Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
BMC Biol ; 18(1): 197, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33317522

ABSTRACT

BACKGROUND: The mechanism underlying the pain symptoms associated with chemotherapeutic-induced peripheral neuropathy (CIPN) is poorly understood. Transient receptor potential ankyrin 1 (TRPA1), TRP vanilloid 4 (TRPV4), TRPV1, and oxidative stress have been implicated in several rodent models of CIPN-evoked allodynia. Thalidomide causes a painful CIPN in patients via an unknown mechanism. Surprisingly, the pathway responsible for such proalgesic response has not yet been investigated in animal models. RESULTS: Here, we reveal that a single systemic administration of thalidomide and its derivatives, lenalidomide and pomalidomide, elicits prolonged (~ 35 days) mechanical and cold hypersensitivity in C57BL/6J mouse hind paw. Pharmacological antagonism or genetic deletion studies indicated that both TRPA1 and TRPV4, but not TRPV1, contribute to mechanical allodynia, whereas cold hypersensitivity was entirely due to TRPA1. Thalidomide per se did not stimulate recombinant and constitutive TRPA1 and TRPV4 channels in vitro, which, however, were activated by the oxidative stress byproduct, hydrogen peroxide. Systemic treatment with an antioxidant attenuated mechanical and cold hypersensitivity, and the increase in oxidative stress in hind paw, sciatic nerve, and lumbar spinal cord produced by thalidomide. Notably, central (intrathecal) or peripheral (intraplantar) treatments with channel antagonists or an antioxidant revealed that oxidative stress-dependent activation of peripheral TRPA1 mediates cold allodynia and part of mechanical allodynia. However, oxidative stress-induced activation of central TRPV4 mediated the residual TRPA1-resistant component of mechanical allodynia. CONCLUSIONS: Targeting of peripheral TRPA1 and central TRPV4 may be required to attenuate pain associated with CIPN elicited by thalidomide and related drugs.


Subject(s)
Hyperalgesia/genetics , Oxidative Stress , Pain/genetics , TRPA1 Cation Channel/genetics , TRPV Cation Channels/genetics , Thalidomide/adverse effects , Animals , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C57BL , Pain/chemically induced , Rats , Rats, Sprague-Dawley , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
2.
Sci Rep ; 10(1): 8632, 2020 05 25.
Article in English | MEDLINE | ID: mdl-32451393

ABSTRACT

Pain evoked by visceral inflammation is often 'referred' to the somatic level. Transient receptor potential ankyrin 1 (TRPA1) has been reported to contribute to visceral pain-like behavior in dextran sulfate sodium (DSS)-evoked colitis. However, the role of TRPA1 in somatic component of hypersensitivity due to visceral inflammation is unknown. The present study investigated the role of TRPA1 in colitis-evoked mechanical hypersensitivity at the somatic level. Colitis was induced in mice by adding DSS to drinking water for one week. Control and DSS-treated mice were tested for various parameters of colitis as well as mechanical pain sensitivity in abdominal and facial regions. DSS treatment caused mechanical hypersensitivity in the abdominal and facial skin. Pharmacological blockade or genetic deletion of TRPA1 prevented the colitis-associated mechanical hypersensitivity in the abdominal and facial skin areas although the severity of colitis remained unaltered. DSS treatment increased expression of TRPA1 mRNA in cultured dorsal root ganglion (DRG) neurons, but not trigeminal ganglion neurons, and selectively enhanced currents evoked by the TRPA1 agonist, allyl isothiocyanate, in cultured DRG neurons. Our findings indicate that the TRPA1 channel contributes to colitis-associated mechanical hypersensitivity in somatic tissues, an effect associated with upregulation of TRPA1 expression and responsiveness in DRG nociceptors.


Subject(s)
Colitis/pathology , Nociceptive Pain/pathology , TRPA1 Cation Channel/metabolism , Acetanilides/pharmacology , Animals , Colitis/chemically induced , Dextran Sulfate/toxicity , Evoked Potentials/drug effects , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Isothiocyanates/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Purines/pharmacology , Stress, Mechanical , TRPA1 Cation Channel/antagonists & inhibitors , TRPA1 Cation Channel/genetics , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism
3.
J Cell Mol Med ; 23(3): 1976-1986, 2019 03.
Article in English | MEDLINE | ID: mdl-30636360

ABSTRACT

Safranal, contained in Crocus sativus L., exerts anti-inflammatory and analgesic effects. However, the underlying mechanisms for such effects are poorly understood. We explored whether safranal targets the transient receptor potential ankyrin 1 (TRPA1) channel, which in nociceptors mediates pain signals. Safranal by binding to specific cysteine/lysine residues, stimulates TRPA1, but not the TRP vanilloid 1 and 4 channels (TRPV1 and TRPV4), evoking calcium responses and currents in human cells and rat and mouse dorsal root ganglion (DRG) neurons. Genetic deletion or pharmacological blockade of TRPA1 attenuated safranal-evoked release of calcitonin gene-related peptide (CGRP) from rat and mouse dorsal spinal cord, and acute nociception in mice. Safranal contracted rat urinary bladder isolated strips in a TRPA1-dependent manner, behaving as a partial agonist. After exposure to safranal the ability of allyl isothiocyanate (TRPA1 agonist), but not that of capsaicin (TRPV1 agonist) or GSK1016790A (TRPV4 agonist), to evoke currents in DRG neurons, contraction of urinary bladder strips and CGRP release from spinal cord slices in rats, and acute nociception in mice underwent desensitization. As previously shown for other herbal extracts, including petasites or parthenolide, safranal might exert analgesic properties by partial agonism and selective desensitization of the TRPA1 channel.


