Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 180(4): 385-400, 2023 02.
Article in English | MEDLINE | ID: mdl-36131381

ABSTRACT

BACKGROUND AND PURPOSE: T-type calcium channels, mainly the Cav 3.2 subtype, are important contributors to the nociceptive signalling pathway. We investigated their involvement in inflammation and related pain-like symptoms. EXPERIMENTAL APPROACH: The involvement of Cav 3.2 and T-type channels was investigated using genetic and pharmacological inhibition to assess mechanical allodynia/hyperalgesia and oedema development in two murine inflammatory pain models. The location of Cav 3.2 channels involved in pain-like symptoms was studied in mice with Cav 3.2 knocked out in C-low threshold mechanoreceptors (C-LTMR) and the use of ABT-639, a peripherally restricted T-type channel inhibitor. The anti-oedema effect of Cav 3.2 channel inhibition was investigated in chimeric mice with immune cells deleted for Cav 3.2. Lymphocytes and macrophages from either green fluorescent protein-targeted Cav 3.2 or KO mice were used to determine the expression of Cav 3.2 protein and the functional status of the cells. KEY RESULTS: Cav 3.2 channels contributed to the development of pain-like symptoms and oedema in the two murine inflammatory pain models. Our results provided evidence of the involvement of Cav 3.2 channels located on C-LTMRs and spinal cord in inflammatory pain. Cav 3.2 channels located in T cells and macrophages contribute to the inflammatory process. CONCLUSION AND IMPLICATIONS: Cav 3.2 channels play crucial roles in inflammation and related pain, implying that targeting of Cav 3.2 channels with pharmacological agents could be an attractive and readily evaluable strategy in clinical trials, to relieve chronic inflammatory pain in patients.


Subject(s)
Chronic Pain , Inflammation , Mice , Animals , Hyperalgesia , CD4-Positive T-Lymphocytes , Mechanoreceptors , Macrophages
2.
Sci Adv ; 8(26): eabo7566, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35767616

ABSTRACT

Affective touch is necessary for proper neurodevelopment and sociability. However, it remains unclear how the neurons innervating the skin detect affective and social behaviors. The C low-threshold mechanoreceptors (C-LTMRs), a specific population of somatosensory neurons in mice, appear particularly well suited, physiologically and anatomically, to perceive affective and social touch. However, their contribution to sociability has not been resolved yet. Our observations revealed that C-LTMR functional deficiency induced social isolation and reduced tactile interactions in adulthood. Conversely, transient increase in C-LTMR excitability in adults, using chemogenetics, was rewarding, promoted touch-seeking behaviors, and had prosocial influences on group dynamics. This work provides the first empirical evidence that specific peripheral inputs alone can drive complex social behaviors. It demonstrates the existence of a specialized neuronal circuit, originating in the skin, wired to promote interactions with other individuals.

3.
ACS Synth Biol ; 7(7): 1685-1693, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29975841

ABSTRACT

Nerve growth factor/tropomyosin receptor kinase A (NGF/TrkA) signaling plays a key role in neuronal development, function, survival, and growth. The pathway is implicated in neurodegenerative disorders including Alzheimer's disease, chronic pain, inflammation, and cancer. NGF binds the extracellular domain of TrkA, leading to the activation of the receptor's intracellular kinase domain. As TrkA signaling is highly dynamic, mechanistic studies would benefit from a tool with high spatial and temporal resolution. Here we present the design and evaluation of four strategies for light-inducible activation of TrkA in the absence of NGF. Our strategies involve the light-sensitive protein Arabidopsis cryptochrome 2 and its binding partner CIB1. We demonstrate successful recapitulation of native NGF/TrkA functions by optical induction of plasma membrane recruitment and homo-interaction of the intracellular domain of TrkA. This approach activates PI3K/AKT and Raf/ERK signaling pathways, promotes neurite growth in PC12 cells, and supports survival of dorsal root ganglion neurons in the absence of NGF. This ability to activate TrkA using light bestows high spatial and temporal resolution for investigating NGF/TrkA signaling.


