Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Osteoarthritis Cartilage ; 26(2): 175-183, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29180098

RESUMO

OBJECTIVE: To outline the role that spontaneous osteoarthritis (OA) in companion animals can play in translational research and therapeutic pharmacological development. OUTLINE: Narrative review summarizing the opportunities and limitations of naturally occurring, spontaneous OA as models of human OA pain, with a focus on companion animal pets. The background leading to considering inserting spontaneous disease models in the translational paradigm is provided. The utility of this model is discussed in terms of outcome measures that have been validated as being related to pain, and in terms of the potential for target discovery is outlined. The limitations to using companion animal pets as models of human disease are discussed. CONCLUSIONS: Although many steps along the translational drug development pathway have been identified as needing improvement, spontaneous painful OA in companion animals offers translational potential. Such 'models' may better reflect the complex genetic, environmental, temporal and physiological influences present in humans and current data suggests the predictive validity of the models are good. The opportunity for target discovery exists but is, as yet, unproven.


Assuntos
Dor Crônica/veterinária , Osteoartrite/veterinária , Animais , Artrite Experimental/tratamento farmacológico , Dor Crônica/tratamento farmacológico , Desenvolvimento de Medicamentos/métodos , Humanos , Osteoartrite/tratamento farmacológico , Manejo da Dor/métodos , Animais de Estimação , Pesquisa Translacional Biomédica/métodos
2.
Osteoarthritis Cartilage ; 18(4): 572-80, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20036347

RESUMO

OBJECTIVE: To characterize pain-related behavior during the course of knee osteoarthritis (OA) induced by destabilization of the medial meniscus (DMM) in wild type (WT) and in ADAMTS-5 null mice. METHODS: DMM surgery was performed in the right knee of CD-1 mice. At regular intervals up to 8 weeks after surgery, mice were assessed for the following parameters: mechanical allodynia (via withdrawal thresholds to von Frey filaments applied to the plantar surface of both hind paws or to the tail), thermal hyperalgesia, locomotor activity and gait analysis. In addition, mechanical allodynia was tested in C57BL/6 WT or ADAMTS-5 null mice following DMM surgery. RESULTS: In CD-1 mice, a robust and progressive decrease in withdrawal threshold was observed in both hind paws after DMM but not sham surgery. Allodynia was apparent as early as 14 days postoperatively. Both sexes developed OA changes after surgery with concurrent mechanical allodynia. No other pain-related behavioral changes were detected up to 8 weeks post-surgery. In C57BL/6 mice, a genetic background in which only males develop OA changes after DMM, males but not females developed allodynia in the ipsilateral hind paw. In contrast, C57BL/6 ADAMTS-5 null mice did not develop OA changes or mechanical allodynia up to 8 weeks post-surgery. CONCLUSION: Joint pathology following DMM surgery in mice is associated with progressive mechanical allodynia. ADAMTS-5 null mice are resistant to DMM-induced OA-like lesions and to the associated mechanical allodynia.


Assuntos
Meniscos Tibiais/cirurgia , Osteoartrite do Joelho/fisiopatologia , Dor/fisiopatologia , Acetaminofen/uso terapêutico , Analgésicos não Narcóticos/uso terapêutico , Analgésicos Opioides/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Feminino , Fêmur/patologia , Marcha/efeitos dos fármacos , Marcha/fisiologia , Masculino , Camundongos , Camundongos Knockout , Morfina/uso terapêutico , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Osteoartrite do Joelho/tratamento farmacológico , Dor/tratamento farmacológico , Tíbia/patologia , Suporte de Carga/fisiologia
3.
J Med Genet ; 42(7): 583-7, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15994880

RESUMO

BACKGROUND: A recent genetic study in mice and humans revealed the modulatory effect of MC1R (melanocortin-1 receptor) gene variants on kappa-opioid receptor mediated analgesia. It is unclear whether this gene affects basal pain sensitivity or the efficacy of analgesics acting at the more clinically relevant mu-opioid receptor. OBJECTIVE: To characterise sensitivity to pain and mu-opioid analgesia in mice and humans with non-functional melanocortin-1 receptors. METHODS: Comparisons of spontaneous mutant C57BL/6-Mc1r(e/e) mice to C57BL/6 wildtype mice, followed by a gene dosage study of pain and morphine-6-glucuronide (M6G) analgesia in humans with MC1R variants. RESULTS: C57BL/6-Mc1r(e/e) mutant mice and human redheads--both with non-functional MC1Rs--display reduced sensitivity to noxious stimuli and increased analgesic responsiveness to the mu-opioid selective morphine metabolite, M6G. In both species the differential analgesia is likely due to pharmacodynamic factors, as plasma levels of M6G are similar across genotype. CONCLUSIONS: Genotype at MC1R similarly affects pain sensitivity and M6G analgesia in mice and humans. These findings confirm the utility of cross species translational strategies in pharmacogenetics.


Assuntos
Analgesia , Variação Genética , Dor/genética , Receptor Tipo 1 de Melanocortina/genética , Receptores Opioides mu/efeitos dos fármacos , Adolescente , Adulto , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Feminino , Genótipo , Cor de Cabelo/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Morfina/farmacocinética , Morfina/uso terapêutico , Derivados da Morfina/farmacocinética , Derivados da Morfina/uso terapêutico , Dor/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Valores de Referência
4.
Genes Brain Behav ; 15(6): 604-15, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27231153

RESUMO

Mechanical sensitivity is commonly affected in chronic pain and other neurological disorders. To discover mechanisms of individual differences in punctate mechanosensation, we performed quantitative trait locus (QTL) mapping of the response to von Frey monofilament stimulation in BXD recombinant inbred (BXD) mice. Significant loci were detected on mouse chromosome (Chr) 5 and 15, indicating the location of underlying polymorphisms that cause heritable variation in von Frey response. Convergent evidence from public gene expression data implicates candidate genes within the loci: von Frey thresholds were strongly correlated with baseline expression of Cacna2d1, Ift27 and Csnk1e in multiple brain regions of BXD strains. Systemic gabapentin and PF-670462, which target the protein products of Cacna2d1 and Csnk1e, respectively, significantly increased von Frey thresholds in a genotype-dependent manner in progenitors and BXD strains. Real-time polymerase chain reaction confirmed differential expression of Cacna2d1 and Csnk1e in multiple brain regions in progenitors and showed differential expression of Cacna2d1 and Csnk1e in the dorsal root ganglia of the progenitors and BXD strains grouped by QTL genotype. Thus, linkage mapping, transcript covariance and pharmacological testing suggest that genetic variation affecting Cacna2d1 and Csnk1e may contribute to individual differences in von Frey filament response. This study implicates Cacna2d1 and Ift27 in basal mechanosensation in line with their previously suspected role in mechanical hypersensitivity. Csnk1e is implicated for von Frey response for the first time. Further investigation is warranted to identify the specific polymorphisms involved and assess the relevance of these findings to clinical conditions of disturbed mechanosensation.


Assuntos
Canais de Cálcio/genética , Caseína Quinase I/genética , Mecanotransdução Celular/genética , Proteínas rab de Ligação ao GTP/genética , Aminas/farmacologia , Analgésicos/farmacologia , Animais , Canais de Cálcio/metabolismo , Caseína Quinase I/metabolismo , Ácidos Cicloexanocarboxílicos/farmacologia , Gabapentina , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiologia , Masculino , Camundongos , Pirimidinas/farmacologia , Locos de Características Quantitativas , Limiar Sensorial , Tato/efeitos dos fármacos , Tato/genética , Ácido gama-Aminobutírico/farmacologia , Proteínas rab de Ligação ao GTP/metabolismo
5.
J Neurosci ; 19(18): RC25, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10479718

RESUMO

Recent experiments in our laboratory have revealed a genetic correlation of the sensitivity of inbred mouse strains to different assays of nociception using noxious heat stimuli. An open question is whether the property of the noxious stimulus underlying the genetic correlation is heat specifically or simply a temperature (hot or cold) in the noxious range. The existing electrophysiological, psychophysical, neurochemical, and functional imaging literatures regarding the relationship of heat pain and cold pain are quite contentious, with a number of similarities and dissociations being documented. In the present study, we tested 12 inbred mouse strains (129/J, A/J, AKR/J, BALB/cJ, C3H/HeJ, C57BL/6J, C57BL/10J, C58/J, CBA/J, DBA/2J, RIIIS/J, and SM/J) for their reflexive withdrawal sensitivity to immersion of the tail in hot (47. 5 degrees C) and cold (-15 degrees C) water and compared the observed latencies with those obtained previously, using a 49 degrees C stimulus. All three traits displayed substantial heritabilities, ranging from 0.41 to 0.50. Strain means on these nociceptive traits displayed a high degree of cross-correlation (r = 0.49-0.77). Genetic correlation of hot and cold nociception implies that similar genes underlie interindividual variability in both traits in mice and further suggests that these nociceptive types share physiological mediation. This finding is discussed in relation to existing data in other mammals including humans.


Assuntos
Temperatura Baixa , Temperatura Alta , Dor/genética , Animais , Imersão , Masculino , Camundongos , Camundongos Endogâmicos , Tempo de Reação/genética , Especificidade da Espécie , Cauda
6.
Neurosci Biobehav Rev ; 24(3): 375-89, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10781697

RESUMO

It has been appreciated for some time that the sexes can differ in their sensitivity to pain and its inhibition. Both the human and rodent literatures remain quite contentious, with many investigators failing to observe sex differences that others document clearly. Recent data from our laboratory have pointed to an interaction between sex and genotype in rodents, such that sex differences are observed in some strains but not others. However, these studies employed inbred mouse strains and are thus not directly relevant to existing data. We presently examined whether the observation of statistically significant sex differences in nociception and morphine antinociception might depend on the particular outbred rodent population chosen for study. Rats of both sexes and three common outbred strains were obtained from three suppliers (Long Evans, Simonsen; Sprague Dawley, Harlan; Wistar Kyoto, Taconic) and tested for nociceptive sensitivity on the 49 degrees C tail-withdrawal assay, and antinociception following morphine (1-10mg/kg, i.p.). In further studies, three outbred populations of mice (CD-1, Harlan; Swiss Webster, Harlan; Swiss Webster, Simonsen) were bred in our vivarium for several generations and tested for tail-withdrawal sensitivity and morphine antinociception (1-20male, and no significant difference. In a separate study in which the estrous cycle was tracked in female mice, we found evidence for an interaction between genotype and estrous phase relevant to morphine antinociception. However, estrous cyclicity did not explain the observed sex differences. These data are discussed with respect to the existing sex difference and pain literature, and also as they pertain to future investigations of these phenomena.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Dor/genética , Dor/psicologia , Animais , Feminino , Genótipo , Temperatura Alta , Humanos , Masculino , Camundongos , Ratos , Caracteres Sexuais
7.
Pharmacogenetics ; 9(5): 619-25, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10591542

RESUMO

The ability to predict interindividual differences in drug efficacy or toxicity, based on genetic factors that influence drug disposition or drug action, is fast becoming a realistic goal. The purpose of the present study was to determine whether epibatidine, a prototypical nicotinic analgesic drug, exhibits pharmacogenetic variability in antinociceptive activity. Eight inbred mouse strains (A, AKR, BALB/c, C3H/He, C57BL/6, C57BL/10, DBA/2, and SM) were surveyed for their sensitivity to the antinociceptive effects of epibatidine. All strains exhibited statistically significant antinociception that peaked between 10 and 20 min following the systemic injection of 50 microg/kg epibatidine. However, there was fourfold variability in the magnitude of peak effect between strains, with DBA/2, BALB/c and A strains showing much greater sensitivity than all others. A return to baseline nociceptive threshold at 30 min post-injection was observed for all but the A strain. In contrast, these mice exhibited significant antinociception for at least 3 h following epibatidine administration. Thus, expressing the data as area under the time-latency curve to take into account both the magnitude and duration of effect, epibatidine displayed approximately 20-fold higher antinociceptive potency in the A strain compared with the C3H/He strain. The effects of epibatidine in both the A and C3H/He strains were dose-dependent and sensitive to antagonism by the selective neuronal nicotinic channel blocker mecamylamine. Taken together, these data demonstrate the existence of pharmacogenetic variability in neuronal nicotinic receptor-mediated antinociception between inbred stains of mice and presage the potential for similar variability in analgesic response to nicotinic-based analgesics among humans. Future studies will seek to identify the chromosomal loci underlying this variability.


Assuntos
Analgésicos/farmacologia , Variação Genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores Nicotínicos/efeitos dos fármacos , Receptores Nicotínicos/genética , Analgésicos/administração & dosagem , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Mecamilamina/farmacologia , Camundongos , Camundongos Endogâmicos , Agonistas Nicotínicos/administração & dosagem , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Farmacogenética , Piridinas/administração & dosagem , Piridinas/farmacologia , Receptores Nicotínicos/metabolismo , Especificidade da Espécie
8.
Pain ; 91(1-2): 5-13, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11240073

