Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 67: 230-245, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28890155

ABSTRACT

The microbiota-gut-brain axis (MGBA) regulates the reciprocal interaction between chronic inflammatory bowel and psychiatric disorders. This interaction involves multiple pathways that are highly debated. We examined the behavioural, biochemical and electrophysiological alterations, as well as gut microbiota composition in a model of antibiotic-induced experimental dysbiosis. Inflammation of the small intestine was also assessed. Mice were exposed to a mixture of antimicrobials for 2weeks. Afterwards, they received Lactobacillus casei DG (LCDG) or a vehicle for up to 7days via oral gavage. Perturbation of microbiota was accompanied by a general inflammatory state and alteration of some endocannabinoidome members in the gut. Behavioural changes, including increased immobility in the tail suspension test and reduced social recognition were observed, and were associated with altered BDNF/TrkB signalling, TRPV1 phosphorylation and neuronal firing in the hippocampus. Moreover, morphological rearrangements of non-neuronal cells in brain areas controlling emotional behaviour were detected. Subsequent probiotic administration, compared with vehicle, counteracted most of these gut inflammatory, behavioural, biochemical and functional alterations. Interestingly, levels of Lachnospiraceae were found to significantly correlate with the behavioural changes observed in dysbiotic mice. Our findings clarify some of the biomolecular and functional modifications leading to the development of affective disorders associated with gut microbiota alterations.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Depression/microbiology , Endocannabinoids/metabolism , Gastrointestinal Microbiome/drug effects , Hippocampus/metabolism , Inflammation/microbiology , Neuroglia/metabolism , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Depression/metabolism , Dysbiosis/complications , Dysbiosis/metabolism , Dysbiosis/microbiology , Hippocampus/drug effects , Inflammation/complications , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/microbiology , Male , Mice, Inbred C57BL , Neuroglia/drug effects , Neurons/drug effects , Neurons/metabolism , Probiotics/administration & dosage
2.
Sci Rep ; 7(1): 375, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28336953

ABSTRACT

The endogenous fatty acid amide palmitoylethanolamide (PEA) has been shown to exert anti-inflammatory actions mainly through inhibition of the release of pro-inflammatory molecules from mast cells, monocytes and macrophages. Indirect activation of the endocannabinoid (eCB) system is among the several mechanisms of action that have been proposed to underlie the different effects of PEA in vivo. In this study, we used cultured rat microglia and human macrophages to evaluate whether PEA affects eCB signaling. PEA was found to increase CB2 mRNA and protein expression through peroxisome proliferator-activated receptor-α (PPAR-α) activation. This novel gene regulation mechanism was demonstrated through: (i) pharmacological PPAR-α manipulation, (ii) PPAR-α mRNA silencing, (iii) chromatin immunoprecipitation. Moreover, exposure to PEA induced morphological changes associated with a reactive microglial phenotype, including increased phagocytosis and migratory activity. Our findings suggest indirect regulation of microglial CB2R expression as a new possible mechanism underlying the effects of PEA. PEA can be explored as a useful tool for preventing/treating the symptoms associated with neuroinflammation in CNS disorders.


Subject(s)
Cell Movement/drug effects , Ethanolamines/pharmacology , Macrophages/drug effects , Microglia/drug effects , Palmitic Acids/pharmacology , Phagocytosis/drug effects , Receptor, Cannabinoid, CB2/metabolism , Amides , Animals , HEK293 Cells , Humans , Macrophages/metabolism , Microglia/metabolism , PPAR alpha/metabolism , RNA, Messenger/metabolism , Rats
3.
Mol Brain ; 8: 47, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26260027

ABSTRACT

BACKGROUND: Enhanced supraspinal glutamate levels following nerve injury are associated with pathophysiological mechanisms responsible for neuropathic pain. Chronic pain can interfere with specific brain areas involved in glutamate-dependent neuropsychological processes, such as cognition, memory, and decision-making. The medial prefrontal cortex (mPFC) is thought to play a critical role in pain-related depression and anxiety, which are frequent co-morbidities of chronic pain. Using an animal model of spared nerve injury (SNI) of the sciatic nerve, we assess bio-molecular modifications in glutamatergic synapses in the mPFC that underlie neuropathic pain-induced plastic changes at 30 days post-surgery. Moreover, we examine the effects of palmitoylethanolamide (PEA) administration on pain-related behaviours, as well as the cortical biochemical and morphological changes that occur in SNI animals. RESULTS: At 1 month, SNI was associated with mechanical and thermal hypersensitivity, as well as depression-like behaviour, cognitive impairments, and obsessive-compulsive activities. Moreover, we observed an overall glutamate synapse modification in the mPFC, characterized by changes in synaptic density proteins and amino acid levels. Finally, with regard to the resolution of pain and depressive-like syndrome in SNI mice, PEA restored the glutamatergic synapse proteins and changes in amino acid release. CONCLUSIONS: Given the potential role of the mPFC in pain mechanisms, our findings may provide novel insights into neuropathic pain forebrain processes and indicate PEA as a new pharmacological tool to treat neuropathic pain and the related negative affective states. Graphical Abstract Palmitoylethanolamide: a new pharmacological tool to treat neuropathic pain and the related negative affective states.


Subject(s)
Behavior, Animal/drug effects , Ethanolamines/therapeutic use , Glutamic Acid/metabolism , Homeostasis/drug effects , Neuralgia/drug therapy , Palmitic Acids/therapeutic use , Prefrontal Cortex/metabolism , Synapses/metabolism , Amides , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Electrophysiological Phenomena/drug effects , Ethanolamines/pharmacology , Immobilization , Male , Mice , Microglia/drug effects , Microglia/metabolism , Microinjections , Neuralgia/metabolism , Neuralgia/pathology , Neuralgia/physiopathology , Neurons/drug effects , Neurons/metabolism , Palmitic Acids/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Prefrontal Cortex/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkB/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Tail
4.
Pharmacol Res ; 91: 36-46, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25434589

ABSTRACT

Peripheral neuropathy is characterized by abnormal pain responses triggered by the release of several mediators and neuronal hyperexcitability at the spinal cord level. Emerging evidence indicates that the enhanced activity of dorsal horn neurons requires communication with glia and microglia, cells that are physiologically involved in neuronal wellbeing. Prokineticins (PKs), which include PK1 and PK2, represent a novel family of chemokines characterized by a unique structural motif comprising five disulfide bonds. They are expressed in the peripheral and central nervous system. PKs bind two G protein coupled receptors, PKR1 and PKR2, and participate in the regulation of several biological processes, including pain sensation. This study aimed to investigate the anti-nociceptive effect of PC1, a non-peptide PKR1-preferring antagonist, in a mouse model of neuropathic pain. To do this, we assessed the activity of spinal cord nociceptive neurons as well as astrocyte and microglia phenotypes after repeated administration of PC1 in vivo. PC1 treatment strongly delayed the development of thermal hyperalgesia and tactile and mechanical allodynia. It also reduced spinal microglial and glial activation 8 days post injury in spared nerve injury (SNI) mice. Neuropathic mice showed an increased level of PK2 protein in the spinal cord, mostly in astrocytes. PC1 treatment completely reversed the increased responsiveness to mechanical stimuli, the decreased threshold of neuronal activation, and the increased spontaneous activity that were observed in nociceptive specific (NS) neurons of SNI mice.


Subject(s)
Analgesics/therapeutic use , Gastrointestinal Hormones/metabolism , Neuralgia/drug therapy , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Spinal Cord/drug effects , Triazines/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Behavior, Animal/drug effects , Gastrointestinal Hormones/genetics , Hot Temperature , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Mice , Neuralgia/metabolism , Neurons/drug effects , Neurons/physiology , Neuropeptides/genetics , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Spinal Cord/metabolism , Spinal Cord/physiology , Triazines/pharmacology
5.
Glia ; 62(1): 122-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24272707

ABSTRACT

The purinergic system is highly involved in the regulation of microglial physiological processes. In addition to the accepted roles for the P2 X4,7 and P2 Y12 receptors activated by adenosine triphosphate (ATP) and adenosine diphosphate, respectively, recent evidence suggests a role for the adenosine A2A receptor in microglial cytoskeletal rearrangements. However, the expression and function of adenosine A1 receptor (A1AR) in microglia is still unclear. Several reports have demonstrated possible expression of A1AR in microglia, but a new study has refuted such evidence. In this study, we investigated the presence and function of A1AR in microglia using biomolecular techniques, live microscopy, live calcium imaging, and in vivo electrophysiological approaches. The aim of this study was to clarify the expression of A1AR in microglia and to highlight its possible roles. We found that microglia express A1AR and that it is highly upregulated upon ATP treatment. Moreover, we observed that selective stimulation of A1AR inhibits the morphological activation of microglia, possibly by suppressing the Ca(2+) influx induced by ATP treatment. Finally, we recorded the spontaneous and evoked activity of spinal nociceptive-specific neuron before and after application of resting or ATP-treated microglia, with or without preincubation with a selective A1AR agonist. We found that the microglial cells, pretreated with the A1AR agonist, exhibit lower capability to facilitate the nociceptive neurons, as compared with the cells treated with ATP alone.


Subject(s)
Microglia/physiology , Receptor, Adenosine A1/metabolism , Action Potentials/drug effects , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Cells, Cultured , Lipopolysaccharides/pharmacology , Mice , Microglia/drug effects , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptor, Adenosine A1/genetics , Spinal Cord/cytology , Spinal Cord/metabolism
6.
Br J Pharmacol ; 166(3): 950-63, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22122547