Subject(s)
Analgesics/pharmacology , Crocus/chemistry , Cyclohexenes/pharmacology , Nociception/drug effects , TRPA1 Cation Channel/metabolism , Terpenes/pharmacology , Animals , Calcium Channels/metabolism , Cell Line , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Isothiocyanates/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Sesquiterpenes/pharmacology , TRPV Cation Channels/metabolism
4.
Brain ; 141(8): 2312-2328, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29985973

ABSTRACT

Glyceryl trinitrate is administered as a provocative test for migraine pain. Glyceryl trinitrate causes prolonged mechanical allodynia in rodents, which temporally correlates with delayed glyceryl trinitrate-evoked migraine attacks in patients. However, the underlying mechanism of the allodynia evoked by glyceryl trinitrate is unknown. The proalgesic transient receptor potential ankyrin 1 (TRPA1) channel, expressed by trigeminal nociceptors, is sensitive to oxidative stress and is targeted by nitric oxide or its by-products. Herein, we explored the role of TRPA1 in glyceryl trinitrate-evoked allodynia. Systemic administration of glyceryl trinitrate elicited in the mouse periorbital area an early and transient vasodilatation and a delayed and prolonged mechanical allodynia. The systemic, intrathecal or local administration of selective enzyme inhibitors revealed that nitric oxide, liberated from the parent drug by aldehyde dehydrogenase 2 (ALDH2), initiates but does not maintain allodynia. The central and the final phases of allodynia were respectively associated with generation of reactive oxygen and carbonyl species within the trigeminal ganglion. Allodynia was absent in TRPA1-deficient mice and was reversed by TRPA1 antagonists. Knockdown of neuronal TRPA1 by intrathecally administered antisense oligonucleotide and selective deletion of TRPA1 from sensory neurons in Advillin-Cre; Trpa1fl/fl mice revealed that nitric oxide-dependent oxidative and carbonylic stress generation is due to TRPA1 stimulation, and resultant NADPH oxidase 1 (NOX1) and NOX2 activation in the soma of trigeminal ganglion neurons. Early periorbital vasodilatation evoked by glyceryl trinitrate was attenuated by ALDH2 inhibition but was unaffected by TRPA1 blockade. Antagonists of the calcitonin gene-related peptide receptor did not affect the vasodilatation but partially inhibited allodynia. Thus, although both periorbital allodynia and vasodilatation evoked by glyceryl trinitrate are initiated by nitric oxide, they are temporally and mechanistically distinct. While vasodilatation is due to a direct nitric oxide action in the vascular smooth muscle, allodynia is a neuronal phenomenon mediated by TRPA1 activation and ensuing oxidative stress. The autocrine pathway, sustained by TRPA1 and NOX1/2 within neuronal cell bodies of trigeminal ganglia, may sensitize meningeal nociceptors and second order trigeminal neurons to elicit periorbital allodynia, and could be of relevance for migraine-like headaches evoked by glyceryl trinitrate in humans.


Subject(s)
NADPH Oxidase 1/physiology , TRPA1 Cation Channel/genetics , Trigeminal Ganglion/physiology , Aldehyde Dehydrogenase, Mitochondrial , Animals , Cell Body , Headache , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Migraine Disorders/chemically induced , Migraine Disorders/physiopathology , NADPH Oxidase 1/genetics , NADPH Oxidase 1/metabolism , Nitroglycerin/adverse effects , Nitroglycerin/pharmacology , Pain/metabolism , Sensory Receptor Cells , TRPA1 Cation Channel/physiology , Transient Receptor Potential Channels/antagonists & inhibitors
5.
Nat Commun ; 8(1): 1887, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29192190

ABSTRACT

It is known that transient receptor potential ankyrin 1 (TRPA1) channels, expressed by nociceptors, contribute to neuropathic pain. Here we show that TRPA1 is also expressed in Schwann cells. We found that in mice with partial sciatic nerve ligation, TRPA1 silencing in nociceptors attenuated mechanical allodynia, without affecting macrophage infiltration and oxidative stress, whereas TRPA1 silencing in Schwann cells reduced both allodynia and neuroinflammation. Activation of Schwann cell TRPA1 evoked NADPH oxidase 1 (NOX1)-dependent H2O2 release, and silencing or blocking Schwann cell NOX1 attenuated nerve injury-induced macrophage infiltration, oxidative stress and allodynia. Furthermore, the NOX2-dependent oxidative burst, produced by macrophages recruited to the perineural space activated the TRPA1-NOX1 pathway in Schwann cells, but not TRPA1 in nociceptors. Schwann cell TRPA1 generates a spatially constrained gradient of oxidative stress, which maintains macrophage infiltration to the injured nerve, and sends paracrine signals to activate TRPA1 of ensheathed nociceptors to sustain mechanical allodynia.


