Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
J Pain ; 25(1): 88-100, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37524219

ABSTRACT

The platinum chemotherapeutic oxaliplatin produces dose-limiting pain, dysesthesia, and cold hypersensitivity in most patients immediately after infusion. An improved understanding of the mechanisms underlying these symptoms is urgently required to facilitate the development of symptomatic or preventative therapies. In this study, we have used skin-saphenous nerve recordings in vitro and behavioral experiments in mice to characterize the direct effects of oxaliplatin on different types of sensory afferent fibers. Our results confirmed that mice injected with oxaliplatin rapidly develop mechanical and cold hypersensitivities. We further noted profound changes to A fiber activity after the application of oxaliplatin to the receptive fields in the skin. Most oxaliplatin-treated Aδ- and rapidly adapting Aß-units lost mechanical sensitivity, but units that retained responsiveness additionally displayed a novel, aberrant cold sensitivity. Slowly adapting Aß-units did not display mechanical tachyphylaxis, and a subset of these fibers was sensitized to mechanical and cold stimulation after oxaliplatin treatment. C fiber afferents were less affected by acute applications of oxaliplatin, but a subset gained cold sensitivity. Taken together, our findings suggest that direct effects on peripheral A fibers play a dominant role in the development of acute oxaliplatin-induced cold hypersensitivity, numbness, and dysesthesia. PERSPECTIVE: The chemotherapeutic drug oxaliplatin rapidly gives rise to dose-limiting cold pain and dysesthesia. Here, we have used behavioral and electrophysiological studies of mice to characterize the responsible neurons. We show that oxaliplatin directly confers aberrant cold responsiveness to subsets of A-fibers while silencing other fibers of the same type.


Subject(s)
Antineoplastic Agents , Cryopyrin-Associated Periodic Syndromes , Humans , Mice , Animals , Oxaliplatin/adverse effects , Paresthesia , Cryopyrin-Associated Periodic Syndromes/chemically induced , Pain , Hyperalgesia/chemically induced , Antineoplastic Agents/adverse effects
2.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34196305

ABSTRACT

Fibromyalgia syndrome (FMS) is characterized by widespread pain and tenderness, and patients typically experience fatigue and emotional distress. The etiology and pathophysiology of fibromyalgia are not fully explained and there are no effective drug treatments. Here we show that IgG from FMS patients produced sensory hypersensitivity by sensitizing nociceptive neurons. Mice treated with IgG from FMS patients displayed increased sensitivity to noxious mechanical and cold stimulation, and nociceptive fibers in skin-nerve preparations from mice treated with FMS IgG displayed an increased responsiveness to cold and mechanical stimulation. These mice also displayed reduced locomotor activity, reduced paw grip strength, and a loss of intraepidermal innervation. In contrast, transfer of IgG-depleted serum from FMS patients or IgG from healthy control subjects had no effect. Patient IgG did not activate naive sensory neurons directly. IgG from FMS patients labeled satellite glial cells and neurons in vivo and in vitro, as well as myelinated fiber tracts and a small number of macrophages and endothelial cells in mouse dorsal root ganglia (DRG), but no cells in the spinal cord. Furthermore, FMS IgG bound to human DRG. Our results demonstrate that IgG from FMS patients produces painful sensory hypersensitivities by sensitizing peripheral nociceptive afferents and suggest that therapies reducing patient IgG titers may be effective for fibromyalgia.


Subject(s)
Fibromyalgia/immunology , Fibromyalgia/physiopathology , Animals , Case-Control Studies , Disease Models, Animal , Female , Fibromyalgia/etiology , Ganglia, Spinal/physiopathology , Humans , Immunization, Passive , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Male , Mice , Mice, Inbred C57BL , Nociceptors/immunology , Nociceptors/physiology , Pain/physiopathology , Pain Threshold/physiology
3.
Diabetes ; 69(12): 2667-2677, 2020 12.
Article in English | MEDLINE | ID: mdl-32994272

ABSTRACT

Animal models are important tools in diabetes research because ethical and logistical constraints limit access to human tissue. ß-Cell dysfunction is a common contributor to the pathogenesis of most types of diabetes. Spontaneous hyperglycemia was developed in a colony of C57BL/6J mice at King's College London (KCL). Sequencing identified a mutation in the Ins2 gene, causing a glycine-to-serine substitution at position 32 on the B chain of the preproinsulin 2 molecule. Mice with the Ins2 +/G32S mutation were named KCL Ins2 G32S (KINGS) mice. The same mutation in humans (rs80356664) causes dominantly inherited neonatal diabetes. Mice were characterized, and ß-cell function was investigated. Male mice became overtly diabetic at ∼5 weeks of age, whereas female mice had only slightly elevated nonfasting glycemia. Islets showed decreased insulin content and impaired glucose-induced insulin secretion, which was more severe in males. Transmission electron microscopy and studies of gene and protein expression showed ß-cell endoplasmic reticulum (ER) stress in both sexes. Despite this, ß-cell numbers were only slightly reduced in older animals. In conclusion, the KINGS mouse is a novel model of a human form of diabetes that may be useful to study ß-cell responses to ER stress.


Subject(s)
Diabetes Mellitus/genetics , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Insulin-Secreting Cells/metabolism , Animals , Ecosystem , Female , Glucose Tolerance Test , Humans , Insulin/blood , Male , Mice , Mice, Inbred Strains , Mutation , Polymorphism, Single Nucleotide
4.
J Neurosci ; 39(40): 7840-7852, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31451581

ABSTRACT

Transient receptor potential melastatin 3 (TRPM3) is a nonselective cation channel that is inhibited by Gßγ subunits liberated following activation of Gαi/o protein-coupled receptors. Here, we demonstrate that TRPM3 channels are also inhibited by Gßγ released from Gαs and Gαq Activation of the Gs-coupled adenosine 2B receptor and the Gq-coupled muscarinic acetylcholine M1 receptor inhibited the activity of TRPM3 heterologously expressed in HEK293 cells. This inhibition was prevented when the Gßγ sink ßARK1-ct (C terminus of ß-adrenergic receptor kinase-1) was coexpressed with TRPM3. In neurons isolated from mouse dorsal root ganglion (DRG), native TRPM3 channels were inhibited by activating Gs-coupled prostaglandin-EP2 and Gq-coupled bradykinin B2 (BK2) receptors. The Gi/o inhibitor pertussis toxin and inhibitors of PKA and PKC had no effect on EP2- and BK2-mediated inhibition of TRPM3, demonstrating that the receptors did not act through Gαi/o or through the major protein kinases activated downstream of G-protein-coupled receptor (GPCR) activation. When DRG neurons were dialyzed with GRK2i, which sequesters free Gßγ protein, TRPM3 inhibition by EP2 and BK2 was significantly reduced. Intraplantar injections of EP2 or BK2 agonists inhibited both the nocifensive response evoked by TRPM3 agonists, and the heat hypersensitivity produced by Freund's Complete Adjuvant (FCA). Furthermore, FCA-induced heat hypersensitivity was completely reversed by the selective TRPM3 antagonist ononetin in WT mice and did not develop in Trpm3-/- mice. Our results demonstrate that TRPM3 is subject to promiscuous inhibition by Gßγ protein in heterologous expression systems, primary neurons and in vivo, and suggest a critical role for this ion channel in inflammatory heat hypersensitivity.SIGNIFICANCE STATEMENT The ion channel TRPM3 is widely expressed in the nervous system. Recent studies showed that Gαi/o-coupled GPCRs inhibit TRPM3 through a direct interaction between Gßγ subunits and TRPM3. Since Gßγ proteins can be liberated from other Gα subunits than Gαi/o, we examined whether activation of Gs- and Gq-coupled receptors also influence TRPM3 via Gßγ. Our results demonstrate that activation of Gs- and Gq-coupled GPCRs in recombinant cells and sensory neurons inhibits TRPM3 via Gßγ liberation. We also demonstrated that Gs- and Gq-coupled receptors inhibit TRPM3 in vivo, thereby reducing pain produced by activation of TRPM3, and inflammatory heat hypersensitivity. Our results identify Gßγ inhibition of TRPM3 as an effector mechanism shared by the major Gα subunits.


Subject(s)
GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , Receptors, G-Protein-Coupled/physiology , TRPM Cation Channels/physiology , Animals , Behavior, Animal , Female , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Nociceptors/drug effects , Pertussis Toxin/pharmacology , Receptor, Adenosine A2B/physiology , Receptor, Muscarinic M1/physiology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/physiology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
5.
Pain ; 160(12): 2855-2865, 2019 12.
Article in English | MEDLINE | ID: mdl-31343542

ABSTRACT

Complex regional pain syndrome (CRPS) is a posttraumatic pain condition with an incompletely understood pathophysiological basis. Here, we have examined the cellular basis of pain in CRPS using behavioral and electrophysiological methods in mice treated with IgG from CRPS patients, in combination with a paw incision. Mice were subjected to a hind paw skin-muscle incision alone, or in combination with administration of IgG purified from either healthy control subjects or patients with persistent CRPS. Nociceptive function was examined behaviorally in vivo, and electrophysiologically in vitro using skin-nerve preparations to study the major classes of mechanosensitive single units. Administration of IgG from CRPS patients exacerbated and prolonged the postsurgical hypersensitivity to noxious mechanical, cold, and heat stimulation, but did not influence tactile sensitivity after a paw incision. Studies of IgG preparations pooled from patient cohorts (n = 26-27) show that pathological autoantibodies are present in the wider population of patients with persistent CRPS, and that patients with more severe pain have higher effective autoantibody titres than patients with moderate pain intensity. Electrophysiological investigation of skin-nerve preparations from mice treated with CRPS IgG from a single patient identified both a significantly increased evoked impulse activity in A and C nociceptors, and an increased spontaneous impulse rate in the intact saphenous nerve. Our results show that painful hypersensitivity in persistent CRPS is maintained by autoantibodies, which act by sensitizing A and C nociceptors.