ABSTRACT

BACKGROUND AND PURPOSE: The amphibian peptide Bv8 induces potent nociceptive sensitization in rodents. Its mammalian homologue, prokineticin 2 (PROK2), is strongly up-regulated in inflamed tissues and is a major determinant in triggering inflammatory pain. Bv8 and PROK2 activate two closely related GPCRs, PK(1) and PK(2) , in a relatively non-selective fashion. To characterize better the roles of the two receptors in hyperalgesia and to obtain ligands whose binding affinity and efficacy differed for the two receptors, we modified the Bv8 molecule in regions essential for receptor recognition and activation. EXPERIMENTAL APPROACH: We modified the Bv8 molecule by substituting Trp in position 24 with Ala (A-24) and compared it with Bv8 for binding and activating PK(1) and PK(2) receptors in cell preparations and in affecting nociceptive thresholds in rodents. KEY RESULTS: A-24 preferentially bound to PK(2) receptors and activated them with a lower potency (5-fold) than Bv8. When systemically injected, A-24 induced Bv8-like hyperalgesia in rats and in mice, at doses 100 times higher than Bv8. Locally and systemically injected at inactive doses, A-24 antagonized Bv8-induced hyperalgesia. In rat and mouse models of inflammatory and post-surgical pain, A-24 showed potent and long-lasting anti-hyperalgesic activity. Unlike Bv8, A-24 increased ß-endorphin levels in mouse brain. CONCLUSIONS AND IMPLICATIONS: A-24 induced its anti-hyperalgesic effect in rodents by directly blocking nociceptor PK(1) receptors and by activating the central opioid system and the descending pain control pathway through brain PK(2) receptors.


Subject(s)
Amphibian Proteins/chemistry , Amphibian Proteins/pharmacology , Analgesics/chemistry , Analgesics/pharmacology , Neuropeptides/chemistry , Neuropeptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Alanine/chemistry , Amino Acid Substitution , Amphibian Proteins/therapeutic use , Analgesics/therapeutic use , Animals , CHO Cells , Chemotaxis/drug effects , Cricetinae , Disease Models, Animal , Dose-Response Relationship, Drug , Edema/drug therapy , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Ligands , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Neuropeptides/therapeutic use , Pain, Postoperative/drug therapy , Pain, Postoperative/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Transfection , Tryptophan/chemistry
8.
Neurobiol Dis ; 37(1): 177-85, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19804829

ABSTRACT

Neuropathic pain is a devastating neurological disease that seriously affects quality of life in patients. The mechanisms leading to the development and maintenance of neuropathic pain are still poorly understood. However, recent evidence points towards a role of spinal microglia in the modulation of neuronal mechanisms. In this context, cannabinoids are thought to modulate synaptic plasticity as well as glial functions. Here, we have investigated the effect of chronic treatment with a selective agonist of cannabinoid type 2 receptor (CB2), 1-(2',4'-dichlorophenyl)-6-methyl-N-cyclohexylamine-1,4-dihydroindeno[1,2-c]pyrazole-3 carboxamide (NESS400), on pain thresholds in the spared nerve injury (SNI) model in the mouse and on the distribution and activation of spinal microglia. Repeated treatment with NESS400 (4 mg/kg) significantly alleviated neuropathic mechanical allodynia and thermal hyperalgesia. In the dorsal horn (L4-L6) of neuropathic mice microglia activation (quantification of the length of microglial processes) and astrocytosis were associated with CB2 receptor over-expression on both cell types. Treatment with NESS400 significantly reduced the number of hypertrophic microglia while leaving microglial cell number unaffected and reduced astrogliosis. Moreover, prolonged administration of NESS400 reduced mRNA expression of pro-inflammatory markers and enhanced anti-inflammatory marker gene expression in dorsal horn extracts. In conclusion, we show that selective CB2 receptor stimulation prevents thermal hyperalgesia, alleviates mechanical allodynia and facilitates the proliferation of anti-inflammatory microglial phenotype in the ipsilateral dorsal horn of the spinal cord in SNI mice.


Subject(s)
Analgesics/pharmacology , Indenes/pharmacology , Microglia/drug effects , Pain/drug therapy , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/agonists , Trauma, Nervous System/drug therapy , Animals , Astrocytes/drug effects , Astrocytes/physiology , Cell Count , Cytokines/metabolism , Gliosis/drug therapy , Gliosis/physiopathology , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/physiology , Pain/physiopathology , Pain Threshold/drug effects , Physical Stimulation , RNA, Messenger/metabolism , Spinal Cord/drug effects , Spinal Cord/physiopathology , Temperature , Time Factors , Trauma, Nervous System/physiopathology
9.
Br J Pharmacol ; 150(6): 766-81, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17279090

