Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Drug Metab Dispos ; 43(9): 1326-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26109562

RESUMO

Cytochrome P450 monooxygenases (P450s), which are well-known drug-metabolizing enzymes, are thought to play a signal transduction role in µ opioid analgesia and may serve as high-affinity (3)H-cimetidine ((3)HCIM) binding sites in the brain. (3)HCIM binding sites may also be related to opioid or nonopioid analgesia. However, of the more than 100 murine P450 enzymes, the specific isoform(s) responsible for either function have not been identified. Presently, three lines of constitutive P450 gene cluster knockout (KO) mice with full-length deletions of 14 Cyp2c, 9 Cyp2d, and 7 Cyp3a genes were studied for deficiencies in (3)HCIM binding and for opioid analgesia. Liver and brain homogenates from all three genotypes showed normal (3)HCIM binding values, indicating that gene products of Cyp2d, Cyp3a, and Cyp2c are not (3)HCIM-binding proteins. Cyp2d KO and Cyp3a KO mice showed normal antinociceptive responses to a moderate systemic dose of morphine (20 mg/kg, s.c.), thereby excluding 16 P450 isoforms as mediators of opioid analgesia. In contrast, Cyp2c KO mice showed a 41% reduction in analgesic responses following systemically (s.c.) administered morphine. However, the significance of brain Cyp2c gene products in opioid analgesia is uncertain because little or no analgesic deficits were noted in Cyp2c KO mice following intracerebroventricular or intrathecalmorphine administration, respectively. These results show that the gene products of Cyp2d and Cyp3a do not contribute to µ opioid analgesia in the central nervous system. A possible role for Cyp2c gene products in opioid analgesia requires further consideration.


Assuntos
Analgésicos Opioides/administração & dosagem , Sistema Enzimático do Citocromo P-450/metabolismo , Isoenzimas/metabolismo , Analgésicos Opioides/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/genética , Isoenzimas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
J Pharmacol Exp Ther ; 347(3): 746-54, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24045421

RESUMO

Nicotine metabolism is believed to affect not only nicotine's pharmacological effects but also nicotine addiction. As a key step toward testing this hypothesis, we have studied nicotine metabolism and nicotine's pharmacological and behavioral effects in a novel knockout mouse model [named Cyp2a(4/5)bgs-null] lacking a number of cytochrome P450 genes known to be or possibly involved in nicotine metabolism, including two Cyp2a and all Cyp2b genes. We found that, compared with wild-type mice, the Cyp2a(4/5)bgs-null mice showed >90% decreases in hepatic microsomal nicotine oxidase activity in vitro, and in rates of systemic nicotine clearance in vivo. Further comparisons of nicotine metabolism between Cyp2a(4/5)bgs-null and Cyp2a5-null mice revealed significant roles of both CYP2A5 and CYP2B enzymes in nicotine clearance. Compared with the behavioral responses in wild-type mice, the decreases in nicotine metabolism in the Cyp2a(4/5)bgs-null mice led to prolonged nicotine-induced acute pharmacological effects, in that null mice showed enhanced nicotine hypothermia and antinociception. Furthermore, we found that the Cyp2a(4/5)bgs-null mice developed a preference for nicotine in a conditioned place preference test, a commonly used test of nicotine's rewarding effects, at a nicotine dose that was 4-fold lower than what was required by wild-type mice. Thus, CYP2A/2B-catalyzed nicotine clearance affects nicotine's behavioral response as well as its acute pharmacological effects in mice. This result provides direct experimental support of the findings of pharmacogenetic studies that suggest linkage between rates of nicotine metabolism and smoking behavior in humans.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Comportamento Animal/efeitos dos fármacos , Nicotina/metabolismo , Nicotina/farmacologia , Agonistas Nicotínicos/metabolismo , Agonistas Nicotínicos/farmacologia , Esteroide Hidroxilases/genética , Animais , Temperatura Corporal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Citocromo P-450 CYP2A6 , Família 2 do Citocromo P450 , Meia-Vida , Imersão , Masculino , Camundongos , Camundongos Knockout , Medição da Dor/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Recompensa
3.
J Neurophysiol ; 108(9): 2393-404, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956800

RESUMO

Respiratory depression is a therapy-limiting side effect of opioid analgesics, yet our understanding of the brain circuits mediating this potentially lethal outcome remains incomplete. Here we studied the contribution of the rostral ventromedial medulla (RVM), a region long implicated in pain modulation and homeostatic regulation, to opioid-induced respiratory depression. Microinjection of the µ-opioid agonist DAMGO in the RVM of lightly anesthetized rats produced both analgesia and respiratory depression, showing that neurons in this region can modulate breathing. Blocking opioid action in the RVM by microinjecting the opioid antagonist naltrexone reversed the analgesic and respiratory effects of systemically administered morphine, showing that this region plays a role in both the analgesic and respiratory-depressant properties of systemically administered morphine. The distribution of neurons directly inhibited by RVM opioid microinjection was determined with a fluorescent opioid peptide, dermorphin-Alexa 594, and found to be concentrated in and around the RVM. The non-opioid analgesic improgan, like DAMGO, produced antinociception but, unlike DAMGO, stimulated breathing when microinjected into the RVM. Concurrent recording of RVM neurons during improgan microinjection showed that this agent activated RVM ON-cells, OFF-cells, and NEUTRAL-cells. Since opioids are known to activate OFF-cells but suppress ON-cell firing, the differential respiratory response to these two analgesic drugs is best explained by their opposing effects on the activity of RVM ON-cells. These findings show that pain relief can be separated pharmacologically from respiratory depression and identify RVM OFF-cells as important central targets for continued development of potent analgesics with fewer side effects.