Subject(s)
Autoantibodies , Complex Regional Pain Syndromes/physiopathology , Hyperalgesia/physiopathology , Nociceptors/physiology , Pain Threshold/physiology , Animals , Disease Models, Animal , Humans , Immunoglobulin G , Mice , Pain Measurement , Skin/innervation
6.
Front Mol Neurosci ; 11: 332, 2018.
Article in English | MEDLINE | ID: mdl-30258352

ABSTRACT

Traditionally, neuroscience has had to rely on mixed tissue analysis to examine transcriptional and epigenetic changes in the context of nervous system function or pathology. However, particularly when studying chronic pain conditions, this approach can be flawed, since it neglects to take into account the shifting contribution of different cell types across experimental conditions. Here, we demonstrate this using the example of DNA methyltransferases (DNMTs) - a group of epigenetic modifiers consisting of Dnmt1, Dnmt3a, and Dnmt3b in mammalian cells. We used sensory neuron-specific knockout mice for Dnmt3a/3b as well as pharmacological blockade of Dnmt1 to study their role in nociception. In contrast to previous analyses on whole tissue, we find that Dnmt3a and 3b protein is not expressed in adult DRG neurons, that none of the DNA methyltransferases are regulated with injury and that interfering with their function has no effect on nociception. Our results therefore currently do not support a role for neuronal DNA methyltransferases in pain processing in adult animals.

7.
Diabetes ; 67(8): 1650-1662, 2018 08.
Article in English | MEDLINE | ID: mdl-29875100

ABSTRACT

The mechanisms responsible for painful and insensate diabetic neuropathy are not completely understood. Here, we have investigated sensory neuropathy in the Ins2+/Akita mouse, a hereditary model of diabetes. Akita mice become diabetic soon after weaning, and we show that this is accompanied by an impaired mechanical and thermal nociception and a significant loss of intraepidermal nerve fibers. Electrophysiological investigations of skin-nerve preparations identified a reduced rate of action potential discharge in Ins2+/Akita mechanonociceptors compared with wild-type littermates, whereas the function of low-threshold A-fibers was essentially intact. Studies of isolated sensory neurons demonstrated a markedly reduced heat responsiveness in Ins2+/Akita dorsal root ganglion (DRG) neurons, but a mostly unchanged function of cold-sensitive neurons. Restoration of normal glucose control by islet transplantation produced a rapid recovery of nociception, which occurred before normoglycemia had been achieved. Islet transplantation also restored Ins2+/Akita intraepidermal nerve fiber density to the same level as wild-type mice, indicating that restored insulin production can reverse both sensory and anatomical abnormalities of diabetic neuropathy in mice. The reduced rate of action potential discharge in nociceptive fibers and the impaired heat responsiveness of Ins2+/Akita DRG neurons suggest that ionic sensory transduction and transmission mechanisms are modified by diabetes.


Subject(s)
Diabetic Neuropathies/metabolism , Epidermis/innervation , Ganglia, Spinal/metabolism , Insulin/metabolism , Nerve Fibers, Unmyelinated/metabolism , Somatosensory Disorders/metabolism , Thermoreceptors/metabolism , Action Potentials , Amino Acid Substitution , Animals , Behavior, Animal , Cells, Cultured , Diabetes Mellitus/blood , Diabetes Mellitus/surgery , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Diabetic Neuropathies/prevention & control , Epidermis/metabolism , Epidermis/pathology , Epidermis/physiopathology , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Heterozygote , Insulin/genetics , Islets of Langerhans Transplantation , Kidney , Male , Mechanoreceptors/metabolism , Mechanoreceptors/pathology , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Fibers, Unmyelinated/pathology , Pain Measurement , Somatosensory Disorders/complications , Somatosensory Disorders/physiopathology , Somatosensory Disorders/prevention & control , Thermoreceptors/pathology , Thermoreceptors/physiopathology , Transplantation, Heterotopic
8.
Elife ; 62017 08 15.
Article in English | MEDLINE | ID: mdl-28826490

ABSTRACT

Transient receptor potential (TRP) ion channels in peripheral sensory neurons are functionally regulated by hydrolysis of the phosphoinositide PI(4,5)P2 and changes in the level of protein kinase mediated phosphorylation following activation of various G protein coupled receptors. We now show that the activity of TRPM3 expressed in mouse dorsal root ganglion (DRG) neurons is inhibited by agonists of the Gi-coupled µ opioid, GABA-B and NPY receptors. These agonist effects are mediated by direct inhibition of TRPM3 by Gßγ subunits, rather than by a canonical cAMP mediated mechanism. The activity of TRPM3 in DRG neurons is also negatively modulated by tonic, constitutive GPCR activity as TRPM3 responses can be potentiated by GPCR inverse agonists. GPCR regulation of TRPM3 is also seen in vivo where Gi/o GPCRs agonists inhibited and inverse agonists potentiated TRPM3 mediated nociceptive behavioural responses.


