Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Publication year range
1.
Cell ; 186(10): 2160-2175.e17, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37137306

ABSTRACT

The serotonin transporter (SERT) removes synaptic serotonin and is the target of anti-depressant drugs. SERT adopts three conformations: outward-open, occluded, and inward-open. All known inhibitors target the outward-open state except ibogaine, which has unusual anti-depressant and substance-withdrawal effects, and stabilizes the inward-open conformation. Unfortunately, ibogaine's promiscuity and cardiotoxicity limit the understanding of inward-open state ligands. We docked over 200 million small molecules against the inward-open state of the SERT. Thirty-six top-ranking compounds were synthesized, and thirteen inhibited; further structure-based optimization led to the selection of two potent (low nanomolar) inhibitors. These stabilized an outward-closed state of the SERT with little activity against common off-targets. A cryo-EM structure of one of these bound to the SERT confirmed the predicted geometry. In mouse behavioral assays, both compounds had anxiolytic- and anti-depressant-like activity, with potencies up to 200-fold better than fluoxetine (Prozac), and one substantially reversed morphine withdrawal effects.


Subject(s)
Ibogaine , Selective Serotonin Reuptake Inhibitors , Serotonin Plasma Membrane Transport Proteins , Small Molecule Libraries , Animals , Mice , Fluoxetine/pharmacology , Ibogaine/chemistry , Ibogaine/pharmacology , Molecular Conformation , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/ultrastructure , Selective Serotonin Reuptake Inhibitors/pharmacology , Small Molecule Libraries/pharmacology
2.
Nature ; 600(7890): 759-764, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34880501

ABSTRACT

The σ2 receptor has attracted intense interest in cancer imaging1, psychiatric disease2, neuropathic pain3-5 and other areas of biology6,7. Here we determined the crystal structure of this receptor in complex with the clinical candidate roluperidone2 and the tool compound PB288. These structures templated a large-scale docking screen of 490 million virtual molecules, of which 484 compounds were synthesized and tested. We identified 127 new chemotypes with affinities superior to 1 µM, 31 of which had affinities superior to 50 nM. The hit rate fell smoothly and monotonically with docking score. We optimized three hits for potency and selectivity, and achieved affinities that ranged from 3 to 48 nM, with up to 250-fold selectivity versus the σ1 receptor. Crystal structures of two ligands bound to the σ2 receptor confirmed the docked poses. To investigate the contribution of the σ2 receptor in pain, two potent σ2-selective ligands and one potent σ1/σ2 non-selective ligand were tested for efficacy in a mouse model of neuropathic pain. All three ligands showed time-dependent decreases in mechanical hypersensitivity in the spared nerve injury model9, suggesting that the σ2 receptor has a role in nociception. This study illustrates the opportunities for rapid discovery of in vivo probes through structure-based screens of ultra large libraries, enabling study of underexplored areas of biology.


Subject(s)
Neuralgia , Receptors, sigma , Animals , Ligands , Mice , Neuralgia/drug therapy , Receptors, sigma/metabolism , Structure-Activity Relationship
3.
Cell ; 139(2): 267-84, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19837031

ABSTRACT

The nervous system detects and interprets a wide range of thermal and mechanical stimuli, as well as environmental and endogenous chemical irritants. When intense, these stimuli generate acute pain, and in the setting of persistent injury, both peripheral and central nervous system components of the pain transmission pathway exhibit tremendous plasticity, enhancing pain signals and producing hypersensitivity. When plasticity facilitates protective reflexes, it can be beneficial, but when the changes persist, a chronic pain condition may result. Genetic, electrophysiological, and pharmacological studies are elucidating the molecular mechanisms that underlie detection, coding, and modulation of noxious stimuli that generate pain.


