首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 0 毫秒
1.
In the presence of physiological cations (in Krebs-4-(2-hydroxyethyl)-1- piperazineethanesulfonic acid buffer) at 37 degrees C the Ki value's of beta-endorphin for mu- and delta-opioid receptor binding sites in rat neocortical membranes, labeled with [3H][D-Ala2,MePhe4,Gly- ol5]enkephalin (DAMGO) and [3H][D-Ala2-D-Leu5]enkephalin (in the presence of unlabeled DAMGO), respectively, amounted to about 9 and 22 nM. Surprisingly, a very different selectivity pattern for the endogenous opioid peptide was found when the affinity of beta-endorphin for functional presynaptic opioid receptors was examined. Thus, beta-endorphin strongly inhibited the electrically evoked release of [3H]NE from rat neocortical slices with an IC50 value of about 0.5 nM, whereas [14C] acetylcholine release from neostriatal slices was inhibited with an IC50 value of about 100 nM. On the other hand, the electrically evoked release of [3H]dopamine from striatal slices was not affected by beta-endorphin. The inhibitory effects of DAMGO and beta-endorphin on [3H]NE release from neocortical slices were equally well antagonized by naloxone. Moreover, 10 nM of the highly selective mu-opioid receptor antagonist D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen- Thr-NH2 antagonized competitively the inhibitory effect of beta-endorphin on [3H]NE release.(ABSTRACT TRUNCATED AT 250 WORDS)  相似文献   

2.
The relative importance of mu and delta opioid receptors in brain regulation of sympathoadrenal, cardiovascular and respiratory function was investigated using highly selective mu and delta opioid peptide analogs. Groups of conscious rats received i.c.v. injections of either the mu-selective agonist, [D-Ala2, MePhe4, Gly-ol5]enkephalin (DAMGO) or the delta-selective agonist, [D-Pen2, D-Pen5]enkephalin (DPDPE). Blood pressure and heart rate were recorded continuously via a chronic catheter in the carotid artery, and arterial blood samples were taken at intervals through the same catheter for determination of blood pH, pCO2, pO2 and plasma catecholamine concentrations. Both DAMGO and DPDPE increased plasma catecholamine levels and blood pressure in a dose-related manner. The slopes of the dose-response lines were parallel, but the delta compound was about 250 times less potent than DAMGO. Only the highest dose of 5 nmol of DAMGO caused a significant bradycardia, mediated by parasympathetic (vagal) activation. DAMGO and DPDPE also induced dose-dependent acidosis, with DAMGO again being much more potent than DPDPE. The effects of both DAMGO and DPDPE on plasma catecholamines, blood pressure and blood gases were antagonized by a mu-selective dose of naloxone (0.4 mg/kg i.a.). Intracerebroventricular administration of the delta-selective antagonist, ICI 174,864, only partially attenuated sympathoadrenal and blood gas responses to DAMGO or DPDPE. The pressor responses to DAMGO or DPDPE were resistant to antagonism by ICI 174,864. These results indicate that brain opioid receptors regulating autonomic outflow, cardiovascular and respiratory function are mainly of the mu type, although a delta opioid system may contribute to sympathoadrenal and respiratory effects of opioids.  相似文献   

3.
4.
We examined the effects of amlodipine, a selective L-type voltage dependent Ca(2+) channel (VDCC) blocker, and mibefradil, a selective T-type VDCC blocker on the antinociceptive effects of morphine, and mu, delta and kappa opioid receptor selective agonist-induced antinociception at the spinal level. Intrathecally administered amlodipine and mibefradil potentiated morphine and [D-Ala(2), N mePhe(4), Gly-ol(5)] enkephalin (DAMGO)-induced antinociception by shifting their dose response curves to the left. However, intrathecally administered amlodipine and mibefradil did not affect [D-Pen(2), D-Pen(5)]enkephalin (DPDPE) and [trans-(+/-)-3,4-dichloro-N-methyl-N-[2-(1-pyrolidinyl)cyclohexyl] benzene acetamide (U-50, 488H)-induced antinociception. These data indicate that L-type and T-type VDCC blockers synergistically potentiate the analgesic effects of mu opioid receptor agonists, but not delta and kappa opioid receptor agonists, at the spinal level. Additionally, these data suggest that there is an important functional interaction between L-type and/ or T-type VDCC and mu opioid receptors in the process of analgesia.  相似文献   