Subject(s)
Receptor, trkA/metabolism , Animals , Cell Membrane/metabolism , Cell Survival/genetics , Cell Survival/physiology , Ganglia, Spinal/metabolism , Nerve Growth Factor/metabolism , PC12 Cells , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/genetics , Phosphorylation/physiology , Rats , Receptor, trkA/genetics , Signal Transduction
4.
Neuron ; 98(1): 90-108.e5, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29576387

ABSTRACT

Cellular interactions between delta and mu opioid receptors (DORs and MORs), including heteromerization, are thought to regulate opioid analgesia. However, the identity of the nociceptive neurons in which such interactions could occur in vivo remains elusive. Here we show that DOR-MOR co-expression is limited to small populations of excitatory interneurons and projection neurons in the spinal cord dorsal horn and unexpectedly predominates in ventral horn motor circuits. Similarly, DOR-MOR co-expression is rare in parabrachial, amygdalar, and cortical brain regions processing nociceptive information. We further demonstrate that in the discrete DOR-MOR co-expressing nociceptive neurons, the two receptors internalize and function independently. Finally, conditional knockout experiments revealed that DORs selectively regulate mechanical pain by controlling the excitability of somatostatin-positive dorsal horn interneurons. Collectively, our results illuminate the functional organization of DORs and MORs in CNS pain circuits and reappraise the importance of DOR-MOR cellular interactions for developing novel opioid analgesics.


Subject(s)
Anterior Horn Cells/metabolism , Nerve Net/metabolism , Pain/metabolism , Posterior Horn Cells/metabolism , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, mu/biosynthesis , Animals , Anterior Horn Cells/chemistry , Anterior Horn Cells/pathology , Central Nervous System/chemistry , Central Nervous System/metabolism , Central Nervous System/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/chemistry , Nerve Net/pathology , Pain/pathology , Pain Measurement/methods , Posterior Horn Cells/chemistry , Posterior Horn Cells/pathology , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/genetics
5.
Handb Exp Pharmacol ; 247: 87-114, 2018.
Article in English | MEDLINE | ID: mdl-28993838

ABSTRACT

The functional diversity of primary afferent neurons of the dorsal root ganglia (DRG) generates a variety of qualitatively and quantitatively distinct somatosensory experiences, from shooting pain to pleasant touch. In recent years, the identification of dozens of genetic markers specifically expressed by subpopulations of DRG neurons has dramatically improved our understanding of this diversity and provided the tools to manipulate their activity and uncover their molecular identity and function. Opioid receptors have long been known to be expressed by discrete populations of DRG neurons, in which they regulate cell excitability and neurotransmitter release. We review recent insights into the identity of the DRG neurons that express the delta opioid receptor (DOR) and the ion channel mechanisms that DOR engages in these cells to regulate sensory input. We highlight recent findings derived from DORGFP reporter mice and from in situ hybridization and RNA sequencing studies in wild-type mice that revealed DOR presence in cutaneous mechanosensory afferents eliciting touch and implicated in tactile allodynia. Mechanistically, we describe how DOR modulates opening of voltage-gated calcium channels (VGCCs) to control glutamatergic neurotransmission between somatosensory neurons and postsynaptic neurons in the spinal cord dorsal horn. We additionally discuss other potential signaling mechanisms, including those involving potassium channels, which DOR may engage to fine tune somatosensation. We conclude by discussing how this knowledge may explain the analgesic properties of DOR agonists against mechanical pain and uncovers an unanticipated specialized function for DOR in cutaneous mechanosensation.


Subject(s)
Neurons, Afferent/metabolism , Receptors, Opioid, delta/biosynthesis , Sensory Receptor Cells/metabolism , Animals , Ganglia, Spinal/metabolism , Humans , Pain/drug therapy , Pain/physiopathology , Receptors, Opioid, delta/agonists
6.
Neuron ; 93(4): 822-839.e6, 2017 Feb 22.
Article in English | MEDLINE | ID: mdl-28162807