Subject(s)
GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Ion Channels/drug effects , Sensory Receptor Cells/drug effects , TRPM Cation Channels/drug effects , Analgesics, Opioid/antagonists & inhibitors , Animals , Baclofen/antagonists & inhibitors , CHO Cells , Calcium/analysis , Capsaicin , Cricetulus , Electrophysiology/methods , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/antagonists & inhibitors , Pain/metabolism , Pain Measurement , Phosphatidylinositols/metabolism , Receptor, Cannabinoid, CB1/agonists , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
9.
Pain ; 158(7): 1314-1322, 2017 07.
Article in English | MEDLINE | ID: mdl-28394852

ABSTRACT

The immune and sensory systems are known for their close proximity and interaction. Indeed, in a variety of pain states, a myriad of different immune cells are activated and recruited, playing a key role in neuronal sensitisation. During inflammatory pain it is thought that mast cells (MC) are one of the immune cell types involved in this process, but so far the evidence outlining their direct effect on neuronal cells remains unclear. To clarify whether MC are involved in inflammatory pain states, we used a transgenic mouse line (Mctp5Cre-iDTR) in which MC could be depleted in an inducible manner by administration of diphtheria toxin. Our results show that ablation of MC in male mice did not result in any change in mechanical and thermal hypersensitivity in the CFA model of inflammatory pain. Similarly, edema and temperature triggered by CFA inflammation at the injection site remained identical in MC depleted mice compared with their littermate controls. In addition, we show that Mctp5Cre-iDTR mice display normal levels of mechanical hypersensitivity after local injection of nerve growth factor (NGF), a factor well characterised to produce peripheral sensitisation and for being upregulated upon injury and inflammation. We also demonstrate that NGF treatment in vitro does not lead to an increased level of tumor necrosis factor-α in bone marrow-derived MC. Furthermore, our qRT-PCR data reveal that MC express negligible levels of NGF receptors, thereby explaining the lack of response to NGF. Together, our data suggest that MC do not play a direct role in peripheral sensitisation during inflammatory conditions.


Subject(s)
Hyperalgesia/immunology , Mast Cells/immunology , Pain/immunology , Animals , Inflammation/immunology , Inflammation/metabolism , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Transgenic , Nerve Growth Factor/pharmacology , Pain/metabolism , Pain Measurement , Pain Threshold/drug effects , Tumor Necrosis Factor-alpha/metabolism
10.
Arthritis Res Ther ; 18: 7, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26754745

ABSTRACT

BACKGROUND: The effect of cold temperature on arthritis symptoms is unclear. The aim of this study was to investigate how environmental cold affects pain and blood flow in mono-arthritic mice, and examine a role for transient receptor potential ankyrin 1 (TRPA1), a ligand-gated cation channel that can act as a cold sensor. METHODS: Mono-arthritis was induced by unilateral intra-articular injection of complete Freund's adjuvant (CFA) in CD1 mice, and in mice either lacking TRPA1 (TRPA1 KO) or respective wildtypes (WT). Two weeks later, nociception and joint blood flow were measured following exposure to 10 °C (1 h) or room temperature (RT). Primary mechanical hyperalgesia in the knee was measured by pressure application apparatus; secondary mechanical hyperalgesia by automated von Frey system; thermal hyperalgesia by Hargreaves technique, and weight bearing by the incapacitance test. Joint blood flow was recorded by full-field laser perfusion imager (FLPI) and using clearance of (99m)Technetium. Blood flow was assessed after pretreatment with antagonists of either TRPA1 (HC-030031), substance P neurokinin 1 (NK1) receptors (SR140333) or calcitonin gene-related peptide (CGRP) (CGRP8-37). TRPA1, TAC-1 and CGRP mRNA levels were examined in dorsal root ganglia, synovial membrane and patellar cartilage samples. RESULTS: Cold exposure caused bilateral primary mechanical hyperalgesia 2 weeks after CFA injection, in a TRPA1-dependent manner. In animals maintained at RT, clearance techniques and FLPI showed that CFA-treated joints exhibited lower blood flow than saline-treated joints. In cold-exposed animals, this reduction in blood flow disappears, and increased blood flow in the CFA-treated joint is observed using FLPI. Cold-induced increased blood flow in CFA-treated joints was blocked by HC-030031 and not observed in TRPA1 KOs. Cold exposure increased TRPA1 mRNA levels in patellar cartilage, whilst reducing it in synovial membranes from CFA-treated joints. CONCLUSIONS: We provide evidence that environmental cold exposure enhances pain and increases blood flow in a mono-arthritis model. These changes are dependent on TRPA1. Thus, TRPA1 may act locally within the joint to influence blood flow via sensory nerves, in addition to its established nociceptive actions.