Subject(s)
Nociceptors/physiology , Pain/physiopathology , Animals , Chronic Disease , Humans , Neuronal Plasticity
4.
Cell ; 137(6): 1148-59, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524516

ABSTRACT

Delta and mu opioid receptors (DORs and MORs) are inhibitory G protein-coupled receptors that reportedly cooperatively regulate the transmission of pain messages by substance P and TRPV1-expressing pain fibers. Using a DOReGFP reporter mouse we now show that the DOR and MOR are, in fact, expressed by different subsets of primary afferents. The MOR is expressed in peptidergic pain fibers, the DOR in myelinated and nonpeptidergic afferents. Contrary to the prevailing view, we demonstrate that the DOR is trafficked to the cell surface under resting conditions, independently of substance P, and internalized following activation by DOR agonists. Finally, we show that the segregated DOR and MOR distribution is paralleled by a remarkably selective functional contribution of the two receptors to the control of mechanical and heat pain, respectively. These results demonstrate that behaviorally relevant pain modalities can be selectively regulated through the targeting of distinct subsets of primary afferent pain fibers.


Subject(s)
Pain/physiopathology , Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/physiology , Analgesia , Analgesics, Opioid/pharmacology , Animals , Gene Knock-In Techniques , Hot Temperature , Male , Mechanoreceptors/physiology , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Nociceptors/physiology , Pain/chemically induced , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Spinal Cord/pathology , Spinal Cord/physiology , Substance P/metabolism , TRPV Cation Channels/metabolism
5.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34234018

ABSTRACT

A remarkable molecular and functional heterogeneity of the primary sensory neurons and dorsal horn interneurons transmits pain- and or itch-relevant information, but the molecular signature of the projection neurons that convey the messages to the brain is unclear. Here, using retro-TRAP (translating ribosome affinity purification) and RNA sequencing, we reveal extensive molecular diversity of spino- and trigeminoparabrachial projection neurons. Among the many genes identified, we highlight distinct subsets of Cck+ -, Nptx2+ -, Nmb+ -, and Crh+ -expressing projection neurons. By combining in situ hybridization of retrogradely labeled neurons with Fos-based assays, we also demonstrate significant functional heterogeneity, including both convergence and segregation of pain- and itch-provoking inputs into molecularly diverse subsets of NK1R- and non-NK1R-expressing projection neurons.


Subject(s)
Neurons/pathology , Pain/complications , Pain/pathology , Pruritus/complications , Pruritus/pathology , Spinal Cord/pathology , Trigeminal Nerve/pathology , Animals , Chloroquine/pharmacology , Female , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Pain/genetics , Physical Stimulation , Pruritus/genetics , RNA/isolation & purification , RNA/metabolism , Receptors, Neurokinin-1/metabolism , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism
6.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33602818

ABSTRACT

Pruritus is a common symptom of inflammatory skin conditions, including atopic dermatitis (AD). Although primary sensory neurons that transmit pruritic signals are well-cataloged, little is known about the neuronal alterations that occur as a result of skin disruption in AD. To address this question, we examined the molecular and behavioral consequences of challenging Grhl3PAR2/+ mice, which overexpress PAR2 in suprabasal keratinocytes, with serial topical application of the environmental allergen house dust mite (HDM). We monitored behavior and used RNA sequencing, qPCR, and in situ hybridization to evaluate gene expression in trigeminal ganglia (TG), before and after HDM. We found that neither Grhl3PAR2/+ nor wild-type (WT) mice exhibited spontaneous scratching, and pruritogen-induced acute scratching did not differ. In contrast, HDM exacerbated scratching in Grhl3PAR2/+ mice. Despite the absence of scratching in untreated Grhl3PAR2/+ mice, several TG genes in these mice were up-regulated compared to WT. HDM treatment of the Grhl3PAR2/+ mice enhanced up-regulation of this set of genes and induced additional genes, many within the subset of TG neurons that express TRPV1. The same set of genes was up-regulated in HDM-treated Grhl3PAR2/+ mice that did not scratch, but at lesser magnitude. Finally, we recorded comparable transcriptional changes in IL31Tg mice, demonstrating that a common genetic program is induced in two AD models. Taken together, we conclude that transcriptional changes that occur in primary sensory neurons in dermatitis-susceptible animals underlie a genetic priming that not only sensitizes the animal to chronic allergens but also contributes to pruritus in atopic skin disease.


