of this receptor to NT.

11.
EMBO J ; 35(4): 414-28, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26772186

ABSTRACT

Extracellular pH variations are seen as the principal endogenous signal that triggers activation of Acid-Sensing Ion Channels (ASICs), which are basically considered as proton sensors, and are involved in various processes associated with tissue acidification. Here, we show that human painful inflammatory exudates, displaying non-acidic pH, induce a slow constitutive activation of human ASIC3 channels. This effect is largely driven by lipids, and we identify lysophosphatidylcholine (LPC) and arachidonic acid (AA) as endogenous activators of ASIC3 in the absence of any extracellular acidification. The combination of LPC and AA evokes robust depolarizing current in DRG neurons at physiological pH 7.4, increases nociceptive C-fiber firing, and induces pain behavior in rats, effects that are all prevented by ASIC3 blockers. Lipid-induced pain is also significantly reduced in ASIC3 knockout mice. These findings open new perspectives on the roles of ASIC3 in the absence of tissue pH variation, as well as on the contribution of those channels to lipid-mediated signaling.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Arachidonic Acid/metabolism , Lysophosphatidylcholines/metabolism , Nociceptors/physiology , Animals , Cell Line , Ganglia, Spinal/cytology , Humans , Mice, Knockout , Pain , Rats
12.
Neuropharmacology ; 94: 49-57, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25724084

ABSTRACT

Since their molecular cloning in the late 90's, Acid-Sensing Ion Channels (ASICs) have been shown to be involved in many aspects of nociception, both in peripheral and central neurons. In rodents, the combination of specific or non-specific pharmacological modulators of ASICs, together with in vivo knockdown and/or knockout animals has revealed their contribution to the detection, the modulation and the sensitization of the pain message by primary and secondary sensory neurons. Functional ASICs are homo or heterotrimers of different homologous subunits (ASIC1-3). Channels containing ASIC3 or ASIC1 subunits, appear to be important in peripheral nociceptors, where they are subject to intense regulation, while ASIC1a-containing channels also have a prominent role in central neurons, including spinal cord neurons that modulate and transmit the pain signal to the brain. In humans, experiments performed in healthy volunteers using drugs already used in the clinic and acting as poorly-selective inhibitors of ASICs, together with recent in vitro data obtained from stem cell-derived sensory neurons both support a role for these channels in nociception. These data thus suggest a real translational potential in the development of inhibitory strategies of ASICs for the treatment of pain. This article is part of the Special Issue entitled 'Acid-Sensing Ion Channels in the Nervous System'.


Subject(s)
Acid Sensing Ion Channels/metabolism , Central Nervous System/metabolism , Nociception/physiology , Peripheral Nervous System/metabolism , Animals , Humans
13.
Nat Commun ; 5: 5113, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25311713

ABSTRACT

Sumoylation plays important roles in the modulation of protein function, neurotransmission and plasticity, but the mechanisms regulating this post-translational system in neurons remain largely unknown. Here we demonstrate that the synaptic diffusion of Ubc9, the sole conjugating enzyme of the sumoylation pathway, is regulated by synaptic activity. We use restricted photobleaching/photoconversion of individual hippocampal spines to measure the diffusion properties of Ubc9 and show that it is regulated through an mGlu5R-dependent signalling pathway. Increasing synaptic activity with a GABAA receptor antagonist or directly activating mGlu5R increases the synaptic residency time of Ubc9 via a Gαq/PLC/Ca(2+)/PKC cascade. This activation promotes a transient synaptic trapping of Ubc9 through a PKC phosphorylation-dependent increase of Ubc9 recognition to phosphorylated substrates and consequently leads to the modulation of synaptic sumoylation. Our data demonstrate that Ubc9 diffusion is subject to activity-dependent regulatory processes and provide a mechanism for the dynamic changes in sumoylation occurring during synaptic transmission.