ABSTRACT

Pain thresholds are, in part, set as a function of emotional and internal states by descending modulation of nociceptive transmission in the spinal cord. Neurons of the rostral ventromedial medulla (RVM) are thought to critically contribute to this process; however, the neural circuits and synaptic mechanisms by which distinct populations of RVM neurons facilitate or diminish pain remain elusive. Here we used in vivo opto/chemogenetic manipulations and trans-synaptic tracing of genetically identified dorsal horn and RVM neurons to uncover an RVM-spinal cord-primary afferent circuit controlling pain thresholds. Unexpectedly, we found that RVM GABAergic neurons facilitate mechanical pain by inhibiting dorsal horn enkephalinergic/GABAergic interneurons. We further demonstrate that these interneurons gate sensory inputs and control pain through temporally coordinated enkephalin- and GABA-mediated presynaptic inhibition of somatosensory neurons. Our results uncover a descending disynaptic inhibitory circuit that facilitates mechanical pain, is engaged during stress, and could be targeted to establish higher pain thresholds. VIDEO ABSTRACT.


Subject(s)
Enkephalins/metabolism , GABAergic Neurons/metabolism , Interneurons/metabolism , Neural Pathways/physiology , Pain/physiopathology , Spinal Cord/metabolism , Animals , Brain Stem/metabolism , Brain Stem/physiopathology , Medulla Oblongata/metabolism , Mice, Transgenic , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/physiopathology
7.
Cell Rep ; 17(6): 1699-1710, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27806306

ABSTRACT

Spinal dorsal horn circuits receive, process, and transmit somatosensory information. To understand how specific components of these circuits contribute to behavior, it is critical to be able to directly modulate their activity in unanesthetized in vivo conditions. Here, we develop experimental tools that enable optogenetic control of spinal circuitry in freely moving mice using commonly available materials. We use these tools to examine mechanosensory processing in the spinal cord and observe that optogenetic activation of somatostatin-positive interneurons facilitates both mechanosensory and itch-related behavior, while reversible chemogenetic inhibition of these neurons suppresses mechanosensation. These results extend recent findings regarding the processing of mechanosensory information in the spinal cord and indicate the potential for activity-induced release of the somatostatin neuropeptide to affect processing of itch. The spinal implant approach we describe here is likely to enable a wide range of studies to elucidate spinal circuits underlying pain, touch, itch, and movement.


Subject(s)
Mechanotransduction, Cellular , Spinal Cord/physiology , Animals , Female , Histamine , Interneurons/physiology , Light , Mice, Inbred C57BL , Optical Fibers , Optogenetics , Proto-Oncogene Proteins c-fos/metabolism , Pruritus/pathology , Pruritus/physiopathology , Somatostatin/metabolism
8.
Pain ; 157 Suppl 1: S15-S22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26785151

ABSTRACT

Pain is a quite frequent complaint accompanying numerous pathologies. Among these pathological cases, numerous neuropathies are retrieved with identified etiologies (chemotherapies, diabetes, surgeries…) and also more diffuse syndromes such as fibromyalgia. More broadly, pain is one of the first consequences of most inherited diseases. Despite its importance for the quality of life, current pain management is limited to drugs that are either old or with a limited efficacy or that possess a bad risk benefit ratio. As no new pharmacological concept has led to new analgesics in the last decades, the discovery of new medications is needed, and to this aim, the identification of new druggable targets in pain transmission is a first step. Therefore, studies of ion channels in pain pathways are extremely active. This is particularly true with ion channels in peripheral sensory neurons in dorsal root ganglia known how to express unique sets of these channels. Moreover, both spinal and supraspinal levels are clearly important in pain modulation. Among these ion channels, we and others revealed the important role of low voltage-gated calcium channels in cellular excitability in different steps of the pain pathways. These channels, by being activated nearby resting membrane potential, have biophysical characteristics suited to facilitate action potential generation and rhythmicity. In this review, we will present the current knowledge on the role of these channels in the perception and modulation of pain.