Assuntos
Analgésicos Opioides/toxicidade , Bulbo/efeitos dos fármacos , Neurônios/fisiologia , Dor Nociceptiva/fisiopatologia , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/antagonistas & inibidores , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/antagonistas & inibidores , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Masculino , Bulbo/citologia , Bulbo/fisiologia , Morfina/antagonistas & inibidores , Morfina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/fisiopatologia
4.
J Pharmacol Exp Ther ; 336(1): 30-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20864501

RESUMO

Histamine H(3) receptors (H(3)Rs), distributed within the brain, the spinal cord, and on specific types of primary sensory neurons, can modulate pain transmission by several mechanisms. In the skin, H(3)Rs are found on certain Aß fibers, and on keratinocytes and Merkel cells, as well as on deep dermal, peptidergic Aδ fibers terminating on deep dermal blood vessels. Activation of H(3)Rs on the latter in the skin, heart, lung, and dura mater reduces calcitonin gene-related peptide and substance P release, leading to anti-inflammatory (but not antinociceptive) actions. However, activation of H(3)Rs on the spinal terminals of these sensory fibers reduces nociceptive responding to low-intensity mechanical stimuli and inflammatory stimuli such as formalin. These findings suggest that H(3)R agonists might be useful analgesics, but these drugs have not been tested in clinically relevant pain models. Paradoxically, H(3) antagonists/inverse agonists have also been reported to attenuate several types of pain responses, including phase II responses to formalin. In the periaqueductal gray (an important pain regulatory center), the H(3) inverse agonist thioperamide releases neuronal histamine and mimics histamine's biphasic modulatory effects in thermal nociceptive tests. Newer H(3) inverse agonists with potent, selective, and brain-penetrating properties show efficacy in several neuropathic and arthritis pain models, but the sites and mechanisms for these actions remain poorly understood.


Assuntos
Encéfalo/metabolismo , Nociceptores/metabolismo , Dor/metabolismo , Receptores Histamínicos H3/fisiologia , Medula Espinal/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Agonistas dos Receptores Histamínicos/metabolismo , Agonistas dos Receptores Histamínicos/farmacologia , Agonistas dos Receptores Histamínicos/uso terapêutico , Humanos , Dor/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Medula Espinal/efeitos dos fármacos
5.
Drug Metab Dispos ; 39(7): 1221-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21460231

RESUMO

Arachidonate epoxidation, which mediates important biological functions in several tissues, is catalyzed by specific cytochrome P450 (P450) enzymes. Two fatty acid derivatives [2-(2-propynyloxy)-benzenehexanoic acid (PPOH) and N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide (MS-PPOH)] are used as general, mechanism-based P450 epoxygenase inactivators, but the effects of these drugs on nearly all P450 isoforms are unknown. Here, the activity of these compounds on nine human and three rat recombinant P450s was studied. As expected, PPOH inhibited five known epoxygenases [CYP2B1, 2B6, 2C6, 2C9, and 2C11 (IC(50) = 23-161 µM)] but had little or no activity on P450s typically not considered to be epoxygenases (CYP1A1, 1A2, 1B1, 2A6, 2D6, and 2E1). PPOH was only a very weak inhibitor (IC(50) = ∼300 µM) of CYP2C19, an important human expoxygenase. An unexpected finding was that MS-PPOH (a metabolically stable congener of PPOH) potently inhibited only two P450 epoxygenases (2C9 and 2C11, IC(50) = 11-16 µM) and showed considerably lower activity (IC(50) = >90 µM) on all other P450s tested, including three epoxygenases (CYP2B1, 2B6, and 2C19). In addition, PPOH and MS-PPOH displayed time- and NADPH-dependent inhibition of CYP2C9 and other epoxygenases. These results support the putative mechanism of action of PPOH and MS-PPOH on recombinant P450s and (with one exception) confirm a general epoxygenase inhibitory profile for PPOH. However, the heterogeneity of inhibitory potencies for MS-PPOH on these enzymes suggests caution in the use of this drug as a general epoxygenase inhibitor. These results will facilitate the judicious use of PPOH and MS-PPOH for epoxygenase research.


Assuntos
Acetileno/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Animais , Humanos , Concentração Inibidora 50 , NADP/metabolismo , Ratos , Proteínas Recombinantes/antagonistas & inibidores
6.
J Neurophysiol ; 104(6): 3222-30, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926616