Subject(s)
Neurons/metabolism , Protein Kinase C/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Synapses/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/enzymology , Hippocampus/metabolism , Mice , Neurons/enzymology , Protein Kinase C/genetics , Receptor, Metabotropic Glutamate 5/genetics , Sumoylation , Synapses/enzymology , Synapses/genetics , Synaptic Transmission , Ubiquitin-Conjugating Enzymes/genetics
14.
Nat Commun ; 4: 2941, 2013.
Article in English | MEDLINE | ID: mdl-24346231

ABSTRACT

Morphine is the gold-standard pain reliever for severe acute or chronic pain but it also produces adverse side effects that can alter the quality of life of patients and, in some rare cases, jeopardize the vital prognosis. Morphine elicits both therapeutic and adverse effects primarily through the same µ opioid receptor subtype, which makes it difficult to separate the two types of effects. Here we show that beneficial and deleterious effects of morphine are mediated through different signalling pathways downstream from µ opioid receptor. We demonstrate that the TREK-1 K(+) channel is a crucial contributor of morphine-induced analgesia in mice, while it is not involved in morphine-induced constipation, respiratory depression and dependence-three main adverse effects of opioid analgesic therapy. These observations suggest that direct activation of the TREK-1 K(+) channel, acting downstream from the µ opioid receptor, might have strong analgesic effects without opioid-like adverse effects.


Subject(s)
Analgesia/methods , Morphine/adverse effects , Morphine/therapeutic use , Potassium Channels, Tandem Pore Domain/metabolism , Analgesics, Opioid/adverse effects , Analgesics, Opioid/therapeutic use , Animals , COS Cells , Chlorocebus aethiops , Constipation , Crosses, Genetic , Dose-Response Relationship, Drug , Drug Tolerance , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Naloxone/chemistry , Pain/drug therapy , Pain Management , Receptors, Opioid, mu/metabolism , Respiratory Insufficiency , Signal Transduction , Time Factors
15.
Toxicon ; 75: 187-204, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23624383

ABSTRACT

Acid-sensing ion channels (ASICs) are voltage-independent proton-gated cation channels that are largely expressed in the nervous system as well as in some non-neuronal tissues. In rodents, six different isoforms (ASIC1a, 1b, 2a, 2b, 3 and 4) can associate into homo- or hetero-trimers to form a functional channel. Specific polypeptide toxins targeting ASIC channels have been isolated from the venoms of spider (PcTx1), sea anemone (APETx2) and snakes (MitTx and mambalgins). They exhibit different and sometimes partially overlapping pharmacological profiles and are usually blockers of ASIC channels, except for MitTx, which is a potent activator. This review focuses on the use of these toxins to explore the structure-function relationships, the physiological and the pathophysiological roles of ASIC channels, illustrating at the same time the therapeutic potential of some of these natural compounds.


Subject(s)
Acid Sensing Ion Channels/physiology , Toxins, Biological/pharmacology , Venoms/chemistry , Amino Acid Sequence , Amygdala/drug effects , Amygdala/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Peptides/chemistry , Protein Conformation , Rodentia , Sea Anemones , Snakes , Spiders , Structure-Activity Relationship , Toxins, Biological/isolation & purification , Vasoconstriction/drug effects
16.
Proc Natl Acad Sci U S A ; 109(32): 13124-9, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22829666

ABSTRACT

In rodent sensory neurons, acid-sensing ion channel 3 (ASIC3) has recently emerged as a particularly important sensor of nonadaptive pain associated with tissue acidosis. However, little is known about the human ASIC3 channel, which includes three splice variants differing in their C-terminal domain (hASIC3a, hASIC3b, and hASIC3c). hASIC3a transcripts represent the main mRNAs expressed in both peripheral and central neuronal tissues (dorsal root ganglia [DRG], spinal cord, and brain), where a small proportion of hASIC3c transcripts is also detected. We show that hASIC3 channels (hASIC3a, hASIC3b, or hASIC3c) are able to directly sense extracellular pH changes not only during acidification (up to pH 5.0), but also during alkalization (up to pH 8.0), an original and inducible property yet unknown. When the external pH decreases, hASIC3 display a transient acid mode with brief activation that is relevant to the classical ASIC currents, as previously described. On the other hand, an external pH increase activates a sustained alkaline mode leading to a constitutive activity at resting pH. Both modes are inhibited by the APETx2 toxin, an ASIC3-type channel inhibitor. The alkaline sensitivity of hASIC3 is an intrinsic property of the channel, which is supported by the extracellular loop and involves two arginines (R68 and R83) only present in the human clone. hASIC3 is thus able to sense the extracellular pH in both directions and therefore to dynamically adapt its activity between pH 5.0 and 8.0, a property likely to participate in the fine tuning of neuronal membrane potential and to neuron sensitization in various pH environments.