Subject(s)
Allergens/toxicity , DNA-Binding Proteins/physiology , Dermatitis, Atopic/pathology , Receptor, PAR-2/metabolism , Sensory Receptor Cells/pathology , Skin/pathology , Transcription Factors/physiology , Animals , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , RNA-Seq , Receptor, PAR-2/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Skin/drug effects , Skin/innervation , Skin/metabolism
7.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33972431

ABSTRACT

Febrile seizures (FSs) are the most common convulsion in infancy and childhood. Considering the limitations of current treatments, it is important to examine the mechanistic cause of FSs. Prompted by a genome-wide association study identifying TMEM16C (also known as ANO3) as a risk factor of FSs, we showed previously that loss of TMEM16C function causes hippocampal neuronal hyperexcitability [Feenstra et al., Nat. Genet. 46, 1274-1282 (2014)]. Our previous study further revealed a reduction in the number of warm-sensitive neurons that increase their action potential firing rate with rising temperature of the brain region harboring these hypothalamic neurons. Whereas central neuronal hyperexcitability has been implicated in FSs, it is unclear whether the maximal temperature reached during fever or the rate of body temperature rise affects FSs. Here we report that mutant rodent pups with TMEM16C eliminated from all or a subset of their central neurons serve as FS models with deficient thermoregulation. Tmem16c knockout (KO) rat pups at postnatal day 10 (P10) are more susceptible to hyperthermia-induced seizures. Moreover, they display a more rapid rise of body temperature upon heat exposure. In addition, conditional knockout (cKO) mouse pups (P11) with TMEM16C deletion from the brain display greater susceptibility of hyperthermia-induced seizures as well as deficiency in thermoregulation. We also found similar phenotypes in P11 cKO mouse pups with TMEM16C deletion from Ptgds-expressing cells, including temperature-sensitive neurons in the preoptic area (POA) of the anterior hypothalamus, the brain region that controls body temperature. These findings suggest that homeostatic thermoregulation plays an important role in FSs.


Subject(s)
Body Temperature Regulation/genetics , Chloride Channels/genetics , Fever/genetics , Hyperthermia/genetics , Preoptic Area/metabolism , Seizures, Febrile/genetics , Action Potentials/physiology , Animals , Animals, Newborn , Body Temperature/drug effects , Body Temperature/physiology , Chloride Channels/deficiency , Female , Fever/chemically induced , Fever/metabolism , Fever/physiopathology , Gene Expression , Hippocampus/metabolism , Hippocampus/physiopathology , Hyperthermia/metabolism , Hyperthermia/physiopathology , Kainic Acid/administration & dosage , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Preoptic Area/physiopathology , Protein Isoforms/deficiency , Protein Isoforms/genetics , Rats , Seizures, Febrile/chemically induced , Seizures, Febrile/metabolism , Seizures, Febrile/physiopathology
8.
Nature ; 534(7608): 494-9, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27281198

ABSTRACT

Voltage-gated sodium (Nav) channels initiate action potentials in most neurons, including primary afferent nerve fibres of the pain pathway. Local anaesthetics block pain through non-specific actions at all Nav channels, but the discovery of selective modulators would facilitate the analysis of individual subtypes of these channels and their contributions to chemical, mechanical, or thermal pain. Here we identify and characterize spider (Heteroscodra maculata) toxins that selectively activate the Nav1.1 subtype, the role of which in nociception and pain has not been elucidated. We use these probes to show that Nav1.1-expressing fibres are modality-specific nociceptors: their activation elicits robust pain behaviours without neurogenic inflammation and produces profound hypersensitivity to mechanical, but not thermal, stimuli. In the gut, high-threshold mechanosensitive fibres also express Nav1.1 and show enhanced toxin sensitivity in a mouse model of irritable bowel syndrome. Together, these findings establish an unexpected role for Nav1.1 channels in regulating the excitability of sensory nerve fibres that mediate mechanical pain.


