Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Br J Pharmacol ; 163(5): 912-26, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21306580

ABSTRACT

BACKGROUND AND PURPOSE: The two longest C-termini of the purinergic P2X receptors occur in the P2X2 and P2X7 receptors and are thought to interact with multiple cytoplasmic proteins, among which are members of the cytoskeleton, including microtubules. In this work we asked whether disrupting the microtubule cytoskeleton might affect the functions of these receptors. EXPERIMENTAL APPROACH: Functions of heterologously expressed P2X2 and P2X7 receptors were evaluated with electrophysiology and dye uptake following ATP application. Permeabilization and secretion of pro-inflammatory agents were quantified from fresh or cultured peritoneal mouse macrophages, treated in vitro or in vivo with colchicine. KEY RESULTS: Disrupting the microtubule network with colchicine did not affect currents generated by ATP in P2X2 and P2X7 receptor-expressing cells but inhibited uptake of the dye Yo-Pro-1 in Xenopus oocytes and HEK293 cells expressing these channels. Peritoneal mouse macrophages showed less ATP-induced permeabilization to ethidium bromide in the presence of colchicine, and less reactive oxygen species (ROS) formation, nitric oxide (NO) and interleukin (IL)-1ß release. Colchicine treatment did not affect ATP-evoked currents in macrophages. Finally, in vivo assays with mice inoculated with lipopolysaccharide and ATP showed diminished ROS, IL-1ß, interferon-γ and NO production after colchicine treatment. CONCLUSIONS AND IMPLICATIONS: Colchicine has known anti-inflammatory actions and is used to treat several conditions involving innate immunity, including gout and familial Mediterranean fever. Here we propose a new mechanism of action - inhibition of pore formation induced by activation of P2X receptors - which could explain some of the anti-inflammatory effects of colchicine.


Subject(s)
Adenosine Triphosphate/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colchicine/pharmacology , Ion Channel Gating/drug effects , Receptors, Purinergic P2X2/physiology , Receptors, Purinergic P2X7/physiology , Adenosine Triphosphate/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Death/drug effects , Colchicine/therapeutic use , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Female , Fluorescent Dyes/pharmacokinetics , HEK293 Cells , Humans , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/ultrastructure , Male , Mice , Microscopy, Fluorescence , Microtubules/drug effects , Microtubules/ultrastructure , Nitric Oxide/metabolism , Oocytes/drug effects , Oocytes/metabolism , Oocytes/ultrastructure , Rats , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2X2/genetics , Receptors, Purinergic P2X7/genetics , Transfection , Xenopus laevis
2.
Br J Pharmacol ; 159(8): 1716-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20401963

ABSTRACT

BACKGROUND AND PURPOSE: Compound LASSBio-881 is an orally effective antinociceptive that binds to cannabinoid receptors and is active mainly on the neurogenic component of pain models. We investigated whether transient receptor potential vanilloid subfamily type 1 (TRPV1) channels are involved in the effects of LASSBio-881. EXPERIMENTAL APPROACH: Modulation of capsaicin (CAP)- and low pH-induced currents was evaluated in TRPV1-expressing Xenopus oocytes. In vivo effects were evaluated in CAP-induced acute and inflammatory changes in nociception, as well as in partial sciatic ligation-induced thermal hypernociception. KEY RESULTS: LASSBio-881 inhibited TRPV1 currents elicited by CAP with an IC(50) of 14 microM, and inhibited proton-gated currents by 70% at 20 microM. Functional interaction with CAP was surmountable. Locally applied LASSBio-881 decreased time spent in CAP-elicited nocifensive behaviour by 30%, and given orally it reduced measures of CAP- or carrageenan-evoked thermal hypernociception by 60 and 40% respectively. In addition, LASSBio-881 decreased the paw withdrawal responses to thermal stimuli of animals with sciatic neuropathy 7-11 days after nerve ligation, at a dose of 300 micromol*kg(-1)*day(-1) p.o. At this dose, hyperthermia was not observed within 4 h following oral administration. CONCLUSIONS AND IMPLICATIONS: LASSBio-881 is a TRPV1 antagonist that apparently competes with CAP. Accordingly, LASSBio- 881 inhibited nociception in models of acute, inflammatory and neuropathic pain presumed to involve TRPV1 signalling. These in vivo actions were not hindered by hyperthermia, a common side effect of other TRPV1 antagonists. We propose that the antinociceptive properties of LASSBio-881 are due to TRPV1 antagonism, although other molecular interactions may contribute to the effects of this multi-target drug candidate.


Subject(s)
Analgesics/therapeutic use , Capsaicin/pharmacology , Hydrazines/therapeutic use , Pain/drug therapy , Sciatic Nerve/surgery , TRPV Cation Channels/antagonists & inhibitors , Administration, Oral , Analgesics/administration & dosage , Animals , Female , Hydrazines/administration & dosage , Mice , Pain/chemically induced , Pain/etiology , Rats , Rats, Wistar , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...