Subject(s)
Arthritis, Experimental/metabolism , Blood Flow Velocity/physiology , Cold Temperature/adverse effects , Freund's Adjuvant/toxicity , Joints/metabolism , Transient Receptor Potential Channels/biosynthesis , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Blood Flow Velocity/drug effects , Freund's Adjuvant/administration & dosage , Hindlimb/drug effects , Hindlimb/metabolism , Hindlimb/pathology , Injections, Intra-Articular , Joints/drug effects , Joints/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/drug effects , Pain Measurement/methods , Pain Threshold/drug effects , Pain Threshold/physiology , TRPA1 Cation Channel , Transient Receptor Potential Channels/deficiency
11.
Arthritis Rheumatol ; 68(4): 857-67, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26605536

ABSTRACT

OBJECTIVE: Pain is the most common symptom of osteoarthritis (OA), yet where it originates in the joint and how it is driven are unknown. The aim of this study was to identify pain-sensitizing molecules that are regulated in the joint when mice subjected to surgical joint destabilization develop OA-related pain behavior, the tissues in which these molecules are being regulated, and the factors that control their regulation. METHODS: Ten-week-old mice underwent sham surgery, partial meniscectomy, or surgical destabilization of the medial meniscus (DMM). Pain-related behavior as determined by a variety of methods (testing of responses to von Frey filaments, cold plate testing for cold sensitivity, analgesiometry, incapacitance testing, and forced flexion testing) was assessed weekly. Once pain-related behavior was established, RNA was extracted from either whole joints or microdissected tissue samples (articular cartilage, meniscus, and bone). Reverse transcription-polymerase chain reaction analysis was performed to analyze the expression of 54 genes known to regulate pain sensitization. Cartilage injury assays were performed using avulsed immature hips from wild-type or genetically modified mice or by explanting articular cartilage from porcine joints preinjected with pharmacologic inhibitors. Levels of nerve growth factor (NGF) protein were measured by enzyme-linked immunosorbent assay. RESULTS: Mice developed pain-related behavior 8 weeks after undergoing partial meniscectomy or 12 weeks after undergoing DMM. NGF, bradykinin receptors B1 and B2, tachykinin, and tachykinin receptor 1 were significantly regulated in the joints of mice displaying pain-related behavior. Little regulation of inflammatory cytokines, leukocyte activation markers, or chemokines was observed. When tissue samples from articular cartilage, meniscus, and bone were analyzed separately, NGF was consistently regulated in the articular cartilage. The other pain sensitizers were also largely regulated in the articular cartilage, although there were some differences between the 2 models. NGF and tachykinin were strongly regulated by simple mechanical injury of cartilage in vitro in a transforming growth factor ß-activated kinase 1-, fibroblast growth factor 2-, and Src kinase-dependent manner. CONCLUSION: Damaged joint tissues produce proalgesic molecules, including NGF, in murine OA.


Subject(s)
Behavior, Animal , Bone and Bones/metabolism , Cartilage, Articular/metabolism , Menisci, Tibial/metabolism , Nociceptive Pain/genetics , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2 , Gene Expression Regulation , MAP Kinase Kinase Kinases , Mice , Nerve Growth Factor/genetics , Nociceptive Pain/metabolism , Osteoarthritis, Knee , Pain/genetics , Pain/metabolism , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/genetics , Receptors, Neurokinin-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Swine , Tachykinins/genetics , Tibial Meniscus Injuries , src-Family Kinases
12.
Sci Rep ; 5: 12771, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26227887

ABSTRACT

Acetaminophen (APAP) is an effective antipyretic and one of the most commonly used analgesic drugs. Unlike antipyretic non-steroidal anti-inflammatory drugs, APAP elicits hypothermia in addition to its antipyretic effect. Here we have examined the mechanisms responsible for the hypothermic activity of APAP. Subcutaneous, but not intrathecal, administration of APAP elicited a dose dependent decrease in body temperature in wildtype mice. Hypothermia was abolished in mice pre-treated with resiniferatoxin to destroy or defunctionalize peripheral TRPV1-expressing terminals, but resistant to inhibition of cyclo-oxygenases. The hypothermic activity was independent of TRPV1 since APAP evoked hypothermia was identical in wildtype and Trpv1(-/-) mice, and not reduced by administration of a maximally effective dose of a TRPV1 antagonist. In contrast, a TRPA1 antagonist inhibited APAP induced hypothermia and APAP was without effect on body temperature in Trpa1(-/-) mice. In a model of yeast induced pyrexia, administration of APAP evoked a marked hypothermia in wildtype and Trpv1(-/-) mice, but only restored normal body temperature in Trpa1(-/-) and Trpa1(-/-)/Trpv1(-/-) mice. We conclude that TRPA1 mediates APAP evoked hypothermia.