RESUMO

Individual differences in sensitivity to pain and analgesia are well appreciated, and increasing evidence has pointed towards a role of inherited genetic factors in explaining some proportion of such variability. It has long been known by practitioners of acupuncture, an ancient modality of analgesia, that some patients are 'responders' and others 'non-responders.' The present research was aimed at defining the inherited genetic influence on acupuncture analgesia in the mouse, using 10 common inbred strains. Two pairs of metallic needles were inserted into acupoints ST 36 and SP 6, fixed in situ and then connected to the output channel of an electric pulse generator. Electroacupuncture (EA) parameters were set as constant current output (intensity: 1.0-1.5-2.0 mA, 10 min each; frequency: 2 or 100 Hz) with alteration of a positive and negative square wave, 0.3 ms in pulse width. Tail-flick latencies evoked by radiant heat were measured before, during and after EA stimulation. Narrow-sense heritability estimates of 2 and 100 Hz EA were 0.37 and 0.16, respectively. We found that the C57BL/10 strain was the most sensitive, and the SM strain was the least sensitive to both 2 and 100 Hz EA. However, the relative sensitivities of other strains to these two EA frequencies suggested some genetic dissociation between them as well. These results demonstrate a role of inherited genetic factors in EA sensitivity in the mouse, although the low-to-moderate heritability estimates suggest that environmental factors may be of greater importance in predicting who will benefit from this analgesic modality.


Assuntos
Analgesia por Acupuntura , Eletroacupuntura , Camundongos/genética , Animais , Feminino , Genótipo , Masculino , Camundongos/fisiologia , Camundongos Endogâmicos/genética , Camundongos Endogâmicos/fisiologia , Nociceptores/fisiologia , Limiar da Dor/fisiologia , Caracteres Sexuais , Especificidade da Espécie
9.
Pain ; 76(1-2): 115-25, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9696464

RESUMO

The important role of genetic factors in the mediation of sensitivity to pain and pain inhibition is being increasingly appreciated. In an attempt to systematically study the genotypic influences on inflammatory nociception, we conducted a survey of the nociceptive responsivity of three common outbred mouse strains and 11 inbred mouse strains on the formalin test. The formalin test is known to display a biphasic temporal pattern of behavioral and electrophysiological activity, defined by an acute/early phase and a tonic/late phase. Nociceptive sensitivity (licking/biting of the affected area) to a subcutaneous injection of 5% formalin (25 microl volume) into the plantar surface of the right hindpaw displayed moderate heritability in both phases (0.38 and 0.46, respectively). One strain, A/J, was identified as extremely resistant to formalin nociception, displaying total licking in the acute and tonic phases that was 60% and 87% lower, respectively, than the grand mean of all strains. A subsequent series of experiments were performed to characterize the difference between A/J and C57BL/6J mice. The findings establish this inbred strain comparison as a useful genetic model of nociceptive sensitivity.


Assuntos
Inflamação/genética , Inflamação/psicologia , Dor/genética , Dor/psicologia , Animais , Formaldeído , Genótipo , Inflamação/complicações , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Dor/etiologia , Medição da Dor/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Especificidade da Espécie , Natação/psicologia
10.
Pain ; 81(1-2): 179-86, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10353506

RESUMO

Abdominal contractions are a viscerosomatic reflex response to noxious colorectal irritation in rats. In this study we characterize the modulating effect of chemical and mechanical colonic irritation on this reflex response to peritoneal irritation induced by diluted acetic acid (HAc) in conscious C57BL/6N mice. Pain responses were scored by counting the number of abdominal contractions during the 30-min period after intraperitoneal (i.p.) injection of either vehicle or HAc. Abdominal contractions were induced by 0.6% but not by 0.3% HAc. Chemical irritation of the colon by intraluminal 25% turpentine did not produce abdominal contractions by itself, but significantly increased the effect of both 0.3 and 0.6% i.p. HAc, administered 60 min after the luminal stimulus. Mechanical stimulation of the anorectum and colon by insertion of a balloon did not modify the effect of 0.6% HAc, while the insertion plus the inflation to 0.1 and 0.2 ml (30 s on/30 s off for 10 min) reduced the response to i.p. HAc by 35 and 88%, respectively. This inhibitory effect was reversed by naloxone (5 mg/kg, s.c.) pretreatment, while naloxone alone did not modify the effect of 0.6% HAc. These results demonstrate that chemical irritation of visceral afferents in the colonic mucosa and peritoneum of mice interact to enhance viscerosomatic pain responses, while the activation of colonic mechanoreceptors inhibits peritoneal irritation-induced pain responses and induces a freezing behavior by a naloxone-sensitive mechanism.


Assuntos
Comportamento Animal/fisiologia , Doenças do Colo/fisiopatologia , Dor/fisiopatologia , Dor/psicologia , Músculos Abdominais/efeitos dos fármacos , Músculos Abdominais/fisiopatologia , Ácido Acético/farmacologia , Animais , Cateterismo , Doenças do Colo/induzido quimicamente , Doenças do Colo/etiologia , Injeções Intraperitoneais , Irritantes/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/efeitos dos fármacos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Estimulação Física , Estimulação Química , Terebintina/farmacologia
11.
Pain ; 70(2-3): 267-77, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9150302

RESUMO

The inbred mouse strains, DBA/2J (D2) and C57BL/6J (B6), display differential sensitivity to acute, thermal nociception as measured on the hot-plate (HP) assay. In an ongoing quantitative trait locus (QTL) mapping study designed to reveal genomic loci showing genetic linkage to HP sensitivity, a putative QTL on chromosome 4 (50-80 cM from the centromere) has been identified that appears to account for variability in this trait in male, but not female mice. An obvious candidate gene located in this same chromosomal region is Oprd1, which encodes the murine delta-opioid receptor. In an attempt to evaluate whether Oprd1 represents this sex-specific QTL for HP sensitivity, we tested D2 and B6 mice of both sexes for HP latencies (hindpaw-lift, -lick or -flutter) following systemic injections of saline, or the opioid receptor antagonists naloxone (NAL; 0.1 and 10 mg/kg), nor-binaltorphimine (nor-BNI; 5 mg/kg), naltrindole (NTI; 5 mg/kg), 7-benzylidenenaltrexone (BNTX; 0.7 mg/kg), or naltriben (NTB; 1 mg/kg). High-dose (10 mg/kg) NAL lowered HP latencies in D2, but not B6 mice, suggesting that the higher HP latencies exhibited by D2 mice reflect opioid mechanisms. HP latencies in both strains and both sexes were unaffected by pretreatment with low-dose (0.1 mg/kg) NAL or nor-BNI, suggesting that neither mu nor kappa receptors affect basal nociceptive sensitivity. The delta-receptor antagonist, NTI, and the delta2-specific antagonist, NTB, (but not the delta1-specific antagonist, BNTX) effectively lowered HP latencies in a strain- and sex-dependent manner: D2 male > B6 male > D2 female > B6 female. These data support the possibility that Oprd1 is a QTL mediating HP sensitivity in mice, and more generally illustrate the important roles of genetic background and gender in the perception of pain.


Assuntos
Camundongos Endogâmicos C57BL/genética , Camundongos Endogâmicos C57BL/fisiologia , Camundongos Endogâmicos DBA/genética , Camundongos Endogâmicos DBA/fisiologia , Nociceptores/fisiologia , Receptores Opioides delta/fisiologia , Caracteres Sexuais , Animais , Mapeamento Cromossômico , Feminino , Temperatura Alta , Masculino , Camundongos , Camundongos Endogâmicos/genética , Camundongos Endogâmicos/fisiologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/métodos , Tempo de Reação/efeitos dos fármacos
12.
Pain ; 80(1-2): 67-82, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10204719

RESUMO

It is generally acknowledged that humans display highly variable sensitivity to pain, including variable responses to identical injuries or pathologies. The possible contribution of genetic factors has, however, been largely overlooked. An emerging rodent literature documents the importance of genotype in mediating basal nociceptive sensitivity, in establishing a predisposition to neuropathic pain following neural injury, and in determining sensitivity to pharmacological agents and endogenous antinociception. One clear finding from these studies is that the effect of genotype is at least partially specific to the nociceptive assay being considered. In this report we begin to systematically describe and characterize genetic variability of nociception in a mammalian species, Mus musculus. We tested 11 readily-available inbred mouse strains (129/J, A/J, AKR/J, BALB/cJ, C3H/HeJ, C57BL/6J, C58/J, CBA/J, DBA/2J, RIIIS/J and SM/J) using 12 common measures of nociception. These included assays for thermal nociception (hot plate, Hargreaves' test, tail withdrawal), mechanical nociception (von Frey filaments), chemical nociception (abdominal constriction, carrageenan, formalin), and neuropathic pain (autotomy, Chung model peripheral nerve injury). We demonstrate the existence of clear strain differences in each assay, with 1.2 to 54-fold ranges of sensitivity. All nociceptive assays display moderate-to-high heritability (h2 = 0.30-0.76) and mediation by a limited number of apparent genetic loci. Data comparing inbred strains have considerable utility as a tool for understanding the genetics of nociception, and a particular relevance to transgenic studies.


Assuntos
Nociceptores/fisiologia , Medição da Dor , Dor/genética , Animais , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos , Dor/fisiopatologia , Tempo de Reação , Especificidade da Espécie
13.
Pain ; 80(1-2): 83-93, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10204720

RESUMO

Clinical pain syndromes, and experimental assays of nociception, are differentially affected by manipulations such as drug administration and exposure to environmental stress. This suggests that there are different 'types' of pain. We exploited genetic differences among inbred strains of mice in an attempt to define these primary 'types'; that is, to identify the fundamental parameters of pain processing. Eleven randomly-chosen inbred mouse strains were tested for their basal sensitivity on 12 common measures of nociception. These measures provided for a range of different nociceptive dimensions including noxious stimulus modality, location, duration and etiology, among others. Since individual members of inbred strains are identical at all genetic loci, the observation of correlated strain means in any given pair of nociceptive assays is an index of genetic correlation between these assays, and hence an indication of common physiological mediation. Obtained correlation matrices were subjected to multivariate analyses to identify constellations of nociceptive assays with common genetic mediation. This analysis revealed three major clusters of nociception: (1) baseline thermal nociception, (2) spontaneously-emitted responses to chemical stimuli, and (3) baseline mechanical sensitivity and cutaneous hypersensitivity. Many other nociceptive parameters that might a priori have been considered closely related proved to be genetically divergent.


Assuntos
Medição da Dor , Dor/genética , Animais , Análise por Conglomerados , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos , Dor/fisiopatologia , Tempo de Reação , Especificidade da Espécie
14.
Neuropharmacology ; 44(3): 304-10, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12604090

RESUMO

In the present study, we studied the interaction between serotonergic (5-HTergic) and gamma-aminobutyric acid (GABA)-ergic systems in the modulation of analgesia from morphine, a mu-opioid agonist, and U50,488, a kappa-opioid agonist. All experiments were performed in mice using the 49 degrees C tail-withdrawal assay. The benzodiazepine receptor agonist, diazepam, the serotonin synthesis inhibitor, para-chlorophenylalanine (p-CPA), and the 5-HT(1A) receptor agonist, 8-OH-DPAT, were all found to attenuate morphine and U50,488 analgesia. In each case, the attenuation was itself blocked by treatment with L-5-HTP, a serotonin precursor, bicuculline, a GABA(A) receptor antagonist or picrotoxin, a GABA(A)-gated chloride channel blocker. Neither L-5-HTP nor the GABA(A) receptor antagonists were found to affect morphine or U50,488 analgesia per se. Thus, these findings indicate that a benzodiazepine-GABAergic agent (diazepam) attenuates opioid analgesia through the serotonergic system, and antiserotonergic agents (8-OH-DPAT, p-CPA) attenuate opioid analgesia through the GABAergic system. The intimate interactions between GABA and serotonin in the present study further suggest that these neurotransmitters work in complex ways together rather than alone in the modulation of opioid analgesia.


Assuntos
Analgesia , Analgésicos Opioides , Receptores Opioides/fisiologia , Serotonina/fisiologia , Ácido gama-Aminobutírico/fisiologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , 5,7-Di-Hidroxitriptamina/farmacologia , 8-Hidroxi-2-(di-n-propilamino)tetralina/farmacologia , Analgésicos não Narcóticos/farmacologia , Animais , Bicuculina/farmacologia , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Fenclonina/farmacologia , Antagonistas GABAérgicos/farmacologia , Moduladores GABAérgicos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Morfina , Limiar da Dor/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Receptores Opioides kappa , Receptores Opioides mu , Serotoninérgicos/farmacologia , Antagonistas da Serotonina/farmacologia
15.
Neuroscience ; 104(1): 217-22, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11311544

RESUMO

It has been hypothesized that morphine tolerance and dependence in mice following chronic exposure may reflect increased compensatory activity of antiopioid systems. The endogenous peptide nociceptin/orphanin FQ has been shown to have anti-opioid effects, for example antagonizing morphine analgesia. Moreover, chronic morphine administration increases synthesis of the peptide, and morphine tolerance and dependence can be attenuated or reversed by antagonists and agonists of the nociceptin/orphanin FQ receptor, respectively. The present study seeks to confirm a role for nociceptin/orphanin FQ in opioid tolerance and dependence by comparing morphine ED(50) values and naloxone-precipitated withdrawal jumping in mice homozygous (knock-out) and heterozygous for a null mutation of the Npnc1 gene encoding the nociceptin/orphanin FQ propeptide, and their wild type littermates, following chronic morphine exposure. Relative to morphine-naive control mice, significant rightward shifts in the morphine dose-response curve, resulting in increased morphine ED(50) values (approximately two to three-fold), was observed for all genotypes following three days of repeated systemic morphine injections. However, no differences between genotypes in the magnitude of tolerance were observed. In contrast, knock-out mice displayed significantly increased naloxone-precipitated withdrawal jumping relative to heterozygous and wild-type mice following implantation with a morphine pellet (25mg) for 72h. Use of nociception/orphaninFQ transgenic knock-out mice thus demonstrate the differential involvement of nociceptin/orphanin FQ in morphine tolerance and dependence.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Tolerância a Medicamentos/genética , Camundongos Knockout/metabolismo , Dependência de Morfina/genética , Peptídeos Opioides/deficiência , Analgésicos Opioides/farmacologia , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Masculino , Camundongos , Camundongos Knockout/genética , Morfina/farmacologia , Dependência de Morfina/metabolismo , Dependência de Morfina/fisiopatologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Peptídeos Opioides/genética , Nociceptina
16.
Neuroscience ; 115(2): 463-9, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12421612

RESUMO

Physical dependence is a widely known consequence of morphine intake. Although commonly associated with prolonged or repeated morphine administration, withdrawal symptoms can be elicited even after a single prior morphine exposure. What remains contentious is the extent to which physical dependence following acute and chronic morphine treatment is mediated by common physiological substrates and, accordingly, represent distinct syndromes. The genetic relationship between acute and chronic morphine dependence was thus presently studied by comparing mice of 11 inbred strains (129P3, A, AKR, BALB/c, C3H/He, C57BL/6, CBA, DBA/2, LP, SJL, and SWR) for naloxone-precipitated withdrawal jumping responses using three subcutaneous morphine administration paradigms: acute (single injection) or chronic (three daily morphine injections for 4 days) injection, or chronic infusion (7 days via implanted osmotic minipumps). Although there were differences in the magnitude of withdrawal jumping between the three different morphine administration paradigms, large and significant strain differences were observed for each. In addition, the same strains were unusually sensitive or, conversely, altogether refractory to withdrawal jumping across all morphine treatment conditions. Overall, strain jumping means between acute and chronic dependence paradigms displayed a high degree of genetic correlation (r=0.87-0.95). The significant correlation between chronic morphine injection and continuous morphine infusion discounts the possible confounding effect of contextual learning and spontaneous withdrawal between chronic injections on the assessment of naloxone-precipitated withdrawal. Substantial heritability was also observed for acute and both paradigms of chronic dependence, with estimates ranging from h(2)=0.53 to 0.70. The present demonstration of a strong genetic correlation between physical dependence to morphine following acute and chronic treatment implies that genes associated with variable sensitivity in the two traits are the same, and is suggestive of shared physiological substrates. The data also demonstrate that the differential genetic liability to morphine physical dependence begins with, and is predicted by, the first morphine exposure.


Assuntos
Dependência de Morfina/genética , Dependência de Morfina/fisiopatologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Síndrome de Abstinência a Substâncias/genética , Síndrome de Abstinência a Substâncias/fisiopatologia , Doença Aguda , Analgésicos Opioides/farmacologia , Animais , Doença Crônica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos AKR , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Morfina/farmacologia , Especificidade da Espécie
17.
Neuroscience ; 101(3): 709-17, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11113319

RESUMO

The role of endogenous opioid systems in the analgesic response to exogenous opiates remains controversial. We previously reported that mice lacking the peptide neurotransmitter beta-endorphin, although unable to produce opioid-mediated stress-induced antinociception, nevertheless displayed intact antinociception after systemic administration of the exogenous opiate morphine. Morphine administered by a peripheral route can activate opioid receptors in both the spinal cord and brain. However, beta-endorphin neuronal projections are confined predominantly to supraspinal nociceptive nuclei. Therefore, we questioned whether the absence of beta-endorphin would differentially affect antinociceptive responses depending on the route of opiate administration. Time- and dose-response curves were obtained in beta-endorphin-deficient and matched wild-type C57BL/6 congenic control mice using the tail-immersion/withdrawal assay. Null mutant mice were found to be more sensitive to supraspinal (i.c.v.) injection of the micro-opioid receptor-selective agonists, morphine and D-Ala(2)-MePhe(4)-Gly-ol(5) enkephalin. In contrast, the mutant mice were less sensitive to spinal (i.t.) injection of these same drugs. Quantitative receptor autoradiography revealed no differences between genotypes in the density of mu, delta, or kappa opioid receptor binding sites in either the spinal cord or pain-relevant supraspinal areas. Thus we report that the absence of a putative endogenous ligand for the mu-opioid receptor results in opposite changes in morphine sensitivity between discrete areas of the nervous system, which are not simply caused by changes in opioid receptor expression.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Vias Eferentes/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , beta-Endorfina/deficiência , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Vias Eferentes/citologia , Vias Eferentes/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL/anatomia & histologia , Camundongos Endogâmicos C57BL/genética , Camundongos Endogâmicos C57BL/metabolismo , Camundongos Knockout , Morfina/farmacologia , Neurônios/citologia , Neurônios/metabolismo , Nociceptores/citologia , Nociceptores/metabolismo , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Ensaio Radioligante/estatística & dados numéricos , Medula Espinal/citologia , Medula Espinal/metabolismo , beta-Endorfina/genética
18.
Neuroscience ; 75(2): 333-7, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8930999

RESUMO

The heptadecapeptide orphanin FQ has recently been shown to be the endogenous agonist for the orphan opioid-like receptor, LC132. The molecular evidence that LC132 and orphanin FQ are evolutionarily related to other opioid receptors and their ligands suggests that these proteins may also play a role in modulating opiate actions. We now report that orphanin FQ (0.5-10 nmol), injected intracerebroventricularly in mice, does not produce hyperalgesia as suggested previously but rather reverses opioid-mediated (i.e. naloxone-sensitive) stress-induced antinociception in three different algesiometric assays. In addition to its antagonism of endogenous opioid antinociception, orphanin FQ dose-dependently (2.5-25 nmol) reverses systemic morphine antinociception (5 mg/kg, s.c.). Based on these data, we propose that orphanin FQ is a functional anti-opioid peptide.


Assuntos
Antagonistas de Entorpecentes , Peptídeos Opioides/farmacologia , Acetatos , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Hiperalgesia/induzido quimicamente , Injeções Intraventriculares , Masculino , Camundongos , Morfina/antagonistas & inibidores , Morfina/farmacologia , Peptídeos Opioides/administração & dosagem , Medição da Dor , Estresse Psicológico/complicações , Nociceptina
19.
Br J Pharmacol ; 120(4): 676-80, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9051307

RESUMO

1. The present study was designed to investigate further the effects of the newly discovered orphanin FQ (OFQ)-the endogenous ligand for the orphan opioid receptor (called, e.g., ORL, and LC132)-on pain modulation in the rat. We used the tail-flick assay as a nociceptive index. 2. When injected into a cerebral ventricle, OFQ (4 fmol-10 nmol) has no effect on basal tail-flick latency by itself at any dose, but dose-dependently antagonizes systemic morphine analgesia (400 fmol 50 nmol). 3. Injected intrathecally, OFQ (3 and 10 nmol) displayed an analgesic effect without producing motor dysfunction, and potentiated morphine analgesia (1 and 10 nmol). 4. The anti-opioid effect of OFQ in rat brain and the high level of expression of LC132/ORL, receptor in the locus coeruleus indicated a possible role of OFQ in the precipitation of opiate withdrawal symptoms. However, no such precipitation was observed by OFQ in morphine-dependent rats.


Assuntos
Analgesia , Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Morfina/farmacologia , Peptídeos Opioides/administração & dosagem , Receptores Opioides/agonistas , Medula Espinal/efeitos dos fármacos , Animais , Interações Medicamentosas , Injeções Intraventriculares , Injeções Espinhais , Masculino , Medição da Dor , Ratos , Ratos Wistar , Receptor de Nociceptina , Nociceptina
20.
Pharmacogenomics ; 2(3): 177-94, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11535108

RESUMO

Interindividual differences in the experience of pain have been appreciated clinically for over a century. More recently, there has been a growing body of evidence demonstrating differences in analgesic response to various pharmacotherapies, although the source of this variability largely remains to be explained. To this end, basic science research is beginning to identify the allelic variants that underlie such antinociceptive variability using a multiplicity of animal models, and powerful genetic approaches are being exploited to accelerate this process. Although the vast majority of these studies have focused on the pharmacogenetics of opioids, owing to their prominent status as analgesics, the number of pharmacotherapies evincing genetically-based variability is rapidly expanding. In addition, analogous studies have been undertaken in humans, as a small but growing number of clinical trials have begun to evaluate prospectively the existence, if oftentimes not the origin, of interindividual differences in analgesic drug response. Importantly, with a few notable exceptions, such efforts have primarily identified differences in analgesic efficacy and/or potency between male and female human subjects. Looking toward the future development of one or more widely utilised, pharmacogenetic screens that would lead to modifications in treatment planning, at least with respect to the pharmacologic management of pain, this review will document the breadth of genetically-based variability in drug-mediated antinociception in animals. Specific examples in which the gene or genes underlying such variability have been postulated or identified will be given, while highlighting the effect of sex and its interactions with other genetic backgrounds. Finally, we will summarise and evaluate the literature on pharmacogenetic differences in human analgesic drug response, for which the influence of sex has served as one of the better studied and heuristically insightful examples.


Assuntos
Analgésicos/uso terapêutico , Dor/tratamento farmacológico , Dor/genética , Farmacogenética , Animais , Citocromo P-450 CYP2D6/genética , Feminino , Humanos , Masculino , Polimorfismo Genético , Característica Quantitativa Herdável , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA