Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Life (Basel) ; 12(4)2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35454990

ABSTRACT

Formyl peptide receptor type 2 (FPR2/ALX) belongs to the formyl peptide receptors (FPRs) family clustered on chromosome 19 and encodes a family of three Class A of G protein-coupled receptors (GPCRs). A short N-terminal region, an NPXXY motif in transmembrane (TM) region 7 and an E/DRY motif that bridges TM3 and TM6 stabilizing inactive receptor conformations characterize this class of receptors. In recognizing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), FPRs play a crucial role in innate immune responses. FPR2/ALX is highly expressed in myeloid cells, as well as in chondrocytes, fibroblasts, endothelial, epithelial and smooth muscle cells. FPR2/ALX mRNA expression was recently reported in the rat brainstem, spinal cord, thalamus/hypothalamus, cerebral neocortex, hippocampus, cerebellum and striatum. The central nervous system (CNS) distribution of FPR2/ALX suggests important functions in nociception. Thus, the present study was carried out to investigate the possible role of FPR2/ALX in nociception in mice. Intrathecal administration of the formyl peptide receptor type 1 (FPR1) agonist fMLF and the FPR2/ALX agonist BML-111 relieved nociception and these effects were reduced by contemporary administration of the FPR2/ALX antagonist WRW4. Furthermore, measurement of cytokines and brain-derived neurotrophic factor (BDNF) in the spinal cord of neuropathic mice demonstrated that the antinociceptive effects of BML-111 might depend on the reduction in cytokine release and BDNF in the spinal cord. These results suggest a possible role of FPR2/ALX for pain control in the spinal cord.

2.
Eur J Pharmacol ; 888: 173572, 2020 Dec 05.
Article in English | MEDLINE | ID: mdl-32946866

ABSTRACT

Pyridazine derivatives, such as arylpiperazinylalkyl pyridazinones, display antinociceptive effects to thermal and chemical stimuli. Here, we extended our previous knowledge on the pharmacological profile of 4-amino-6-methyl-2-(3-(4-(4-methylcyclohexa-1,3-dien-1-yl)piperazin-1-yl)propyl)-5-vinylpyridazin-3(2H)-one, here referred as ET1, paving the way for the comprehension of its complete mechanism of action. To this aim, we have evaluated the mouse behavioural responses in several animal models of pain, the effect of ET1 in the murine model of zymosan-induced paw oedema and air-pouch, assessing the cytokines and the cellular phenotype and finally, an in vitro radioligand binding study was performed on a panel of 30 different receptors. In the formalin test, ET1 reduced both neurogenic and inflammatory phase of nociception induced by the aldehyde. Similarly, ET1 strongly reduced paw licking response in the capsaicin test, the abdominal stretching in the writhing test and the carrageenan-induced thermal hyperalgesia. ET1 also evoked a long-lasting reduction of thermal hyperalgesia. Furthermore, ET1 produced a long-lasting anti-inflammatory effect in the zymosan-induced mouse paw oedema and air-pouch through the selective inhibition of inflammatory monocytes recruitment and the modulation of IL-1ß, IL-6, TNF-α and MCP-1. Binding experiments confirmed an inhibitory effect on adrenergic α1A, α1B and α2A receptors subtypes and, for the first time, a moderate affinity was observed for the following receptors: histamine H1, imidazoline I2, sigma non-opioid intracellular receptor 1 and σ2. These results prompt ET1 as a potent analgesic and anti-inflammatory agent, and support the possibility that it may be suitable for clinical applications in a wide-range of inflammatory-based diseases.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Pain Measurement/drug effects , Pain/drug therapy , Pyridazines/therapeutic use , Analgesics/chemistry , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , CHO Cells , Carrageenan/toxicity , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Male , Mice , Pain/chemically induced , Pain/pathology , Pain Measurement/methods , Pyridazines/chemistry , Pyridazines/pharmacology , Rats, Wistar
3.
Peptides ; 32(2): 266-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21126546