Subject(s)
Macrophages/immunology , Neuralgia/immunology , Schwann Cells/immunology , TRPA1 Cation Channel/immunology , Animals , Humans , Male , Mice , Mice, Inbred C57BL , NADPH Oxidase 1/genetics , NADPH Oxidase 1/immunology , NADPH Oxidase 2/genetics , NADPH Oxidase 2/immunology , Neuralgia/genetics , Oxidative Stress , Sciatic Nerve/immunology , TRPA1 Cation Channel/genetics
6.
Br J Pharmacol ; 174(17): 2897-2911, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28622417

ABSTRACT

BACKGROUND AND PURPOSE: The mechanism of the anti-migraine action of extracts of butterbur [Petasites hybridus (L.) Gaertn.] is unknown. Here, we investigated the ability of isopetasin, a major constituent of these extracts, to specifically target TRPA1 channel and to affect functional responses relevant to migraine. EXPERIMENTAL APPROACH: Single-cell calcium imaging and patch-clamp recordings in human and rodent TRPA1-expressing cells, neurogenic motor responses in rodent isolated urinary bladder, release of CGRP from mouse spinal cord in vitro and facial rubbing in mice and meningeal blood flow in rats were examined. KEY RESULTS: Isopetasin induced (i) calcium responses and currents in rat/mouse trigeminal ganglion (TG) neurons and in cells expressing the human TRPA1, (ii) substance P-mediated contractions of rat isolated urinary bladders and (iii) CGRP release from mouse dorsal spinal cord, responses that were selectively abolished by genetic deletion or pharmacological antagonism of TRPA1 channels. Pre-exposure to isopetasin produced marked desensitization of allyl isothiocyanate (AITC, TRPA1 channel agonist)- or capsaicin (TRPV1 channel agonist)-evoked currents in rat TG neurons, contractions of rat or mouse bladder and CGRP release from mouse central terminals of primary sensory neurons. Repeated intragastric administration of isopetasin attenuated mouse facial rubbing, evoked by local AITC or capsaicin, and dilation of rat meningeal arteries by acrolein or ethanol (TRPA1 and TRPV1 channel agonists respectively). CONCLUSION AND IMPLICATIONS: Activation of TRPA1 channels by isopetasin results in excitation of neuropeptide-containing nociceptors, followed by marked heterologous neuronal desensitization. Such atten uation in pain and neurogenic inflammation may account for the anti-migraine action of butterbur.


Subject(s)
Petasites , Plant Extracts/chemistry , Sesquiterpenes/pharmacology , TRPA1 Cation Channel/physiology , Animals , Cells, Cultured , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Migraine Disorders/drug therapy , Nociceptors/metabolism , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Spinal Cord/drug effects , Spinal Cord/physiology , Urinary Bladder/drug effects , Urinary Bladder/physiology
7.
Sci Transl Med ; 9(392)2017 05 31.
Article in English | MEDLINE | ID: mdl-28566424

ABSTRACT

Typically considered to be cell surface sensors of extracellular signals, heterotrimeric GTP-binding protein (G protein)-coupled receptors (GPCRs) control many pathophysiological processes and are the target of 30% of therapeutic drugs. Activated receptors redistribute to endosomes, but researchers have yet to explore whether endosomal receptors generate signals that control complex processes in vivo and are viable therapeutic targets. We report that the substance P (SP) neurokinin 1 receptor (NK1R) signals from endosomes to induce sustained excitation of spinal neurons and pain transmission and that specific antagonism of the NK1R in endosomes with membrane-anchored drug conjugates provides more effective and sustained pain relief than conventional plasma membrane-targeted antagonists. Pharmacological and genetic disruption of clathrin, dynamin, and ß-arrestin blocked SP-induced NK1R endocytosis and prevented SP-stimulated activation of cytosolic protein kinase C and nuclear extracellular signal-regulated kinase, as well as transcription. Endocytosis inhibitors prevented sustained SP-induced excitation of neurons in spinal cord slices in vitro and attenuated nociception in vivo. When conjugated to cholestanol to promote endosomal targeting, NK1R antagonists selectively inhibited endosomal signaling and sustained neuronal excitation. Cholestanol conjugation amplified and prolonged the antinociceptive actions of NK1R antagonists. These results reveal a critical role for endosomal signaling of the NK1R in the complex pathophysiology of pain and demonstrate the use of endosomally targeted GPCR antagonists.


Subject(s)
Endosomes/metabolism , Molecular Targeted Therapy , Nociception , Pain/drug therapy , Receptors, Neurokinin-1/metabolism , Signal Transduction , Animals , Cell Compartmentation , Clathrin/metabolism , Dynamins/metabolism , Endocytosis/drug effects , Endosomes/drug effects , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Lipids/chemistry , Models, Biological , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nociception/drug effects , Pain/pathology , Protein Binding/drug effects , Rats , Signal Transduction/drug effects , Spinal Cord/pathology , Subcellular Fractions/metabolism , Substance P/metabolism , beta-Arrestins/metabolism
8.
Microvasc Res ; 109: 38-44, 2017 01.
Article in English | MEDLINE | ID: mdl-27818253