Subject(s)
Acetaminophen/pharmacology , Hypothermia, Induced , Transient Receptor Potential Channels/metabolism , Acrolein/analogs & derivatives , Acrolein/pharmacology , Animals , Antipyretics/pharmacology , Benzoquinones/pharmacokinetics , Diterpenes/pharmacology , Female , Hypothermia/metabolism , Imines/pharmacokinetics , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Sensory Receptor Cells/drug effects , TRPA1 Cation Channel , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/genetics
13.
J Neurosci ; 35(23): 8959-69, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26063927

ABSTRACT

G-protein receptor 84 (GPR84) is an orphan receptor that is induced markedly in monocytes/macrophages and microglia during inflammation, but its pathophysiological function is unknown. Here, we investigate the role of GPR84 in a murine model of traumatic nerve injury. Naive GPR84 knock-out (KO) mice exhibited normal behavioral responses to acute noxious stimuli, but subsequent to partial sciatic nerve ligation (PNL), KOs did not develop mechanical or thermal hypersensitivity, in contrast to wild-type (WT) littermates. Nerve injury increased ionized calcium binding adapter molecule 1 (Iba1) and phosphorylated p38 MAPK immunoreactivity in the dorsal horn and Iba1 and cluster of differentiation 45 expression in the sciatic nerve, with no difference between genotypes. PCR array analysis revealed that Gpr84 expression was upregulated in the spinal cord and sciatic nerve of WT mice. In addition, the expression of arginase-1, a marker for anti-inflammatory macrophages, was upregulated in KO sciatic nerve. Based on this evidence, we investigated whether peripheral macrophages behave differently in the absence of GPR84. We found that lipopolysaccharide-stimulated KO macrophages exhibited attenuated expression of several proinflammatory mediators, including IL-1ß, IL-6, and TNF-α. Forskolin-stimulated KO macrophages also showed greater cAMP induction, a second messenger associated with immunosuppression. In summary, our results demonstrate that GPR84 is a proinflammatory receptor that contributes to nociceptive signaling via the modulation of macrophages, whereas in its absence the response of these cells to an inflammatory insult is impaired.


Subject(s)
Gene Expression Regulation/genetics , Pain Threshold/physiology , Receptors, G-Protein-Coupled/metabolism , Sciatica/metabolism , Sciatica/physiopathology , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Hypersensitivity/etiology , Hypersensitivity/genetics , Inflammation/etiology , Inflammation/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Pain Measurement , Physical Stimulation/adverse effects , Receptors, G-Protein-Coupled/genetics , Sciatica/pathology , Spinal Cord/metabolism , Temperature , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Invest Dermatol ; 135(10): 2484-2491, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25955385

ABSTRACT

A role for proteinase-activated receptor-4 (PAR-4) was recently suggested in itch sensation. Here, we investigated the mechanisms underlying the pruriceptive actions of the selective PAR-4 agonist AYPGKF-NH2 (AYP) in mice. Dorsal intradermal (i.d.) administration of AYP elicited intense scratching behavior in mice, which was prevented by the selective PAR-4 antagonist (pepducin P4pal-10). PAR-4 was found to be coexpressed in 32% of tryptase-positive skin mast cells, and AYP caused a 2-fold increase in mast cell degranulation. However, neither the treatment with cromolyn nor the deficiency of mast cells (WBB6F1-Kit(W/Wv) mice) was able to affect AYP-induced itch. PAR-4 was also found on gastrin-releasing peptide (GRP)-positive neurons (pruriceptive fibers), and AYP-induced itch was reduced by the selective GRP receptor antagonist RC-3095. In addition, AYP evoked calcium influx in ∼1.5% of cultured DRG neurons also sensitive to TRPV1 (capsaicin) and/or TRPA1 (AITC) agonists. Importantly, AYP-induced itch was reduced by treatment with either the selective TRPV1 (SB366791), TRPA1 (HC-030031), or NK1 (FK888) receptor antagonists. However, genetic loss of TRPV1, but not of TRPA1, diminished AYP-induced calcium influx in DRG neurons and the scratching behavior in mice. These findings provide evidence that PAR-4 activation by AYP causes pruriceptive itch in mice via a TRPV1/TRPA1-dependent mechanism.