5.
Sulfated cholecystokinin octapeptide (CCK-8; Asp-Tyr-SO3H-Met-Gly-Trp-Met-Asp-Phe-NH2) produced analgesia in mice when administered i.c.v. and tested in the acetic acid-induced writhing assay. The ED50 was found to be 0.03 nmol/mouse which was about 3, 24 and 714 times more potent than morphine. [D-Pen2,D-Pen5]enkephalin and U50,488H [trans-(+/-)-3,4-dichloro-N-methyl-N-[2-(1-pyrolidinyl)cyclohexyl] benzeneacetamidel], respectively. When administered i.t., CCK-8 produced partial analgesia of up to 22 to 23% at low doses ranging from 15 to 60 ng/mouse and hyperalgesia at doses over 120 ng/mouse. Naloxone, an opioid antagonist, inhibited the analgesia induced by CCK-8 (i.c.v. and i.t.) but potentiated hyperalgesia induced by CCK-8 (i.t.). Apparent pA2 value for CCK-8 (i.c.v.) against naloxone (s.c.) was 5.88 which was significantly different from those for morphine-naloxone and U50,488H-naloxone but was not significantly different from that for [D-Pen2,D-Pen5]enkephalin-naloxone. Studies using highly selective opioid antagonists showed that CCK-8-induced analgesia was significantly antagonized by the delta receptor antagonist, ICI154,129 [(Allyl)2-Tyr-gly-gly-psi-(CH2S)-Phe-Leu] but not by beta-funaltrexamine, a highly selective mu receptor antagonist or nor-binaltorphimine, a highly selective kappa receptor antagonist. Opioid receptor binding study using [3H]-[D-Ala2,D-Leu5]enkephalin (+unlabeled [D-Ala2,MePhe4,Gly-ol5]enkephalin) in mouse brain membrane preparations revealed that there were no changes in the maximum binding or Kd of delta opioid binding sites in the presence of CCK-8 (1 microM) in vitro.(ABSTRACT TRUNCATED AT 250 WORDS)  相似文献   

6.
Opioid receptors exist in at least three forms: mu, delta and kappa. Agonists at mu receptors produce orthostatic hypotension in man by a mechanism involving a reduction in baroreflex sensitivity. We describe here the cardiovascular properties of metkephamid, a relatively selective delta opioid receptor agonist. Blood pressure, heart rate and plasma noradrenaline concentration were measured over a 7 h period in eight normal young male volunteers in the supine position and after 70 degrees 5 min head-up tilt, after receiving metkephamid (50 mg intramuscularly) or placebo. Metkephamid increased heart rate in the supine position with no change in blood pressure or plasma noradrenaline concentration. This was accompanied by symptoms consistent with an anti-muscarinic anticholinergic effect. Head-up tilt resulted in substantial hypotension after metkephamid with an attenuated change in heart rate and no increase in noradrenaline concentration. We conclude that delta as well as mu opioid receptor agonists can produce orthostatic hypotension with attenuation of heart rate response. Metkephamid possesses anticholinergic properties not seen with mu receptor agonists, suggesting a possible role of delta opioid receptors in cholinergic activity.  相似文献   

