Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
J Biosci ; 492024.
Article in English | MEDLINE | ID: mdl-38920106

ABSTRACT

Noradrenaline (NA) and serotonin (5-HT) induce nociception and antinociception. This antagonistic effect can be explained by the dose and type of activated receptors. We investigated the existence of synergism between the noradrenergic and serotonergic systems during peripheral antinociception. The paw pressure test was performed in mice that had increased sensitivity by intraplantar injection of prostaglandin E2 (PGE2). Noradrenaline (80 ng) administered intraplantarly induced an antinociceptive effect, that was reversed by the administration of selective antagonists of serotoninergic receptors 5-HT1B isamoltan, 5-HT1D BRL15572, 5-HT2A ketanserin, 5-HT3 ondansetron, but not by selective receptor antagonist 5-HT7 SB-269970. The administration of escitalopram, a serotonin reuptake inhibitor, potentiated the antinociceptive effect at a submaximal dose of NA. These results, indicate the existence of synergism between the noradrenergic and serotonergic systems in peripheral antinociception in mice.


Subject(s)
Norepinephrine , Receptors, Serotonin , Serotonin Antagonists , Serotonin , Animals , Mice , Norepinephrine/metabolism , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Male , Receptors, Serotonin/metabolism , Dinoprostone/metabolism , Citalopram/pharmacology , Nociception/drug effects , Analgesics/pharmacology , Ondansetron/pharmacology , Ketanserin/pharmacology , Pain/drug therapy , Pain/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
2.
Article in English | MEDLINE | ID: mdl-38753048

ABSTRACT

The present study aimed to evaluate the possible peripheral H2O2-induced antinociception and determine the involvement of opioidergic, cannabinoidergic and nitrergic systems, besides potassium channels in its antinociceptive effect. Prostaglandin E2 was used to induce hyperalgesia in male Swiss mice using the mechanical paw pressure test. H2O2 (0.1, 0.2, 0.3 µg/paw) promoted a dose-dependent antinociceptive effect that was not observed in contralateral paw. Female mice also showed antinociception in the model. The partial H2O2-induced antinociception was potentiated by the inhibitor of catalase enzyme, aminotriazole (40, 60, 80 µg/paw). The antinociception was not reversed by opioid and cannabinoid receptor antagonists naloxone, AM 251 and AM 630. The involvement of nitric oxide (NO) was observed by the reversal of H2O2-induced antinociception using the non-selective inhibitor of nitric oxide synthases L-NOarg and by inhibition of iNOS (L-NIL), eNOS (L-NIO) and nNOS (L-NPA). ODQ, a cGMP-forming enzyme selective inhibitor, also reversed the antinociception. The blockers of potassium channels voltage-gated (TEA), ATP-sensitive (glibenclamide), large (paxillin) and small (dequalinium) conductance calcium-activated were able to revert H2O2 antinociception. Our data suggest that H2O2 induced a peripheral antinociception in mice and the NO pathway and potassium channels (voltage-gated, ATP-sensitive, calcium-activated) are involved in this mechanism. However, the role of the opioid and cannabinoid systems was not evidenced.

3.
Pharmacology ; : 1-18, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38643765

ABSTRACT

INTRODUCTION: Tissue injury results in the release of inflammatory mediators, including a cascade of algogenic substances, which contribute to the development of hyperalgesia. During this process, endogenous analgesic substances are peripherally released to counterbalance hyperalgesia. The present study aimed to investigate whether inflammatory mediators TNF-α, IL-1ß, CXCL1, norepinephrine (NE), and prostaglandin E2 (PGE2) may be involved in the deflagration of peripheral endogenous modulation of inflammatory pain by activation of the cholinergic system. METHODS: Male Swiss mice were subjected to paw withdrawal test. All the substances were injected via the intraplantar route. RESULTS: The main findings of this study were as follows: (1) carrageenan (Cg), TNF-α, CXCL-1, IL1-ß, NE, and PGE2 induced hyperalgesia; (2) the acetylcholinesterase enzyme inhibitor, neostigmine, reversed the hyperalgesia observed after Cg, TNF-α, CXCL-1, and IL1-ß injection; (3) the non-selective muscarinic receptor antagonist, atropine, and the selective muscarinic type 1 receptor (m1AChr) antagonist, telenzepine, potentiated the hyperalgesia induced by Cg and CXCL-1; (4) mecamylamine, a non-selective nicotinic receptor antagonist, potentiated the hyperalgesia induced by Cg, TNF-α, CXCL-1, and IL1-ß; (5) Cg, CXCL-1, and PGE2 increased the expression of the m1AChr and nicotinic receptor subunit α4protein. CONCLUSION: These results suggest that the cholinergic system may modulate the inflammatory pain induced by Cg, PGE2, TNF-α, CXCL-1, and IL1-ß.