Subject(s)
Calcium Channels, T-Type/metabolism , Neuralgia/metabolism , Animals , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/genetics , Humans , Neuralgia/drug therapy , Neuralgia/genetics
9.
Cell Rep ; 10(3): 370-382, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25600872

ABSTRACT

The T-type calcium channel Cav3.2 emerges as a key regulator of sensory functions, but its expression pattern within primary afferent neurons and its contribution to modality-specific signaling remain obscure. Here, we elucidate this issue using a unique knockin/flox mouse strain wherein Cav3.2 is replaced by a functional Cav3.2-surface-ecliptic GFP fusion. We demonstrate that Cav3.2 is a selective marker of two major low-threshold mechanoreceptors (LTMRs), Aδ- and C-LTMRs, innervating the most abundant skin hair follicles. The presence of Cav3.2 along LTMR-fiber trajectories is consistent with critical roles at multiple sites, setting their strong excitability. Strikingly, the C-LTMR-specific knockout uncovers that Cav3.2 regulates light-touch perception and noxious mechanical cold and chemical sensations and is essential to build up that debilitates allodynic symptoms of neuropathic pain, a mechanism thought to be entirely A-LTMR specific. Collectively, our findings support a fundamental role for Cav3.2 in touch/pain pathophysiology, validating their critic pharmacological relevance to relieve mechanical and cold allodynia.

10.
Neuron ; 83(5): 1144-58, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25189210

ABSTRACT

T-type calcium channels are essential contributors to the transmission of nociceptive signals in the primary afferent pain pathway. Here, we show that T-type calcium channels are ubiquitinated by WWP1, a plasma-membrane-associated ubiquitin ligase that binds to the intracellular domain III-IV linker region of the Cav3.2 T-type channel and modifies specific lysine residues in this region. A proteomic screen identified the deubiquitinating enzyme USP5 as a Cav3.2 III-IV linker interacting partner. Knockdown of USP5 via shRNA increases Cav3.2 ubiquitination, decreases Cav3.2 protein levels, and reduces Cav3.2 whole-cell currents. In vivo knockdown of USP5 or uncoupling USP5 from native Cav3.2 channels via intrathecal delivery of Tat peptides mediates analgesia in both inflammatory and neuropathic mouse models of mechanical hypersensitivity. Altogether, our experiments reveal a cell signaling pathway that regulates T-type channel activity and their role in nociceptive signaling.


Subject(s)
Calcium Channels, T-Type/metabolism , Endopeptidases/metabolism , Inflammation/physiopathology , Neuralgia/enzymology , Animals , Calcium Channels, T-Type/genetics , Cells, Cultured , Disease Models, Animal , Endopeptidases/genetics , Freund's Adjuvant/toxicity , Humans , Hyperalgesia/diagnosis , Hyperalgesia/physiopathology , In Vitro Techniques , Inflammation/chemically induced , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuralgia/drug therapy , Pain Threshold/drug effects , Pain Threshold/physiology , Peptides/therapeutic use , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Spinal Cord/cytology , Transfection , Ubiquitination/genetics , Ubiquitination/physiology
11.
Neuron ; 81(6): 1312-1327, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24583022

ABSTRACT

Cutaneous mechanosensory neurons detect mechanical stimuli that generate touch and pain sensation. Although opioids are generally associated only with the control of pain, here we report that the opioid system in fact broadly regulates cutaneous mechanosensation, including touch. This function is predominantly subserved by the delta opioid receptor (DOR), which is expressed by myelinated mechanoreceptors that form Meissner corpuscles, Merkel cell-neurite complexes, and circumferential hair follicle endings. These afferents also include a small population of CGRP-expressing myelinated nociceptors that we now identify as the somatosensory neurons that coexpress mu and delta opioid receptors. We further demonstrate that DOR activation at the central terminals of myelinated mechanoreceptors depresses synaptic input to the spinal dorsal horn, via the inhibition of voltage-gated calcium channels. Collectively our results uncover a molecular mechanism by which opioids modulate cutaneous mechanosensation and provide a rationale for targeting DOR to alleviate injury-induced mechanical hypersensitivity.