Subject(s)
NAV1.1 Voltage-Gated Sodium Channel/metabolism , Nociception/drug effects , Nociceptors/drug effects , Nociceptors/metabolism , Spider Venoms/pharmacology , Stress, Mechanical , Animals , Disease Models, Animal , Female , Ganglia, Sensory/cytology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Irritable Bowel Syndrome/metabolism , Male , Myelin Sheath/metabolism , NAV1.1 Voltage-Gated Sodium Channel/chemistry , Nerve Fibers/drug effects , Nerve Fibers/metabolism , Oocytes/metabolism , Pain/chemically induced , Pain/metabolism , Protein Structure, Tertiary , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Spiders/chemistry , Substrate Specificity/drug effects , Temperature
9.
Brain ; 142(9): 2655-2669, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31321411

ABSTRACT

Dysfunction of inhibitory circuits in the rostral anterior cingulate cortex underlies the affective (aversive), but not the sensory-discriminative features (hypersensitivity) of the pain experience. To restore inhibitory controls, we transplanted inhibitory interneuron progenitor cells into the rostral anterior cingulate cortex in a chemotherapy-induced neuropathic pain model. The transplants integrated, exerted a GABA-A mediated inhibition of host pyramidal cells and blocked gabapentin preference (i.e. relieved ongoing pain) in a conditioned place preference paradigm. Surprisingly, pain aversiveness persisted when the transplants populated both the rostral and posterior anterior cingulate cortex. We conclude that selective and long lasting inhibition of the rostral anterior cingulate cortex, in the mouse, has a profound pain relieving effect against nerve injury-induced neuropathic pain. However, the interplay between the rostral and posterior anterior cingulate cortices must be considered when examining circuits that influence ongoing pain and pain aversiveness.


Subject(s)
GABAergic Neurons/metabolism , GABAergic Neurons/transplantation , Gyrus Cinguli/metabolism , Neuralgia/metabolism , Neuralgia/therapy , Sciatic Neuropathy/therapy , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuralgia/pathology , Receptors, GABA-B/metabolism , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology
10.
Proc Natl Acad Sci U S A ; 114(12): E2524-E2532, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28265084

ABSTRACT

Pain-producing animal venoms contain evolutionarily honed toxins that can be exploited to study and manipulate somatosensory and nociceptive signaling pathways. From a functional screen, we have identified a secreted phospholipase A2 (sPLA2)-like protein, BomoTx, from the Brazilian lancehead pit viper (Bothrops moojeni). BomoTx is closely related to a group of Lys49 myotoxins that have been shown to promote ATP release from myotubes through an unknown mechanism. Here we show that BomoTx excites a cohort of sensory neurons via ATP release and consequent activation of P2X2 and/or P2X3 purinergic receptors. We provide pharmacological and electrophysiological evidence to support pannexin hemichannels as downstream mediators of toxin-evoked ATP release. At the behavioral level, BomoTx elicits nonneurogenic inflammatory pain, thermal hyperalgesia, and mechanical allodynia, of which the latter is completely dependent on purinergic signaling. Thus, we reveal a role of regulated endogenous nucleotide release in nociception and provide a detailed mechanism of a pain-inducing Lys49 myotoxin from Bothrops species, which are responsible for the majority of snake-related deaths and injuries in Latin America.


Subject(s)
Adenosine Triphosphate/metabolism , Bothrops/physiology , Group II Phospholipases A2/toxicity , Pain/metabolism , Reptilian Proteins/toxicity , Sensory Receptor Cells/drug effects , Snake Bites/metabolism , Toxins, Biological/toxicity , Viper Venoms/enzymology , Animals , Bothrops/genetics , Brazil , Female , Group II Phospholipases A2/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Pain/etiology , Pain/genetics , Pain/parasitology , Rats , Receptors, Purinergic/metabolism , Reptilian Proteins/genetics , Sensory Receptor Cells/metabolism , Signal Transduction , Snake Bites/genetics , Snake Bites/parasitology , Viper Venoms/toxicity
12.
J Allergy Clin Immunol ; 140(2): 454-464.e2, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28232084

