Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.765
Filter
1.
Sci Rep ; 14(1): 18077, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103432

ABSTRACT

Insulin has been shown to modulate neuronal processes through insulin receptors. The ion channels located on neurons may be important targets for insulin/insulin receptor signaling. Both insulin receptors and acid-sensing ion channels (ASICs) are expressed in dorsal root ganglia (DRG) neurons. However, it is still unclear whether there is an interaction between them. Therefore, the purpose of this investigation was to determine the effects of insulin on the functional activity of ASICs. A 5 min application of insulin rapidly enhanced acid-evoked ASIC currents in rat DRG neurons in a concentration-dependent manner. Insulin shifted the concentration-response plot for ASIC currents upward, with an increase of 46.2 ± 7.6% in the maximal current response. The insulin-induced increase in ASIC currents was eliminated by the insulin receptor antagonist GSK1838705, the tyrosine kinase inhibitor lavendustin A, and the phosphatidylinositol-3 kinase antagonist wortmannin. Moreover, insulin increased the number of acid-triggered action potentials by activating insulin receptors. Finally, local administration of insulin exacerbated the spontaneous nociceptive behaviors induced by intraplantar acid injection and the mechanical hyperalgesia induced by intramuscular acid injections through peripheral insulin receptors. These results suggested that insulin/insulin receptor signaling enhanced the functional activity of ASICs via tyrosine kinase and phosphatidylinositol-3 kinase pathways. Our findings revealed that ASICs were targets in primary sensory neurons for insulin receptor signaling, which may underlie insulin modulation of pain.


Subject(s)
Acid Sensing Ion Channels , Ganglia, Spinal , Insulin , Receptor, Insulin , Sensory Receptor Cells , Animals , Acid Sensing Ion Channels/metabolism , Insulin/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/cytology , Rats , Receptor, Insulin/metabolism , Male , Signal Transduction/drug effects , Action Potentials/drug effects , Rats, Sprague-Dawley , Hyperalgesia/metabolism , Cells, Cultured
2.
Sci Adv ; 10(32): eadn0367, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121219

ABSTRACT

The development of noninvasive approaches to precisely control neural activity in mammals is highly desirable. Here, we used the ion channel transient receptor potential ankyrin-repeat 1 (TRPA1) as a proof of principle, demonstrating remote near-infrared (NIR) activation of endogenous neuronal channels in mice through an engineered nanoagonist. This achievement enables specific neurostimulation in nongenetically modified mice. Initially, target-based screening identified flavins as photopharmacological agonists, allowing for the photoactivation of TRPA1 in sensory neurons upon ultraviolet A/blue light illumination. Subsequently, upconversion nanoparticles (UCNPs) were customized with an emission spectrum aligned to flavin absorption and conjugated with flavin adenine dinucleotide, creating a nanoagonist capable of NIR activation of TRPA1. Following the intrathecal injection of the nanoagonist, noninvasive NIR stimulation allows precise bidirectional control of nociception in mice through remote activation of spinal TRPA1. This study demonstrates a noninvasive NIR neurostimulation method with the potential for adaptation to various endogenous ion channels and neural processes by combining photochemical toolboxes with customized UCNPs.


Subject(s)
Infrared Rays , Nanoparticles , TRPA1 Cation Channel , Animals , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/agonists , Mice , Nanoparticles/chemistry , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Sensory Receptor Cells/drug effects , Ion Channels/metabolism , Nociception/drug effects
3.
Proc Natl Acad Sci U S A ; 121(34): e2405465121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39145932

ABSTRACT

Over half of spinal cord injury (SCI) patients develop opioid-resistant chronic neuropathic pain. Safer alternatives to opioids for treatment of neuropathic pain are gabapentinoids (e.g., pregabalin and gabapentin). Clinically, gabapentinoids appear to amplify opioid effects, increasing analgesia and overdose-related adverse outcomes, but in vitro proof of this amplification and its mechanism are lacking. We previously showed that after SCI, sensitivity to opioids is reduced by fourfold to sixfold in rat sensory neurons. Here, we demonstrate that after injury, gabapentinoids restore normal sensitivity of opioid inhibition of cyclic AMP (cAMP) generation, while reducing nociceptor hyperexcitability by inhibiting voltage-gated calcium channels (VGCCs). Increasing intracellular Ca2+ or activation of L-type VGCCs (L-VGCCs) suffices to mimic SCI effects on opioid sensitivity, in a manner dependent on the activity of the Raf1 proto-oncogene, serine/threonine-protein kinase C-Raf, but independent of neuronal depolarization. Together, our results provide a mechanism for potentiation of opioid effects by gabapentinoids after injury, via reduction of calcium influx through L-VGCCs, and suggest that other inhibitors targeting these channels may similarly enhance opioid treatment of neuropathic pain.