Subject(s)
Capsaicin/pharmacology , Pruritus/physiopathology , Receptors, Bombesin/metabolism , Receptors, Thrombin/metabolism , Transient Receptor Potential Channels/drug effects , Animals , Behavior, Animal , Cells, Cultured , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Immunohistochemistry , Injections, Intradermal , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pruritus/chemically induced , Pruritus/psychology , Random Allocation , Reference Values , Signal Transduction , Transient Receptor Potential Channels/metabolism
15.
Nat Commun ; 6: 7150, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25998021

ABSTRACT

Specific peripheral sensory neurons respond to increases in extracellular osmolality but the mechanism responsible for excitation is unknown. Here we show that small increases in osmolality excite isolated mouse dorsal root ganglion (DRG) and trigeminal ganglion (TG) neurons expressing the cold-sensitive TRPM8 channel (transient receptor potential channel, subfamily M, member 8). Hyperosmotic responses were abolished by TRPM8 antagonists, and were absent in DRG and TG neurons isolated from Trpm8(-/-) mice. Heterologously expressed TRPM8 was activated by increased osmolality around physiological levels and inhibited by reduced osmolality. Electrophysiological studies in a mouse corneal preparation demonstrated that osmolality regulated the electrical activity of TRPM8-expressing corneal afferent neurons. Finally, the frequency of eye blinks was reduced in Trpm8(-/-) compared with wild-type mice and topical administration of a TRPM8 antagonist reduced blinking in wild-type mice. Our findings identify TRPM8 as a peripheral osmosensor responsible for the regulation of normal eye-blinking in mice.


Subject(s)
Blinking , Sensory Receptor Cells/physiology , TRPM Cation Channels/physiology , Action Potentials , Animals , CHO Cells , Cold Temperature , Cornea/physiology , Cricetinae , Cricetulus , Female , Male , Mice , Mice, Knockout , Osmolar Concentration
16.
J Biol Chem ; 290(24): 15185-96, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25903127

ABSTRACT

Streptozotocin (STZ)-induced diabetes is the most commonly used animal model of diabetes. Here, we have demonstrated that intraplantar injections of low dose STZ evoked acute polymodal hypersensitivities in mice. These hypersensitivities were inhibited by a TRPA1 antagonist and were absent in TRPA1-null mice. In wild type mice, systemic STZ treatment (180 mg/kg) evoked a loss of cold and mechanical sensitivity within an hour of injection, which lasted for at least 10 days. In contrast, Trpa1(-/-) mice developed mechanical, cold, and heat hypersensitivity 24 h after STZ. The TRPA1-dependent sensory loss produced by STZ occurs before the onset of diabetes and may thus not be readily distinguished from the similar sensory abnormalities produced by the ensuing diabetic neuropathy. In vitro, STZ activated TRPA1 in isolated sensory neurons, TRPA1 cell lines, and membrane patches. Mass spectrometry studies revealed that STZ oxidizes TRPA1 cysteines to disulfides and sulfenic acids. Furthermore, incubation of tyrosine with STZ resulted in formation of dityrosine, suggesting formation of peroxynitrite. Functional analysis of TRPA1 mutants showed that cysteine residues that were oxidized by STZ were important for TRPA1 responsiveness to STZ. Our results have identified oxidation of TRPA1 cysteine residues, most likely by peroxynitrite, as a novel mechanism of action of STZ. Direct stimulation of TRPA1 complicates the interpretation of results from STZ models of diabetic sensory neuropathy and strongly argues that more refined models of diabetic neuropathy should replace the use of STZ.


Subject(s)
Peroxynitrous Acid/metabolism , Streptozocin/pharmacology , Transient Receptor Potential Channels/drug effects , Analgesics/pharmacology , Animals , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , TRPA1 Cation Channel , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/genetics
17.
Pharmacol Res Perspect ; 3(6): e00191, 2015 Dec.
Article in English | MEDLINE | ID: mdl-27022465

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a sensor of nociceptive stimuli, expressed predominantly in a subpopulation of peptidergic sensory neurons which co-express the noxious heat-sensor transient receptor potential vanilloid 1. In this study, we describe a spinal cord synaptosome-calcitonin gene-related peptide (CGRP) release assay for examining activation of TRPA1 natively expressed on the central terminals of dorsal root ganglion neurons. We have shown for the first time that activation of TRPA1 channels expressed on spinal cord synaptosomes by a selection of agonists evokes a concentration-dependent release of CGRP which is inhibited by TRPA1 antagonists. In addition, our results demonstrate that depolarization of spinal cord synaptosomes by a high concentration of KCl induces CGRP release via a T-type calcium channel-dependent mechanism whilst TRPA1-induced CGRP release functions independently of voltage-gated calcium channel activation. Finally, we have shown that pre-treatment of synaptosomes with the opioid agonist, morphine, results in a reduction of depolarization-induced CGRP release. This study has demonstrated the use of a dorsal spinal cord homogenate assay for investigation of natively expressed TRPA1 channels and for modulation of depolarizing stimuli at the level of the dorsal spinal cord.