ABSTRACT

BACKGROUND: Despite recent insights into the pathophysiology of acute and chronic itch, chronic itch remains an often intractable condition. Among major contributors to chronic itch is dysfunction of spinal cord gamma aminobutyric acidergic (GABAergic) inhibitory controls. OBJECTIVES: We sought to test the hypothesis that selective GABA agonists as well as cell transplant-derived GABA are antipruritic against acute itch and in a transgenic mouse model of atopic dermatitis produced by overexpression of the TH2 cell-associated cytokine, IL-31 (IL-31Tg mice). METHODS: We injected wild-type and IL-31Tg mice with combinations of GABA-A (muscimol) or GABA-B (baclofen) receptor agonists 15 to 20 minutes prior to injection of various pruritogens (histamine, chloroquine, or endothelin-1) and recorded spontaneous scratching before and after drug administration. We also tested the antipruritic properties of intraspinal transplantation of precursors of GABAergic interneurons in the IL-31Tg mice. RESULTS: Systemic muscimol or baclofen are antipruritic against both histamine-dependent and -independent pruritogens, but the therapeutic window using either ligand alone was very small. In contrast, combined subthreshold doses of baclofen and muscimol produced a significant synergistic antipruritic effect, with no sedation. Finally, transplant-mediated long-term enhancement of GABAergic signaling not only reduced spontaneous scratching in the IL-31Tg mice but also dramatically resolved the associated skin lesions. CONCLUSIONS: Although additional research is clearly needed, existing approved GABA agonists should be considered in the management of chronic itch, notably atopic dermatitis.


Subject(s)
Antipruritics/therapeutic use , Baclofen/therapeutic use , Dermatitis, Atopic/drug therapy , GABA-A Receptor Agonists/therapeutic use , GABA-B Receptor Agonists/therapeutic use , Muscimol/therapeutic use , Animals , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/therapy , Disease Models, Animal , Drug Synergism , Gastrin-Releasing Peptide/genetics , Glutamate Decarboxylase/genetics , Interleukins/genetics , Interneurons/drug effects , Male , Median Eminence/cytology , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Receptors, Bombesin/genetics , Receptors, GABA-A/genetics , Receptors, GABA-B/genetics , Receptors, Neurokinin-1/genetics , Skin/drug effects , Skin/pathology , Spinal Cord/drug effects , Spinal Cord/metabolism , Stem Cell Transplantation
13.
J Neurosci ; 36(46): 11634-11645, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27852772

ABSTRACT

Spinal cord transplants of embryonic cortical GABAergic progenitor cells derived from the medial ganglionic eminence (MGE) can reverse mechanical hypersensitivity in the mouse models of peripheral nerve injury- and paclitaxel-induced neuropathic pain. Here, we used electrophysiology, immunohistochemistry, and electron microscopy to examine the extent to which MGE cells integrate into host circuitry and recapitulate endogenous inhibitory circuits. Whether the transplants were performed before or after nerve injury, the MGE cells developed into mature neurons and exhibited firing patterns characteristic of subpopulations of cortical and spinal cord inhibitory interneurons. Conversely, the transplanted cells preserved cortical morphological and neurochemical properties. We also observed a robust anatomical and functional synaptic integration of the transplanted cells into host circuitry in both injured and uninjured animals. The MGE cells were activated by primary afferents, including TRPV1-expressing nociceptors, and formed GABAergic, bicuculline-sensitive, synapses onto host neurons. Unexpectedly, MGE cells transplanted before injury prevented the development of mechanical hypersensitivity. Together, our findings provide direct confirmation of an extensive, functional synaptic integration of MGE cells into host spinal cord circuits. This integration underlies normalization of the dorsal horn inhibitory tone after injury and may be responsible for the prophylactic effect of preinjury transplants. SIGNIFICANCE STATEMENT: Spinal cord transplants of embryonic cortical GABAergic interneuron progenitors from the medial ganglionic eminence (MGE), can overcome the mechanical hypersensitivity produced in different neuropathic pain models in adult mice. Here, we examined the properties of transplanted MGE cells and the extent to which they integrate into spinal cord circuitry. Using electrophysiology, immunohistochemistry, and electron microscopy, we demonstrate that MGE cells, whether transplanted before or after nerve injury, develop into inhibitory neurons, are activated by nociceptive primary afferents, and form GABA-A-mediated inhibitory synapses with the host. Unexpectedly, cells transplanted into naive spinal cord prevented the development of nerve-injury-induced mechanical hypersensitivity. These results illustrate the remarkable plasticity of adult spinal cord and the potential of cell-based therapies against neuropathic pain.