Subject(s)
Analgesics, Opioid , Cyclic AMP , Gabapentin , Neuralgia , Signal Transduction , Spinal Cord Injuries , Animals , Neuralgia/drug therapy , Neuralgia/metabolism , Cyclic AMP/metabolism , Rats , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Analgesics, Opioid/pharmacology , Gabapentin/pharmacology , Signal Transduction/drug effects , Rats, Sprague-Dawley , Male , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Pregabalin/pharmacology , Pregabalin/therapeutic use , Drug Synergism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects
4.
Mol Pain ; 20: 17448069241272149, 2024.
Article in English | MEDLINE | ID: mdl-39079948

ABSTRACT

Cadaverine is an endogenous metabolite produced by the gut microbiome with various activity in physiological and pathological conditions. However, whether cadaverine regulates pain or itch remains unclear. In this study, we first found that cadaverine may bind to histamine 4 receptor (H4R) with higher docking energy score using molecular docking simulations, suggesting cadaverine may act as an endogenous ligand for H4R. We subsequently found intradermal injection of cadaverine into the nape or cheek of mice induces a dose-dependent scratching response in mice, which was suppressed by a selective H4R antagonist JNJ-7777120, transient receptor potential vanilloid 1 (TRPV1) antagonist capsazepine and PLC inhibitor U73122, but not H1R antagonist or TRPA1 antagonist or TRPV4 antagonist. Consistently, cadaverine-induced itch was abolished in Trpv1-/- but not Trpa1-/- mice. Pharmacological analysis indicated that mast cells and opioid receptors were also involved in cadaverine-induced itch in mice. scRNA-Seq data analysis showed that H4R and TRPV1 are mainly co-expressed on NP2, NP3 and PEP1 DRG neurons. Calcium imaging analysis showed that cadaverine perfusion enhanced calcium influx in the dissociated dorsal root ganglion (DRG) neurons, which was suppressed by JNJ-7777120 and capsazepine, as well as in the DRG neurons from Trpv1-/- mice. Patch-clamp recordings found that cadaverine perfusion significantly increased the excitability of small diameter DRG neurons, and JNJ-7777120 abolished this effect, indicating involvement of H4R. Together, these results provide evidences that cadaverine is a novel endogenous pruritogens, which activates H4R/TRPV1 signaling pathways in the primary sensory neurons.


Subject(s)
Cadaverine , Ganglia, Spinal , Mice, Inbred C57BL , Pruritus , TRPV Cation Channels , Animals , Pruritus/metabolism , Pruritus/chemically induced , TRPV Cation Channels/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Male , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Cadaverine/metabolism , Mice , Mice, Knockout , Humans , Mast Cells/metabolism , Mast Cells/drug effects , TRPA1 Cation Channel/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Receptors, G-Protein-Coupled/metabolism , Capsaicin/analogs & derivatives
5.
Sci Rep ; 14(1): 17360, 2024 07 29.
Article in English | MEDLINE | ID: mdl-39075089

ABSTRACT

Prostaglandin E2 (PGE2) is a major contributor to inflammatory pain hyperalgesia, however, the extent to which it modulates the activity of nociceptive axons is incompletely understood. We developed and characterized a microfluidic cell culture model to investigate sensitisation of the axons of dorsal root ganglia neurons. We show that application of PGE2 to fluidically isolated axons leads to sensitisation of their responses to depolarising stimuli. Interestingly the application of PGE2 to the DRG axons elicited a direct and persistent spiking activity propagated to the soma. Both the persistent activity and the membrane depolarisation in the axons are abolished by the EP4 receptor inhibitor and a blocker of cAMP synthesis. Further investigated into the mechanisms of the spiking activity showed that the PGE2 evoked depolarisation was inhibited by Nav1.8 sodium channel blockers but was refractory to the application of TTX or zatebradine. Interestingly, the depolarisation of axons was blocked by blocking ANO1 channels with T16Ainh-A01. We further show that PGE2-elicited axonal responses are altered by the changes in chloride gradient within the axons following treatment with bumetanide a Na-K-2Cl cotransporter NKCC1 inhibitor, but not by VU01240551 an inhibitor of potassium-chloride transporter KCC2. Our data demonstrate a novel role for PGE2/EP4/cAMP pathway which culminates in a sustained depolarisation of sensory axons mediated by a chloride current through ANO1 channels. Therefore, using a microfluidic culture model, we provide evidence for a potential dual function of PGE2 in inflammatory pain: it sensitises depolarisation-evoked responses in nociceptive axons and directly triggers action potentials by activating ANO1 and Nav1.8 channels.