18.
Diabetes ; 64(4): 1202-10, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25325736

ABSTRACT

Stimulus-coupled incretin secretion from enteroendocrine cells plays a fundamental role in glucose homeostasis and could be targeted for the treatment of type 2 diabetes. Here, we investigated the expression and function of transient receptor potential (TRP) ion channels in enteroendocrine L cells producing GLP-1. By microarray and quantitative PCR analysis, we identified trpa1 as an L cell-enriched transcript in the small intestine. Calcium imaging of primary L cells and the model cell line GLUTag revealed responses triggered by the TRPA1 agonists allyl-isothiocyanate (mustard oil), carvacrol, and polyunsaturated fatty acids, which were blocked by TRPA1 antagonists. Electrophysiology in GLUTag cells showed that carvacrol induced a current with characteristics typical of TRPA1 and triggered the firing of action potentials. TRPA1 activation caused an increase in GLP-1 secretion from primary murine intestinal cultures and GLUTag cells, an effect that was abolished in cultures from trpa1(-/-) mice or by pharmacological TRPA1 inhibition. These findings present TRPA1 as a novel sensory mechanism in enteroendocrine L cells, coupled to the facilitation of GLP-1 release, which may be exploitable as a target for treating diabetes.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Intestine, Small/metabolism , Signal Transduction/physiology , Transient Receptor Potential Channels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Enteroendocrine Cells/cytology , Intestine, Small/cytology , Mice , Mice, Knockout , TRPA1 Cation Channel , Transient Receptor Potential Channels/genetics
19.
Nat Commun ; 5: 5732, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25501034

ABSTRACT

The cold-induced vascular response, consisting of vasoconstriction followed by vasodilatation, is critical for protecting the cutaneous tissues against cold injury. Whilst this physiological reflex response is historic knowledge, the mechanisms involved are unclear. Here by using a murine model of local environmental cold exposure, we show that TRPA1 acts as a primary vascular cold sensor, as determined through TRPA1 pharmacological antagonism or gene deletion. The initial cold-induced vasoconstriction is mediated via TRPA1-dependent superoxide production that stimulates α2C-adrenoceptors and Rho-kinase-mediated MLC phosphorylation, downstream of TRPA1 activation. The subsequent restorative blood flow component is also dependent on TRPA1 activation being mediated by sensory nerve-derived dilator neuropeptides CGRP and substance P, and also nNOS-derived NO. The results allow a new understanding of the importance of TRPA1 in cold exposure and provide impetus for further research into developing therapeutic agents aimed at the local protection of the skin in disease and adverse climates.


Subject(s)
Hypothermia/metabolism , Receptors, Adrenergic, alpha/genetics , Skin/blood supply , Transient Receptor Potential Channels/genetics , Vasoconstriction/genetics , Acetanilides/pharmacology , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Cold Temperature/adverse effects , Gene Expression Regulation , Hindlimb , Hypothermia/etiology , Hypothermia/genetics , Hypothermia/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Phosphorylation , Purines/pharmacology , Receptors, Adrenergic, alpha/metabolism , Signal Transduction , Skin/metabolism , Skin/pathology , Substance P/genetics , Substance P/metabolism , Superoxides/metabolism , TRPA1 Cation Channel , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/deficiency , Vasodilation/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
20.
J Clin Invest ; 124(5): 2023-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24743146

ABSTRACT

A major dose-limiting side effect associated with cancer-treating antineoplastic drugs is the development of neuropathic pain, which is not readily relieved by available analgesics. A better understanding of the mechanisms that underlie pain generation has potential to provide targets for prophylactic management of chemotherapy pain. Here, we delineate a pathway for pain that is induced by the chemotherapeutic drug vincristine sulfate (VCR). In a murine model of chemotherapy-induced allodynia, VCR treatment induced upregulation of endothelial cell adhesion properties, resulting in the infiltration of circulating CX3CR1⁺ monocytes into the sciatic nerve. At the endothelial-nerve interface, CX3CR1⁺ monocytes were activated by the chemokine CX3CL1 (also known as fractalkine [FKN]), which promoted production of reactive oxygen species that in turn activated the receptor TRPA1 in sensory neurons and evoked the pain response. Furthermore, mice lacking CX3CR1 exhibited a delay in the development of allodynia following VCR administration. Together, our data suggest that CX3CR1 antagonists and inhibition of FKN proteolytic shedding, possibly by targeting ADAM10/17 and/or cathepsin S, have potential as peripheral approaches for the prophylactic treatment of chemotherapy-induced pain.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Gene Expression Regulation/drug effects , Monocytes/metabolism , Pain/metabolism , Receptors, Chemokine/metabolism , Vincristine/adverse effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , CX3C Chemokine Receptor 1 , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Gene Expression Regulation/genetics , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/pathology , Mice , Mice, Knockout , Monocytes/pathology , Pain/chemically induced , Pain/genetics , Pain/pathology , Receptors, Chemokine/genetics , Vincristine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...