7.
In many cases, body temperature is altered in response to opioid agonists, but the direction, magnitude and time course of alteration vary with a number of factors. Body temperature may be subject to differential modification by different opioid receptor types. The authors examined the effect (i.c.v.) of the selective mu, delta and kappa opioid agonists, [D-Ala2, MePhe4, Gly5-ol] enkephalin (DAGO), [D-Pen2, D-Pen5] enkephalin and U50488H, respectively, on the body temperature of restrained and unrestrained rats. Each of the three opioid agonists produced a differentiable profile of body temperature changes. DAGO caused a primary decrease in body temperature of restrained rats and an increase in body temperature of unrestrained rats. The pretreatment dose of naloxone necessary to attenuate the hyperthermic response to DAGO of unrestrained rats was 10 times higher than that required to block the hypothermic response to DAGO in restrained rats. Low doses of both [D-Pen2, D-Pen5]enkephalin and U50488H caused a decrease in body temperature of both restrained and unrestrained rats. Hypothermic responses to U50488H were not blocked by naloxone, whereas hypothermic responses to [D-Pen2, D-Pen5]enkephalin in unrestrained rats were potentiated by naloxone. The results indicate that the three compounds modified body temperature by different means, suggesting activation of different opioid, and perhaps nonopioid, receptors. This may reflect a differential modulation of body temperature by endogenous opioids depending on the specific peptide released and the receptor type activated. Besides the physiologic implications, body temperature responses provided a sensitive pharmacologic measure for distinguishing the in vivo activity of different selective opioid agonists.  相似文献   

8.
The role of mu and delta opioid receptors in the spinal and supraspinal analgesic actions of morphine and [D-Pen2, L-Pen5] enkephalin were examined in the tail-flick test utilizing the mu opioid receptor deficient CXBK mouse and BALB/cBy and C57BL/6By, the progenitor strains of CXBK. The analgesic effects of i.c.v. administered morphine were equivalent in the CRS-CD1 (Swiss) standard laboratory mice and the progenitor strains of CXBK. Morphine did not, however, produce analgesia in the CXBK mice at doses greater than 10 times the ED50 dose in the progenitor strains. Similarly, the analgesic effect of i.c.v. [D-Ala2, NMePhe4, Gly-ol]enkephalin, a highly selective mu receptor peptide agonist, also was reduced greatly in the CXBK mice. These data are consistent with the deficiency in mu opioid receptors observed autoradiographically in this strain. In contrast, the highly selective delta opioid receptor peptide agonist [D-Pen2, L-Pen5]enkephalin was equipotent i.c.v. in the CXBK mice and in the progenitor strains of CXBK. In contrast to the effects produced by i.c.v. administration, the analgesic effects of intrathecally administered morphine were similar between CRS-CD1 and CXBX strains of mice. These results suggest that 1) both mu and delta opioid receptors can mediate supraspinal analgesia and 2) that the receptor(s) involved in spinally mediated analgesia is (are) quite distinct from those involved supraspinally.  相似文献   

9.
U-50,488: a selective and structurally novel non-Mu (kappa) opioid agonist   总被引:21,自引:0,他引:21  
U-50,488 (trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]-benzeneacetamide) displays analgesic actions in a variety (thermal, pressure and irritant) of assays in mice and rats. Naloxone and MR-2266 block this analgesic effect; thus it is mediated by opioid receptors. However, when compared to morphine analgesia, the naloxone and MR-2266 pA2 values for U-50,488 analgesia were much lower and higher, respectively. Likewise, although tolerance occurs to both morphine and U-50,488 analgesia, there was no cross-tolerance between these drugs, and U-50,488 does not cause morphine-type physical dependence. These observations suggest that different opioid receptors mediate the analgesic effects of morphine and U-50,488. The effects of U-50,488 appear to be mediated by the so-called kappa opioid receptor. In contrast to U-50,488, other reputed kappa opioid agonists displayed varying degrees of mu agonist (ketazocine and ethylketocyclazocine) and narcotic antagonist (bremazocine) activities. Thus U-50,488 is a more selective kappa agonist. This conclusion is further supported by binding studies; of all compounds tested, U-59,488 displacement of [3H]ethylketocyclazocine binding was uniquely not blocked by high concentrations of dihydromorphine. In addition to analgesia, this selective kappa agonist also causes opioid receptor-mediated sedation, diuresis and corticosteroid elevations. U-50,488 is a useful tool for studying contrasting kappa and mu opioid receptor-mediated effects.  相似文献   