RESUMO

Many analgesic drugs, including µ-opioids, cannabinoids, and the novel nonopioid analgesic improgan, produce antinociception by actions in the rostral ventromedial medulla (RVM). There they activate pain-inhibiting neurons, termed "OFF-cells," defined by a nociceptive reflex-related pause in activity. Based on recent functional evidence that neuronal P450 epoxygenases are important for the central antinociceptive actions of morphine and improgan, we explored the convergence of opioid and nonopioid analgesic drug actions in RVM by studying the effects of the P450 epoxygenase inhibitor CC12 on the analgesic drug-induced activation of these OFF-cells and on behavioral antinociception. In rats lightly anesthetized with isoflurane, we recorded the effects of intraventricular morphine and improgan, with and without CC12 pretreatment, on tail flick latency and activity of identified RVM neurons: OFF-cells, ON-cells (pronociceptive neurons), and neutral cells (unresponsive to analgesic drugs). CC12 pretreatment preserved reflex-related changes in OFF-cell firing and blocked the analgesic actions of both drugs, without interfering with the increase in spontaneous firing induced by improgan or morphine. CC12 blocked suppression of evoked ON-cell firing by improgan, but not morphine. CC12 pretreatment had no effect by itself on RVM neurons or behavior. These data show that the epoxygenase inhibitor CC12 works downstream from receptors for both µ-opioid and improgan, at the inhibitory input mediating the OFF-cell pause. This circuit-level analysis thus provides a cellular basis for the convergence of opioid and nonopioid analgesic actions in the RVM. A presynaptic P450 epoxygenase may therefore be an important target for development of clinically useful nonopioid analgesic drugs.


Assuntos
Analgésicos/antagonistas & inibidores , Cimetidina/análogos & derivados , Imidazóis/farmacologia , Bulbo/efeitos dos fármacos , Morfina/antagonistas & inibidores , Percepção da Dor/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Sulfetos/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Cimetidina/antagonistas & inibidores , Citocromo P-450 CYP2J2 , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450 , Masculino , Bulbo/citologia , Bulbo/fisiologia , Modelos Neurológicos , Percepção da Dor/fisiologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptor CB1 de Canabinoide/fisiologia , Receptores Opioides mu/fisiologia , Receptores Pré-Sinápticos/efeitos dos fármacos , Receptores Pré-Sinápticos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia
7.
Bioorg Med Chem Lett ; 19(18): 5452-7, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19666223

RESUMO

A new flow cytometry method that uses an optimized DNA and RNA staining strategy to monitor the growth and development of the Plasmodium falciparum strain W2mef has been used in a pilot study and has identified Bay 43-9006 1, SU 11274 2, and TMC 125 5 as compounds that exhibit potent (<1 microM) overall and ring stage in vitro antimalarial activity.


Assuntos
Antimaláricos/farmacologia , DNA de Protozoário/análise , Citometria de Fluxo/métodos , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , RNA de Protozoário/análise , Animais , Antimaláricos/química , Descoberta de Drogas , Resistência a Medicamentos , Eritrócitos/parasitologia , Citometria de Fluxo/economia , Testes de Sensibilidade Parasitária , Plasmodium falciparum/crescimento & desenvolvimento , Relação Estrutura-Atividade
8.
Drug Metab Dispos ; 36(3): 614-21, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18094038

RESUMO

[(3)H]Cimetidine (3HCIM) specifically binds to an unidentified site in the rat brain. Because recently described ligands for this site have pharmacological activity, 3HCIM binding was characterized. 3HCIM binding was saturable, heat-labile, and distinct from the histamine H(2) receptor. To test the hypothesis that 3HCIM binds to a cytochrome P450 (P450), the effects of nonselective and isoform-selective P450 inhibitors were studied. The heme inhibitor KCN and the nonselective P450 inhibitor metyrapone both produced complete, concentration-dependent inhibition of 3HCIM binding (K(i) = 1.3 mM and 11.9 muM, respectively). Binding was largely unaffected by inhibitors of CYP1A2, 2B6, 2C8, 2C9, 2D6, 2E1, and 19A1 but was eliminated by inhibitors of CYP2C19 (tranylcypromine) and CYP3A4 (ketoconazole). Synthesis and testing of CC11 [4(5)-(benzylthiomethyl)-1H-imidazole] and CC12 [4(5)-((4-iodobenzyl)-thiomethyl)-1H-imidazole] confirmed both drugs to be high-affinity inhibitors of 3HCIM binding. On recombinant human P450s, CC12 was a potent inhibitor of CYP2B6 (IC(50) = 11.7 nM), CYP2C19 (51.4 nM), and CYP19A1 (140.7 nM) and had a range of activities (100-494 nM) on nine other isoforms. Although the 3HCIM binding site pharmacologically resembles some P450s, eight recombinant human P450s and three recombinant rat P450s did not exhibit 3HCIM binding. Inhibition by KCN and metyrapone suggests that 3HCIM binds to a heme-containing brain protein (possibly a P450). However, results with selective P450 inhibitors, recombinant P450 isoforms, and a P450 antibody did not identify a 3HCIM-binding P450 isoform. Finally, CC12 is a new, potent inhibitor of CYP2B6 and CYP2C19 that may be a valuable tool for P450 research.