Subject(s)
Anoctamin-1 , Axons , Dinoprostone , Ganglia, Spinal , NAV1.8 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Animals , Dinoprostone/pharmacology , Dinoprostone/metabolism , Axons/metabolism , Axons/drug effects , Axons/physiology , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Rats , Anoctamin-1/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Action Potentials/drug effects , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Rats, Sprague-Dawley , Cells, Cultured , Solute Carrier Family 12, Member 2/metabolism , Cyclic AMP/metabolism
6.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000275

ABSTRACT

In tumor cells, interleukin-6 (IL-6) signaling can lead to activation of the epidermal growth factor receptor (EGFR), which prolongs Stat3 activation. In the present experiments, we tested the hypothesis that IL-6 signaling activates EGFR signaling in peripheral and spinal nociception and examined whether EGFR localization and activation coincide with pain-related behaviors in arthritis. In vivo in anesthetized rats, spinal application of the EGFR receptor blocker gefitinib reduced the responses of spinal cord neurons to noxious joint stimulation, but only after spinal pretreatment with IL-6 and soluble IL-6 receptor. Using Western blots, we found that IL-6-induced Stat3 activation was reduced by gefitinib in microglial cells of the BV2 cell line, but not in cultured DRG neurons. Immunohistochemistry showed EGFR localization in most DRG neurons from normal rats, but significant downregulation in the acute and most painful arthritis phase. In the spinal cord of mice, EGFR was highly activated mainly in the chronic phase of inflammation, with localization in neurons. These data suggest that spinal IL-6 signaling may activate spinal EGFR signaling. Downregulation of EGFR in DRG neurons in acute arthritis may limit nociception, but pronounced delayed activation of EGFR in the spinal cord may be involved in chronic inflammatory pain.


Subject(s)
ErbB Receptors , Interleukin-6 , Sensory Receptor Cells , Spinal Cord , Animals , Female , Mice , Rats , Arthritis/metabolism , Arthritis, Experimental/metabolism , Cell Line , ErbB Receptors/metabolism , Ganglia, Spinal/metabolism , Gefitinib/pharmacology , Interleukin-6/metabolism , Receptors, Interleukin-6/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction , Spinal Cord/metabolism , STAT3 Transcription Factor/metabolism
7.
Brain Behav Immun ; 120: 471-487, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38925417

ABSTRACT

Activity-induced muscle pain increases interleukin-1ß (IL-1ß) release from muscle macrophages and the development of hyperalgesia is prevented by blockade of IL-1ß in muscle. Brain derived neurotrophic factor (BDNF) is released from sensory neurons in response to IL-1ß and mediates both inflammatory and neuropathic pain. Thus, we hypothesize that in activity-induced pain, fatigue metabolites combined with IL-1ß activate sensory neurons to increase BDNF release, peripherally in muscle and centrally in the spinal dorsal horn, to produce hyperalgesia. We tested the effect of intrathecal or intramuscular injection of BDNF-Tropomyosin receptor kinase B (TrkB) inhibitors, ANA-12 or TrkB-Fc, on development of activity-induced pain. Both inhibitors prevented the hyperalgesia when given before or 24hr after induction of the model in male but not female mice. BDNF messenger ribonucleic acid (mRNA) and protein were significantly increased in dorsal root ganglion (DRG) 24hr after induction of the model in both male and female mice. Blockade of IL-1ß in muscle had no effect on the increased BNDF mRNA observed in the activity-induced pain model, while IL-1ß applied to cultured DRG significantly induced BDNF expression, suggesting IL-1ß is sufficient but not necessary to induce BNDF. Thus, fatigue metabolites, combined with IL-1ß, upregulate BDNF in primary DRG neurons in both male and female mice, but contribute to activity-induced pain only in males.


Subject(s)
Brain-Derived Neurotrophic Factor , Ganglia, Spinal , Hyperalgesia , Interleukin-1beta , Myalgia , Animals , Brain-Derived Neurotrophic Factor/metabolism , Male , Female , Mice , Ganglia, Spinal/metabolism , Interleukin-1beta/metabolism , Myalgia/metabolism , Hyperalgesia/metabolism , Mice, Inbred C57BL , Receptor, trkB/metabolism , Muscle, Skeletal/metabolism , Sex Factors , Sex Characteristics , Benzamides/pharmacology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Azepines
8.
Biomed Pharmacother ; 176: 116879, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38850666

ABSTRACT

Cannabinoid CB2 agonists show therapeutic efficacy without unwanted CB1-mediated side effects. The G protein-biased CB2 receptor agonist LY2828360 attenuates the maintenance of chemotherapy-induced neuropathic nociception in male mice and blocks development of morphine tolerance in this model. However, the cell types involved in this phenomenon are unknown and whether this therapeutic profile is observed in female mice has never been investigated. We used conditional deletion of CB2 receptors to determine the cell population(s) mediating the anti-allodynic and morphine-sparing effects of CB2 agonists. Anti-allodynic effects of structurally distinct CB2 agonists (LY2828360 and AM1710) were present in paclitaxel-treated CB2f/f mice and in mice lacking CB2 receptors in CX3CR1 expressing microglia/macrophages (CX3CR1CRE/+; CB2f/f), but were absent in mice lacking CB2 receptors in peripheral sensory neurons (AdvillinCRE/+; CB2f/f). The morphine-sparing effect of LY28282360 occurred in a sexually-dimorphic manner, being present in male, but not female, mice. LY2828360 treatment (3 mg/kg per day i.p. x 12 days) blocked the development of morphine tolerance in male CB2f/f and CX3CR1CRE/+; CB2f/f mice with established paclitaxel-induced neuropathy but was absent in male (or female) AdvillinCRE/+; CB2f/f mice. Co-administration of morphine with a low dose of LY2828360 (0.1 mg/kg per day i.p. x 6 days) reversed morphine tolerance in paclitaxel-treated male CB2f/f mice, but not AdvillinCRE/+; CB2f/f mice of either sex. LY2828360 (3 mg/kg per day i.p. x 8 days) delayed, but did not prevent, the development of paclitaxel-induced mechanical or cold allodynia in either CB2f/f or CX3CR1CRE/+; CB2f/f mice of either sex. Our findings have potential clinical implications.