10.
Neuropathic (NP) pain is a debilitating chronic pain disorder considered by some to be inherently resistant to therapy with traditional analgesics. Indeed, micro opioid receptor (OR) agonists show reduced therapeutic benefit and their long term use is hindered by the high incidence of adverse effects. However, pharmacological and physiological evidence increasingly suggests a role for deltaOR agonists in modulating NP pain symptoms. In this study, we examined the antihyperalgesic and antiallodynic effects of the spinally administered deltaOR agonist, d-[Ala(2), Glu(4)]deltorphin II (deltorphin II), as well as the changes in deltaOR expression, in rats following chronic constriction injury (CCI) of the sciatic nerve. Rats with CCI exhibited cold hyperalgesia and mechanical allodynia over a 14-day testing period. Intrathecal administration of deltorphin II reversed cold hyperalgesia on day 14 and dose-dependently attenuated mechanical allodynia. The effects of deltorphin II were mediated via activation of the deltaOR as the effect was antagonized by co-treatment with the delta-selective antagonist, naltrindole. Western blotting experiments revealed no changes in deltaOR protein in the dorsal spinal cord following CCI. Taken together, these data demonstrate the antihyperalgesic and antiallodynic effectiveness of a spinally administered deltaOR agonist following peripheral nerve injury and support further investigation of deltaORs as potential therapeutic targets in the treatment of NP pain.  相似文献   

11.
OBJECTIVES: We sought to evaluate whether there is a sex difference in the analgesic response to mu versus kappa opioids in the management of acute moderate to severe pain of injury in the emergency department. METHODS: The study was a randomized, double-blind, clinical trial comparing the prototypical mu-receptor agonist, morphine sulfate, to the prototypical kappa agonist, butorphanol. The primary endpoints were degree of relief by visual analog scores at 30 and 60 minutes. Statistical analysis was performed using Mann-Whitney Utest for nonparametric analysis and repeated-measures analysis of variance. RESULTS: Ninety-four patients were entered in the study, with 49 (52%) males and 45 (48%) females. Forty-six received morphine sulfate and 48 received butorphanol. There was no difference in demographics in the two groups. At 60 minutes, females had significantly lower visual analog scores with butorphanol compared with morphine (P = 0.046). At 60 minutes, there was a trend for a difference in response of males versus females to morphine, with males responding better than females (P = 0.06). CONCLUSION: Females had better pain scores with butorphanol than morphine at 60 minutes.  相似文献   

12.
The cyclic, conformationally restricted octapeptide [3H]-[H-D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2] ([3H]CTOP) was synthesized and its binding to mu opioid receptors was characterized in rat brain membrane preparations. Association rates (k+1) of 1.25 x 10(8) M-1 min-1 and 2.49 x 10(8) M-1 min-1 at 25 and 37 degrees C, respectively, were obtained, whereas dissociation rates (k-1) at the same temperatures were 1.93 x 10(-2) min-1 and 1.03 x 10(-1) min-1 at 25 and 37 degrees C, respectively. Saturation isotherms of [3H]CTOP binding to rat brain membranes gave apparent Kd values of 0.16 and 0.41 nM at 25 and 37 degrees C, respectively. Maximal number of binding sites in rat brain membranes were found to be 94 and 81 fmol/mg of protein at 25 and 37 degrees C, respectively. [3H]CTOP binding over a concentration range of 0.1 to 10 nM was best fit by a one site model consistent with binding to a single site. The general effect of different metal ions and guanyl-5'-yl-imidodiphosphate on [3H]CTOP binding was to reduce its affinity. High concentrations (100 mM) of sodium also produced a reduction of the apparent mu receptor density. Utilizing the delta opioid receptor specific peptide [3H]-[D-Pen2,D-Pen5]enkephalin, CTOP appeared to be about 2000-fold more specific for mu vs. delta opioid receptor than naloxone. Specific [3H]CTOP binding was inhibited by a large number of opioid or opiate ligands.(ABSTRACT TRUNCATED AT 250 WORDS)  相似文献   