ABSTRACT

In guinea-pig ileum (GPI), the chemotactic peptide N-formyl-Met-Leu-Phe-OH (fMLF) possesses spasmogenic properties through the activation of formyl peptide receptors (FPRs). Despite this, the mediators involved remain to be elucidated. fMLF (1nM-1µM) induced a dose-dependent contraction of GPI (EC(50)=24nM), that is blocked by pre-treatment with the FPRs antagonist Boc(2). The pre-treatment with tetrodotoxin (TTX) atropine or with SR140333 reduced the fMLF-induced contraction, whereas with hexamethonium, MEN10627, SB222200, mepyramine, cimetidine, thioperamide or methysergide did not produce any effect. With DuP697 pre-treatment, but not with piroxicam, reduced the fMLF-induced contraction. After stimulation with 24nM fMLF, a strong increase in the PGE(2) levels was observed. Finally, the concomitant blocking of the NK(1) receptor, the muscarinic receptors and COX-2 abolished the GPI contractions induced by fMLF. fMLF induced a concentration-dependent contraction of guinea-pig jejunum (EC(50)=11nM), proximal colon (EC(50)=3.5nM) and distal colon (EC(50)=2.2nM), with a time-course similar to that observed in GPI. In these preparations as well, the co-administration of atropine, SR140333 and DuP697 abolished the contractions induced by fMLF. Intraperitoneal injection of fMLF (0.1 or 1µmol/kg) enhanced the gastrointestinal motility in mice, abolished by the co-administration of atropine, SR140333 and DuP697. In conclusion, we showed that fMLF exerts spasmogenic actions on guinea-pig intestine both in vitro and in vivo through the release of acetylcholine and substance P from myenteric motorneurons and through prostanoids, probably from the inflammatory cells of the enteric immune system.


Subject(s)
Gastrointestinal Motility/physiology , Ileum/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neurotransmitter Agents/metabolism , Prostaglandins/metabolism , Animals , Atropine/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/metabolism , Gastrointestinal Motility/drug effects , Gastrointestinal Transit/drug effects , Gastrointestinal Transit/physiology , Guinea Pigs , Ileum/drug effects , Lower Gastrointestinal Tract/drug effects , Lower Gastrointestinal Tract/physiology , Male , Mice , Mice, Inbred Strains , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , N-Formylmethionine Leucyl-Phenylalanine/administration & dosage , N-Formylmethionine Leucyl-Phenylalanine/antagonists & inhibitors , Neurokinin-1 Receptor Antagonists , Oligopeptides/pharmacology , Piperidines/pharmacology , Piroxicam/pharmacology , Quinuclidines/pharmacology , Tetrodotoxin/pharmacology , Thiophenes/pharmacology , Upper Gastrointestinal Tract/drug effects , Upper Gastrointestinal Tract/physiology
4.
J Med Chem ; 52(23): 7397-409, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19788200

ABSTRACT

A number of pyridazinone derivatives bearing an arylpiperazinylalkyl chain were synthesized and tested icv in a model of acute nociception induced by thermal stimuli in mice (tail flick). The most interesting and potent compound in this series was 6a, which showed an ED(50) = 3.5 microg, a value about 3-fold higher with respect to morphine by the same route of administration. When administered per os, 6a was 4-fold more potent than morphine in the same test, suggesting a significant bioavailability. The same compound also showed high potency in the hot plate test. The antinociceptive effect of 6a was completely reversed by pretreatment with yohimbine both in the hot plate test and in the tail flick test. This demonstrated the involvement of the adrenergic system, which was confirmed by in vitro radioligand binding studies.


Subject(s)
Analgesics/administration & dosage , Analgesics/pharmacology , Hot Temperature/adverse effects , Pain/drug therapy , Piperazines/chemistry , Pyridazines/administration & dosage , Pyridazines/pharmacology , Administration, Oral , Analgesics/chemistry , Analgesics/metabolism , Animals , Cell Line , Drug Discovery , Humans , Male , Mice , Pain/etiology , Pain/metabolism , Piperazine , Pyridazines/chemistry , Pyridazines/metabolism , Radioligand Assay , Rats , Receptors, Adrenergic/metabolism , Structure-Activity Relationship
5.
Nat Prod Commun ; 4(4): 469-72, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19475986

ABSTRACT

The potential anxiolytic and anti-depressive activity of CMP1 was studied in the elevated plus-maze test and in the forced swimming test. Furthermore, CMP1 sedative activity was evaluated in pentobarbital treated animals; the effect of CMP1 on spontaneous motor activity (total locomotion) was also evaluated. Our data show that CMP1, at doses that did not affect locomotion, was able to induce anxiolytic and sedative, but not anti-depressive effects. In conclusion, our results represent first evidence for an anxiolytic activity of this diterpenoid from Salvia cinnabarina.


Subject(s)
Anti-Anxiety Agents/pharmacology , Diterpenes/pharmacology , Salvia/chemistry , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/isolation & purification , Diterpenes/chemistry , Diterpenes/isolation & purification , Male , Mice
6.
Pharmacol Res ; 57(6): 419-25, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18508278

ABSTRACT

DMSO is one of the most common solvents used experimentally to dissolve hydrophobic substances for in vivo and in vitro purposes. A wide range of pharmacological effects exerted by DMSO has been documented in both animal and human experimental models. However, only a few and sometimes contrasting data about the effects of DMSO in animal models of nociception and inflammation are presently available. In this study, we evaluated the effects induced by DMSO and a DMSO-containing saline on thermal and chemical nociception, inflammation and locomotor activity in CD1 mice. We demonstrated that centrally or orally administered DMSO displayed anti-nociceptive effects to thermal (hot plate and tail-flick test) and chemical (formalin test) stimuli. Conversely, DMSO was able to increase both nociceptive phases in the formalin test when applied subcutaneously in the dorsal surface of the mouse hind paws 10 min before formalin administration. Oral administration of DMSO produced anti-inflammatory effects on zymosan-induced edema in the mouse paw, whereas local administration potentiated the inflammatory action exerted by zymosan. Oral and central, but not local, administration of DMSO improved the mouse locomotor activity. These results suggest that DMSO displayed opposite effects on nociception and inflammation, depending on the route of administration. New and helpful evidence about DMSO laboratory applications need to be considered in the in vivo studies to assess correctly the pharmacological properties of investigated drugs.


Subject(s)
Anti-Infective Agents, Local/therapeutic use , Dimethyl Sulfoxide/therapeutic use , Inflammation/drug therapy , Pain/drug therapy , Administration, Oral , Animals , Anti-Infective Agents, Local/administration & dosage , Dimethyl Sulfoxide/administration & dosage , Edema/chemically induced , Edema/prevention & control , Formaldehyde , Hot Temperature , Inflammation/pathology , Injections, Intraventricular , Injections, Subcutaneous , Male , Mice , Motor Activity/drug effects , Pain/pathology , Pain Measurement/drug effects , Reaction Time/drug effects
7.
J Neurosci ; 26(25): 6716-27, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16793879

ABSTRACT

Bv8, prokineticin-1 or EG-VEGF (endocrine gland-derived vascular endothelial growth factor), and prokineticin-2, are naturally occurring peptide agonists of two G-protein-coupled receptors (GPCRs), prokineticin receptor 1 (PKR1) and PKR2. PKRs are expressed in neurons in the CNS and peripheral nervous system and many dorsal root ganglion (DRG) cells expressing PKRs also express transient receptor potential vanilloid receptor-1 (TRPV1). Mice lacking the pkr1 gene were generated to explore the role of the PKR1 receptor in nociceptive signaling and in nociceptor sensitization. When compared with wild-type littermates, mice lacking the pkr1 gene showed impaired responsiveness to noxious heat, mechanical stimuli, capsaicin, and protons. In wild-type mice, activation of PKRs by the PKR agonist Bv8 caused hyperalgesia and sensitized to the actions of capsaicin. pkr1-null mice exhibited impaired responses to Bv8 but showed normal hyperalgesic responses to bradykinin and PGE2 (prostaglandin E2). Conversely, trpv1-null mice showed a reduced pronociceptive response to Bv8. Additionally, pkr1-null mice showed diminished thermal hyperalgesia after acute inflammation elicited by mustard oil and reduced pain behavior after chronic inflammation produced by complete Freund's adjuvant. The number of neurons that responded with a [Ca2+]i increase to Bv8 exposure was five times lower in pkr1-null DRG cultures than in wild-type cultures. Furthermore, Bv8-responsive neurons from pkr1-null mice showed a significant reduction in the [Ca2+]i response to capsaicin. These findings indicate a modulatory role of PKR1 in acute nociception and inflammatory pain and disclose a pharmacological interaction between PKR1 and TRPV1 in nociceptor activation and sensitization.


Subject(s)
Nociceptors/physiology , Pain/physiopathology , Receptors, G-Protein-Coupled/physiology , TRPV Cation Channels/metabolism , Animals , Behavior, Animal , Body Temperature/drug effects , Body Temperature/physiology , Calcium/metabolism , Capsaicin/adverse effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Ganglia, Spinal/cytology , Gastrointestinal Hormones/adverse effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hyperalgesia/genetics , Hyperalgesia/physiopathology , In Situ Hybridization/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Neurons, Afferent/pathology , Neuropeptides/adverse effects , Pain/chemically induced , Pain/genetics , Pain Measurement/methods , Pain Threshold/drug effects , Pain Threshold/physiology , Physical Stimulation/methods , Reaction Time/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
8.
J Med Chem ; 49(12): 3653-8, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16759107

ABSTRACT

A new environment-sensitive fluorophore, 6-N,N-(dimethylamino)-2,3-naphthalimide (6DMN) was introduced in the delta-selective opioid peptide agonist H-Dmt-Tic-Glu-NH(2) and in the mu-selective opioid peptide agonist endomorphin-2 (H-Tyr-Pro-Phe-Phe-NH(2)). Environment-sensitive fluorophores are a special class of chromophores that generally exhibit a low quantum yield in aqueous solution but become highly fluorescent in nonpolar solvents or when bound to hydrophobic sites in proteins or membranes. New fluorescent delta-selective irreversible antagonists (H-Dmt-Tic-Glu-NH-(CH(2))(5)-CO-Dap(6DMN)-NH(2) (1) and H-Dmt-Tic-Glu-Dap(6DMN)-NH(2) (2)) were identified as potential fluorescent probes showing good properties for use in studies of distribution and internalization of delta receptors by confocal laser scanning microscopy.


Subject(s)
Fluorescent Dyes/chemical synthesis , Imides/chemistry , Naphthalenes/chemistry , Oligopeptides/chemical synthesis , Opioid Peptides/chemical synthesis , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Animals , Binding, Competitive , Cell Line, Tumor , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Guinea Pigs , In Vitro Techniques , Male , Mice , Muscle Contraction , Naphthalimides , Neuroblastoma , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Radioligand Assay , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
9.
J Neurosci ; 26(19): 5109-16, 2006 May 10.
Article in English | MEDLINE | ID: mdl-16687502

ABSTRACT

Small mammalian proteins called the prokineticins [prokineticin 1 (PK1) and PK2] and two corresponding G-protein-coupled receptors [prokineticin receptor 1 (PKR1) and PKR2] have been identified recently, but the physiological role of the PK/PKR system remains mostly unexplored. Bv8, a protein extracted from frog skin, is a convenient and potent agonist for both PKR1 and PKR2, and injection of Bv8 in vivo causes a potent and long-lasting hyperalgesia. Here, we investigate the cellular basis of hyperalgesia caused by activation of PKRs. Bv8 caused increases in [Ca]i in a population of isolated dorsal root ganglion (DRG) neurons, which we identified as nociceptors, or sensors for painful stimuli, from their responses to capsaicin, bradykinin, mustard oil, or proteases. Bv8 enhanced the inward current carried by the heat and capsaicin receptor, transient receptor potential vanilloid 1 (TRPV1) via a pathway involving activation of protein kinase Cepsilon (PKCepsilon), because Bv8 caused translocation of PKCepsilon to the neuronal membrane and because PKC antagonists reduced both the enhancement of current carried by TRPV1 and behavioral hyperalgesia in rodents. The neuronal population expressing PKRs consisted partly of small peptidergic neurons and partly of neurons expressing the N52 marker for myelinated fibers. Using single-cell reverse transcriptase-PCR, we found that mRNA for PKR1 was mainly expressed in small DRG neurons. Exposure to GDNF (glial cell line-derived neurotrophic factor) induced de novo expression of functional receptors for Bv8 in a nonpeptidergic population of neurons. These results show that prokineticin receptors are expressed in nociceptors and cause heat hyperalgesia by sensitizing TRPV1 through activation of PKCepsilon. The results suggest a role for prokineticins in physiological inflammation and hyperalgesia.


Subject(s)
Amphibian Proteins/pharmacology , Calcium/metabolism , Ganglia, Spinal/metabolism , Gastrointestinal Hormones/agonists , Membrane Potentials/physiology , Neurons/metabolism , Neuropeptides/pharmacology , TRPV Cation Channels/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/agonists , Animals , Animals, Newborn , Cells, Cultured , Ganglia, Spinal/drug effects , Membrane Potentials/drug effects , Mice , Neurons/drug effects , Rats , Rats, Sprague-Dawley
10.
Br J Pharmacol ; 137(8): 1147-54, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12466223

ABSTRACT

1 The small protein Bv8, isolated from amphibian skin, belongs to a novel family of secretory proteins (Bv8-Prokineticin family, SWISS-PROT: Q9PW66) whose orthologues have been conserved throughout evolution, from invertebrates to humans. 2 When injected intravenously or subcutaneously (from 0.06 to 500 pmol kg(-1)) or intrathecally (from 6 fmol to 250 pmol) in rats, Bv8 produced an intense systemic nociceptive sensitization to mechanical and thermal stimuli applied to the tail and paws. 3 Topically delivered into one rat paw, 50 fmol of Bv8 decreased by 50% the nociceptive threshold to pressure in the injected paw without affecting the threshold in the contralateral paw. 4 The two G-protein coupled prokineticin receptors, PK-R1 and PK-R2, were expressed in rat dorsal root ganglia (DRG) and in dorsal quadrants of spinal cord (DSC) and bound Bv8 and the mammalian orthologue, EG-VEGF, with high affinity. In DSC, PK-R1 was more abundant than PK-R2, whereas both receptors were equally expressed in DRG. IC(50) of Bv8 and EG-VEGF to inhibit [(125)I]-Bv8 binding to rat DRG and DSC were 4.1+/-0.4 nM Bv8 and 76.4+/-7.6 nM EG-VEGF, in DRG; 7.3+/-0.9 nM Bv8 and 330+/-41 nM EG-VEGF, in DSC. 5 In the small diameter neurons (<30 microm) of rat DRG cultures, Bv8 concentrations, ranging from 0.2 to 10 nM, raised [Ca(2+)](i) in a dose-dependent manner. 6 These data suggest that Bv8, through binding to PK receptors of DSC and primary sensitive neurons, results in intense sensitization of peripheral nociceptors to thermal and mechanical stimuli.


Subject(s)
Amphibian Proteins , Neuropeptides , Pain Threshold/drug effects , Proteins/pharmacology , Receptors, G-Protein-Coupled , Animals , Anura , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Pain Measurement/drug effects , Pain Measurement/methods , Pain Threshold/physiology , Proteins/isolation & purification , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Peptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...