4.
Nitric Oxide ; 146: 1-9, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38428514

ABSTRACT

BACKGROUND: Cannabidiol (CBD) is the second most abundant pharmacologically active component present in Cannabis sp. Unlike Δ-9-tetrahydrocannabinol (THC), it has no psychotomimetic effects and has recently received significant interest from the scientific community due to its potential to treat anxiety and epilepsy. CBD has excellent anti-inflammatory potential and can be used to treat some types of inflammatory and neuropathic pain. In this context, the present study aimed to evaluate the analgesic mechanism of cannabidiol administered systemically for the treatment of neuropathic pain and determine the endogenous mechanisms involved with this analgesia. METHODS: Neuropathic pain was induced by sciatic nerve constriction surgery, and the nociceptive threshold was measured using the paw compression test in mice. RESULTS: CBD produced dose-dependent antinociception after intraperitoneal injection. Selective inhibition of PI3Kγ dose-dependently reversed CBD-induced antinociception. Selective inhibition of nNOS enzymes reversed the antinociception induced by CBD, while selective inhibition of iNOS and eNOS did not alter this antinociception. However, the inhibition of cGMP production by guanylyl cyclase did not alter CBD-mediated antinociception, but selective blockade of ATP-sensitive K+ channels dose-dependently reversed CBD-induced antinociception. Inhibition of S-nitrosylation dose-dependently and completely reversed CBD-mediated antinociception. CONCLUSION: Cannabidiol has an antinociceptive effect when administered systemically and this effect is mediated by the activation of PI3Kγ as well as by nitric oxide and subsequent direct S-nitrosylation of KATP channels on peripheral nociceptors.


Subject(s)
Analgesics , Cannabidiol , Class Ib Phosphatidylinositol 3-Kinase , KATP Channels , Neuralgia , Nitric Oxide Synthase Type I , Nitric Oxide , Signal Transduction , Animals , Cannabidiol/pharmacology , KATP Channels/metabolism , Male , Signal Transduction/drug effects , Neuralgia/drug therapy , Neuralgia/metabolism , Mice , Nitric Oxide/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Nitric Oxide Synthase Type I/metabolism , Analgesics/pharmacology , Analgesia
5.
Can J Physiol Pharmacol ; 102(3): 218-227, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37976474

ABSTRACT

Tramadol, an analgesic classified as an "atypical opioid", exhibits both opioid and non-opioid mechanisms of action. This study aimed to explore these mechanisms, specifically the opioid-, cannabinoid-, nitric oxide-, and potassium channel-based mechanisms, which contribute to the peripheral antinociception effect of tramadol, in an experimental rat model. The nociceptive threshold was determined using paw pressure withdrawal. To examine the mechanisms of action, several substances were administered intraplantarly: naloxone, a non-selective opioid antagonist (50 µg/paw); AM251 (80 µg/paw) and AM630 (100 µg/paw) as the selective antagonists for types 1 and 2 cannabinoid receptors, respectively; nitric oxide synthase inhibitors L-NOArg, L-NIO, L-NPA, and L-NIL (24 µg/paw); and the enzyme inhibitors of guanylatocyclase and phosphodiesterase of cGMP, ODQ, and zaprinast. Additionally, potassium channel blockers glibenclamide, tetraethylammonium, dequalinium, and paxillin were used. The results showed that opioid and cannabinoid receptor antagonists did not reverse tramadol's effects. L-NOarg, L-NIO, and L-NPA partially reversed antinociception, while ODQ completely reversed, and zaprinast enhanced tramadol's antinociception effect. Notably, glibenclamide blocked tramadol's antinociception in a dose-dependent manner. These findings suggest that tramadol's peripheral antinociception effect is likely mediated by the nitrergic pathway and sensitive ATP potassium channels, rather than the opioid and cannabinoid pathways.


Subject(s)
Cannabinoids , Tramadol , Rats , Animals , Analgesics, Opioid/pharmacology , Tramadol/pharmacology , Tramadol/therapeutic use , Nitric Oxide/metabolism , Rats, Wistar , Potassium Channels/metabolism , Hyperalgesia/metabolism , Nitroarginine , Receptors, Cannabinoid/metabolism , Glyburide , Analgesics/pharmacology , Analgesics/therapeutic use , Cyclic GMP/metabolism , Cannabinoids/adverse effects
6.
Neurosci Lett ; 818: 137536, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37898181

ABSTRACT

It has already been shown that serotonin can release endocannabinoids at the spinal cord level, culminating in inhibition of the dorsal horn. At the peripheral level, cannabinoid receptors modulate primary afferent neurons by inhibiting calcium conductance and increasing potassium conductance. Studies have shown that after the activation of opioid receptors and cannabinoids, there is also the activation of the NO/cGMP/KATP pathway, inducing cellular hyperpolarization. In this study, we evaluated the participation of the cannabinoid system with subsequent activation of the NO/cGMP/KATP pathway in the peripheral antinociceptive effect of serotonin. The paw pressure test of mice was used in animals that had their sensitivity to pain increased due to an intraplantar injection of PGE2 (2 µg). Serotonin (250 ng/paw), administered locally in the right hind paw, induced antinociceptive effect. CB1 and CB2 cannabinoid receptors antagonists, AM251 (20, 40 and 80 µg) and AM630 (25, 50 and 100 µg), respectively, reversed the serotonin-induced antinociceptive effect. MAFP (0.5 µg), an inhibitor of the FAAH enzyme that degrades anandamide, and JZL184 (3.75 µg), an inhibitor of the enzyme MAGL that degrades 2-AG, as well as the VDM11 (2.5 µg) inhibitor of anandamide reuptake, potentiated the antinociceptive effect induced by a low dose (62. 5 ng) of serotonin. In the evaluation of the participation of the NO/cGMP/KATP pathway, the antinociceptive effect of serotonin was reversed by the administration of the non-selective inhibitor of NOS isoforms L-NOarg (12.5, 25 and 50 µg) and by the selective inhibitor for the neuronal isoform LNPA (24 µg), as well as by the soluble guanylate cyclase inhibitor ODQ (25, 50 and 100 µg). Among potassium channel blockers, only Glibenclamide (20, 40 and 80 µg), an ATP-sensitive potassium channel blocker, reversed the effect of serotonin. In addition, intraplantar administration of serotonin (250 ng) was shown to induce a significant increase in nitrite levels in the homogenate of the plantar surface of the paw of mice. Taken together, these data suggest that the antinociceptive effect of serotonin occurs by activation of the cannabinoid system with subsequent activation of the NO/cGMP/KATP pathway.


Subject(s)
Cannabinoids , Mice , Animals , Cannabinoids/metabolism , Analgesics/pharmacology , Serotonin/pharmacology , Potassium Channel Blockers , Receptors, Cannabinoid , Adenosine Triphosphate , Hyperalgesia/metabolism
7.
Eur J Pharmacol ; 956: 175932, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37536622

ABSTRACT

Graft-versus-host disease (GVHD) is a serious inflammatory illness that often occurs as a secondary complication of bone marrow transplantation. Current therapies have limited effectiveness and fail to achieve a balance between inflammation and the graft-versus-tumor effect. In this study, we investigate the effects of the endocannabinoid anandamide on the complex pathology of GVHD. We assess the effects of an irreversible inhibitor of fatty acid amine hydrolase or exogenous anandamide and find that they increase survival and reduce clinical signs in GVHD mice. In the intestine of GVHD mice, treatment with exogenous anandamide also leads to a reduction in the number of CD3+, CD3+CD4+, and CD3+CD8+ cells, which reduces the activation of CD3+CD4+ and CD3+CD8+ cells, as assessed by enhanced CD28 expression, a T cell co-stimulatory molecule. Exogenous AEA was also able to reduce TNF-α and increase IL-10 in the intestine of GVHD mice. In the liver, exogenous AEA reduces injury, TNF-α levels, and the number of CD3+CD8+ cells. Interestingly, anandamide reduces Mac-1α, which lowers the adhesion of transplanted cells in mesenteric veins. These effects are mimicked by JWH133-a CB2 selective agonist-and abolished by treatment with a CB2 antagonist. Furthermore, the effects caused by anandamide treatment on survival were related to the CB2 receptor, as the CB2 antagonist abolished it. This study shows the critical role of the CB2 receptor in the modulation of the inflammatory response of GVHD by treatment with anandamide, the most prominent endocannabinoid.


Subject(s)
Endocannabinoids , Graft vs Host Disease , Animals , Mice , Endocannabinoids/pharmacology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/prevention & control , Intestines , Lymphocytes/metabolism , Polyunsaturated Alkamides/pharmacology , Receptor, Cannabinoid, CB1 , Receptor, Cannabinoid, CB2 , Tumor Necrosis Factor-alpha
8.
Biochem Biophys Res Commun ; 660: 58-64, 2023 06 11.
Article in English | MEDLINE | ID: mdl-37068389

ABSTRACT

Cannabidiol (CBD) is the most abundant non-psychoactive component found in plants of the genus Cannabis. Its analgesic effect for the treatment of neuropathy has been widely studied. However, little is known about its effects in the acute treatment when Cannabidiol is administered peripherally. Because of that, this research was aimed to evaluate the antinociceptive effects of the CBD when administered peripherally for the treatment of acute neuropathic pain and check the involvement of the 5-HT1A and the TRPV1 receptors in this event. Neuropathic pain was induced with the constriction of the sciatic nerve while the nociceptive threshold was measured using the pressure test of the mouse paw. The technique used proved to be efficient to induce neuropathy, and the CBD (5, 10 and 30 µg/paw) induced the antinociception in a dosage-dependent manner. The dosage used that induced a more potent effect (30 µg/paw), did not induce a systemic response, as demonstrated by both the motor coordination assessment test (RotaRod) and the antinociceptive effect restricted to the paw treated with CBD. The administration of NAN-190 (10 µg/paw), a selective 5-HT1A receptor antagonist, and SB-366791 (16 µg/paw), a selective TRPV1 antagonist, partially reversed the CBD-induced antinociception. The results of the research suggest that the CBD produces the peripheral antinociception during the acute treatment of the neuropathic pain and it partially involved the participation of the 5-HT1A and TRPV1 receptors.


Subject(s)
Cannabidiol , Neuralgia , Mice , Animals , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Serotonin , Neuralgia/drug therapy , Disease Models, Animal , Analgesics/pharmacology , Analgesics/therapeutic use , Receptor, Serotonin, 5-HT1A , TRPV Cation Channels
9.
Magnes Res ; 35(1): 1-10, 2022 01 01.
Article in English | MEDLINE | ID: mdl-36214549

ABSTRACT

In the present study, we investigated whether magnesium sulphate activates the L-arginine/NO/cGMP pathway and elicits peripheral antinociception. The male Swiss mice paw pressure test was performed with hyperalgesia induced by intraplantar injection of prostaglandin E2. All drugs were administered locally into the right hind paw of animals. Magnesium sulphate (20, 40, 80 and 160 µg/paw) induced an antinociceptive effect. The dose of 80 µg/paw elicited a local antinociceptive effect that was antagonized by the non-selective NOS inhibitor, L-NOArg, and by the selective neuronal NOS inhibitor, L-NPA. The inhibitors, L-NIO and L-NIL, selectively inhibited endothelial and inducible NOS, respectively, but were ineffective regarding peripheral magnesium sulphate injection. The soluble guanylyl cyclase inhibitor, ODQ, blocked the action of magnesium sulphate, and the cGMP-phosphodiesterase inhibitor, zaprinast, enhanced the antinociceptive effects of intermediate dose of magnesium sulphate. Our results suggest that magnesium sulphate stimulates the NO/cGMP pathway via neuronal NO synthase to induce peripheral antinociceptive effects.


Subject(s)
Dinoprostone , Magnesium Sulfate , Analgesics/pharmacology , Animals , Arginine/metabolism , Cyclic GMP/metabolism , Dinoprostone/adverse effects , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Magnesium Sulfate/pharmacology , Male , Mice , Nitric Oxide , Nitroarginine , Phosphodiesterase Inhibitors/pharmacology , Soluble Guanylyl Cyclase/antagonists & inhibitors
10.
Biochem Pharmacol ; 198: 114965, 2022 04.
Article in English | MEDLINE | ID: mdl-35182520

ABSTRACT

BACKGROUND: Bradykinin (BK) is an endogenous peptide involved in vascular permeability and inflammation. It has opposite effects (inducing hyperalgesia or antinociception) when administered directly in the central nervous system. The aim of this study was to evaluate whether BK may also present this dual effect when injected peripherally in a PGE2-induced nociceptive pain model, as well as to investigate the possible mechanisms of action involved in this event in mice. METHODS: Male Swiss and C57BL/6 knockout mice for B1 or B2 bradykinin receptors were submitted to a mechanical paw pressure test and hyperalgesia was induced by intraplantar prostaglandin E2 (2 µg/paw) injection. RESULTS: Bradykinin (20, 40 and 80 ng/paw) produced dose-dependent peripheral antinociception against PGE2-induced hyperalgesia. This effect was antagonized by bradyzide (8, 16 and 32 µg/paw), naloxone (12.5, 25 and 50 µg/paw), nor-binaltorphimine (50, 100 and 200 µg/paw) and AM251 (20, 40 and 80 µg/paw). Bestatin (400 µg/paw), MAFP (0.5 µg/paw) and VDM11 (2.5 µg/paw) potentiated the antinociception of a lower 20 ng BK dose. The knockout of B1 or B2 bradykinin receptors partially abolished the antinociceptive action of BK (80 ng/paw), bremazocine (1 µg/paw) and anandamide (40 ng/paw) when compared with wild-type animals, which show complete antinociception with the same dose of each drug. CONCLUSION: The present study is the first to demonstrate BK-induced antinociception in peripheral tissues against PGE2-induced nociception in mice and the involvement of κ-opioid and CB1 cannabinoid receptors in this effect.


Subject(s)
Bradykinin , Hyperalgesia , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Bradykinin/pharmacology , Dinoprostone , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Male , Mice , Mice, Inbred C57BL , Receptors, Bradykinin
11.
Eur J Pain ; 26(4): 825-834, 2022 04.
Article in English | MEDLINE | ID: mdl-35044019

ABSTRACT

BCKGROUND: Aripiprazole is an antipsychotic drug used to treat schizophrenia and bipolar disorder. Recently, its peripheral analgesic component was evaluated, however, the mechanism involved in this effect is not fully established. Therefore, the aim of the study was to obtain pharmacological evidence for the involvement of the nitric oxide system in the peripheral antinociceptive effect induced by aripiprazole. METHODS: The hyperalgesia was induced via intraplantar injection of prostaglandin E2 in mice and the nociceptive thresholds were evaluated using the paw pressure test. All drugs were injected locally into the right hind paw. RESULTS: The PI3K inhibitor (AS605240), but not rapamycin (mTOR kinase inhibitor), reversed the peripheral antinociceptive effect induced by Aripiprazole. Antinociception was antagonized by the non-selective inhibitor of the nitric oxide synthase (L-NOarg). The same response was observed using the selective iNOS, but not with the selective nNOS inhibitors. The selective guanylyl cyclase enzyme inhibitor (ODQ) and the non-selective potassium channel blocker tetraethylammonium were able to reverse the antinociceptive effect of aripiprazole. The same was seen using glibenclamide, an ATP-dependent K+ channel blocker. However, calcium-activated potassium channel blockers of small and high conductance, dequalinium chloride and paxilline, respectively, did not reverse this effect. The injection of cGMP-specific phosphodiesterase type 5 inhibitor zaprinast, potentiated the antinociceptive effect induced by a low dose of aripiprazole. CONCLUSION: The results provide evidence that aripiprazole induces peripheral antinociceptive effects via PI3K/NO/cGMP/KATP pathway activation.


Subject(s)
Analgesics , Antipsychotic Agents , Aripiprazole , Adenosine Triphosphate , Analgesics/therapeutic use , Animals , Antipsychotic Agents/pharmacology , Aripiprazole/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Cyclic GMP/metabolism , Mice , Nitric Oxide/metabolism
12.
Life Sci ; 293: 120279, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35032552

ABSTRACT

BACKGROUND: Curcumin is one of the compounds present in plants of the genus Curcuma sp., being very used not only as condiment but also with medicinal purposes. As an analgesic, papers highlight the efficacy of curcumin in the treatment of various types of pain. AIMS: In this study we evaluated the peripheral antinociceptive effect of curcumin and by which mechanisms this effect is induced. MAIN METHODS: The mice paw pressure test was used on animals which had increased pain sensitivity by intraplantar injection of carrageenan. All the drugs were administered in the right hind paw. KEY FINDINGS: Curcumin was administered to the right hind paw animals induced antinociceptive effect. Non -selective antagonist of opioid receptors naloxone reverted the antinociceptive effect induced by curcumin. Selective antagonists for µ, δ and κ opioid receptors clocinnamox, naltrindole and nor- binaltorphimine, respectively, reverted the antinociceptive effect induced by curcumin. Bestatin, enkephalinases inhibitor that degrade peptides opioids, did not change the nociceptive response. Selective antagonists for CB1 and CB2 cannabinoid receptors, AM251 and AM630, respectively, reversed the antinociceptive effect induced by curcumin. The MAFP inhibitor of the enzyme FAAH which breaks down anandamide, JZL184, enzyme inhibitor MAGL which breaks down the 2-AG, as well as the VDM11 anandamide reuptake inhibitor potentiated the antinociceptive effect of curcumin. SIGNIFICANCE: These results suggest that curcumin possibly peripheral antinociception induced by opioid and cannabinoid systems activation and possibly for endocannabinoids and opioids release.


Subject(s)
Analgesics/therapeutic use , Cannabinoid Receptor Agonists/therapeutic use , Curcumin/therapeutic use , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Receptors, Opioid/metabolism , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arachidonic Acids/pharmacology , Arachidonic Acids/therapeutic use , Cannabinoid Receptor Agonists/pharmacology , Carrageenan/toxicity , Cinnamates/pharmacology , Curcumin/pharmacology , Dose-Response Relationship, Drug , Endocannabinoids/pharmacology , Endocannabinoids/therapeutic use , Hyperalgesia/chemically induced , Male , Mice , Morphine Derivatives/pharmacology , Narcotic Antagonists/pharmacology , Pain/chemically induced , Pain/drug therapy , Pain/metabolism , Polyunsaturated Alkamides/pharmacology , Polyunsaturated Alkamides/therapeutic use
13.
Eur J Pharmacol ; 901: 174089, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-33826922

ABSTRACT

The participation of the peripheral opioid and cannabinoid endogenous systems in modulating muscle pain and inflammation has not been fully explored. Thus, the aim of this study was to investigate the involvement of these endogenous systems during muscular-tissue hyperalgesia induced by inflammation. Hyperalgesia was induced by carrageenan injection into the tibialis anterior muscles of male Wistar rats. We padronized an available Randal-Sellito test adaptation to evaluate nociceptive behavior elicited by mechanical insult in muscles. Western blot analysis was performed to evaluate the expression levels of opioid and cannabinoid receptors in the dorsal root ganglia. The non-selective opioid peptide receptor antagonist (naloxone) and the selective mu opioid receptor MOP (clocinnamox) and kappa opioid receptor KOP (nor-binaltorphimine) antagonists were able to intensify carrageenan-induced muscular hyperalgesia. On the other hand, the selective delta opioid receptor (DOP) antagonist (naltrindole) did not present any effect on nociceptive behavior. Moreover, the selective inhibitor of aminopeptidases (Bestatin) provoked considerable dose-dependent analgesia when intramuscularly injected into the hyperalgesic muscle. The CB1 receptor antagonist (AM251), but not the CB2 receptor antagonist (AM630), intensified muscle hyperalgesia. All irreversible inhibitors of anandamide hydrolase (MAFP), the inhibitor for monoacylglycerol lipase (JZL184) and the anandamide reuptake inhibitor (VDM11) decreased carrageenan-induced hyperalgesia in muscular tissue. Lastly, MOP, KOP and CB1 expression levels in DRG were baseline even after muscular injection with carrageenan. The endogenous opioid and cannabinoid systems participate in peripheral muscle pain control through the activation of MOP, KOP and CB1 receptors.


Subject(s)
Myalgia/drug therapy , Receptors, Cannabinoid/physiology , Receptors, Opioid/physiology , Animals , Arachidonic Acids/antagonists & inhibitors , Carrageenan , Cinnamates/pharmacology , Endocannabinoids/antagonists & inhibitors , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/psychology , Male , Monoacylglycerol Lipases/antagonists & inhibitors , Morphine Derivatives/pharmacology , Myalgia/chemically induced , Myalgia/psychology , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Pain Measurement/drug effects , Polyunsaturated Alkamides/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, Cannabinoid/drug effects , Receptors, Opioid/drug effects , Receptors, Opioid, delta/drug effects , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects
14.
J Pharmacol Exp Ther ; 377(2): 273-283, 2021 05.
Article in English | MEDLINE | ID: mdl-33658314

ABSTRACT

Cannabidiol (CBD) is a highly lipidic phytocannabinoid with remarkable anti-inflammatory effects. The aim of this study was to evaluate CBD's effects and mechanisms of action in the treatment of mice subjected to acute graft-versus-host disease (aGVHD). aGVHD was induced by the transplantation of bone marrow cells and splenocytes from C57BL-6j to Balb-c mice. The recipient mice were treated daily with CBD, and the treatment reduced mouse mortality by decreasing inflammation and injury and promoting immune regulation in the jejunum, ileum, and liver. Analysis of the jejunum and ileum showed that CBD treatment reduced the levels of C-C motif chemokine ligand (CCL) 2, CCL3, CCL5, tumor necrosis factor α, and interferon γ (IFNγ). CCL3 and IFNγ levels were also decreased in the liver. Mechanistically, CBD also increased the number of cannabinoid receptor type 2 (CB2) receptors on CD4+ and forkhead box P3+ cells in the intestine, which may explain the reduction in proinflammatory cytokines and chemokines. Antagonists of the CB2 receptor reduced the survival rates of CBD-treated mice, suggesting the participation of this receptor in the effects of CBD. Furthermore, treatment with CBD did not interfere with the graft-versus-leukemia response. CBD treatment appears to protect aGVHD mice by anti-inflammatory and immunomodulatory effects partially mediated by CB2 receptor interaction. Altogether, our study suggests that CBD represents an interesting approach in the treatment of aGVHD, with potential therapeutic applications in patients undergoing bone marrow transplantation. SIGNIFICANCE STATEMENT: This study provides for the first time a mechanism by which cannabidiol, a phytocannabinoid with no psychoactive effect, induces immunomodulation in the graft-versus-host disease. Enhancing intestinal cannabinoid receptor type 2 (CB2) receptor expression on CD4+ and forkhead box P3+ cells and increasing the number of these regulatory cells, cannabidiol decreases proinflammatory cytokines and increases graft-versus-host disease mice survival. This effect is dependent of CB2 receptor activation. Besides, cannabidiol did not interfere with graft-versus-leukemia response, a central response to avoid primary disease relapse.


Subject(s)
Cannabidiol/therapeutic use , Graft vs Host Disease/drug therapy , Intestinal Mucosa/metabolism , Leukemia/therapy , Receptor, Cannabinoid, CB2/metabolism , Animals , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/methods , Cannabidiol/pharmacology , Cells, Cultured , Chemokines, CC/genetics , Chemokines, CC/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Intestinal Mucosa/drug effects , Leukemia/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Eur J Pharmacol ; 896: 173900, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33545158

ABSTRACT

Tissue injury results in the release of inflammatory mediators, including a cascade of nociceptive substances, which contribute to development of hyperalgesia. In addition, during this process endogenous analgesic substances are also peripherally released with the aim of controlling the hyperalgesia. Thus, the present study aimed to investigate whether inflammatory mediators TNF-α, IL-1ß, CXCL1, norepinephrine (NE) and prostaglandin E2 (PGE2) may be involved in the deflagration of peripheral endogenous modulation of inflammatory pain by activation of the opioid system. Thus, male Swiss mice and the paw withdrawal test were used. All substances were injected by the intraplantar route. Carrageenan, TNF-α, CXCL-1, IL1-ß, NE and PGE2 induced hyperalgesia. Selectives µ (clocinamox), δ (naltrindole) and κ (norbinaltorphimine, nor-BNI) and non-selective (naloxone) opioid receptor antagonists potentiated the hyperalgesia induced by carrageenan, TNF-α, CXCL-1 and IL1-ß. In contrast, when the enzyme N-aminopeptidase involved in the degradation of endogenous opioid peptides was inhibited by bestatin, the hyperalgesia was significantly reduced. In addition, the western blotting assay indicated that the expression of the opioid δ receptor was increased after intraplantar injection of carrageenan. The data obtained in this work corroborate the hypothesis that TNF-α, CXCL-1 and IL-ß cause, in addition to hyperalgesia, the release of endogenous substances such as opioid peptides, which in turn exert endogenous control over peripheral inflammatory pain.


Subject(s)
Chemokine CXCL1 , Hyperalgesia/chemically induced , Interleukin-1beta , Nociception , Nociceptive Pain/chemically induced , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Tumor Necrosis Factor-alpha , Animals , Carrageenan , Dinoprostone , Disease Models, Animal , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Male , Mice , Narcotic Antagonists/pharmacology , Nociception/drug effects , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Nociceptive Pain/prevention & control , Norepinephrine , Receptors, Opioid/drug effects , Signal Transduction
16.
Biomed Pharmacother ; 135: 111210, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33453675

ABSTRACT

Serotonin (5-HT) acts as a neurotransmitter in the central nervous system (CNS) and as a mediator released by enterochromaffin cells to regulate intestinal motility. However, this amine also plays an important role as an inflammatory mediator and induces phenotypic changes of nociceptors. Despite the wide knowledge of the role of 5-HT in nociception, most studies have focused on its role in the CNS, while a clear information about its role in peripheral tissues is still lacking. In the present study, we investigated the role of peripheral 5-HT receptors in the nociceptive response induced by 5-HT or carrageenan in mice by using antagonists that target different 5-HT receptors. Mechanical nociceptive threshold was measured with an analgesimeter and evaluated after intraplantar (i.pl.) injection of 5-HT or carrageenan. 5-HT antagonists were injected via the i.pl. route. 5-HT (10, 20, 40 or 80 µg/paw) or carrageenan (100 µg/paw) induced mechanical allodynia. Pretreatment with isamoltane (5 µg; 5-HT1B antagonist) or ketanserine (1 µg; 5-HT2A antagonist) did not affect the mechanical allodynia induced by 5-HT. This response was inhibited by BRL 15572 (10 µg; 5-HT1D antagonist) or SB 269970 (25 µg; 5-HT7 antagonist). On the other hand, mechanical allodynia induced by 5-HT or carrageenan was exacerbated by ondansetron (10, 20 or 40 µg; 5-HT3 antagonist). The results indicate that activation of 5-HT1D and 5-HT7 receptors plays a role in the mechanical allodynia induced by 5-HT in mice. This study also demonstrates the inhibitory role of peripheral 5-HT3 receptors in the nociceptive response induced by 5-HT or carrageenan.


Subject(s)
Hyperalgesia/metabolism , Receptor, Serotonin, 5-HT1D/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Receptors, Serotonin/metabolism , Animals , Carrageenan , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Male , Mice , Nociception , Ondansetron/pharmacology , Pain Threshold , Serotonin , Serotonin Antagonists/pharmacology , Signal Transduction
17.
Sci Total Environ ; 744: 140981, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-32755787

ABSTRACT

Accelerated changes in land use in the regions of the Brazilian Amazon and Cerrado in the last four decades have raised questions about the possible consequences for the regional hydrology. Our study area is the Tocantins-Araguaia River Basin (TAW), focusing on the Tucuruí Hydropower Plant (THP), downstream of the TAW. In this study, we evaluated four scenarios of change in land use and cover for the TAW in which forest areas were replaced by pasture, then by agriculture, then by reforestation vegetation and, finally, by regenerated forest to investigate the impacts on the hydrological components of the basin and the hydropower potential of the THP according to these scenarios. For this evaluation, the SWAT model was used to simulate the streamflow of each scenario, so it was possible to predict the hydropower potential in the TAW under different environmental perspectives. Nonparametric statistics were used to identify the efficiency of turbines in converting the streamflow into energy at the 5% significance level. A reduction was observed in the annual evapotranspiration rate and increments were observed in the surface runoff and streamflow, but despite the increase in flow, there was no increase in the energy produced at the THP due to the inability of the turbines to convert excess water into energy, with losses in the energy production of up to 30% per month and 65% in the annual balance. Our results emphasize the importance of the sustainable management of hydrological basins located in tropical regions and aid in planning and decision-making to create public policies that better meet the demand for the exploitation of natural resources.

18.
Pharmacol Rep ; 72(1): 96-103, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32016857

ABSTRACT

BACKGROUND: Xylazine is an α2 adrenoceptor agonist that is extensively used in veterinary medicine and animal experimentation procedures to produce analgesia, sedation and muscle relaxation without causing general anesthesia. Considering the lack of knowledge of the mechanisms involved in peripheral antinociception induced by xylazine and the potential interactions between the adrenergic and endocannabinoid systems, the present study investigated the contribution of the latter system in the mechanism of xylazine. METHODS: The rat paw pressure test, in which hyperalgesia was induced by the intraplantar injection of prostaglandin E2, was performed. RESULTS: Xylazine administered via an intraplantar injection (25, 50 and 100 µg) induced a peripheral antinociceptive effect against prostaglandin E2 (2 µg)-induced hyperalgesia. This effect was blocked by treatment with the selective CB1 cannabinoid antagonist AM251 (20, 40 and 80 µg) but not by the selective CB2 cannabinoid antagonist AM630 (100 µg). The anandamide reuptake inhibitor VDM11 (2.5 µg) intensified the peripheral antinociceptive effect of a submaximal dose of xylazine (25 µg), and the inhibitor of endocannabinoid enzymatic hydrolysis, MAFP (0.5 µg), showed a tendency towards this same effect. In addition, liquid-chromatography mass spectrometric analysis indicated that xylazine (100 µg) treatment was associated with an increase in anandamide levels in the rat paws treated with PGE2. CONCLUSIONS: The present results provides evidence that the peripheral antinociceptive effect of the α2 adrenoceptor agonist xylazine probably results from anandamide release and subsequent CB1 cannabinoid receptor activation.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Hyperalgesia/drug therapy , Polyunsaturated Alkamides/metabolism , Xylazine/pharmacology , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Chromatography, Liquid , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mass Spectrometry , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Xylazine/administration & dosage
19.
Eur J Pharmacol ; 865: 172808, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31738939

ABSTRACT

Ketamine is a drug largely used in clinical practice as an anesthetic and it can also be used as an analgesic to manage chronic pain symptoms. Despite its interactions with several other signaling systems such as cholinergic, serotoninergic and adrenergic, it is accepted that NMDA receptor antagonism is the main mechanism of action of this drug. In this study we investigated the actions of endogenous opioids in the mechanism of peripheral analgesia induced by ketamine. The nociceptive threshold for mechanical stimuli was measured in Swiss mice using the Randall and Selitto test. The drugs used in this study were administered via intraplantar injection. Our results demonstrated that non selective opioid receptor antagonism (naloxone), selective µ- and δ-opioid receptors antagonism (clocinamox and naltrindole, respectively) but not κ-opioid receptor antagonism (nor-binaltorphimine NORBNI) antagonized ketamine-induced peripheral antinociception in a dose-dependent manner. In addition, administration of aminopeptidase inhibitor bestatin significantly potentiated ketamine-induced peripheral antinociception. Ketamine injection in the right hind paw induced ß-endorphine synthesis in the epithelial tissue of the hindpaw. Together these results indicate a role for µ- and δ-opioid receptors and for the endogenous opioid ß-endorphine increased synthesis in ketamine-induced peripheral analgesia mechanism of action.


Subject(s)
Analgesics/therapeutic use , Ketamine/therapeutic use , Pain/drug therapy , Receptors, Opioid, delta , Receptors, Opioid, mu , Analgesics/pharmacology , Animals , Cinnamates/pharmacology , Dinoprostone , Ketamine/pharmacology , Male , Mice , Morphine Derivatives/pharmacology , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain/chemically induced , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors
20.
Toxicol Appl Pharmacol ; 369: 30-38, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30763598

ABSTRACT

Despite all the development of modern medicine, around 100 compounds derived from natural products were undergoing clinical trials only at the end of 2013. Among these natural substances in clinical trials, we found the resveratrol (RES), a pharmacological multi-target drug. RES analgesic properties have been demonstrated, although the bases of these mechanisms have not been fully elucidated. The aim of this study was to evaluate the involvement of opioid and cannabinoid systems in RES-induced peripheral antinociception. Paw withdrawal method was used and hyperalgesia was induced by carrageenan (200 µg/paw). All drugs were given by intraplantar injection in male Swiss mice (n = 5). RES (100 µg/paw) administered in the right hind paw induced local antinociception that was antagonized by naloxone, non-selective opioid receptor antagonist, and clocinnamox, µOR selective antagonist. Naltrindole and nor-binaltorfimine, selective antagonists for δOR and kOR, respectively, did not reverse RES-induced peripheral antinociception. CB1R antagonist AM251, but not CB2R antagonist AM630, antagonized RES-induced peripheral antinociception. Peripheral antinociception of RES intermediate-dose (50 µg/paw) was increased by: (i) bestatin, inhibitor of endogenous opioid degradation involved-enzymes; (ii) MAFP, inhibitor of anandamide amidase; (iii) JZL184, inhibitor of 2-arachidonoylglycerol degradation involved-enzyme; (iv) VDM11, endocannabinoid reuptake inhibitor. Acute and peripheral administration of RES failed to affect the amount of µOR, CB1R and CB2R. Experimental data suggest that RES induces peripheral antinociception through µOR and CB1R activation by endogenous opioid and endocannabinoid releasing.


Subject(s)
Analgesics/pharmacology , Endocannabinoids/metabolism , Hyperalgesia/prevention & control , Nociceptive Pain/prevention & control , Opioid Peptides/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptors, Opioid, mu/agonists , Resveratrol/pharmacology , Animals , Behavior, Animal/drug effects , Cannabinoid Receptor Antagonists/pharmacology , Carrageenan , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/psychology , Male , Mice , Narcotic Antagonists/pharmacology , Nociceptive Pain/chemically induced , Nociceptive Pain/metabolism , Nociceptive Pain/psychology , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...