13.
Opioid agonists produce analgesia in mammals through the activation of mu, kappa, or delta opioid receptors. Previous behavioral and binding studies from our laboratory using an amphibian model suggested that mu, kappa, or delta opioid agonists may activate a single type of opioid receptor in the grass frog, Rana pipiens. In the present study, kinetic, saturation, and competitive binding profiles for three opioid radioligands, [(3)H]DAMGO ([D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin) (mu-selective), [(3)H]U65953 [(5 alpha, 7 alpha,8 beta)-(+)-N-methyl-N-[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl]-benzeneacetamide] (kappa-selective), and [(3)H]DPDPE ([D-Pen(2),D-Pen(5)]-enkephalin) (delta-selective) were determined using frog whole brain homogenates. Kinetic analyses and experimentally derived values from saturation experiments gave affinity constants (K(D)) in the low nanomolar range. The density of opioid binding sites (B(max)) was 224.4, 118.6, and 268.9 fmol/mg for mu, kappa, and delta opioid radioligands, respectively. The affinity values did not significantly differ among the three opioid radioligands, but the kappa radioligand bound to significantly fewer sites than did the mu or delta radioligands. K(i) values for unlabeled mu, kappa, and delta competitors, including highly selective opioid antagonists, were consistent with each radioligand selectivity profile. The present data suggest that mu, kappa, and delta opioid radioligands bind to distinct opioid receptors in amphibians that are surprisingly similar to those found in mammalian brain.  相似文献   

14.
CI-977 is a selective, nonpeptide kappa opioid agonist. In rhesus monkeys, CI-977 is a potent antinociceptive agent against thermal stimuli after i.m. administration. Increasing the intensity of the nociceptive stimulus can reduce the analgesic activity of CI-977. Antinociceptive activity also was seen when PD 126212, containing CI-977 as the (-)-enantiomer, was administered sublingually. Naloxone antagonized the antinociceptive action of CI-977, demonstrating opiate receptor involvement in this activity. Monkeys treated with CI-977 also showed sedation at doses close to those required to produce antinociception. As with morphine, the sedative properties of CI-977 were associated with impaired cognitive performance. Aged monkeys appeared more sensitive than young monkeys to the performance-impairing effect of CI-977. Tolerance developed to the antinociceptive and response-suppressing effects. CI-977 was approximately 1000 times more potent than morphine as an analgesic when tested against a moderate (50 degrees C) thermal stimulus but less effective than morphine against a strong (55 degrees C) thermal stimulus.  相似文献   

15.
U50,488, a highly selective kappa opioid: anticonvulsant profile in rats   总被引:1,自引:0,他引:1  
Subcutaneous or i.c.v. administration of U50,488, a highly selective kappa opioid agonist, resulted in a dose-and time-dependent anticonvulsant action in rats. The anticonvulsant effect was seizure-specific; thus, U50,488 protected against supramaximal electroshock seizures but failed to raise the threshold of flurothyl-induced convulsions. The ED50 for s.c. U50,488 was 8.6 mg/kg, with a duration of action longer than 8 hr. In contrast, the ED50 for i.c.v. U50,488 was 103.8 micrograms, lasting approximately 1 hr. The anticonvulsant effect of U50,488 was partially antagonized by high (10.0 mg/kg), but not low (1.0 mg/kg), doses of s.c. administered naloxone. Results indicate that U50,488 is an efficacious, long-acting anticonvulsant against supramaximal, but not chemical threshold, seizures in rats. Furthermore, the results with naloxone suggest that this effect of U50,488 is mediated by non-mu (probably kappa) binding sites. This structurally novel nonpeptide opioid may offer new insights into the development of therapeutically effective agents in the treatment of grand mal or partial epilepsies.  相似文献   