Subject(s)
Mechanoreceptors/physiology , Neurons/physiology , Nociceptors/physiology , Receptors, Opioid, delta/metabolism , Spinal Cord/metabolism , Analgesics, Opioid/pharmacology , Animals , Calcium Channels/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Mechanoreceptors/drug effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , Nociceptors/drug effects , Pain/physiopathology , Spinal Cord/drug effects
12.
Pflugers Arch ; 466(4): 707-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24590509

ABSTRACT

Pain is a quite frequent complaint accompanying numerous pathologies. Among these pathological cases, neuropathies are retrieved with identified etiologies (chemotherapies, diabetes, surgeries…) and also more diffuse syndromes such as fibromyalgia. More broadly, pain is one of the first consequences of the majority of inherited diseases. Despite its importance for the quality of life, current pain management is limited to drugs that are either old or with a limited efficacy or that possess a bad benefit/risk ratio. As no new pharmacological concept has led to new analgesics in the last decades, the discovery of medications is needed, and to this aim the identification of new druggable targets in pain transmission is a first step. Therefore, studies of ion channels in pain pathways are extremely active. This is particularly true with ion channels in peripheral sensory neurons in dorsal root ganglia (DRG) known now to express unique sets of these channels. Moreover, both spinal and supraspinal levels are clearly important in pain modulation. Among these ion channels, we and others revealed the important role of low voltage-gated calcium channels in cellular excitability in different steps of the pain pathways. These channels, by being activated nearby resting membrane potential have biophysical characteristics suited to facilitate action potential generation and rhythmicity. In this review, we will review the current knowledge on the role of these channels in the perception and modulation of pain.


Subject(s)
Analgesics/administration & dosage , Calcium Channel Blockers/administration & dosage , Calcium Channels, T-Type/physiology , Chronic Pain/metabolism , Disease Models, Animal , Animals , Chronic Pain/drug therapy , Drug Delivery Systems/trends , Humans , Mice , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Pain ; 155(4): 764-772, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24447516

ABSTRACT

To exert its analgesic action, paracetamol requires complex metabolism to produce a brain-specific lipoamino acid compound, AM404, which targets central transient receptor potential vanilloid receptors (TRPV1). Lipoamino acids are also known to induce analgesia through T-type calcium-channel inhibition (Ca(v)3.2). In this study we show that the antinociceptive effect of paracetamol in mice is lost when supraspinal Ca(v)3.2 channels are inhibited. Therefore, we hypothesized a relationship between supraspinal Ca(v)3.2 and TRPV1, via AM404, which mediates the analgesic effect of paracetamol. AM404 is able to activate TRPV1 and weakly inhibits Ca(v)3.2. Interestingly, activation of TRPV1 induces a strong inhibition of Ca(v)3.2 current. Supporting this, intracerebroventricular administration of AM404 or capsaicin produces antinociception that is lost in Ca(v)3.2(-/-) mice. Our study, for the first time, (1) provides a molecular mechanism for the supraspinal antinociceptive effect of paracetamol; (2) identifies the relationship between TRPV1 and the Ca(v)3.2 channel; and (3) suggests supraspinal Ca(v)3.2 inhibition as a potential pharmacological strategy to alleviate pain.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Calcium Channels, T-Type/deficiency , Spinal Cord/drug effects , 4-Aminopyridine/pharmacology , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Arthritis/chemically induced , Arthritis/drug therapy , Calcium Channels, T-Type/genetics , Cells, Cultured , Drug Administration Routes , Drug Interactions , Ganglia, Spinal/cytology , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Knockout , Morphine/pharmacology , Neurons/drug effects , Pain Measurement/drug effects , Potassium Channel Blockers/pharmacology , Spinal Cord/cytology , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics , Tetraethylammonium/pharmacology
15.
Cell Rep ; 5(2): 378-88, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24139797

ABSTRACT

C-low-threshold mechanoreceptors (C-LTMRs) are unique among C-unmyelinated primary sensory neurons. These neurons convey two opposite aspects of touch sensation: a sensation of pleasantness, and a sensation of injury-induced mechanical pain. Here, we show that TAFA4 is a specific marker of C-LTMRs. Genetic labeling in combination with electrophysiological recordings show that TAFA4+ neurons have intrinsic properties of mechano-nociceptors. TAFA4-null mice exhibit enhanced mechanical and chemical hypersensitivity following inflammation and nerve injury as well as increased excitability of spinal cord lamina IIi neurons, which could be reversed by intrathecal or bath application of recombinant TAFA4 protein. In wild-type C57/Bl6 mice, intrathecal administration of TAFA4 strongly reversed carrageenan-induced mechanical hypersensitivity, suggesting a potent analgesic role of TAFA4 in pain relief. Our data provide insights into how C-LTMR-derived TAFA4 modulates neuronal excitability and controls the threshold of somatic sensation.