Subject(s)
Extracellular Fluid/chemistry , Membrane Potentials/physiology , Neurons/physiology , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , COS Cells , Chlorocebus aethiops , Fluorescence , Humans , Hydrogen-Ion Concentration , Neurons/metabolism , Patch-Clamp Techniques
17.
J Neurosci ; 31(16): 6059-66, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21508231

ABSTRACT

Iatrogenic pain consecutive to a large number of surgical procedures has become a growing health concern. The etiology and pathophysiology of postoperative pain are still poorly understood, but hydrogen ions appear to be important in this process. We have investigated the role of peripheral acid-sensing ion channels (ASICs), which form depolarizing channels activated by extracellular protons, in a rat model of postoperative pain (i.e., hindpaw skin/muscle incision). We report high levels of ASIC-type currents (∼ 77%) in sensory neurons innervating the hindpaw muscles, with a prevalence of ASIC3-like currents. The ASIC3 protein is largely expressed in lumbar DRG neurons innervating the plantar muscle, and its mRNA and protein levels are increased by plantar incision 24 h after surgery. Pharmacological inhibition of ASIC3 channels with the specific toxin APETx2 or in vivo knockdown of ASIC3 subunit by small interfering RNA led to a significant reduction of postoperative spontaneous, thermal, and postural pain behaviors (spontaneous flinching, heat hyperalgesia, and weight bearing). ASIC3 appears to have an important role in deep tissue but also affects prolonged pain evoked by skin incision alone. The specific homomeric ASIC1a blocker PcTx1 has no effect on spontaneous flinching, when applied peripherally. Together, these data demonstrate a significant role for peripheral ASIC3-containing channels in postoperative pain.


Subject(s)
Hyperalgesia/metabolism , Nerve Tissue Proteins/metabolism , Pain, Postoperative/metabolism , Sensory Receptor Cells/physiology , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Electrophysiology , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Hindlimb/innervation , Hindlimb/metabolism , Hyperalgesia/physiopathology , Male , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Pain Measurement , Pain, Postoperative/physiopathology , RNA, Small Interfering , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
18.
Pharmacol Ther ; 128(3): 549-58, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20807551

ABSTRACT

Tissue acidosis is a common feature of many painful conditions. Protons are indeed among the first factors released by injured tissues, inducing a local pH fall that depolarizes peripheral free terminals of nociceptors and leads to pain. ASICs are excitatory cation channels directly gated by extracellular protons that are expressed in the nervous system. In sensory neurons, they act as "chemo-electrical" transducers and are involved in somatic and visceral nociception. Two highly specific inhibitory peptides isolated from animal venoms have considerably helped in the understanding of the physiological roles of these channels in pain. At the peripheral level, ASIC3 is important for inflammatory pain. Its expression and its activity are potentiated by several pain mediators present in the "inflammatory soup" that sensitize nociceptors. ASICs have also been involved in some aspects of mechanosensation and mechanonociception, notably in the gastrointestinal tract, but the underlying mechanisms remain to be determined. At the central level, ASIC1a is largely expressed in spinal cord neurons where it has been proposed to participate in the processing of noxious stimuli and in central sensitization. Blocking ASIC1a in the spinal cord also produces a potent analgesia in a broad range of pain conditions through activation of the opiate system. Targeting ASIC channels at different levels of the nervous system could therefore be an interesting strategy for the relief of pain.