16.
We have investigated the roles of peripheral and central mu, delta, and kappa opioid receptors and their subtypes in opioid-induced hypothermia in mice. Measuring rectal temperature after i.p. injection, opioid agonists [morphine, fentanyl, SNC80 ((+)-4-[(alphaR)-alpha-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)3-methoybenzyl]-N,N-diethylbenzamide), U50,488H ((trans-(dl)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]-benzeneacetamide), and loperamide)] were tested alone or with opioid antagonists [naloxone, beta-funaltrexamine, naloxonazine, naltrindole, 7-benzylidenenaltrexone (BNTX), naltriben, nor-binaltorphimine, 2-(3,4-dichlorophenyl)-N-methyl-N-[(1S)-1-(3-isothiocyanatophenyl)-2-(1-pyrrolidinyl)ethyl]acetamide (DIPPA), and methyl-naltrexone] given 15 min after the agonist. All agonists produced dose-related hypothermia, although at low doses, morphine and U50,488H produced hyperthermia. The effects of morphine and fentanyl were antagonized by naloxone and by the mu(1) antagonist naloxonazine. The delta(2) antagonist naltriben potentiated the hypothermic effect of mu agonists. SNC80-induced hypothermia was blocked by the delta antagonist naltrindole but not by the delta(1) antagonist BNTX. Depending on the dose, the delta(2) antagonist naltriben produced either a potentiation or an attenuation of the effect of SNC80. U50,488H-induced hypothermia was antagonized by the kappa antagonist nor-binaltorphimine but not by acute treatment with the irreversible kappa antagonist DIPPA. The peripherally acting opioid loperamide produced hypothermia that could be blocked by several mu-, delta-, or kappa-selective antagonists as well as the peripherally acting antagonist methyl-naltrexone. Methyl-naltrexone produced a weak potentiation of morphine-, fentanyl-, and U50,488H-induced hypothermia, whereas a significant attenuation of SNC80-induced hypothermia was observed. In conclusion, at high doses, morphine- and fentanyl-induced hypothermia may involve composite action on mu, kappa, and possibly delta opioid receptors after initial activation. In the mediation of delta opioid-induced hypothermia, no clear selectivity between the delta(1) and delta(2) subtypes was defined. The studies provide new evidence that maintenance of the initial effects of agonist/receptor activation vary with the agonist and the receptor. The existence of both central and peripheral components of opioid-induced hypothermia is also emphasized.  相似文献   

17.
Light microscopic autoradiography was used to visualize the neuroanatomical distribution of rat brain delta opioid receptors. Slide-mounted sections of rat brain were labeled with [3H]-[2-D-penicillamine, 5-D-penicillamine]enkephalin([3H]DPDPE), a highly selective delta opioid agonist. Saturation isotherms of [3H]DPDPE binding to thaw-mounted brain slices gave a maximal number of binding sites of 79.9 fmol/mg of protein and an apparent dissociation constant (Kd) of 6.3 nM. DPDPE and met-enkephalin inhibited [3H]DPDPE binding with high affinity (lC50 values of 6.3 and 13.8 nM, respectively). Putative mu opioid receptor selective ligands such as morphine sulfate, Tyr-D-Ala-Gly-NMePhe-Gyl-ol and [N-MePhe3, D-Pro4]morphiceptin (PL017) were less potent inhibitors of [3H]DPDPE binding. The rat brain areas containing the highest densities of receptors were the claustrum, basolateral amygdaloid nucleus, the caudate-putamen and nucleus accumbens, the external plexiform layer of the olfactory bulb and the olfactory tubercle. Moderate receptor density was characteristic of the hippocampal formation in which grains were seen over the molecular layer of the dentate gyrus and stratum oriens (CA1), and of the different layers of cerebral cortex. Generally, low density of binding was found over the thalamus and the septal nuclei. Low specific binding was also seen in the cerebellum, medulla oblongata and in the dorsal horn of the spinal cord. There was little specific [3H]DPDPE binding over the white matter areas.  相似文献   

18.
In this study naltrindole (NTI) and its benzofuran derivative (NTB) were studied for their antagonist activity against various delta opioid receptor agonists in the tail-flick antinociceptive assay in mice. The antinociceptive ED50 of i.c.v. administered DSLET [(D-Ser2, Leu5, Thr6)enkephalin] was shifted about 4-fold by either s.c. NTB or i.c.v. NTI injection. On the other hand, the antinociceptive ED50 of i.c.v. administered DPDPE [(D-Pen2,D-Pen5)enkephalin] was shifted 1.4- and 1.8-fold with s.c. NTB and i.c.v. NTI administration, respectively, which were significantly lower than the shifts observed with DSLET. NTB did not alter the antinociceptive action of i.c.v. administered [(D-Ala2,D-Leu5)enkephalin], morphine sulfate, [(D-Ala2,MePhe4,Gly-ol5)enkephalin] or U-50,488H (trans(+/-)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl) cyclohexyl]benzeneacetamide). At spinal sites, the antinociceptive ED50 of intrathecal (i.t.) administered DSLET was increased by 12.5-fold by s.c. NTB injection, whereas that of DPDPE was unaffected. NTB injection at this site also did not alter the antinociceptive action of i.t. administered [D-Ala2, D-Leu5]enkephalin, [(D-Ala2,MePhe4,Gly-ol5)enkephalin] or morphine sulfate. Pretreatment of animals with beta-funaltrexamine caused a large increase in the capacity of NTB to antagonize the antinociceptive activity of i.t. administered DSLET with little change in that of i.t. administered DPDPE. When cross-tolerance between DSLET and DPDPE was studied by i.c.v. injection of a single large dose of either DSLET or DPDPE 24 hr before the antinociceptive assay, there was no development of cross-tolerance between the two peptides. Based on these results, it was concluded that the antinociceptive action of DSLET and DPDPE may be mediated by different receptors, possibly delta opioid subtypes.  相似文献   

19.
The i.c.v. administration of opioid peptides having selectivity for the mu receptor (D-Ala2-NMePhe4-Gly5(ol)enkephalin and FK 33,824) produced effects on the locomotor activity of nondependent and morphine-dependent rats that differed both quantitatively and qualitatively from those effects produced by peptides having selectivity for the delta receptor (D-Ala2-D-Leu5enkephalin and metkephamid) and beta-endorphin, which has similar affinity for both receptors. Peptides selective for the mu receptor: had a biphasic effect on locomotor activity of nondependent rats, inducing an increase at low doses and an initial decrease followed by a later increase at higher doses and had an enhanced stimulant effect on locomotor activity with tolerance to the depressant effect in morphine-dependent rats. Peptides selective for the delta receptor and beta-endorphin: induced only a dose-related increase in the locomotor activity of nondependent rats and had effects on the locomotor activity of morphine-dependent rats that did not differ substantially from those in nondependent rats. Naltrexone (0.1 mg/kg s.c.) and beta-funaltrexamine (5.0 micrograms/rat i.c.v.), an irreversible antagonist, each blocked to a comparable extent the effects of D-Ala2-NMePhe4-Gly5(ol)enkephalin and DAla2-D-Leu5enkephalin on the locomotor activity of nondependent rats. Thus, effects of opioid peptides that act predominantly at mu or delta receptors on locomotor activity cannot be differentiated in nondependent rats by antagonists but can be differentiated in morphine-dependent rats. These results suggest that the depressant and stimulant effects of opioid peptides on locomotor activity are mediated by distinct neuronal sites.  相似文献   

20.
Extracellular recording of population spike amplitudes in the hippocampal CA1 region were compared in slices from control and chronically morphine-treated rats. Morphine treatment resulted in a decrease in the maximal excitation produced by both mu and delta selective agonists, [N-MePhe3,D-Pro4]-morphiceptin and [D-Pen2,L-Pen5]-enkephalin. The opioid antagonist naloxone did not produce apparent signs of withdrawal in hippocampal slices from morphine-treated rats as shown by a lack of change in the evoked population spike in the presence of 500 nM naloxone. Brain slices from morphine-treated rats that were maintained in the absence of morphine showed signs of tolerance reversal when monitored over an 8-hr period after dissection. If morphine-tolerant slices were maintained in vitro in the presence of 5 microM morphine (the concentration found by high-performance liquid chromatography to be present in the cerebrospinal fluid of morphine-treated rats), there was no significant reversal of tolerance. Furchgott analysis of the [N-MePhe3,D-Pro4]-morphiceptin concentration-response curve shifts induced by the irreversible opioid receptor antagonist beta-chlornaltrexamine revealed an apparent 50% spare receptor reserve for the mu selective agonist in slices from drug-naive rats. beta-Chlornaltrexamine (20 nM) treatment and chronic morphine exposure resulted in a similar reduction in the maximal response to [N-MePhe3,D-Pro4]morphiceptin. This observation indicates that the development of morphine tolerance resulted in an elimination of the spare opioid receptors.(ABSTRACT TRUNCATED AT 250 WORDS)  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号