Assuntos
Encéfalo/metabolismo , Cimetidina/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Cimetidina/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Antagonistas dos Receptores H2 da Histamina/metabolismo , Antagonistas dos Receptores H2 da Histamina/farmacologia , Isoenzimas , Cinética , Ligantes , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Trítio
9.
Neuropharmacology ; 52(5): 1244-55, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17336343

RESUMO

Improgan, a chemical congener of cimetidine, is a highly effective non-opioid analgesic when injected into the CNS. Despite extensive characterization, neither the improgan receptor, nor a pharmacological antagonist of improgan has been previously described. Presently, the specific binding of [(3)H]cimetidine (3HCIM) in brain fractions was used to discover 4(5)-((4-iodobenzyl)thiomethyl)-1H-imidazole, which behaved in vivo as the first improgan antagonist. The synthesis and pharmacological properties of this drug (named CC12) are described herein. In rats, CC12 (50-500nmol, i.c.v.) produced dose-dependent inhibition of improgan (200-400nmol) antinociception on the tail flick and hot plate tests. When given alone to rats, CC12 had no effects on nociceptive latencies, or on other observable behavioral or motor functions. Maximal inhibitory effects of CC12 (500nmol) were fully surmounted with a large i.c.v. dose of improgan (800nmol), demonstrating competitive antagonism. In mice, CC12 (200-400nmol, i.c.v.) behaved as a partial agonist, producing incomplete improgan antagonism, but also limited antinociception when given alone. Radioligand binding, receptor autoradiography, and electrophysiology experiments showed that CC12's antagonist properties are not explained by activity at 25 sites relevant to analgesia, including known receptors for cannabinoids, opioids or histamine. The use of CC12 as an improgan antagonist will facilitate the characterization of improgan analgesia. Furthermore, because CC12 was also found presently to inhibit opioid and cannabinoid antinociception, it is suggested that this drug modifies a biochemical mechanism shared by several classes of analgesics. Elucidation of this mechanism will enhance understanding of the biochemistry of pain relief.


Assuntos
Cimetidina/análogos & derivados , Antagonistas dos Receptores H2 da Histamina/metabolismo , Imidazóis/farmacologia , Receptores Histamínicos H2/efeitos dos fármacos , Sulfetos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Autorradiografia , Benzoxazinas/farmacologia , Sítios de Ligação/efeitos dos fármacos , Cimetidina/antagonistas & inibidores , Cimetidina/metabolismo , Cimetidina/farmacologia , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Histamina/farmacologia , Imidazóis/síntese química , Indicadores e Reagentes , Injeções Intraventriculares , Ligantes , Masculino , Membranas/efeitos dos fármacos , Membranas/metabolismo , Camundongos , Morfolinas/farmacologia , Naloxona/farmacologia , Naftalenos/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sulfetos/síntese química
10.
Brain Res ; 1152: 42-8, 2007 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-17433267

RESUMO

Improgan, a congener of the H(2) antagonist cimetidine, produces non-opioid antinociception which is blocked by the CB(1) antagonist rimonabant, implying a cannabinoid mechanism of action. Since cannabinoids produce hypothermia as well as antinociception in rodents, the present study investigated the pharmacological activity of improgan on core body temperature and nociceptive (tail flick) responses. Improgan (60, 100 and 140 microg, intraventricular [ivt]) elicited significant decreases in core temperature 3-30 min following injection with a maximal hypothermic effect of -1.3 degrees C. Pretreatment with rimonabant (50 microg, ivt) produced a statistically significant but incomplete (29-42%) antagonism of improgan hypothermia. In control experiments, the CB(1) agonist CP-55,940 (37.9 microg, ivt) induced significant decreases in core temperature (-1.8 degrees C) 3-30 min following injection. However, unlike the case with improgan, pretreatment with rimonabant completely blocked CP-55,940 hypothermia. Furthermore, CP-55,940 and improgan elicited maximal antinociception over the same time course and dose ranges, and both effects were attenuated by rimonabant. These results show that, like cannabinoid agonists in the rat, improgan produces antinociception and hypothermia which is blocked by a CB(1) antagonist. Unlike cannabinoid agonists, however, improgan does not produce locomotor inhibition at antinociceptive doses. Additional experiments were performed to determine the effect of CC12, a recently discovered improgan antagonist which lacks affinity at CB(1) receptors. Pretreatment with CC12 (183 microg, ivt) produced complete inhibition of both the antinociception and the hypothermia produced by improgan, suggesting the possible role of an unknown improgan receptor in both of these effects.


Assuntos
Analgésicos/farmacologia , Temperatura Corporal/efeitos dos fármacos , Cimetidina/análogos & derivados , Limiar da Dor/efeitos dos fármacos , Receptor CB1 de Canabinoide/fisiologia , Animais , Cimetidina/antagonistas & inibidores , Cimetidina/farmacologia , Imidazóis/farmacologia , Masculino , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto , Sulfetos/farmacologia
11.
J Pain ; 8(11): 850-60, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17644043

RESUMO

UNLABELLED: Improgan is a congener of the H(2) antagonist cimetidine, which produces potent antinociception. Because a) the mechanism of action of improgan remains unknown and b) this drug may indirectly activate cannabinoid CB(1) receptors, the effects of the CB(1) antagonist/inverse agonist rimonabant (SR141716A) and 3 congeners with varying CB(1) potencies were studied on improgan antinociception after intracerebroventricular (icv) dosing in rats. Consistent with blockade of brain CB(1) receptors, rimonabant (K(d) = 0.23 nM), and O-1691 (K(d) = 0.22 nM) inhibited improgan antinociception by 48% and 70% after icv doses of 43 nmol and 25 nmol, respectively. However, 2 other derivatives with much lower CB(1) affinity (O-1876, K(d) = 139 nM and O-848, K(d) = 352 nM) unexpectedly blocked improgan antinociception by 65% and 50% after icv doses of 300 nmol and 30 nmol, respectively. These derivatives have 600-fold to 1500-fold lower CB(1) potencies than that of rimonabant, yet they retained improgan antagonist activity in vivo. In vitro dose-response curves with (35)S-GTPgammaS on CB(1) receptor-containing membranes confirmed the approximate relative potency of the derivatives at the CB(1) receptor. Although antagonism of improgan antinociception by rimonabant has previously implicated a mechanistic role for the CB(1) receptor, current findings with rimonabant congeners suggest that receptors other than, or in addition to CB(1) may participate in the pain-relieving mechanisms activated by this drug. The use of congeners such as O-848, which lack relevant CB(1)-blocking properties, will help to identify these cannabinoid-like, non-CB(1) mechanisms. PERSPECTIVE: This article describes new pharmacological characteristics of improgan, a pain-relieving drug that acts by an unknown mechanism. Improgan may use a marijuana-like (cannabinoid) pain-relieving mechanism, but it is shown presently that the principal cannabinoid receptor in the brain (CB(1)) is not solely responsible for improgan analgesia.