ABSTRACT

The role of endogenous H2S has been highlighted as a gaseous transmitter. The vascular smooth muscle inhibitory effects of H2S have been characterized in isolated aorta and mesenteric arteries in rats and mice. Our study was aimed at investigating the vascular effects of H2S on human isolated mesenteric arteries and examining the underlying mechanisms involved. All experiments were performed on rings (4-8mm long) of human mesenteric arteries obtained from patients undergoing abdominal surgery. Ethical approval was obtained from the Ethics Committee of the University Hospital of the University of Florence (app. N. 2015/0024947). The effect of NaHS, an H2S donor, was determined using noradrenaline pre-contracted human isolated mesenteric rings. NaHS evoked a concentration-dependent relaxation (EC50 57µM). In contrast, homocysteine, an endogenous precursor of H2S, failed to affect human isolated mesenteric rings. Vasorelaxant response to NaHS was reduced by endothelium removal, application of the nitric oxide synthase inhibitor L-NAME and ODQ inhibitor of cyclic GMP. SQ 22536, an adenylate-cyclase inhibitor, failed to block NaHS-induced vasorelaxation. Inhibition of endogenous prostanoid production by indomethacin significantly reduced NaHS induced vasorelaxation. The role of potassium channels was also examined: blockers of the Ca2+-dependent potassium channel, charybdotoxin and apamin, failed to have any influence on the relaxant response to NaHS on this vascular tissue. In summary, H2S induced relaxation of isolated rings of human mesenteric arteries. Endothelium-dependent related mechanisms with the stimulation of ATP-sensitive potassium channels represents important cellular mechanisms for H2S effect on human mesenteric arteries.


Subject(s)
Hydrogen Sulfide/pharmacology , Mesenteric Arteries/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Oxadiazoles/pharmacology , Quinoxalines/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Calcium/metabolism , Endothelium, Vascular/metabolism , Humans , In Vitro Techniques , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels, Calcium-Activated/metabolism , Prostaglandins/metabolism
9.
Br J Pharmacol ; 174(1): 57-69, 2017 01.
Article in English | MEDLINE | ID: mdl-27759880

ABSTRACT

BACKGROUND AND PURPOSE: Peptides from venomous animals have long been important for understanding pain mechanisms and for the discovery of pain treatments. Here, we hypothesized that Phα1ß, a peptide from the venom of the armed spider Phoneutria nigriventer, produces analgesia by blocking the TRPA1 channel. EXPERIMENTAL APPROACH: Cultured rat dorsal root ganglion (DRG) neurons, human fetal lung fibroblasts (IMR90) or HEK293 cells expressing the human TRPA1 (hTRPA1-HEK293), human TRPV1 (hTRPV1-HEK293) or human TRPV4 channels (hTRPV4-HEK293), were used for calcium imaging and electrophysiology. Nociceptive responses induced by TRPA1, TRPV1 or TRPV4 agonists or by bortezomib were investigated in mice. KEY RESULTS: Phα1ß selectively inhibited calcium responses and currents evoked by the TRPA1 agonist, allyl isothiocyanate (AITC), on hTRPA1-HEK293, IMR90 fibroblasts and DRG neurons. Phα1ß did not affect calcium responses evoked by selective TRPV1 (capsaicin) or TRPV4 (GSK 1016790A) agonists on the various cell types. Intrathecal (i.t.) and intraplantar (i.pl.) administration of low doses of Phα1ß (up to 300 pmol per paw) attenuated acute nociception and mechanical and cold hyperalgesia evoked by AITC (i.t. or i.pl.), without affecting responses produced by capsaicin or hypotonic solution. Notably, Phα1ß abated the TRPA1-dependent neuropathic pain-like responses induced by bortezomib. In vitro and in vivo inhibition of TRPA1 by Phα1ß was reproduced by a recombinant form of the peptide, CTK 01512-2. CONCLUSIONS AND IMPLICATIONS: Phα1ß and CTK 01512-2 selectively target TRPA1, but not other TRP channels. This specific action underlines the potential of Phα1ß and CTK 01512-2 for pain treatment.


Subject(s)
Analgesics/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nociception/drug effects , Spider Venoms/chemistry , Transient Receptor Potential Channels/antagonists & inhibitors , Analgesics/chemistry , Animals , Calcium Channels/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuralgia/drug therapy , Neurons/drug effects , Rats , Spider Venoms/pharmacology , Spiders , Structure-Activity Relationship , TRPA1 Cation Channel , Transient Receptor Potential Channels/metabolism
10.
Cancer Res ; 76(23): 7024-7035, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27758889

ABSTRACT

Aromatase inhibitors (AI) induce painful musculoskeletal symptoms (AIMSS), which are dependent upon the pain transducing receptor TRPA1. However, as the AI concentrations required to engage TRPA1 in mice are higher than those found in the plasma of patients, we hypothesized that additional factors may cooperate to induce AIMSS. Here we report that the aromatase substrate androstenedione, unique among several steroid hormones, targeted TRPA1 in peptidergic primary sensory neurons in rodent and human cells expressing the native or recombinant channel. Androstenedione dramatically lowered the concentration of letrozole required to engage TRPA1. Notably, addition of a minimal dose of androstenedione to physiologically ineffective doses of letrozole and oxidative stress byproducts produces AIMSS-like behaviors and neurogenic inflammatory responses in mice. Elevated androstenedione levels cooperated with low letrozole concentrations and inflammatory mediators were sufficient to provoke AIMSS-like behaviors. The generation of such painful conditions by small quantities of simultaneously administered TRPA1 agonists justifies previous failure to identify a precise link between AIs and AIMSS, underscoring the potential of channel antagonists to treat AIMSS. Cancer Res; 76(23); 7024-35. ©2016 AACR.


Subject(s)
Androstenedione/adverse effects , Aromatase Inhibitors/adverse effects , Transient Receptor Potential Channels/chemistry , Animals , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Transfection
12.
Am J Respir Cell Mol Biol ; 55(5): 645-656, 2016 11.
Article in English | MEDLINE | ID: mdl-27281024

ABSTRACT

Pseudomonas aeruginosa colonization, prominent inflammation with massive expression of the neutrophil chemokine IL-8, and luminal infiltrates of neutrophils are hallmarks of chronic lung disease in patients with cystic fibrosis (CF). The nociceptive transient receptor potential ankyrin (TRPA) 1 calcium channels have been recently found to be involved in nonneurogenic inflammation. Here, we investigate the role of TRPA1 in CF respiratory inflammatory models in vitro. Expression of TRPA1 was evaluated in CF lung tissue sections and cells by immunohistochemistry and immunofluorescence. Epithelial cell lines (A549, IB3-1, CuFi-1, CFBE41o-) and primary cells from patients with CF were used to: (1) check TRPA1 function modulation, by Fura-2 calcium imaging; (2) down-modulate TRPA1 function and expression, by pharmacological inhibitors (HC-030031 and A-967079) and small interfering RNA silencing; and (3) assess the effect of TRPA1 down-modulation on expression and release of cytokines upon exposure to proinflammatory challenges, by quantitative RT-PCR and 27-protein Bioplex assay. TRPA1 channels are expressed in the CF pseudostratified columnar epithelium facing the bronchial lumina exposed to bacteria, where IL-8 is coexpressed. Inhibition of TRPA1 expression results in a relevant reduction of release of several cytokines, including IL-8 and the proinflammatory cytokines IL-1ß and TNF-α, in CF primary bronchial epithelial cells exposed to P. aeruginosa and to the supernatant of mucopurulent material derived from the chronically infected airways of patients with CF. In conclusion, TRPA1 channels are involved in regulating the extent of airway inflammation driven by CF bronchial epithelial cells.


Subject(s)
Calcium Channels/metabolism , Cystic Fibrosis/complications , Lung/pathology , Nerve Tissue Proteins/metabolism , Pneumonia/complications , Pneumonia/pathology , Transient Receptor Potential Channels/metabolism , A549 Cells , Adult , Bronchi/pathology , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cytokines/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Silencing , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Male , Middle Aged , Models, Biological , Nerve Tissue Proteins/antagonists & inhibitors , Pneumonia/genetics , Pseudomonas aeruginosa/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , TRPA1 Cation Channel , Tissue Donors , Transcription, Genetic , Transient Receptor Potential Channels/antagonists & inhibitors , Young Adult
13.
Brain ; 139(Pt 5): 1361-77, 2016 05.
Article in English | MEDLINE | ID: mdl-26984186

ABSTRACT

Despite intense investigation, the mechanisms of the different forms of trigeminal neuropathic pain remain substantially unidentified. The transient receptor potential ankyrin 1 channel (encoded by TRPA1) has been reported to contribute to allodynia or hyperalgesia in some neuropathic pain models, including those produced by sciatic nerve constriction. However, the role of TRPA1 and the processes that cause trigeminal pain-like behaviours from nerve insult are poorly understood. The role of TRPA1, monocytes and macrophages, and oxidative stress in pain-like behaviour evoked by the constriction of the infraorbital nerve in mice were explored. C57BL/6 and wild-type (Trpa1(+/+)) mice that underwent constriction of the infraorbital nerve exhibited prolonged (20 days) non-evoked nociceptive behaviour and mechanical, cold and chemical hypersensitivity in comparison to sham-operated mice (P < 0.05-P < 0.001). Both genetic deletion of Trpa1 (Trpa1(-/-)) and pharmacological blockade (HC-030031 and A-967079) abrogated pain-like behaviours (both P < 0.001), which were abated by the antioxidant, α-lipoic acid, and the nicotinamide adenine dinucleotide phosphate oxidase inhibitor, apocynin (both P < 0.001). Nociception and hypersensitivity evoked by constriction of the infraorbital nerve was associated with intra- and perineural monocytic and macrophagic invasion and increased levels of oxidative stress by-products (hydrogen peroxide and 4-hydroxynonenal). Attenuation of monocyte/macrophage increase by systemic treatment with an antibody against the monocyte chemoattractant chemokine (C-C motif) ligand 2 (CCL2) or the macrophage-depleting agent, clodronate (both P < 0.05), was associated with reduced hydrogen peroxide and 4-hydroxynonenal perineural levels and pain-like behaviours (all P < 0.01), which were abated by perineural administration of HC-030031, α-lipoic acid or the anti-CCL2 antibody (all P < 0.001). The present findings propose that, in the constriction of the infraorbital nerve model of trigeminal neuropathic pain, pain-like behaviours are entirely mediated by the TRPA1 channel, targeted by increased oxidative stress by-products released from monocytes and macrophages clumping at the site of nerve injury.


Subject(s)
Hyperalgesia/physiopathology , Macrophages/drug effects , Monocytes/drug effects , Neuralgia/physiopathology , Oxidative Stress/drug effects , Transient Receptor Potential Channels/physiology , Acetanilides/antagonists & inhibitors , Acetanilides/pharmacology , Acetophenones/pharmacology , Animals , Chemokine CCL2/antagonists & inhibitors , Clodronic Acid/pharmacology , Hyperalgesia/metabolism , Macrophages/metabolism , Male , Mice , Mice, Knockout , Monocytes/metabolism , Neuralgia/metabolism , Oximes/antagonists & inhibitors , Oximes/pharmacology , Purines/antagonists & inhibitors , Purines/pharmacology , TRPA1 Cation Channel , Thioctic Acid/pharmacology , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
14.
PLoS One ; 10(9): e0136425, 2015.
Article in English | MEDLINE | ID: mdl-26371475

ABSTRACT

BACKGROUND: Nerve Growth Factor (NGF) holds a great therapeutic promise for Alzheimer's disease, diabetic neuropathies, ophthalmic diseases, dermatological ulcers. However, the necessity for systemic delivery has hampered the clinical applications of NGF due to its potent pro-nociceptive action. A "painless" human NGF (hNGF R100E) mutant has been engineered. It has equal neurotrophic potency to hNGF but a lower nociceptive activity. We previously described and characterized the neurotrophic and nociceptive properties also of the hNGF P61S and P61SR100E mutants, selectively detectable against wild type hNGF. However, the reduced pain-sensitizing potency of the "painless" hNGF mutants has not been quantified. OBJECTIVES AND RESULTS: Aiming at the therapeutic application of the "painless" hNGF mutants, we report on the comparative functional characterization of the precursor and mature forms of the mutants hNGF R100E and hNGF P61SR100E as therapeutic candidates, also in comparison to wild type hNGF and to hNGF P61S. The mutants were assessed by a number of biochemical, biophysical methods and assayed by cellular assays. Moreover, a highly sensitive ELISA for the detection of the P61S-tagged mutants in biological samples has been developed. Finally, we explored the pro-nociceptive effects elicited by hNGF mutants in vivo, demonstrating an expanded therapeutic window with a ten-fold increase in potency. CONCLUSIONS: This structure-activity relationship study has led to validate the concept of developing painless NGF as a therapeutic, targeting the NGF receptor system and supporting the choice of hNGF P61S R100E as the best candidate to advance in clinical development. Moreover, this study contributes to the identification of the molecular determinants modulating the properties of the hNGF "painless" mutants.


Subject(s)
Mutation , Nerve Growth Factor/adverse effects , Nerve Growth Factor/genetics , Pain/chemically induced , Protein Engineering , Protein Precursors/adverse effects , Protein Precursors/genetics , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Escherichia coli/genetics , Humans , Kinetics , Nerve Growth Factor/metabolism , Nerve Growth Factor/therapeutic use , Oligodendroglia/cytology , Oligodendroglia/drug effects , Protein Precursors/metabolism , Protein Precursors/therapeutic use , Protein Stability , Proteolysis , Rats , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Temperature
15.
Int J Urol ; 22(9): 878-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26105144

ABSTRACT

OBJECTIVES: To evaluate the influence of hypotonic solutions on ureteral relaxation mediated by the release of calcitonin gene-related peptide from intramural sensory nerve endings. METHODS: Urine osmolarity of Sprague-Dawley rats drinking water low in salt content (Fiuggi water) or a reference water for 7 days was measured. Release of calcitonin gene-related peptide-like immunoreactivity from slices of rat ureter and urinary bladder by hypotonic solutions was assessed by an immunometric assay. The mechanism through which hypotonic solutions inhibit neurokinin A-induced phasic contractions of isolated rat ureters was evaluated by organ bath studies. RESULTS: A 7-day consumption of Fiuggi water in rats reduced urine osmolarity by ~40%. Exposure to hypotonic solutions released calcitonin gene-related peptide-like immunoreactivity from slices of rat ureter. This response was abated in a calcium-free medium, after capsaicin desensitization, and in the presence of the unselective transient receptor potential channel antagonist, ruthenium red. Exposure of isolated rat ureteral preparations to a hypotonic solution inhibited neurokinin A-evoked phasic contraction. This response was attenuated by capsaicin desensitization and in the presence of the calcitonin gene-related peptide receptor antagonist, calcitonin gene-related peptide8-37 . Transient receptor potential vanilloid 1 or transient receptor potential vanilloid 4 antagonists did not affect the neurogenic and calcitonin gene-related peptide-dependent relaxation. CONCLUSION: Present data show that hypotonic solution evokes calcitonin gene-related peptide release from capsaicin-sensitive intramural sensory nerves, thus inhibiting ureteral contractility, through a transient receptor potential-dependent mechanism. However, this mechanism does not involve transient receptor potential vanilloid 1 or transient receptor potential vanilloid 4. Future studies with appropriate in vivo models should investigate the hypothesis that hypostenuric urine diffusing into the ureteral tissue might favor ureteral relaxation through this novel mechanism.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Hypotonic Solutions/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Sensory Receptor Cells/metabolism , Ureter/drug effects , Animals , Calcitonin Gene-Related Peptide/pharmacology , Capsaicin/pharmacology , Male , Neurokinin A/pharmacology , Neurotransmitter Agents/pharmacology , Osmolar Concentration , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sensory System Agents/pharmacology , TRPV Cation Channels/antagonists & inhibitors , Tissue Culture Techniques , Ureter/innervation , Ureter/metabolism , Urinalysis
16.
Br J Pharmacol ; 172(13): 3397-411, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25765567

ABSTRACT

BACKGROUND AND PURPOSE: Although still used by hundreds of millions of people worldwide, the mechanism of the analgesic action of the pyrazolone derivatives (PDs), dipyrone, propyphenazone and antipyrine remains unknown. The transient receptor potential ankyrin 1 (TRPA1) channel, expressed by nociceptors, is emerging as a major pain transduction pathway. We hypothesized that PDs target the TRPA1 channel and by this mechanism produce their analgesic effect. EXPERIMENTAL APPROACH: Calcium responses and currents were studied in cultured TRPA1-expressing rodent dorsal root ganglion neurons and human cells. Acute nociception and mechanical hypersensitivity were investigated in naïve and genetically manipulated mice. KEY RESULTS: Pyrazolone and PDs selectively inhibited calcium responses and currents in TRPA1-expressing cells and acute nocifensor responses in mice evoked by reactive channel agonists (allyl isothiocyanate, acrolein and H2 O2 ). In line with recent results obtained with TRPA1 antagonists and TRPA1 gene deletion, the two most largely used PDs, dipyrone and propyphenazone, attenuated TRPA1-mediated nociception and mechanical allodynia in models of inflammatory and neuropathic pain (formalin, carrageenan, partial sciatic nerve ligation and the chemotherapeutic drug, bortezomib). Notably, dipyrone and propyphenazone attenuated carrageenan-evoked mechanical allodynia, without affecting PGE2 levels. The main metabolites of PDs did not target TRPA1 and did not affect TRPA1-dependent nociception and allodynia. CONCLUSIONS AND IMPLICATIONS: Evidence that in rodents the nociceptive/hyperalgesic effect produced by TRPA1 activation is blocked by PDs suggests that a similar pathway is attenuated by PDs in humans and that TRPA1 antagonists could be novel analgesics, devoid of the adverse haematological effects of PDs.


Subject(s)
Calcium Channels/metabolism , Hyperalgesia/metabolism , Nerve Tissue Proteins/metabolism , Nociception/physiology , Pain/metabolism , TRPC Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Dipyrone/pharmacology , Dipyrone/therapeutic use , HEK293 Cells , Humans , Hyperalgesia/drug therapy , Male , Mice, Inbred C57BL , Nociception/drug effects , Pain/drug therapy , Pyrazolones/pharmacology , Pyrazolones/therapeutic use , Rats, Sprague-Dawley , TRPA1 Cation Channel
17.
Nat Commun ; 5: 5736, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25484020

ABSTRACT

Use of aromatase inhibitors (AIs), exemestane, letrozole and anastrozole, for breast cancer therapy is associated with severe pain symptoms, the underlying mechanism of which is unknown. The electrophilic nature of AIs suggests that they may target the transient receptor potential ankyrin 1 (TRPA1) channel, a major pathway in pain transmission and neurogenic inflammation. AIs evoke TRPA1-mediated calcium response and current in rodent nociceptors and human cells expressing the recombinant channel. In mice, AIs produce acute nociception, which is exaggerated by pre-exposure to proalgesic stimuli, and, by releasing sensory neuropeptides, neurogenic inflammation in peripheral tissues. AIs also evoke mechanical allodynia and decreased grip strength, which do not undergo desensitization on prolonged AI administration. These effects are markedly attenuated by TRPA1 pharmacological blockade or in TRPA1-deficient mice. TRPA1 is a major mediator of the proinflammatory/proalgesic actions of AIs, thus suggesting TRPA1 antagonists for the treatment of pain symptoms associated with AI use.


Subject(s)
Aromatase Inhibitors/chemistry , Calcium Channels/metabolism , Nerve Tissue Proteins/metabolism , Pain/chemically induced , Steroids/chemistry , TRPC Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Anastrozole , Androstadienes/chemistry , Animals , Behavior, Animal , Calcium/chemistry , Cysteine/chemistry , HEK293 Cells , Humans , Inflammation , Letrozole , Male , Mice , Mice, Inbred C57BL , Neuropeptides/chemistry , Nitriles/chemistry , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , TRPA1 Cation Channel , Triazoles/chemistry
18.
Gastroenterology ; 147(6): 1417-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25194674

ABSTRACT

BACKGROUND & AIMS: Patients with cholestatic disease have increased systemic concentrations of bile acids (BAs) and profound pruritus. The G-protein-coupled BA receptor 1 TGR5 (encoded by GPBAR1) is expressed by primary sensory neurons; its activation induces neuronal hyperexcitability and scratching by unknown mechanisms. We investigated whether the transient receptor potential ankyrin 1 (TRPA1) is involved in BA-evoked, TGR5-dependent pruritus in mice. METHODS: Co-expression of TGR5 and TRPA1 in cutaneous afferent neurons isolated from mice was analyzed by immunofluorescence, in situ hybridization, and single-cell polymerase chain reaction. TGR5-induced activation of TRPA1 was studied in in HEK293 cells, Xenopus laevis oocytes, and primary sensory neurons by measuring Ca(2+) signals. The contribution of TRPA1 to TGR5-induced release of pruritogenic neuropeptides, activation of spinal neurons, and scratching behavior were studied using TRPA1 antagonists or Trpa1(-/-) mice. RESULTS: TGR5 and TRPA1 protein and messenger RNA were expressed by cutaneous afferent neurons. In HEK cells, oocytes, and neurons co-expressing TGR5 and TRPA1, BAs caused TGR5-dependent activation and sensitization of TRPA1 by mechanisms that required Gßγ, protein kinase C, and Ca(2+). Antagonists or deletion of TRPA1 prevented BA-stimulated release of the pruritogenic neuropeptides gastrin-releasing peptide and atrial natriuretic peptide B in the spinal cord. Disruption of Trpa1 in mice blocked BA-induced expression of Fos in spinal neurons and prevented BA-stimulated scratching. Spontaneous scratching was exacerbated in transgenic mice that overexpressed TRG5. Administration of a TRPA1 antagonist or the BA sequestrant colestipol, which lowered circulating levels of BAs, prevented exacerbated spontaneous scratching in TGR5 overexpressing mice. CONCLUSIONS: BAs induce pruritus in mice by co-activation of TGR5 and TRPA1. Antagonists of TGR5 and TRPA1, or inhibitors of the signaling mechanism by which TGR5 activates TRPA1, might be developed for treatment of cholestatic pruritus.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/metabolism , Pruritus/metabolism , Receptors, G-Protein-Coupled/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Cholestasis/complications , Disease Models, Animal , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gastrin-Releasing Peptide/metabolism , HEK293 Cells , Humans , Mice, Knockout , Natriuretic Peptides/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Nociceptors/metabolism , Oocytes/cytology , Oocytes/metabolism , Primary Cell Culture , Pruritus/etiology , Receptors, G-Protein-Coupled/genetics , TRPA1 Cation Channel , Transient Receptor Potential Channels/genetics , Xenopus laevis
19.
Br J Pharmacol ; 171(18): 4289-99, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24846744