Subject(s)
GABAergic Neurons/pathology , Hyperalgesia/physiopathology , Hyperalgesia/therapy , Neural Stem Cells/transplantation , Spinal Cord Regeneration/physiology , Spinal Cord/physiology , Synapses/pathology , Animals , GABAergic Neurons/metabolism , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Prosencephalon/cytology , Stem Cell Transplantation/methods , Synapses/metabolism , Treatment Outcome
14.
Nature ; 479(7373): 410-4, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22094702

ABSTRACT

Natural products that elicit discomfort or pain represent invaluable tools for probing molecular mechanisms underlying pain sensation. Plant-derived irritants have predominated in this regard, but animal venoms have also evolved to avert predators by targeting neurons and receptors whose activation produces noxious sensations. As such, venoms provide a rich and varied source of small molecule and protein pharmacophores that can be exploited to characterize and manipulate key components of the pain-signalling pathway. With this in mind, here we perform an unbiased in vitro screen to identify snake venoms capable of activating somatosensory neurons. Venom from the Texas coral snake (Micrurus tener tener), whose bite produces intense and unremitting pain, excites a large cohort of sensory neurons. The purified active species (MitTx) consists of a heteromeric complex between Kunitz- and phospholipase-A2-like proteins that together function as a potent, persistent and selective agonist for acid-sensing ion channels (ASICs), showing equal or greater efficacy compared with acidic pH. MitTx is highly selective for the ASIC1 subtype at neutral pH; under more acidic conditions (pH < 6.5), MitTx massively potentiates (>100-fold) proton-evoked activation of ASIC2a channels. These observations raise the possibility that ASIC channels function as coincidence detectors for extracellular protons and other, as yet unidentified, endogenous factors. Purified MitTx elicits robust pain-related behaviour in mice by activation of ASIC1 channels on capsaicin-sensitive nerve fibres. These findings reveal a mechanism whereby snake venoms produce pain, and highlight an unexpected contribution of ASIC1 channels to nociception.


Subject(s)
Elapid Venoms/chemistry , Elapid Venoms/pharmacology , Elapidae , Nerve Tissue Proteins/metabolism , Pain/chemically induced , Protein Multimerization , Sodium Channels/metabolism , Acid Sensing Ion Channels , Amino Acid Sequence , Animals , Capsaicin/pharmacology , Cells, Cultured , Hindlimb/drug effects , Hindlimb/physiopathology , Humans , Hydrogen-Ion Concentration , Ion Channel Gating/drug effects , Male , Mice , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nociception/drug effects , Nociception/physiology , Oocytes , Pain/metabolism , Pain/physiopathology , Protein Structure, Quaternary , Protons , Rats , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sodium Channel Agonists , Sodium Channels/deficiency , Sodium Channels/genetics , TRPV Cation Channels/metabolism , Xenopus laevis
15.
J Neurosci ; 35(2): 648-57, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25589759

ABSTRACT

There is continuing controversy relating to the primary afferent neurotransmitter that conveys itch signals to the spinal cord. Here, we investigated the DRG and spinal cord expression of the putative primary afferent-derived "itch" neurotransmitter, gastrin-releasing peptide (GRP). Using ISH, qPCR, and immunohistochemistry, we conclude that GRP is expressed abundantly in spinal cord, but not in DRG neurons. Titration of the most commonly used GRP antiserum in tissues from wild-type and GRP mutant mice indicates that the antiserum is only selective for GRP at high dilutions. Paralleling these observations, we found that a GRPeGFP transgenic reporter mouse has abundant expression in superficial dorsal horn neurons, but not in the DRG. In contrast to previous studies, neither dorsal rhizotomy nor an intrathecal injection of capsaicin, which completely eliminated spinal cord TRPV1-immunoreactive terminals, altered dorsal horn GRP immunoreactivity. Unexpectedly, however, peripheral nerve injury induced significant GRP expression in a heterogeneous population of DRG neurons. Finally, dual labeling and retrograde tracing studies showed that GRP-expressing neurons of the superficial dorsal horn are predominantly interneurons, that a small number coexpress protein kinase C gamma (PKCγ), but that none coexpress the GRP receptor (GRPR). Our studies support the view that pruritogens engage spinal cord "itch" circuits via excitatory superficial dorsal horn interneurons that express GRP and that likely target GRPR-expressing interneurons. The fact that peripheral nerve injury induced de novo GRP expression in DRG neurons points to a novel contribution of this peptide to pruritoceptive processing in neuropathic itch conditions.