Assuntos
Analgésicos/administração & dosagem , Cimetidina/análogos & derivados , Limiar da Dor/efeitos dos fármacos , Dor/tratamento farmacológico , Receptor CB1 de Canabinoide/fisiologia , Análise de Variância , Animais , Cimetidina/administração & dosagem , Cimetidina/química , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Injeções Intraventriculares/métodos , Masculino , Medição da Dor/métodos , Piperidinas/administração & dosagem , Piperidinas/química , Pirazóis/administração & dosagem , Pirazóis/química , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto , Fatores de Tempo
12.
Pharmacol Biochem Behav ; 88(1): 122-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17719621

RESUMO

Pharmacological activation of histamine H3 receptors is known to reduce the release of inflammatory peptides, thereby reducing pain and inflammation, but the site(s) and mechanism(s) of these effects are currently unknown. The present study addressed these questions by examining the effects of the H3 agonist immepip and the H3 antagonist thioperamide on nociceptive behaviors and swelling produced during the rat formalin test. Systemic administration of immepip (5 and 30 mg/kg, s.c.) significantly attenuated formalin-induced flinching but not licking responses during both phases. This attenuation was reversed by either systemic (15 mg/kg, i.p.) or intrathecal (20 or 50 microg) administration of thioperamide. Furthermore, immepip (30 mg/kg, s.c.) significantly inhibited formalin-induced swelling, an action which was completely reversed by systemic (15 mg/kg, i.p.), but not intrathecal (50 microg) thioperamide. Also consistent with this pattern, intrathecal immepip (50 microg) reduced flinching responses, but had no effect on formalin-induced paw swelling. The present findings suggest that activation of H3 receptors located on peripheral and spinal terminals of deep dermal fibers attenuates formalin-induced swelling and flinching, respectively. Pharmacological stimulation of H3 receptors could be an important therapeutic approach for many disorders related to deep dermal or inflammatory pain.


Assuntos
Formaldeído , Agonistas dos Receptores Histamínicos/farmacologia , Antagonistas dos Receptores Histamínicos/farmacologia , Imidazóis/farmacologia , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Medição da Dor/efeitos dos fármacos , Sistema Nervoso Periférico/metabolismo , Piperidinas/farmacologia , Receptores Histamínicos H3/efeitos dos fármacos , Medula Espinal/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Edema/induzido quimicamente , Edema/prevenção & controle , Injeções Espinhais , Masculino , Terminações Nervosas/efeitos dos fármacos , Fibras Nervosas/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Sistema Nervoso Periférico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Pele/inervação , Medula Espinal/efeitos dos fármacos
13.
Neuropharmacology ; 51(3): 447-56, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16806300

RESUMO

Improgan is a chemical congener of the H2 antagonist cimetidine which shows the profile of a highly effective analgesic when administered directly into the CNS. Although the improgan receptor is unknown, improgan activates analgesic pathways which are independent of opioids, but may utilize cannabinoid mechanisms. To discover selective, potent, improgan-like drugs, seven compounds chemically related to improgan were synthesized and tested for antinociceptive activity in rats after intracerebroventricular (icv) administration. Among a series of improgan congeners in which the alkyl chain length of improgan ((-CH2)3-) was varied, five compounds showed full agonist antinociceptive activity with potencies greater than that of improgan. VUF5420 (containing (-CH2)4-, EC50 = 86.1 nmol) produced maximal antinociceptive activity after doses which showed no motor impairment or other obvious toxicity, and was 2.3-fold more potent than improgan (EC50 = 199.5 nmol). As found previously with improgan, VUF5420-induced antinociception was unaffected by administration of the opioid antagonist naltrexone, but was inhibited by the CB1 antagonist SR141716A, suggesting a non-opioid, cannabinoid-related analgesic action. However, VUF5420 showed very low affinity (Kd approximately 10 microM) on CB1-receptor activation of 35S-GTPgammaS binding, indicating that this drug does not directly interact with the CB1 receptor in vivo. The present results show that VUF5420 is a high potency, improgan-like, non-opioid analgesic which may indirectly activate cannabinoid pain-relieving mechanisms.


Assuntos
Analgésicos não Narcóticos/química , Analgésicos não Narcóticos/farmacologia , Cimetidina/análogos & derivados , Medição da Dor/efeitos dos fármacos , Analgésicos/síntese química , Analgésicos/química , Analgésicos/farmacologia , Analgésicos não Narcóticos/síntese química , Animais , Comportamento Animal/efeitos dos fármacos , Antagonistas de Receptores de Canabinoides , Linhagem Celular , Cimetidina/síntese química , Cimetidina/química , Cimetidina/farmacologia , Relação Dose-Resposta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Humanos , Injeções Intraventriculares/métodos , Masculino , Limiar da Dor/efeitos dos fármacos , Piperidinas/farmacologia , Ligação Proteica/efeitos dos fármacos , Pirazóis/farmacologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Receptores de Canabinoides/efeitos dos fármacos , Receptores de Canabinoides/fisiologia , Rimonabanto , Isótopos de Enxofre/farmacocinética
14.
Eur J Pharmacol ; 549(1-3): 79-83, 2006 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16989809

RESUMO

Improgan is a non-opioid analgesic which does not act at known histamine or cannabinoid receptors. Because improgan antinociception is blocked by low doses of a cannabinoid CB1 antagonist, the present experiments determined if development of cannabinoid tolerance in mice would alter improgan antinociception. Twice-daily injections of Delta9-tetrahydrocannabinol (THC, 10 mg/kg, s.c.) for 3.5 days induced 47-54% and 42-56% reductions in cannabinoid (WIN 55,212-2, 20 microg, i.c.v.) and improgan (30 microg, i.c.v.) antinociception, respectively, as compared with responses from vehicle-treated groups. Because improgan lacks cannabinoid-like side effects in rats, and does not act directly on cannabinoid CB1 receptors, the finding that development of cannabinoid tolerance reduces improgan antinociception suggests that this drug may release endocannabinoids, or activate novel cannabinoid sites. Either possibility offers the potential for developing new types of analgesics.


Assuntos
Analgésicos/farmacologia , Canabinoides/farmacologia , Cimetidina/análogos & derivados , Tolerância a Medicamentos , Analgésicos/administração & dosagem , Análise de Variância , Animais , Benzoxazinas , Ligação Competitiva , Canabinoides/administração & dosagem , Cimetidina/administração & dosagem , Cimetidina/farmacologia , Dronabinol/administração & dosagem , Dronabinol/farmacologia , Injeções Intraventriculares , Injeções Subcutâneas , Masculino , Camundongos , Morfolinas/administração & dosagem , Morfolinas/farmacologia , Naftalenos/administração & dosagem , Naftalenos/farmacologia , Dor/fisiopatologia , Dor/prevenção & controle , Medição da Dor/métodos , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/metabolismo , Fatores de Tempo
15.
Neuropharmacology ; 108: 252-63, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27140694

RESUMO

Histamine is an important neurotransmitter that exerts its physiological actions through H1-4 metabotropic receptors in mammals. It also directly activates ionotropic GABAA receptor (GABAAR) ß3 homooligomers and potentiates GABA responses in αß heterooligomers in vitro, but the respective histamine binding sites in GABAARs are unknown. We hypothesized that histamine binds at the extracellular ß+ß- interface at a position homologous to the GABA binding site of heterooligomeric GABAARs. To test this, we individually mutated several residues at the putative ligand binding minus side of a rat GABAAR ß3 wild type subunit and of a ß3 subunit that was made insensitive to trace Zn(2+) inhibition [ß3(H267A); called (Z)ß3]. (Z)ß3, (Z)ß3(Y62L), (Z)ß3(Q64A), (Z)ß3(Q64E), α1(Z)ß3, or α1(Z)ß3(Y62L) receptors were studied in HEK293T cells using whole cell voltage clamp recording. ß3, ß3(Y62C), ß3(Q64C), ß3(N41C), ß3(D43C), ß3(A45C) or ß3(M115C) receptors were examined in Xenopus oocytes using two-electrode voltage clamp. Histamine directly activated (Z)ß3 and ß3 homooligomers and potentiated GABA actions in α1(Z)ß3 heterooligomers. Receptors containing (Z)ß3(Y62L), ß3(Y62C) and ß3(D43C) showed markedly reduced histamine potency, but homo- and heterooligomers with (Z)ß3(Q64E) exhibited increased potency. The GABAAR αß(γ) competitive antagonist bicuculline elicited sub-maximal agonist currents through (Z)ß3 homooligomers, the potency of which was strongly decreased by (Z)ß3(Y62L). Mutations ß3(N41C), ß3(A45C) and ß3(M115C) disturbed receptor expression or assembly. Computational docking into the crystal structure of homooligomeric ß3 receptors resulted in a histamine pose highly consistent with the experimental findings, suggesting that histamine activates ß3 receptors via a site homologous to the GABA site in αßγ receptors.


Assuntos
Histamina/metabolismo , Simulação de Acoplamento Molecular/métodos , Mutagênese/fisiologia , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Animais , Sítios de Ligação/fisiologia , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Histamina/química , Histamina/farmacologia , Humanos , Estrutura Secundária de Proteína , Ratos , Receptores de GABA-A/química , Xenopus laevis
16.
Eur J Pharmacol ; 522(1-3): 38-46, 2005 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-16216240

RESUMO

The antinociceptive profile of selected histamine H(2) and histamine H(3) receptor antagonists led to the discovery of improgan, a non-brain-penetrating analgesic agent which does not act on known histamine receptors. Because no chemical congener of improgan has yet been discovered which has both antinociceptive and brain-penetrating properties, the present study investigated the antinociceptive effects of a series of chemical compounds related to zolantidine, a brain-penetrating histamine H(2) receptor antagonist. The drugs studied presently contain the piperidinomethylphenoxy (PMPO) moiety, hypothesized to introduce brain-penetrating characteristics. Following intracerebroventricular (i.c.v.) dosing in rats, six of eight drugs produced dose- and time-related antinociception on both the tail flick and hot plate tests over a nearly eight-fold range of potencies. Ataxia and other motor side effects were observed after high doses of these drugs, but two of the compounds (SKF94674 and loxtidine) produced maximal antinociception at doses which were completely devoid of these motor effects. Consistent with the hypothesis that PMPO-containing drugs are brain-penetrating analgesics, SKF94674 and another derivative (JB-9322) showed dose-dependent antinociceptive activity 15 to 30 min after systemic dosing in mice, but these effects were accompanied by seizures and death beginning 45 min after dosing. Other drugs showed a similar pattern of antinociceptive and toxic effects. In addition, loxtidine produced seizures without antinociception, whereas zolantidine produced neither effect after systemic dosing in mice. Although several of the drugs tested have histamine H(2) receptor antagonist activity, neither the antinociception nor the toxicity was correlated with histamine H(2) receptor activity. The present results are the first to demonstrate the existence of brain-penetrating antinociceptive agents chemically related to zolantidine and improgan, but further studies are needed to understand the mechanisms of both the pain relief and toxicity produced by these agents.


Assuntos
Analgésicos/farmacologia , Encéfalo/metabolismo , Cimetidina/análogos & derivados , Algoritmos , Analgésicos/química , Analgésicos/farmacocinética , Animais , Comportamento Animal/efeitos dos fármacos , Benzotiazóis , Cimetidina/química , Cimetidina/farmacologia , Relação Dose-Resposta a Droga , Antagonistas dos Receptores H2 da Histamina/química , Antagonistas dos Receptores H2 da Histamina/farmacologia , Injeções Intraventriculares , Masculino , Camundongos , Estrutura Molecular , Dor/prevenção & controle , Medição da Dor/métodos , Fenoxipropanolaminas/química , Fenoxipropanolaminas/farmacologia , Piperidinas/química , Piperidinas/farmacologia , Quinazolinas/química , Quinazolinas/farmacologia , Quinazolinonas , Ratos , Ratos Sprague-Dawley , Tiazóis/química , Tiazóis/farmacologia , Fatores de Tempo , Triazóis/química , Triazóis/farmacologia
17.
J Pain ; 6(3): 193-200, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15772913

RESUMO

Histamine H 3 receptors have been suggested to inhibit the activity of a variety of central and peripheral neurons. Recent studies revealed that activation of spinal histamine H 3 receptors attenuates tail pinch, but not tail flick, nociception. To determine whether H 3 receptor-mediated antinociception is truly modality-specific, the effects of the selective H 3 agonist immepip were evaluated on nociceptive responses in rats induced by a range of thermal and mechanical intensities applied to the hind paw and the tail. In addition, the modulation of chemical nociceptive (ie, formalin) responses by immepip was evaluated. Immepip (5 to 30 mg/kg, subcutaneous) attenuated responses to low-intensity mechanical pinch, but not to high-intensity mechanical pressure applied to either the hind paw or the tail. The same doses of immepip had no effect on thermal nociceptive responses, regardless of the stimulus intensity. These results suggest that immepip-induced antinociception is modality- and intensity-specific. It is likely that immepip inhibits low-intensity mechanical nociception by activation of H 3 receptors located on the spinal terminals of Adelta and possibly C high-threshold mechanoreceptors. In addition, immepip (5 mg/kg, subcutaneous) significantly attenuated formalin-induced flinching, but not formalin-induced licking, during both phase 1 and phase 2, suggesting that H 3 agonists might be effective in treating some forms of clinically relevant pain. Certain classes of pain-transmitting fibers possess histamine H 3 receptors, but the localization and functional significance of these inhibitory receptors was not known. The present study shows that drugs that stimulate H 3 receptors can reduce behavioral responses produced by some, but not all, painful stimuli. Thus, H 3 agonists could be a new type of therapy for certain kinds of pain disorders.


Assuntos
Agonistas dos Receptores Histamínicos/farmacologia , Imidazóis/farmacologia , Nociceptores/efeitos dos fármacos , Dor/tratamento farmacológico , Piperidinas/farmacologia , Receptores Histamínicos H3/metabolismo , Animais , Temperatura Alta , Masculino , Nociceptores/fisiologia , Dor/fisiopatologia , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Estimulação Física , Ratos , Ratos Sprague-Dawley , Estimulação Química
18.
Brain Res ; 1616: 10-8, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25935691

RESUMO

Recent studies suggest a functional role for neuronal cytochrome P450 monooxygenase (P450) activity in opioid analgesia. To characterize the relevant receptors, brain areas, and circuits, detailed in vitro and in vivo studies were performed with the highly selective µ opioid receptor agonist DAMGO in neuronal P450-deficient mutant (Null) and control mice. Homogenates of brain regions and spinal cord showed no differences in DAMGO-induced activation of [(35)S]- GTPγS binding between Null and control mice, indicating no genotype differences in µ opioid receptor signaling, receptor affinities or receptor densities. Intracerebroventricular (icv) DAMGO produced robust, near-maximal, analgesic responses in control mice which were attenuated by 50% in Null mice, confirming a role for µ opioid receptors in activating P450-associated responses. Intra-periaqueductal gray (PAG) and intra-rostral ventromedial medulla (RVM) injections of DAMGO revealed deficits in Null (vs. control) analgesic responses, yet no such genotype differences were observed after intrathecal DAMGO administration. Taken with earlier published findings, the present results suggest that activation of µ opioid receptors in both the PAG and in the RVM relieves pain by mechanisms which include nerve-terminal P450 enzymes within inhibitory PAG-RVM projections. Spinal opioid analgesia, however, does not seem to require such P450 enzyme activity.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Neurônios/metabolismo , Nociceptividade/efeitos dos fármacos , Análise de Variância , Animais , Sistema Enzimático do Citocromo P-450/genética , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Masculino , Camundongos , Camundongos Transgênicos , Microinjeções , Neurônios/efeitos dos fármacos , Substância Cinzenta Periaquedutal/citologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Fatores de Tempo
19.
Pain ; 58(1): 29-37, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7970837

RESUMO

Because previous studies suggest an antinociceptive role for the neuromodulator histamine (HA) in the periaqueductal grey or the nearby dorsal raphe (PAG/DR), a detailed pharmacological investigation of the effects of intracerebral HA on the hot-plate nociceptive test was performed in rats. Intracerebral microinjections of HA (1 microgram) into the PAG/DR or into the median raphe evoked a mild, reversible antinociceptive response; injections into lateral or dorsal midbrain evoked either a delayed response or no response, respectively. In the PAG/DR, the HA dose-response curve had an inverted U-shape, showing that HA can induce both antinociceptive (0.3-3 micrograms) and pro-nociceptive (10-30 micrograms) responses. Larger doses of HA (e.g., 100 micrograms) produced irreversible and highly variable antinociceptive responses that were accompanied by behavioral and histopathological changes; such effects, indicative of toxicity, were not observed after 0.3 microgram of HA, the peak antinociceptive dose. HA (0.3 microgram) antinociception was completely inhibited by intracerebral co-administration of the opiate antagonist naloxone (1 ng), the H1-receptor antagonist temelastine (20 pg), and the H2-receptor antagonist tiotidine (1 ng); none of these drugs altered nociceptive scores in the absence of HA. These results show that: (1) HA, a neurotransmitter in the PAG, can evoke antinociception in the absence of other behavioral or toxic effects; and (2) HA antinociception depends on the activation of both opiate and HA receptors in the PAG/DR.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Histamina/farmacologia , Nociceptores/efeitos dos fármacos , Animais , Cimetidina/análogos & derivados , Cimetidina/farmacologia , Relação Dose-Resposta a Droga , Histamina/administração & dosagem , Antagonistas dos Receptores Histamínicos/farmacologia , Antagonistas dos Receptores Histamínicos H1/farmacologia , Antagonistas dos Receptores H2 da Histamina/farmacologia , Masculino , Mesencéfalo , Microinjeções , Naloxona/farmacologia , Medição da Dor/efeitos dos fármacos , Substância Cinzenta Periaquedutal/anatomia & histologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Substância Cinzenta Periaquedutal/fisiologia , Pirimidinonas/farmacologia , Núcleos da Rafe/anatomia & histologia , Núcleos da Rafe/efeitos dos fármacos , Núcleos da Rafe/fisiologia , Ratos , Ratos Sprague-Dawley
20.
Brain Res ; 974(1-2): 146-52, 2003 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-12742632

RESUMO

Improgan, a chemical congener of the H(2) antagonist cimetidine, induces antinociception following intracerebroventricular (i.c.v.) administration in rodents, but the mechanism of action of this compound remains unknown. Because the chemical structure of improgan closely resembles those of histamine and certain histamine blockers, and because neuronal histamine is known to participate in pain-relieving responses, the antinociceptive actions of improgan were evaluated in mice containing null mutations in the genes for three histamine receptors (H(1), H(2), and H(3)) and also in the gene for histidine decarboxylase (the histamine biosynthetic enzyme). Similar to earlier findings in Swiss-Webster mice, improgan induced maximal, reversible, dose-related reductions in thermal nociceptive responses in ICR mice, but neither pre-improgan (baseline) nor post-improgan nociceptive latencies were changed in any of the mutant mice as compared with wild-type controls. Improgan also had weak inhibitory activity in vitro (pK(i)=4.7-4.9) on specific binding to three recently-discovered, recombinant isoforms of the rat H(3) receptor (H(3A), H(3B), and H(3C)). The present findings strongly support the hypothesis that neuronal histamine and its receptors fail to play a role in improgan-induced antinociception.


Assuntos
Analgésicos/farmacologia , Cimetidina/análogos & derivados , Cimetidina/farmacologia , Histamina/fisiologia , Neurônios/fisiologia , Receptores Histamínicos/fisiologia , Animais , Relação Dose-Resposta a Droga , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Microinjeções , Medição da Dor , Receptores Histamínicos/genética , Receptores Histamínicos H2/genética , Receptores Histamínicos H2/fisiologia , Receptores Histamínicos H3/genética , Receptores Histamínicos H3/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa