Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Free Radic Biol Med ; 89: 972-81, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26476010

ABSTRACT

The analysis of the global thiol-disulfide redox status in tissues and cells is a challenging task since thiols and disulfides can undergo artificial oxido-reductions during sample manipulation. Because of this, the measured values, in particular for disulfides, can have a significant bias. Whereas this methodological problem has already been addressed in samples of red blood cells and solid tissues, a reliable method to measure thiols and disulfides in cell cultures has not been previously reported. Here, we demonstrate that the major artifact occurring during thiol and disulfide analysis in cultured cells is represented by glutathione disulfide (GSSG) and S-glutathionylated proteins (PSSG) overestimation, due to artificial oxidation of glutathione (GSH) during sample manipulation, and that this methodological problem can be solved by the addition of N-ethylmaleimide (NEM) immediately after culture medium removal. Basal levels of GSSG and PSSG in different lines of cultured cells were 3-5 and 10-20 folds higher, respectively, when the cells were processed without NEM. NEM pre-treatment also prevented the artificial reduction of disulfides that occurs during the pre-analytical phase when cells are exposed to an oxidant stimulus. In fact, in the absence of NEM, after medium removal, GSH, GSSG and PSSG levels restored their initial values within 15-30 min, due to the activity of reductases and the lack of the oxidant. The newly developed protocol was used to measure the thiol-disulfide redox status in 16 different line cells routinely used for biomedical research both under basal conditions and after treatment with disulfiram, a thiol-specific oxidant (0-200 µM concentration range). Our data indicate that, in most cell lines, treatment with disulfiram affected the levels of GSH and GSSG only at the highest concentration. On the other hand, PSSG levels increased significantly also at the lower concentrations of the drug, and the rise was remarkable (from 100 to 1000 folds at 200 µM concentration) and dose-dependent for almost all the cell lines. These data support the suitability of the analysis of PSSG in cultured cells as a biomarker of oxidative stress.


Subject(s)
Glutathione Disulfide/metabolism , Glutathione/metabolism , Neoplasms/pathology , Oxidative Stress , Protein Processing, Post-Translational , Proteins/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Ethylmaleimide/pharmacology , Humans , Oxidation-Reduction , Proteins/chemistry
7.
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
8.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...