ABSTRACT

BACKGROUND AND PURPOSE: N-arachidonoyl-serotonin (AA-5-HT) is an inhibitor of fatty acid amide hydrolase (FAAH)-catalysed hydrolysis of the endocannabinoid/ endovanilloid compound, anandamide (AEA). We investigated if AA-5-HT antagonizes the transient receptor potential vanilloid-1 (TRPV1) channel and, as FAAH and TRPV1 are targets for analgesic compounds, if it exerts analgesia in rodent models of hyperalgesia. EXPERIMENTAL APPROACH: AA-5-HT was tested in vitro, on HEK-293 cells overexpressing the human or the rat recombinant TRPV1 receptor, and in vivo, in rats and mice treated with formalin and in rats with chronic constriction injury of the sciatic nerve. The levels of the endocannabinoids, AEA and 2-arachidonoylglycerol, in supraspinal (periaqueductal grey, rostral ventromedial medulla), spinal or peripheral (skin) tissues were measured. KEY RESULTS: AA-5-HT behaved as an antagonist at both rat and human TRPV1 receptors (IC(50)=37-40 nM against 100 nM capsaicin). It exerted strong analgesic activity in all pain models used here. This activity was partly due to FAAH inhibition, elevation of AEA tissue levels and indirect activation of cannabinoid CB(1) receptors, as it was reversed by AM251, a CB(1) antagonist. AA-5-HT also appeared to act either via activation/desensitization of TRPV1, following elevation of AEA, or as a direct TRPV1 antagonist, as suggested by the fact that its effects were either reversed by capsazepine and 5'-iodo-resiniferatoxin, two TRPV1 antagonists, or mimicked by these compounds administered alone. CONCLUSIONS AND IMPLICATIONS: Possibly due to its dual activity as a FAAH inhibitor and TRPV1 antagonist, AA-5-HT was highly effective against both acute and chronic peripheral pain.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics, Non-Narcotic/pharmacology , Arachidonic Acids/pharmacology , Serotonin/analogs & derivatives , TRPV Cation Channels/antagonists & inhibitors , Amides , Analgesics, Non-Narcotic/administration & dosage , Animals , Arachidonic Acids/administration & dosage , Cannabinoid Receptor Modulators/metabolism , Cell Line , Endocannabinoids , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Ethanolamines , Injections, Subcutaneous , Male , Mice , Pain Measurement , Palmitic Acids/metabolism , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Serotonin/administration & dosage , Serotonin/pharmacology , TRPV Cation Channels/genetics
10.
Pain ; 131(3): 281-292, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17317007

ABSTRACT

This study was undertaken in order to investigate the effect of 2-chloro-2'-C-methyl-N(6)-cyclopentyladenosine (2'-Me-CCPA), a potent and highly selective adenosine A(1) receptor agonist, on nociceptive responses and on the ongoing or tail flick-related changes of rostral ventromedial medulla (RVM) ON- and OFF-cell activities. Systemic administrations of 2'-Me-CCPA (2.5-5 mg/kg, i.p.) reduced the nociceptive response in the plantar and formalin tests, in a way prevented by DPCPX (3 mg/kg, i.p.), a selective A(1) receptor antagonist. Similarly, intra-periaqueductal grey (PAG) 2'-Me-CCPA (0.5-1-2 nmol/rat) reduced pain behaviour in the plantar and formalin tests, in a way inhibited by DPCPX (0.5 nmol/rat). Moreover, when administered systemically (2.5-5 mg/kg, i.p.) or intra-PAG (0.5-1 nmol/rat) 2'-Me-CCPA increased the tail flick latencies, delayed the tail flick-related onset of the ON-cell burst and decreased the duration of the OFF-cell pause in a dose dependent manner. Furthermore, it decreased RVM ON-cell and increased OFF-cell ongoing activities. The in vivo electrophysiological effects were all prevented by DPCPX (0.5 nmol/rat). This study confirms the role of adenosine A(1) receptors in modulating pain and suggests a critical involvement of these receptors within PAG-RVM descending pathway for the processing of pain.


Subject(s)
Adenosine A1 Receptor Agonists , Adenosine/analogs & derivatives , Analgesics/administration & dosage , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Pain/drug therapy , Pain/physiopathology , Adenosine/administration & dosage , Animals , Dose-Response Relationship, Drug , Male , Pain Threshold/drug effects , Rats , Rats, Wistar
11.
Neuropharmacology ; 52(2): 253-62, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17113112

ABSTRACT

In this study, the effect of (S)-3,4-dicarboxyphenylglycine (DCPG), a selective mGlu8 receptor agonist, has been investigated in inflammatory and neuropathic pain models in order to elucidate the role of mGlu8 receptor in modulating pain perception. Inflammatory pain was induced by the peripheral injection of formalin or carrageenan in awake mice. Systemic administration of (S)-3,4-DCPG, performed 15 min before formalin, decreased both early and delayed nociceptive responses of the formalin test. When this treatment was carried out 15 min after the peripheral injection of formalin it still reduced the late hyperalgesic phase. Similarly, systemic (S)-3,4-DCPG reduced carrageenan-induced thermal hyperalgesia and mechanical allodynia when administered 15 min before carrageenan, but no effect on pain behaviour was observed when (S)-3,4-DCPG was given after the development of carrageenan-induced inflammatory pain. When microinjected into the lateral PAG (RS)-alpha-methylserine-O-phoshate (MSOP), a group III receptor antagonist, antagonised the analgesic effect induced by systemic administration of (S)-3,4-DCPG in both of the inflammatory pain models. Intra-lateral PAG (S)-3,4-DCPG reduced pain behaviour when administered 10 min before formalin or carrageenan; both the effects were blocked by intra-lateral PAG MSOP. (S)-3,4-DCPG was ineffective in alleviating thermal hyperalgesia and mechanical allodynia 7 days after the chronic constriction injury of the sciatic nerve, whereas it proved effective 3 days after surgery. Taken together these results suggest that stimulation of mGlu8 receptors relieve formalin and carrageenan-induced hyperalgesia in inflammatory pain, whereas it would seem less effective in established inflammatory or neuropathic pain.


