Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Inflammopharmacology ; 27(3): 573-586, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30820720

ABSTRACT

Tumour necrosis factor (TNF) and kinins have been associated with neuropathic pain-like behaviour in numerous animal models. However, the way that they interact to cause neuron sensitisation remains unclear. This study assessed the interaction of kinin receptors and TNF receptor TNFR1/p55 in mechanical hypersensitivity induced by an intraneural (i.n.) injection of rm-TNF into the lower trunk of brachial plexus in mice. The i.n. injection of rm-TNF reduced the mechanical withdrawal threshold of the right forepaw from the 3rd to the 10th day after the injection, indicating that TNF1/p55 displays a critical role in the onset of TNF-elicited neuropathic pain. The connection between TNF1/p55 and kinin B1 and B2 receptors (B1R and B2R) was confirmed using both knockout mice and mRNAs quantification in the injected nerve, DRG and spinal cord. The treatment with the B2R antagonist HOE 140 or with B1R antagonist des-Arg9-Leu8-BK reduced both BK- and DABK-induced hypersensitivity. The experiments using kinin receptor antagonists and CPM inhibitor (thiorphan) suggest that BK does not only activate B2R as an orthosteric agonist, but also seems to be converted into DABK that consequently activates B1R. These results indicate a connection between TNF and the kinin system, suggesting a relevant role for B1R and B2R in the process of sensitisation of the central nervous systems by the cross talk between the receptor and CPM after i.n. injection of rm-TNF.


Subject(s)
Brachial Plexus/metabolism , Neuralgia/metabolism , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Brachial Plexus/drug effects , Bradykinin B1 Receptor Antagonists/pharmacology , Bradykinin B2 Receptor Antagonists/pharmacology , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/drug therapy
3.
PLoS One ; 6(9): e24034, 2011.
Article in English | MEDLINE | ID: mdl-21931637

ABSTRACT

BACKGROUND: Many studies have shown the antinociceptive effects of cannabinoid (CB) agonists in different models of pain. Herein, we have investigated their relevance in neuropathic pain induced by brachial plexus avulsion (BPA) in mice. METHODOLOGY/PRINCIPAL FINDINGS: Mice underwent BPA or sham surgery. The mRNA levels and protein expression of CB(1) and CB(2) receptors were assessed by RT-PCR and immunohistochemistry, respectively. The activation of glial cells, MAP kinases and transcription factors were evaluated by immunohistochemistry. The antinociceptive properties induced by cannabinoid agonists were assessed on the 5(th) and 30(th) days after surgery. We observed a marked increase in CB(1) and CB(2) receptor mRNA and protein expression in the spinal cord and dorsal root ganglion, either at the 5(th) or 30(th) day after surgery. BPA also induced a marked activation of p38 and JNK MAP kinases (on the 30(th) day), glial cells, such as microglia and astrocytes, and the transcription factors CREB and NF-κB (at the 5(th) and 30(th) days) in the spinal cord. Systemic treatment with cannabinoid agonists reduced mechanical allodynia on both the 5(th) and 30(th) days after surgery, but the greatest results were observed by using central routes of administration, especially at the 30(th) day. Treatment with WIN 55,212-2 prevented the activation of both glial cells and MAP kinases, associated with an enhancement of CREB and NF-κB activation. CONCLUSIONS/SIGNIFICANCE: Our results indicate a relevant role for cannabinoid agonists in BPA, reinforcing their potential therapeutic relevance for the management of chronic pain states.


Subject(s)
Analgesics/pharmacology , Brachial Plexus/injuries , Cannabinoids/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Neuralgia/enzymology , Neuralgia/pathology , Neuroglia/drug effects , Animals , Behavior, Animal/drug effects , Benzoxazines/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation/drug effects , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Knockdown Techniques , Hyperalgesia/complications , Mice , Morpholines/pharmacology , NF-kappa B/metabolism , Naphthalenes/pharmacology , Neuralgia/etiology , Neuralgia/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Nociception/drug effects , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/deficiency , Receptor, Cannabinoid, CB2/genetics , Signal Transduction/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Up-Regulation/drug effects
4.
Pain ; 152(8): 1872-1887, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21620566

ABSTRACT

In this study, we report that α,ß-amyrin, a plant-derived pentacyclic triterpene, reduced persistent inflammatory and neuropathic hyperalgesia in mice by a direct activation of the CB(1) and CB(2) cannabinoid receptors (CB(1)R and CB(2)R). The oral treatment with α,ß-amyrin (30 mg/kg) significantly reduced mechanical and thermal hyperalgesia and inflammation induced by complete Freund's adjuvant (CFA) and by partial sciatic nerve ligation (PSNL). The pretreatment with either CB(1)R or CB(2)R antagonists and the knockdown gene of the receptors significantly reverted the antinociceptive effect of α,ß-amyrin. Of note, binding studies showed that α,ß-amyrin directly bound with very high affinity to CB(1)R (K(i)=0.133 nM) and with a lower affinity to CB(2)R (K(i)=1989 nM). Interestingly, α,ß-amyrin, ACEA (CB(1)R agonist), or JWH-133 (CB(2)R agonist), at doses that caused antinociception, failed to provoke any behavioral disturbance, as measured in the tetrad assay. In addition, α,ß-amyrin largely decreased interleukin-1ß (IL-1ß), tumor necrosis factor α (TNF-α), keratinocyte-derived chemokine (KC) and interleukin 6 (IL-6) levels, and myeloperoxidase activity. Likewise, α,ß-amyrin prevented the activation of the transcriptional factors: nuclear factor κB (NF-κB) and cyclic adenosine monophosphate response element binding (CREB) and the expression of cyclooxygenase 2 in mice footpads and spinal cords. The present results demonstrated that α,ß-amyrin exhibits long-lasting antinociceptive and anti-inflammatory properties in 2 models of persistent nociception via activation of cannabinoid receptors and by inhibiting the production of cytokines and expression of NF-κB, CREB and cyclooxygenase 2.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Neuralgia/drug therapy , Oleanolic Acid/analogs & derivatives , Pentacyclic Triterpenes/therapeutic use , Receptors, Cannabinoid/metabolism , Analysis of Variance , Animals , Anti-Inflammatory Agents/chemistry , Area Under Curve , Body Weight/drug effects , Body Weight/physiology , Cyclohexanols/pharmacokinetics , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Edema/diagnosis , Edema/drug therapy , Edema/etiology , Enzyme-Linked Immunosorbent Assay , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Inflammation/physiopathology , Locomotion/drug effects , Male , Mice , Neuralgia/physiopathology , Oleanolic Acid/chemistry , Oleanolic Acid/therapeutic use , Oligodeoxyribonucleotides, Antisense/pharmacology , Pain Threshold/drug effects , Pentacyclic Triterpenes/chemistry , Peroxidase/metabolism , Protein Binding/drug effects , Rats , Rats, Wistar , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/genetics , Tritium/pharmacokinetics
5.
J Ethnopharmacol ; 128(3): 567-74, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20152893

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Dioscorea bulbifera var sativa is a medicinal plant commonly used in Cameroonian traditional medicine to treat pain and inflammation. AIM: The present work evaluated the effects of the methanol extract of the bulbs of Dioscorea bulbifera in inflammatory and neuropathic models of pain and further investigated its possible mechanism of action. MATERIALS AND METHODS: The effects of Dioscorea bulbifera administered orally at the doses of 250 and 500mg/kg were tested in mechanical hypernociception induced by intraplantar (i.pl.) injection of complete Freund's adjuvant (CFA), lipopolysaccharides (LPS) or prostaglandin-E(2) (PGE(2)), as well as in partial ligation sciatic nerve (PLSN), nociception induced by capsaicin and thermal hyperalgesia induced by i.pl. injection of CFA. The therapeutic effects of Dioscorea bulbifera on PGE(2)-induced hyperalgesia were evaluated in the absence and in the presence of l-NAME, an inhibitor of nitric oxide synthase (NOS) and glibenclamide, an inhibitor of ATP-sensitive potassium channels. RESULTS: The extract showed significant antinociceptive effects in persistent pain induced by CFA and on neuropathic pain induced by PLSN. The effects of Dioscorea bulbifera persisted for 5 days after two administrations in CFA-induced hypernociception. Dioscorea bulbifera significantly inhibited acute LPS-induced pain but failed to reduce thermal hypernociception and capsaicin-induced spontaneous nociception. The antinociceptive effects of this plant extract in PGE(2) model was antagonized by either l-NAME or glibenclamide. CONCLUSION: Present demonstrate the antinociceptive activities of Dioscorea bulbifera both in inflammatory and neuropathic models of pain and these effects may result, at least partially, from its ability to activate the NO-cGMP-ATP-sensitive potassium channels pathway.


Subject(s)
Hyperalgesia/drug therapy , Inflammation/drug therapy , Neuralgia/drug therapy , Pain/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Animals , Cyclic GMP/metabolism , Cyclic GMP/therapeutic use , Female , Freund's Adjuvant/adverse effects , Freund's Adjuvant/therapeutic use , Glyburide/adverse effects , Glyburide/therapeutic use , Hyperalgesia/chemically induced , Inflammation/chemically induced , Inflammation/metabolism , KATP Channels/metabolism , Male , Methanol/adverse effects , Methanol/therapeutic use , Mice , NG-Nitroarginine Methyl Ester/adverse effects , NG-Nitroarginine Methyl Ester/therapeutic use , Nitric Oxide Synthase/metabolism , Pain/chemically induced , Plant Extracts/adverse effects , Plant Extracts/pharmacology , Plant Roots/metabolism
6.
J Neurosci ; 28(11): 2856-63, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18337416

ABSTRACT

The relevance of kinin B(1) (B(1)R) and B(2) (B(2)R) receptors in the brachial plexus avulsion (BPA) model was evaluated in mice, by means of genetic and pharmacological tools. BPA-induced hypernociception was absent in B(1)R, but not in B(2)R, knock-out mice. Local or intraperitoneal administration of the B(2)R antagonist Hoe 140 failed to affect BPA-induced mechanical hypernociception. Interestingly, local or intraperitoneal treatment with B(1)R antagonists, R-715 or SSR240612, dosed at the time of surgery, significantly reduced BPA-evoked mechanical hypernociception. Intrathecal or intracerebroventricular administration of these antagonists, at the surgery moment, did not prevent the hypernociception. Both antagonists, dosed by intraperitoneal or intrathecal routes (but not intracerebroventricularly) 4 d after the surgery, significantly inhibited the mechanical hypernociception. At 30 d after the BPA, only the intracerebroventricular treatment effectively reduced the hypernociception. A marked increase in B(1)R mRNA was observed in the hypothalamus, hippocampus, thalamus, and cortex at 4 d after BPA and only in the hypothalamus and cortex at 30 d. In the spinal cord, a slight increase in B(1)R mRNA expression was observed as early as at 2 d. Finally, an enhancement of B(1)R protein expression was found in all the analyzed brain structures at 4 and 30 d after the BPA, whereas in the spinal cord, this parameter was augmented only at 4 d. The data provide new evidence on the role of peripheral and central kinin B(1)R in the BPA model of neuropathic pain. Selective B(1)R antagonists might well represent valuable tools for the management of neuropathic pain.


Subject(s)
Brachial Plexus/injuries , Brachial Plexus/physiology , Neuralgia/physiopathology , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Animals , Brachial Plexus/drug effects , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin/therapeutic use , Bradykinin B1 Receptor Antagonists , Bradykinin B2 Receptor Antagonists , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/drug therapy , Pain Measurement/drug effects , Pain Measurement/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...