Subject(s)
Cytokines/metabolism , Nociceptors/metabolism , Pain/physiopathology , Stress, Mechanical , Animals , Carrageenan/toxicity , Cytokines/genetics , Cytokines/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Pain/metabolism , Pain Threshold/drug effects , Patch-Clamp Techniques , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
16.
Pain ; 154(2): 283-293, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23257507

ABSTRACT

T-type calcium channels encoded by the Ca(V)3.2 isoform are expressed in nociceptive primary afferent neurons where they contribute to hyperalgesia and thus are considered as a potential therapeutic target to treat pathological pain. Here we report that the small organic state-dependent T-type channel antagonist TTA-A2 efficiently inhibits recombinant and native Ca(V)3.2 currents. Although TTA-A2 is a pan Ca(V)3 blocker, it demonstrates a higher potency for Ca(V)3.2 compared to Ca(V)3.1. TTA-A2 selectivity for T-type currents was demonstrated in sensory neurons where it lowered cell excitability uniquely on neurons expressing T-type channels. In vivo pharmacology in Ca(V)3.2 knockout and wild type mice reveal that TTA-A2-mediated antinociception critically depends on Ca(V)3.2 expression. The pathophysiology of irritable bowel syndrome (IBS) was recently demonstrated to involve Ca(V)3.2 in a rat model of this disease. Oral administration of TTA-A2 produced a dose-dependent reduction of hypersensitivity in an IBS model, demonstrating its therapeutic potential for the treatment of pathological pain. Overall, our results suggest that the high potency of TTA-A2 in the depolarized state strengthen its analgesic efficacy and selectivity toward pathological pain syndromes. This characteristic would be beneficial for the development of analgesics targeting T-type channels, in particular for the treatment of pain associated with IBS.


Subject(s)
Benzeneacetamides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Hyperalgesia/drug therapy , Neurons/drug effects , Pyridines/pharmacology , Animals , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/genetics , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hyperalgesia/genetics , Hyperalgesia/metabolism , Male , Mice , Mice, Knockout , Neurons/metabolism , Rats , Rats, Sprague-Dawley
17.
EMBO Mol Med ; 3(5): 266-78, 2011 May.
Article in English | MEDLINE | ID: mdl-21438154

ABSTRACT

Cold hypersensitivity is the hallmark of oxaliplatin-induced neuropathy, which develops in nearly all patients under this chemotherapy. To date, pain management strategies have failed to alleviate these symptoms, hence development of adapted analgesics is needed. Here, we report that oxaliplatin exaggerates cold perception in mice as well as in patients. These symptoms are mediated by primary afferent sensory neurons expressing the thermoreceptor TRPM8. Mechanistically, oxaliplatin promotes over-excitability by drastically lowering the expression of distinct potassium channels (TREK1, TRAAK) and by increasing the expression of pro-excitatory channels such as the hyperpolarization-activated channels (HCNs). These findings are corroborated by the analysis of TREK1-TRAAK null mice and use of the specific HCN inhibitor ivabradine, which abolishes the oxaliplatin-induced cold hypersensibility. These results suggest that oxaliplatin exacerbates cold perception by modulating the transcription of distinct ionic conductances that together shape sensory neuron responses to cold. The translational and clinical implication of these findings would be that ivabradine may represent a tailored treatment for oxaliplatin-induced neuropathy.


Subject(s)
Antineoplastic Agents/adverse effects , Cold Temperature , Hyperalgesia/chemically induced , Nociceptors/drug effects , Organoplatinum Compounds/adverse effects , TRPM Cation Channels/metabolism , Animals , Humans , Mice , Nociceptors/metabolism , Oxaliplatin , Potassium Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...