ABSTRACT

BACKGROUND AND PURPOSE: Transient receptor potential vanilloid 1 (TRPV1) and TRP ankyrin 1 (TRPA1) are involved in many biological processes, including nociception and hyperalgesia. Whereas the involvement of TRPV1 in psychiatric disorders such as anxiety and depression has been reported, little is known regarding the role of TRPA1 in these conditions. EXPERIMENTAL APPROACH: We investigated the role of TRPA1 in mice models of depression [forced swimming test (FST)] and anxiety [elevated plus maze (EPM) test]. KEY RESULTS: Administration of the TRPA1 antagonist (HC030031, 30 nmol in 2 µL, i.c.v.) reduced immobility time in the FST. Similar results were obtained after oral administration of HC030031 (30-300 mg·kg(-1) ). The reduction in immobility time in FST induced by HC030031 (100 mg·kg(-1) ) was completely prevented by pretreatment with TRPA1 agonist, cinnamaldehyde (50 mg·kg(-1) , p.o.), which per se was inactive. In the EPM test, pretreatment with cinnamaldehyde (50 mg·kg(-1) , p.o.), which per se did not affect behaviour response, prevented the anxiolytic-like effect (increased open arm exploration) evoked by TRPA1 blockade (HC030031, 100 mg·kg(-1) , p.o.). Treatment with either cinnamaldehyde or HC030031 did not affect spontaneous ambulation. Furthermore, TRPA1-deficient mice showed anxiolytic- and antidepressant-like phenotypes in the FST and EPM test respectively. CONCLUSION AND IMPLICATIONS: The present findings indicate that genetic deletion or pharmacological blockade of TRPA1 produces inhibitory activity in mouse models of anxiety and depression. These results imply that TRPA1 exerts tonic control, promoting anxiety and depression, and that TRPA1 antagonism has potential as an innovative strategy for the treatment of anxiety and mood disorders.


Subject(s)
Anxiety/physiopathology , Depression/physiopathology , Transient Receptor Potential Channels/physiology , Acetanilides/pharmacology , Acrolein/analogs & derivatives , Acrolein/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal , Diazepam/pharmacology , Disease Models, Animal , Male , Mice, Knockout , Nortriptyline/pharmacology , Purines/pharmacology , Signal Transduction , Swimming , TRPA1 Cation Channel , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
20.
Free Radic Biol Med ; 72: 200-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24780252

ABSTRACT

Acute gout attacks produce severe joint pain and inflammation associated with monosodium urate (MSU) crystals leading to oxidative stress production. The transient potential receptor ankyrin 1 (TRPA1) is expressed by a subpopulation of peptidergic nociceptors and, via its activation by endogenous reactive oxygen species, including hydrogen peroxide (H2O2), contributes to pain and neurogenic inflammation. The aim of this study was to investigate the role of TRPA1 in hyperalgesia and inflammation in a model of acute gout attack in rodents. Inflammatory parameters and mechanical hyperalgesia were measured in male Wistar rats and in wild-type (Trpa1(+/+)) or TRPA1-deficient (Trpa1(-/-)) male mice. Animals received intra-articular (ia, ankle) injection of MSU. The role of TRPA1 was assessed by receptor antagonism, gene deletion or expression, sensory fiber defunctionalization, and calcitonin gene-related peptide (CGRP) release. We found that nociceptor defunctionalization, TRPA1 antagonist treatment (via ia or oral administration), and Trpa1 gene ablation abated hyperalgesia and inflammatory responses (edema, H2O2 generation, interleukin-1ß release, and neutrophil infiltration) induced by ia MSU injection. In addition, we showed that MSU evoked generation of H2O2 in synovial tissue, which stimulated TRPA1 producing CGRP release and plasma protein extravasation. The MSU-elicited responses were also reduced by the H2O2-detoxifying enzyme catalase and the reducing agent dithiothreitol. TRPA1 activation by MSU challenge-generated H2O2 mediates the entire inflammatory response in an acute gout attack rodent model, thus strengthening the role of the TRPA1 receptor and H2O2 production as potential targets for treatment of acute gout attacks.


Subject(s)
Gout/metabolism , Hydrogen Peroxide/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , TRPC Cation Channels/metabolism , Animals , Antioxidants/pharmacology , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Male , Mice , Mice, Knockout , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...