Subject(s)
Gastrin-Releasing Peptide/metabolism , Neurons, Afferent/metabolism , Spinal Cord/metabolism , Animals , Antibodies/immunology , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gastrin-Releasing Peptide/genetics , Gastrin-Releasing Peptide/immunology , Immunochemistry/methods , Immunochemistry/standards , Male , Mice , Mice, Inbred C57BL , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sensitivity and Specificity , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
16.
Nature ; 462(7273): 651-5, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-19915548

ABSTRACT

Mechanical pain contributes to the morbidity associated with inflammation and trauma, but primary sensory neurons that convey the sensation of acute and persistent mechanical pain have not been identified. Dorsal root ganglion (DRG) neurons transmit sensory information to the spinal cord using the excitatory transmitter glutamate, a process that depends on glutamate transport into synaptic vesicles for regulated exocytotic release. Here we report that a small subset of cells in the DRG expresses the low abundance vesicular glutamate transporter VGLUT3 (also known as SLC17A8). In the dorsal horn of the spinal cord, these afferents project to lamina I and the innermost layer of lamina II, which has previously been implicated in persistent pain caused by injury. Because the different VGLUT isoforms generally have a non-redundant pattern of expression, we used Vglut3 knockout mice to assess the role of VGLUT3(+) primary afferents in the behavioural response to somatosensory input. The loss of VGLUT3 specifically impairs mechanical pain sensation, and in particular the mechanical hypersensitivity to normally innocuous stimuli that accompanies inflammation, nerve injury and trauma. Direct recording from VGLUT3(+) neurons in the DRG further identifies them as a poorly understood population of unmyelinated, low threshold mechanoreceptors (C-LTMRs). The analysis of Vglut3(-/-) mice now indicates a critical role for C-LTMRs in the mechanical hypersensitivity caused by injury.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Ganglia, Spinal/metabolism , Hypersensitivity/genetics , Hypersensitivity/physiopathology , Mechanoreceptors/physiology , Pain/genetics , Wounds and Injuries/physiopathology , Amino Acid Transport Systems, Acidic/genetics , Animals , Behavior, Animal/physiology , Female , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
J Pain ; 25(1): 53-63, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37482234

ABSTRACT

Most reports agree that aging negatively impacts pain processing and that the prevalence of chronic pain increases significantly with age. To improve current therapies, it is critical that aged animals be included in preclinical studies. Here we compared sensitivities to pain and itch-provoking stimuli in naïve and injured young and aged mice. Surprisingly, we found that in the absence of injury, aged male and female mice are significantly less responsive to mechanical stimuli and, in females, also to noxious thermal (heat) stimuli. In both older male and female mice, compared to younger (6-month-old mice), we also recorded reduced pruritogen-evoked scratching. On the other hand, after nerve injury, aged mice nevertheless developed significant mechanical hypersensitivity. Interestingly, however, and in contrast to young mice, aged mice developed both ipsilateral and contralateral postinjury mechanical allodynia. In a parallel immunohistochemical analysis of microglial and astrocyte markers, we found that the ipsilateral to the contralateral ratio of nerve injury-induced expression decreased with age. That observation is consistent with our finding of contralateral hypersensitivity after nerve injury in the aged but not the young mice. We conclude that aging has opposite effects on baseline versus postinjury pain and itch processing. PERSPECTIVE: Aged male and female mice (22-24 months) are less sensitive to mechanical, thermal (heat), and itch-provoking stimuli than are younger mice (6 months).