Subject(s)
Drug Tolerance , Morphine , Neuralgia , Paclitaxel , Receptor, Cannabinoid, CB2 , Sensory Receptor Cells , Animals , Male , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB2/genetics , Female , Morphine/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Drug Tolerance/physiology , Mice , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/metabolism , Nociception/drug effects , Mice, Inbred C57BL , Sex Characteristics , Mice, Knockout , Cannabinoid Receptor Agonists/pharmacology
9.
Elife ; 122024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896465

ABSTRACT

Spinal pain affects individuals of all ages and is the most common musculoskeletal problem globally. Its clinical management remains a challenge as the underlying mechanisms leading to it are still unclear. Here, we report that significantly increased numbers of senescent osteoclasts (SnOCs) are observed in mouse models of spinal hypersensitivity, like lumbar spine instability (LSI) or aging, compared to controls. The larger population of SnOCs is associated with induced sensory nerve innervation, as well as the growth of H-type vessels, in the porous endplate. We show that deletion of senescent cells by administration of the senolytic drug Navitoclax (ABT263) results in significantly less spinal hypersensitivity, spinal degeneration, porosity of the endplate, sensory nerve innervation, and H-type vessel growth in the endplate. We also show that there is significantly increased SnOC-mediated secretion of Netrin-1 and NGF, two well-established sensory nerve growth factors, compared to non-senescent OCs. These findings suggest that pharmacological elimination of SnOCs may be a potent therapy to treat spinal pain.


Subject(s)
Cellular Senescence , Osteoclasts , Animals , Mice , Osteoclasts/metabolism , Osteoclasts/drug effects , Osteoclasts/physiology , Cellular Senescence/drug effects , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism , Disease Models, Animal , Male , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Netrin-1/metabolism , Netrin-1/genetics , Mice, Inbred C57BL
10.
Biomed Res ; 45(3): 125-133, 2024.
Article in English | MEDLINE | ID: mdl-38839355

ABSTRACT

Clary sage essential oil (CSEO) is utilized in perfumery, aromatherapy, and skincare. Linalyl acetate (LA), a primary component of CSEO, possesses sedative, anxiolytic, and analgesic properties. However, the mechanism of its analgesic action is not clearly understood. Transient receptor potential ankyrin 1 (TRPA1) channel, a non-selective cation channel, is mainly expressed in sensory neurons and serves as a sensor of various irritants. In this study, we investigated the effects of LA on TRPA1 channel using heterologous expression system and isolated sensory neurons. To detect channel activity, we employed Ca2+ imaging and the whole-cell patch-clamp technique. The analgesic action of LA was measured in a pain-related behavioral mouse model. In cells that heterologously expressed TRPA1, LA diminished [Ca2+]i and current responses to allylisothiocyanate (AITC) and carvacrol: exogenous TRPA1 agonists, and the inhibitory effects were more pronounced for the former than for the latter. Moreover, LA suppressed [Ca2+] i and current responses to PGJ2: an endogenous TRPA1 agonist. Similar inhibitory actions were observed in native TRPA1 channels expressed in mouse sensory neurons. Furthermore, LA diminished PGJ2-induced nociceptive behaviors in mice. These findings suggest that analgesic effects of LA exert through inhibition of nociceptive TRPA1, making it a potential candidate for novel analgesic development.


Subject(s)
Analgesics , Monoterpenes , TRPA1 Cation Channel , Animals , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Mice , Analgesics/pharmacology , Monoterpenes/pharmacology , Humans , Male , Calcium/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , HEK293 Cells , Disease Models, Animal , Pain/drug therapy , Pain/metabolism
11.
Cell Commun Signal ; 22(1): 307, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831315