Subject(s)
Benzoates/therapeutic use , Glycine/analogs & derivatives , Inflammation/drug therapy , Pain/drug therapy , Receptors, Metabotropic Glutamate/agonists , Analysis of Variance , Animals , Benzhydryl Compounds/therapeutic use , Carrageenan , Dinucleoside Phosphates , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Interactions , Excitatory Amino Acid Antagonists/administration & dosage , Formaldehyde , Glycine/therapeutic use , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Inflammation/etiology , Male , Mice , Pain/chemically induced , Pain Measurement/drug effects , Pain Threshold/drug effects , Phosphoserine/administration & dosage , Reaction Time/drug effects
12.
Br J Pharmacol ; 149(4): 431-40, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16953186

ABSTRACT

BACKGROUND AND PURPOSE: We have previously reported the development of CB-25 and CB-52, two ligands of CB1 and CB2 cannabinoid receptors. We assessed here their functional activity. EXPERIMENTAL APPROACH: The effect of the two compounds on forskolin-induced cAMP formation in intact cells or GTP-gamma-S binding to cell membranes, and their action on nociception in vivo was determined. KEY RESULTS: CB-25 enhanced forskolin-induced cAMP formation in N18TG2 cells (EC50 approximately 20 nM, max. stimulation = 48%), behaving as an inverse CB1 agonist, but it stimulated GTP-gamma-S binding to mouse brain membranes, behaving as a partial CB1 agonist (EC50 =100 nM, max. stimulation = 48%). At human CB1 receptors, CB-25 inhibited cAMP formation in hCB1-CHO cells (EC50 = 1600 nM, max. inhibition = 68% of CP-55,940 effect). CB-52 inhibited forskolin-induced cAMP formation by N18TG2 cells (IC50 = 450 nM, max. inhibition = 40%) and hCB1-CHO cells (EC50 = 2600 nM, max. inhibition = 62% of CP-55,940 effect), and stimulated GTP-gamma-S binding to mouse brain membranes (EC50 = 11 nM, max. stimulation approximately 16%). Both CB-25 and CB-52 showed no activity in all assays of CB2-coupled functional activity and antagonized CP55940-induced stimulation of GTP-gamma-S binding to hCB2-CHO cell membranes. In vivo, both compounds, administered i.p., produced dose-dependent nociception in the plantar test carried out in healthy rats, and antagonised the anti-nociceptive effect of i.p. WIN55,212-2. In the formalin test in mice, however, the compounds counteracted both phases of formalin-induced nociception. CONCLUSIONS AND IMPLICATIONS: CB-25 and CB-52 behave in vitro mostly as CB1 partial agonists and CB2 neutral antagonists, whereas their activity in vivo might depend on the tonic activity of cannabinoid receptors.


Subject(s)
Amides/pharmacology , Analgesics/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Resorcinols/pharmacology , Action Potentials/drug effects , Adenylyl Cyclases/metabolism , Animals , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/metabolism , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Ligands , Male , Mice , Pain/chemically induced , Pain/metabolism , Pain/prevention & control , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Transfection
13.
Curr Neuropharmacol ; 4(3): 225-31, 2006 Jul.
Article in English | MEDLINE | ID: mdl-18615148

ABSTRACT

Metabotropic glutamate (mGlu) and cannabinoid receptors are G-protein coupled receptors which have shown synaptic co-operation through small lipid messengers in the central nervous system (CNS). A functional interaction between these two receptor families could have a relevant potential in the treatment of CNS disorders, including chronic pain. Indeed, both mGlu and cannabinoid receptors play a crucial role in the neurobiology of pain and their simultaneous manipulation could lead to novel strategies in pain management. In particular, as both mGlu and cannabinoid receptors have been found in the periaqueductal gray (PAG), a crucial station in the pain modulatory system, these receptors could be a substrate for producing analgesia at this level. In this review we aim to briefly illustrate the role of mGlu and can-nabinoid receptors in controlling nociceptive processes, some points of convergence, and their functional interaction in pain processing. Further insights into this functional linkage between the mGlu and cannabinoid receptors could pave the way to a new strategy for pain relief, such as a drug cocktail acting on cannabinoid/metabotropic glutamate receptors.

14.
Neuroscience ; 134(1): 269-81, 2005.
Article in English | MEDLINE | ID: mdl-15953687

ABSTRACT

This study was undertaken to analyze the involvement of periaqueductal gray (PAG) cannabinoid or group I metabotropic glutamate receptors in the formalin-induced changes on the rostral ventromedial medulla (RVM) ON- and OFF-cells activities. S.c. injection of formalin into the hind paw produced a transient decrease (4-6 min) followed by a longer increase (25-35 min) in tail flick latencies. Formalin also increased basal activity in RVM ON-cells (42+/-7%) and decreased it in OFF-cells (35+/-4%). Intra-PAG microinjection of (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl) pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone mesylate (WIN 55,212-2) (2 nmol/rat), a cannabinoid receptor agonist, prevented the formalin-induced changes in RVM cell activities. Higher dosages of WIN 55,212-2 (4-8 nmol/rat) increased the tail flick latencies, delayed the tail flick-related onset to ON-cell burst, and decreased the duration of OFF-cell pause. Furthermore, WIN 55,212-2 at a dosage of 8 nmol/rat decreased RVM ON-cell (57+/-7%) and increased OFF-cell ongoing activities (26+/-4%). These effects were prevented by N-piperidino-5-(4-chlorophenyl)-1-(2,4dichlorophenyl)-4-methyl-3-pyrazolecarboxamide SR141716A, (1 pmol/rat), a CB1 cannabinoid receptor antagonist, or by 2-methyl-6-(phenylethynyl)pyridine (MPEP 20 nmol/rat), a selective mGlu5 glutamate receptor antagonist. T7-(hydroxyimino) cyclopropa[b]chromen-1alpha-carboxylate ethyl ester (CPCOOE/50 nmol/rat) and (S)-(+)-alpha-amino-4-carboxy-2-methylbenzeneacetic acid (LY367385, 20 nmol/rat), selective mGlu1 glutamate receptor antagonists, were ineffective in preventing the WIN-induced effects. This study suggests that s.c. injection of formalin modifies RVM neuronal activities and this effect is prevented by PAG cannabinoid receptor stimulation. Moreover, the physiological stimulation of PAG mGlu5, but not mGlu1 glutamate receptors, seems to be required for the cannabinoid-mediated effect.


Subject(s)
Formaldehyde/pharmacology , Medulla Oblongata/physiology , Periaqueductal Gray/drug effects , Receptor, Cannabinoid, CB1/metabolism , Receptors, Metabotropic Glutamate/metabolism , Action Potentials/drug effects , Analgesics/pharmacology , Animals , Benzoates/pharmacology , Benzoxazines , Chromones/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Excitatory Amino Acid Antagonists/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Injections, Subcutaneous/methods , Male , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/classification , Neurons/drug effects , Pain Measurement/methods , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Reaction Time/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant
15.
Curr Drug Targets ; 6(1): 75-80, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15720215

ABSTRACT

Neuropathic pain is defined as pain initiated or caused by a primary lesion or dysfunction in the nervous system. It is a devastating and difficult to manage consequence of peripheral nerve injury and has a variety of clinical symptoms. Neuropathic pain is a major health problem. It has been estimated that 70% of patients with advanced cancer and inflammatory pathologies are afflicted by chronic pain. About 95% of patients with spinal cord injuries have neuropathic pain problems. Chronic pain is debilitating and cause of depression and decreasing quality of life. Pharmacological treatment for the symptoms of painful neuropathy is difficult, because there has been limited understanding of the underlying causes and systemic levels that an effective dose can have on multiple side effects. The use of molecular methods, such as gene therapy, stem cell therapy and viral vector for delivery of biologic antinociceptive molecules, has led to a better understanding of the underlying mechanisms of the induction of intractable neuropathic pain.


Subject(s)
Genetic Therapy/methods , Neuralgia/therapy , Animals , Drug Therapy/methods , Genetic Therapy/trends , Humans , Neuralgia/etiology , Neuralgia/physiopathology
16.
Neuropharmacology ; 43(7): 1061-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12504911

ABSTRACT

In this study we investigated the effects of type I and II mGlu receptors ligands in glycine extracellular concentrations at the periaqueductal gray (PAG) level by using in vivo microdialysis, in conscious rats. An agonist of type I mGlu receptors, (S)-3,5-DHPG (1 and 5 mM), but not a selective agonist for mGlu5 receptors, CHPG (3 and 5 mM), was noticed to increase the dialysate glycine levels in a concentration-dependent manner (60+/-15% and 136+/-13%, respectively). CPCCOEt (1mM), a selective mGlu1 receptor antagonist, perfused in combination with (S)-3,5-DHPG, counteracted the effect induced by (S)-3,5-DHPG, but did not change per se the extracellular PAG glycine values, even at the highest dosage used (2 mM). MPEP (1 and 2 mM), a selective antagonist of mGlu5 receptor, did not modify extracellular glycine level. An agonist of type II mGlu receptors, 2R,4R-APDC (25 and 50 microM), decreased the dialysate glycine in a concentration-dependent manner (-26+/-4% and -54+/-6%, respectively). The 2R,4R-APDC-induced decrease in extracellular glycine was prevented by EGlu (0.5 mM), a selective type II mGlu receptors antagonist. EGlu (0.5 and 1 mM), per se, led to a significant decrease (-56+/-7% and -57+/-2%, respectively) in extracellular PAG glycine too. This effect was prevented by DPCPX (100 microM), a selective antagonist for A1 adenosine receptors, but was not affected by CPA (1 mM), a selective A1 adenosine receptors agonist. Intra-PAG perfusion of CPA (0.1-1 mM) decreased the extracellular PAG glycine values (-47+/-13%) with 1 mM concentration. The CPA-induced effect was prevented by DPCPX (100 microM), and resulted to be additive with the 2R,4R-APDC-induced decrease in glycine values. DPCPX (1 mM) increased per se extracellular glycine (48+/-7%) at the highest dose used. Dipyridamole (100 microM), an inhibitor of both adenosine reuptake and phosphodiesterases, decreased extracellular glycine (-28+/-7%). Extracellular concentrations of glutamine never changed throughout this study. These data show opposing effects of type I and II mGlu receptors in the regulation of PAG glycine values. Moreover, functional interaction between type II mGlu and adenosine A1 receptors, which possibly operate through a common transductional pathway, may be relevant in the physiological control of glycine release in awake, freely moving rats at the periaqueductal gray matter.