Subject(s)
Pain , Pruritus , Male , Female , Mice , Animals , Hyperalgesia/etiology
18.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798460

ABSTRACT

T cells have emerged as sex-dependent orchestrators of pain chronification but the sexually dimorphic mechanisms by which T cells control pain sensitivity is not resolved. Here, we demonstrate an influence of regulatory T cells (Tregs) on pain processing that is distinct from their canonical functions of immune regulation and tissue repair. Specifically, meningeal Tregs (mTregs) express the endogenous opioid, enkephalin, and mTreg-derived enkephalin exerts an antinociceptive action through a presynaptic opioid receptor signaling mechanism that is dispensable for immunosuppression. mTregs are both necessary and sufficient for suppressing mechanical pain sensitivity in female but not male mice. Notably, the mTreg modulation of pain thresholds depends on sex-hormones and expansion of enkephalinergic mTregs during gestation imparts a remarkable pregnancy-induced analgesia in a pre-existing, chronic, unremitting neuropathic pain model. These results uncover a fundamental sex-specific, pregnancy-pronounced, and immunologically-derived endogenous opioid circuit for nociceptive regulation with critical implications for pain biology and maternal health.

19.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38328157

ABSTRACT

Large library docking can reveal unexpected chemotypes that complement the structures of biological targets. Seeking new agonists for the cannabinoid-1 receptor (CB1R), we docked 74 million tangible molecules, prioritizing 46 high ranking ones for de novo synthesis and testing. Nine were active by radioligand competition, a 20% hit-rate. Structure-based optimization of one of the most potent of these (Ki = 0.7 uM) led to '4042, a 1.9 nM ligand and a full CB1R agonist. A cryo-EM structure of the purified enantiomer of '4042 ('1350) in complex with CB1R-Gi1 confirmed its docked pose. The new agonist was strongly analgesic, with generally a 5-10-fold therapeutic window over sedation and catalepsy and no observable conditioned place preference. These findings suggest that new cannabinoid chemotypes may disentangle characteristic cannabinoid side-effects from their analgesia, supporting the further development of cannabinoids as pain therapeutics.

20.
J Physiol ; 591(4): 1097-110, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23266932

ABSTRACT

We previously demonstrated that genetic and/or pharmacological ablation of the TRPV1+/peptidergic or the MrgprD+/non-peptidergic subset of nociceptors produced selective, modality-specific deficits in the behavioural responses to heat and mechanical stimuli, respectively. To assess whether this modality-specific contribution is also manifest at the level of spinal cord neuron responsiveness, here we made extracellular recordings from lumbar dorsal horn neurons of the mouse in response to graded thermal and mechanical stimulation. We found that, following intrathecal injection of capsaicin to eliminate the central terminals of TRPV1+ nociceptors, neurons in the region of laminae I and V of the spinal cord lost responsiveness to noxious heat (whether generated by a contact heat probe or diode laser), with no change in their response to noxious mechanical stimulation. In contrast, ablation of MrgprD+ afferents did not alter the response to noxious heat, but reduced the firing of superficial dorsal horn nociceptive-specific neurons in response to graded mechanical stimulation and decreased the relative number of wide dynamic range neurons that were exclusively mechanosensitive. Neither ablation procedure reduced the number of dorsal horn neurons that responded to noxious cold. These findings support the conclusion that TRPV1+ nociceptors are necessary and probably sufficient for noxious heat activation of dorsal horn neurons and that, despite their polymodal properties, TRPV1+ and MrgprD+ nociceptors provide modality-specific contributions to the response properties of spinal cord neurons.


Subject(s)
Nociceptors/physiology , Posterior Horn Cells/physiology , Receptors, G-Protein-Coupled/physiology , TRPV Cation Channels/physiology , Animals , Hot Temperature , Mice , Mice, Inbred C57BL , Peptides , Physical Stimulation
SELECTION OF CITATIONS
SEARCH DETAIL