Subject(s)
Nerve Tissue Proteins/metabolism , Nociceptors/physiology , Pain/drug therapy , Pain/physiopathology , Sensory Receptor Cells/physiology , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Hydrogen-Ion Concentration , Inflammation/metabolism , Inflammation/physiopathology , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nociceptors/metabolism , Pain/metabolism , Sensory Receptor Cells/metabolism , Sodium Channel Agonists , Sodium Channels/genetics
19.
PLoS Biol ; 8(4): e1000355, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20405001

ABSTRACT

Current antidepressant treatments are inadequate for many individuals, and when they are effective, they require several weeks of administration before a therapeutic effect can be observed. Improving the treatment of depression is challenging. Recently, the two-pore domain potassium channel TREK-1 has been identified as a new target in depression, and its antagonists might become effective antidepressants. In mice, deletion of the TREK-1 gene results in a depression-resistant phenotype that mimics antidepressant treatments. Here, we validate in mice the antidepressant effects of spadin, a secreted peptide derived from the propeptide generated by the maturation of the neurotensin receptor 3 (NTSR3/Sortilin) and acting through TREK-1 inhibition. NTSR3/Sortilin interacted with the TREK-1 channel, as shown by immunoprecipitation of TREK-1 and NTSR3/Sortilin from COS-7 cells and cortical neurons co-expressing both proteins. TREK-1 and NTSR3/Sortilin were colocalized in mouse cortical neurons. Spadin bound specifically to TREK-1 with an affinity of 10 nM. Electrophysiological studies showed that spadin efficiently blocked the TREK-1 activity in COS-7 cells, cultured hippocampal pyramidal neurons, and CA3 hippocampal neurons in brain slices. Spadin also induced in vivo an increase of the 5-HT neuron firing rate in the Dorsal Raphe Nucleus. In five behavioral tests predicting an antidepressant response, spadin-treated mice showed a resistance to depression as found in TREK-1 deficient mice. More importantly, an intravenous 4-d treatment with spadin not only induced a strong antidepressant effect but also enhanced hippocampal phosphorylation of CREB protein and neurogenesis, considered to be key markers of antidepressant action after chronic treatment with selective serotonin reuptake inhibitors. This work also shows the development of a reliable method for dosing the propeptide in serum of mice by using AlphaScreen technology. These findings point out spadin as a putative antidepressant of new generation with a rapid onset of action. Spadin can be regarded as the first natural antidepressant peptide identified. It corresponds to a new concept to address the treatment of depression.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Antidepressive Agents/chemistry , Peptides/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/pharmacology , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/therapeutic use , COS Cells , Chlorocebus aethiops , Cyclic AMP Response Element-Binding Protein/metabolism , Depressive Disorder/drug therapy , Drug Design , Humans , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Patch-Clamp Techniques , Peptides/chemistry , Peptides/genetics , Peptides/pharmacology , Peptides/therapeutic use , Potassium Channel Blockers/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Raphe Nuclei/drug effects , Serotonin/metabolism , Synaptic Transmission/drug effects
20.
Expert Rev Clin Pharmacol ; 3(3): 331-46, 2010 May.
Article in English | MEDLINE | ID: mdl-22111614

ABSTRACT

Acid-sensing ion channels (ASICs) form a family of voltage-independent cation channels that predominantly conduct Na+ ions, and were identified at the molecular level a little more than a decade ago. ASICs are activated by extracellular acidification within the physiological range, and they form effective proton sensors in both central and peripheral sensory neurons. A combination of genetic and pharmacologic approaches has revealed their implication in an increasing number of physiological and pathophysiological processes--most of them associated with extracellular pH fluctuations, ranging from synaptic plasticity, learning, memory, fear, depression, seizure termination and neuronal degeneration to nociception and mechanosensation. ASICs, therefore, emerge as new potential therapeutic targets in the management of psychiatric disorders, stroke, neurodegenerative diseases and pain.

SELECTION OF CITATIONS
SEARCH DETAIL