Subject(s)
Glycine/metabolism , Periaqueductal Gray/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/physiology , Receptors, Purinergic P1/metabolism , Animals , Male , Microdialysis/methods , Rats , Rats, Wistar
17.
Neuropharmacology ; 40(3): 319-26, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11166324

ABSTRACT

The purpose of this study was to evaluate the possible contribution of metabotropic glutamate receptors (mGluRs) to cannabinoid-induced antinociception in the periaqueductal grey (PAG) matter of rats. Intra-PAG microinjection of WIN 55,212-2, a cannabinoid receptor agonist, increased the latency of the nociceptive reaction (NR) in a dose-dependent fashion in the plantar test. This effect was prevented by pretreatment with SR141716A, a selective antagonist of CB1 receptors. When injected alone, SR141716A produced, with the highest dosage used, a significant reduction in the latency of the NR. CPCCOEt, a selective mGlu1 receptor antagonist, was unable to prevent the analgesia produced by WIN 55,212-2. On the contrary, MPEP, a selective mGlu5 receptor antagonist, completely antagonized the effect of WIN 55,212-2. However, the analgesia induced by CHPG, a selective mGlu5 receptor agonist, was blocked by MPEP but not by SR141716A. When injected alone, CPCOOEt produced no effect, whereas MPEP produced, with the highest dosage used, a significant reduction in the latency of the NR. These data emphasize that mGlu5 receptors, but not mGluR1, may modulate nociception in the PAG. Similarly, a pretreatment with either 2-(S)-alpha-EGlu or (RS)-alpha-MSOP, selective antagonists for group II and III mGluRs, respectively, prevented the WIN 55,212-2-induced analgesia. When the higher dosage of (RS)-alpha-MSOP was used a decrease in the latency of the NR was observed. This was not the case for 2-(S)-alpha-EGlu. Pretreatment with DL-AP5, a selective antagonist of N-methyl-D-aspartate (NMDA) receptors, blocked the effect of WIN 55,212-2, and by increasing the dosage strongly reduced per se the latency of the NR. This study suggests that endogenous glutamate could tonically modulate nociception through mGlu and NMDA receptors in the PAG matter. In particular, the physiological stimulation of these receptors seems to be required for the cannabinoid-induced analgesia in this midbrain area.


Subject(s)
Analgesics/pharmacology , Cannabinoids/pharmacology , Periaqueductal Gray/drug effects , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Benzoxazines , Calcium Channel Blockers/administration & dosage , Cannabinoids/antagonists & inhibitors , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Male , Microinjections , Morpholines/administration & dosage , Morpholines/antagonists & inhibitors , Naphthalenes/administration & dosage , Naphthalenes/antagonists & inhibitors , Pain Measurement/drug effects , Periaqueductal Gray/metabolism , Piperidines/administration & dosage , Pyrazoles/administration & dosage , Rats , Rats, Wistar , Reaction Time/drug effects , Receptor, Metabotropic Glutamate 5 , Receptors, Drug/antagonists & inhibitors , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Rimonabant
18.
Neuroreport ; 10(7): 1403-7, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10380954

ABSTRACT

Glutamate and GABA are important nociception modulating transmitters in specific brain regions, i.e. the spinal cord, the thalamic nuclei and the periaqueductal gray (PAG). However, quantitative and topographical changes in glutamate and GABA release in these brain regions during peripheral inflammation episodes have not been characterized in awake animals. To address this issue, an in vivo microdialysis study was carried out in freely moving rats in order to analyze PAG extracellular glutamate and GABA concentrations following unilateral formalin injection into the dorsal skin of the right hind-paw. Both glutamate and GABA release decreased after the injection of formalin during phase I and phase II of hyperalgesia. Because naloxone prevented the decrease of GABA and glutamate release induced by formalin, this study shows that, in vivo, a nociceptive stimulation may activate opioidergic fibres into the PAG. The increased release of endogenous opioids may, in turn, inhibit the activity of the GABAergic neurons (i.e. opioid disinhibition). Formalin injection also decreased extracellular glutamate concentration. However, we found that intra-PAG perfusion with tetrodotoxin only decreased GABA, but not glutamate dialysate values. Although it should be reasonable to speculate that opioids also inhibit glutamate fibres, further investigation is needed to clarify whether or not the dialysate glutamate we measured reflects change in the metabolism or neurotransmitter pool of this amino acid.


Subject(s)
Formaldehyde/pharmacology , Glutamic Acid/metabolism , Neuritis/metabolism , Periaqueductal Gray/metabolism , gamma-Aminobutyric Acid/metabolism , Analysis of Variance , Animals , Injections, Subcutaneous , Male , Neuritis/chemically induced , Rats , Rats, Wistar
19.
Naunyn Schmiedebergs Arch Pharmacol ; 358(4): 411-7, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9826062

ABSTRACT

The role of metabotropic (mGluRs) and N-methyl-D-aspartate (NMDA) glutamate receptors on 5-hydroxytryptamine (5-HT) release has been studied in rat periaqueductal gray (PAG) matter by using in vivo microdialysis. (1S,3R)-aminocyclopentane- 1,3-dicarboxylic acid [(IS,3R)-ACPD; 0.5 or 1 mM], a group I/group II mGluRs agonist, increased the dialysate 5-HT concentration. (2S)-alpha-ethylglutamic acid (EGlu; 1 mM), an antagonist of group II mGluRs, but not (RS)-1-aminoindan-1,5-dicarboxylic acid (AIDA; 1 mM), an antagonist of group I mGluRs, antagonized the 1S,3R-ACPD-induced effect. (S)-3,5-dihydroxyphenylglycine (DHPG; 0.5 and 1 mM), an agonist of group I mGluRs, did not modify dialysate 5-HT. (2S, 3S, 4S)-alpha-(carboxycyclopropyl)-glycine (L-CCG-I; 0.5 and I mM), an agonist of group II mGluRs, increased extracellular 5-HT. This effect was antagonized by EGlu. Similarly, L-serine-O-phosphate (L-SOP; 1 and 10 mM), an agonist of group III mGluRs, increased extracellular 5-HT and this effect was antagonized by (RS)-(alpha-methylserine O-phosphate (M-SOP; 1 mM), an antagonist of group III mGluRs. Out of the several N-methylD-aspartate concentrations used (NMDA; 10, 50, 100, 500 and 1000 microM) only the 50 microM infusion significantly decreased dialysate 5-HT. The GABA(A) receptor agonist, bicuculline (30 microM), increased 5-HT release on its own and antagonized the decrease caused by the opiate antagonist, naloxone (2 mM), as well as the increases caused by CCG-I or L-SOP. These data show that stimulation of PAG's group II/group II mGluRs increases 5-HT release, while stimulation of NMDA glutamate receptors may decrease it. We speculate that glutamate does not modulate 5-HT release in the PAG directly, but via activation of tonically active GABAergic interneurons.


Subject(s)
Periaqueductal Gray/metabolism , Receptors, Metabotropic Glutamate/physiology , Serotonin/metabolism , Animals , Cycloleucine/pharmacology , Male , Microdialysis , Phosphoserine/pharmacology , Rats , Rats, Wistar , Receptors, GABA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Opioid/physiology
20.
Br J Pharmacol ; 125(3): 437-40, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9806324

ABSTRACT

1. Pharmacological studies have suggested that A3 receptors are present on central neurons. Recently this adenosine receptor subtype has been identified in the rat and its presence in the central nervous system has been confirmed. 2. In this study we investigated the effects of acute intracerebroventricular (i.c.v.) injections of N6-2-(4-aminophenyl)-ethyladenosine (APNEA), a non-selective A3 adenosine receptor agonist, on arterial blood pressure (ABP) and heart rate (HR), after treatment with 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), a selective antagonist of A1 adenosine receptors. 3. Anaesthetized rats, after DPCPX (12 microg(-1) kg i.c.v.), were treated with APNEA (0.4-4 microg kg(-1) i.c.v.) resulting in a transitory and dose-dependent decrease in arterial blood pressure without a change in heart rate. APNEA also induced hypotensive responses after i.c.v. pretreatment with aminophylline, at a dose of 20 microg kg(-1). In contrast, pretreatment 48 h before, with 4 microg kg(-1) i.c.v. of pertussis toxin reduced the hypotensive effect induced by APNEA. Administration of APNEA at a higher dose (20 microg kg(-1) i.c.v.), after DPCPX, induced a decrease in ABP of -66+/-5.4 mmHg and after 3 min a decrease in heart rate of -62+/-6.0 beats min(-1). Transection of the spinal cord abolished this significant fall in ABP, but not the decrease of HR. 4. These results suggest that a population of A3-receptors is present in the CNS, whose activation induces a decrease in blood pressure with no change of heart rate.


Subject(s)
Blood Pressure/physiology , Central Nervous System/physiology , Receptors, Purinergic P1/physiology , Animals , Heart Rate/physiology , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...