ABSTRACT

BACKGROUND: Interleukin 24 (IL-24) has been implicated in the nociceptive signaling. However, direct evidence and the precise molecular mechanism underlying IL-24's role in peripheral nociception remain unclear. METHODS: Using patch clamp recording, molecular biological analysis, immunofluorescence labeling, siRNA-mediated knockdown approach and behavior tests, we elucidated the effects of IL-24 on sensory neuronal excitability and peripheral pain sensitivity mediated by T-type Ca2+ channels (T-type channels). RESULTS: IL-24 enhances T-type channel currents (T-currents) in trigeminal ganglion (TG) neurons in a reversible and dose-dependent manner, primarily by activating the interleukin-22 receptor 1 (IL-22R1). Furthermore, we found that the IL-24-induced T-type channel response is mediated through tyrosine-protein kinase Lyn, but not its common downstream target JAK1. IL-24 application significantly activated protein kinase A; this effect was independent of cAMP and prevented by Lyn antagonism. Inhibition of PKA prevented the IL-24-induced T-current response, whereas inhibition of protein kinase C or MAPK kinases had no effect. Functionally, IL-24 increased TG neuronal excitability and enhanced pain sensitivity to mechanical stimuli in mice, both of which were suppressed by blocking T-type channels. In a trigeminal neuropathic pain model induced by chronic constriction injury of the infraorbital nerve, inhibiting IL-22R1 signaling alleviated mechanical allodynia, which was reversed by blocking T-type channels or knocking down Cav3.2. CONCLUSION: Our findings reveal that IL-24 enhances T-currents by stimulating IL-22R1 coupled to Lyn-dependent PKA signaling, leading to TG neuronal hyperexcitability and pain hypersensitivity. Understanding the mechanism of IL-24/IL-22R1 signaling in sensory neurons may pave the way for innovative therapeutic strategies in pain management.


Subject(s)
Calcium Channels, T-Type , Cyclic AMP-Dependent Protein Kinases , Receptors, Interleukin , Sensory Receptor Cells , Signal Transduction , Trigeminal Ganglion , src-Family Kinases , Animals , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , src-Family Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Trigeminal Ganglion/metabolism , Male , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Receptors, Interleukin/metabolism , Mice , Mice, Inbred C57BL , Interleukins/metabolism
12.
Mol Pain ; 20: 17448069241258106, 2024.
Article in English | MEDLINE | ID: mdl-38752471

ABSTRACT

Transient Receptor Potential Vanilloid 1 (TRPV1) is a nonselective cation channel expressed by pain-sensing neurons and has been an attractive target for the development of drugs to treat pain. Recently, Src homology region two domain-containing phosphatase-1 (SHP-1, encoded by Ptpn6) was shown to dephosphorylate TRPV1 in dorsal root ganglia (DRG) neurons, which was linked with alleviating different pain phenotypes. These previous studies were performed in male rodents only and did not directly investigate the role of SHP-1 in TRPV-1 mediated sensitization. Therefore, our goal was to determine the impact of Ptpn6 overexpression on TRPV1-mediated neuronal responses and capsaicin-induced pain behavior in mice of both sexes. Twelve-week-old male and female mice overexpressing Ptpn6 (Shp1-Tg) and their wild type (WT) littermates were used. Ptpn6 overexpression was confirmed in the DRG of Shp1-Tg mice by RNA in situ hybridization and RT-qPCR. Trpv1 and Ptpn6 were found to be co-expressed in DRG sensory neurons in both genotypes. Functionally, this overexpression resulted in lower magnitude intracellular calcium responses to 200 nM capsaicin stimulation in DRG cultures from Shp1-Tg mice compared to WTs. In vivo, we tested the effects of Ptpn6 overexpression on capsaicin-induced pain through a model of capsaicin footpad injection. While capsaicin injection evoked nocifensive behavior (paw licking) and paw swelling in both genotypes and sexes, only WT mice developed mechanical allodynia after capsaicin injection. We observed similar level of TRPV1 protein expression in the DRG of both genotypes, however, a higher amount of tyrosine phosphorylated TRPV1 was detected in WT DRG. These experiments suggest that, while SHP-1 does not mediate the acute swelling and nocifensive behavior induced by capsaicin, it does mediate a protective effect against capsaicin-induced mechanical allodynia in both sexes. The protective effect of SHP-1 might be mediated by TRPV1 dephosphorylation in capsaicin-sensitive sensory neurons of the DRG.


Subject(s)
Capsaicin , Ganglia, Spinal , Hyperalgesia , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , TRPV Cation Channels , Animals , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Capsaicin/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Male , Female , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Mice , Neurons/metabolism , Neurons/drug effects , Mice, Transgenic , Calcium/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects
13.
Sci Transl Med ; 16(746): eadk8198, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38718132

ABSTRACT

The phosphate modification of drugs is a common chemical strategy to increase solubility and allow for parenteral administration. Unfortunately, phosphate modifications often elicit treatment- or dose-limiting pruritus through an unknown mechanism. Using unbiased high-throughput drug screens, we identified the Mas-related G protein-coupled receptor X4 (MRGPRX4), a primate-specific, sensory neuron receptor previously implicated in itch, as a potential target for phosphate-modified compounds. Using both Gq-mediated calcium mobilization and G protein-independent GPCR assays, we found that phosphate-modified compounds potently activate MRGPRX4. Furthermore, a humanized mouse model expressing MRGPRX4 in sensory neurons exhibited robust phosphomonoester prodrug-evoked itch. To characterize and confirm this interaction, we further determined the structure of MRGPRX4 in complex with a phosphate-modified drug through single-particle cryo-electron microscopy (cryo-EM) and identified critical amino acid residues responsible for the binding of the phosphate group. Together, these findings explain how phosphorylated drugs can elicit treatment-limiting itch and identify MRGPRX4 as a potential therapeutic target to suppress itch and to guide future drug design.


Subject(s)
Disease Models, Animal , Pruritus , Receptors, G-Protein-Coupled , Animals , Pruritus/metabolism , Pruritus/chemically induced , Pruritus/pathology , Pruritus/drug therapy , Humans , Receptors, G-Protein-Coupled/metabolism , Mice , HEK293 Cells , Phosphorylation/drug effects , Phosphates/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Prodrugs/pharmacology , Cryoelectron Microscopy
14.
Br J Pharmacol ; 181(17): 3192-3214, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38741464

ABSTRACT

BACKGROUND AND PURPOSE: The mechanistic target of rapamycin (mTOR) signalling pathway is a key regulator of cell growth and metabolism. Its deregulation is implicated in several diseases. The macrolide rapamycin, a specific inhibitor of mTOR, has immunosuppressive, anti-inflammatory and antiproliferative properties. Recently, we identified tacrolimus, another macrolide immunosuppressant, as a novel activator of TRPM8 ion channels, involved in cold temperature sensing, thermoregulation, tearing and cold pain. We hypothesized that rapamycin may also have agonist activity on TRPM8 channels. EXPERIMENTAL APPROACH: Using calcium imaging and electrophysiology in transfected HEK293 cells and wildtype or Trpm8 KO mouse DRG neurons, we characterized rapamycin's effects on TRPM8 channels. We also examined the effects of rapamycin on tearing in mice. KEY RESULTS: Micromolar concentrations of rapamycin activated rat and mouse TRPM8 channels directly and potentiated cold-evoked responses, effects also observed in human TRPM8 channels. In cultured mouse DRG neurons, rapamycin increased intracellular calcium levels almost exclusively in cold-sensitive neurons. Responses were markedly decreased in Trpm8 KO mice or by TRPM8 channel antagonists. Cutaneous cold thermoreceptor endings were also activated by rapamycin. Topical application of rapamycin to the eye surface evokes tearing in mice by a TRPM8-dependent mechanism. CONCLUSION AND IMPLICATIONS: These results identify TRPM8 cationic channels in sensory neurons as novel molecular targets of the immunosuppressant rapamycin. These findings may help explain some of its therapeutic effects after topical application to the skin and the eye surface. Moreover, rapamycin could be used as an experimental tool in the clinic to explore cold thermoreceptors.


Subject(s)
Immunosuppressive Agents , Mice, Knockout , Sensory Receptor Cells , Sirolimus , TRPM Cation Channels , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/metabolism , Animals , Humans , HEK293 Cells , Sirolimus/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Immunosuppressive Agents/pharmacology , Rats , Mice , Male , Mice, Inbred C57BL , Cells, Cultured , Calcium/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Cold Temperature
15.
Cell ; 187(12): 2935-2951.e19, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38772371

ABSTRACT

Peripheral sensory neurons widely innervate various tissues to continuously monitor and respond to environmental stimuli. Whether peripheral sensory neurons innervate the spleen and modulate splenic immune response remains poorly defined. Here, we demonstrate that nociceptive sensory nerve fibers extensively innervate the spleen along blood vessels and reach B cell zones. The spleen-innervating nociceptors predominantly originate from left T8-T13 dorsal root ganglia (DRGs), promoting the splenic germinal center (GC) response and humoral immunity. Nociceptors can be activated by antigen-induced accumulation of splenic prostaglandin E2 (PGE2) and then release calcitonin gene-related peptide (CGRP), which further promotes the splenic GC response at the early stage. Mechanistically, CGRP directly acts on B cells through its receptor CALCRL-RAMP1 via the cyclic AMP (cAMP) signaling pathway. Activating nociceptors by ingesting capsaicin enhances the splenic GC response and anti-influenza immunity. Collectively, our study establishes a specific DRG-spleen sensory neural connection that promotes humoral immunity, suggesting a promising approach for improving host defense by targeting the nociceptive nervous system.


Subject(s)
Calcitonin Gene-Related Peptide , Germinal Center , Immunity, Humoral , Spleen , Animals , Male , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Cyclic AMP/metabolism , Dinoprostone/metabolism , Ganglia, Spinal/metabolism , Germinal Center/immunology , Mice, Inbred C57BL , Nociceptors/metabolism , Receptor Activity-Modifying Protein 1/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction , Spleen/innervation , Spleen/immunology , Female
16.
Acta Biomater ; 182: 1-13, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38750917

ABSTRACT

Skin denervation has been shown to cause remission of psoriatic lesions in patients, which can reappear if reinnervation occurs. This effect can be induced by the activation of dendritic cells through sensory innervation. However, a direct effect of nerves on the proliferation of keratinocytes involved in the formation of psoriatic plaques has not been investigated. We developed, by tissue engineering, a model of psoriatic skin made of patient skin cells that showed increased keratinocyte proliferation and epidermal thickness compared to healthy controls. When this model was treated with CGRP, a neuropeptide released by sensory neurons, an increased keratinocyte proliferation was observed in the psoriatic skin model, but not in the control. When a sensory nerve network was incorporated in the psoriatic model and treated with capsaicin to induce neuropeptide release, an increase of keratinocyte proliferation was confirmed, which was blocked by a CGRP antagonist while no difference was noticed in the innervated healthy control. We showed that sensory neurons can participate directly to keratinocyte hyperproliferation in the formation of psoriatic lesions through the release of CGRP, independently of the immune system. Our unique tissue-engineered innervated psoriatic skin model could be a valuable tool to better understand the mechanism by which nerves may modulate psoriatic lesion formation in humans. STATEMENT OF SIGNIFICANCE: This study shows that keratinocytes extracted from patients' psoriatic skin retain, at least in part, the disease phenotype. Indeed, when combined in a 3D model of tissue-engineered psoriatic skin, keratinocytes exhibited a higher proliferation rate, and produced a thicker epidermis than a healthy skin control. In addition, their hyperproliferation was aggravated by a treatment with CGRP, a neuropeptide released by sensory nerves. In a innervated model of tissue-engineered psoriatic skin, an increase in keratinocyte hyperproliferation was also observed after inducing neurons to release neuropeptides. This effect was prevented by concomitant treatment with an antagonist to CGRP. Thus, this study shows that sensory nerves can directly participate to affect keratinocyte hyperproliferation in psoriasis through CGRP release.


Subject(s)
Calcitonin Gene-Related Peptide , Cell Proliferation , Keratinocytes , Psoriasis , Sensory Receptor Cells , Tissue Engineering , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Calcitonin Gene-Related Peptide/metabolism , Psoriasis/pathology , Psoriasis/metabolism , Cell Proliferation/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Sensory Receptor Cells/drug effects , Models, Biological , Female , Adult , Male , Skin/innervation , Skin/pathology , Skin/metabolism
17.
J Histochem Cytochem ; 72(5): 275-287, 2024 05.
Article in English | MEDLINE | ID: mdl-38725415

ABSTRACT

The TRPA1 ion channel is a sensitive detector of reactive chemicals, found primarily on sensory neurons. The phenotype exhibited by mice lacking TRPA1 suggests its potential as a target for pharmacological intervention. Antibody-based detection for distribution analysis is a standard technique. In the case of TRPA1, however, there is no antibody with a plausible validation in knockout animals or functional studies, but many that have failed in this regard. To this end we employed the single molecule in situ hybridization technique RNAscope on sensory neurons immediately after detection of calcium responses to the TRPA1 agonist allyl isothiocyanate. There is a clearly positive correlation between TRPA1 calcium imaging and RNAscope detection (R = 0.43), although less than what might have been expected. Thus, the technique of choice should be carefully considered to suit the research question. The marginal correlation between TRPV1 RNAscope and the specific agonist capsaicin indicates that such validation is advisable for every RNAscope target. Given the recent description of a long-awaited TRPA1 reporter mouse, TRPA1 RNAscope detection might still have its use cases, for detection of RNA at particular sites, for example, defined structurally or by other molecular markers.


Subject(s)
Calcium , Isothiocyanates , TRPA1 Cation Channel , Animals , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Isothiocyanates/pharmacology , Mice , Calcium/metabolism , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/agonists , Capsaicin/pharmacology , In Situ Hybridization , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/agonists , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Mice, Inbred C57BL , Calcium Channels/metabolism , Calcium Channels/genetics , Male
18.
Neuropharmacology ; 253: 109967, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38657946

ABSTRACT

Botulinum neurotoxin type A BoNT/A is used off-label as a third line therapy for neuropathic pain. However, the mechanism of action remains unclear. In recent years, the role of voltage-gated sodium channels (Nav) in neuropathic pain became evident and it was suggested that block of sodium channels by BoNT/A would contribute to its analgesic effect. We assessed sodium channel function in the presence of BoNT/A in heterologously expressed Nav1.7, Nav1.3, and the neuronal cell line ND7/23 by high throughput automated and manual patch-clamp. We used both the full protein and the isolated catalytic light chain LC/A for acute or long-term extracellular or intracellular exposure. To assess the toxin's effect in a human cellular system, we differentiated induced pluripotent stem cells (iPSC) into sensory neurons from a healthy control and a patient suffering from a hereditary neuropathic pain syndrome (inherited erythromelalgia) carrying the Nav1.7/p.Q875E-mutation and carried out multielectrode-array measurements. Both BoNT/A and the isolated catalytic light chain LC/A showed limited effects in heterologous expression systems and the neuronal cell line ND7/23. Spontaneous activity in iPSC derived sensory neurons remained unaltered upon BoNT/A exposure both in neurons from the healthy control and the mutation carrying patient. BoNT/A may not specifically be beneficial in pain syndromes linked to sodium channel variants. The favorable effects of BoNT/A in neuropathic pain are likely based on mechanisms other than sodium channel blockage and new approaches to understand BoNT/A's therapeutic effects are necessary.


Subject(s)
Botulinum Toxins, Type A , Induced Pluripotent Stem Cells , NAV1.7 Voltage-Gated Sodium Channel , Neuralgia , Humans , Neuralgia/drug therapy , Botulinum Toxins, Type A/pharmacology , Botulinum Toxins, Type A/therapeutic use , Induced Pluripotent Stem Cells/drug effects , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Analgesics/pharmacology , Animals , NAV1.3 Voltage-Gated Sodium Channel/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , HEK293 Cells , Cell Line
19.
Brain Res Bull ; 212: 110966, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38670469

ABSTRACT

Intraoperative remifentanil administration has been linked to increased postoperative pain sensitivity. Recent studies have identified the involvement of euchromatic histone-lysine N-methyltransferase 2 (Ehmt2/G9a) in neuropathic pain associated with the transcriptional silencing of many potassium ion channel genes. This study investigates whether G9a regulates the potassium sodium-activated channel subfamily T member 1 (Slo2.2) in remifentanil-induced post-incisional hyperalgesia (RIH) in rodents. We performed remifentanil infusion (1 µg·kg-1·min-1 for 60 min) followed by plantar incision to induce RIH in rodents. Our results showed that RIH was accompanied by increased G9a and H3K9me2 production and decreased Slo2.2 expression 48 h postoperatively. Deletion of G9a rescued Slo2.2 expression in DRG and reduced RIH intensity. Slo2.2 overexpression also reversed this hyperalgesia phenotype. G9a overexpression decreased Slo2.2-mediated leak current and increased excitability in the small-diameter DRG neurons and laminal II small-diameter neurons in the spinal dorsal horn, which was implicated in peripheral and central sensitization. These results suggest that G9a contributes to the development of RIH by epigenetically silencing Slo2.2 in DRG neurons, leading to decreased central sensitization in the spinal cord. The findings may have implications for the development of novel therapeutic targets for the treatment of postoperative pain.


Subject(s)
Histone-Lysine N-Methyltransferase , Hyperalgesia , Remifentanil , Sensory Receptor Cells , Animals , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Male , Remifentanil/pharmacology , Hyperalgesia/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Potassium Channels, Sodium-Activated , Mice , Analgesics, Opioid/pharmacology , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Neuralgia/metabolism , Pain, Postoperative/drug therapy , Pain, Postoperative/metabolism , Rats , Pain Threshold/drug effects , Rats, Sprague-Dawley , Mice, Inbred C57BL , Nerve Tissue Proteins
20.
Pain ; 165(7): 1592-1604, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38293826

ABSTRACT

ABSTRACT: Visceral pain is a leading cause of morbidity in inflammatory bowel disease (IBD), contributing significantly to reduced quality of life. Currently available analgesics often lack efficacy or have intolerable side effects, driving the need for a more complete understanding of the mechanisms causing pain. Whole transcriptome gene expression analysis was performed by bulk RNA sequencing of colonic biopsies from patients with ulcerative colitis (UC) and Crohn's disease (CD) reporting abdominal pain and compared with noninflamed control biopsies. Potential pronociceptive mediators were identified based on gene upregulation in IBD biopsy tissue and cognate receptor expression in murine colonic sensory neurons. Pronociceptive activity of identified mediators was assessed in assays of sensory neuron and colonic afferent activity. RNA sequencing analysis highlighted a 7.6-fold increase in the expression of angiotensinogen transcripts, Agt , which encode the precursor to angiotensin II (Ang II), in samples from UC patients ( P = 3.2 × 10 -8 ). Consistent with the marked expression of the angiotensin AT 1 receptor in colonic sensory neurons, Ang II elicited an increase in intracellular Ca 2+ in capsaicin-sensitive, voltage-gated sodium channel subtype Na V 1.8-positive sensory neurons. Ang II also evoked action potential discharge in high-threshold colonic nociceptors. These effects were inhibited by the AT 1 receptor antagonist valsartan. Findings from our study identify AT 1 receptor-mediated colonic nociceptor activation as a novel pathway of visceral nociception in patients with UC. This work highlights the potential utility of angiotensin receptor blockers, such as valsartan, as treatments for pain in IBD.


Subject(s)
Angiotensin II , Gene Expression Profiling , Inflammatory Bowel Diseases , Humans , Animals , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/genetics , Mice , Male , Female , Colon/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Adult , Middle Aged , Mice, Inbred C57BL , Nociceptors/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL