Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Behav Pharmacol ; 32(1): 21-31, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33079734

ABSTRACT

There are sex differences in the development of cocaine addiction. For example, the time that it takes for women from initial use to addiction is significantly shorter than for men. Thus, understanding why females are more vulnerable to cocaine addiction will provide insights into sex differences in the mechanisms underlying cocaine addiction. This study aimed to determine how cocaine demand intensity and elasticity might differ between sexes. In addition, the impact of estrous cycle and cocaine intake on demand was investigated. Male and female rats were trained to self-administer 0.125 mg of cocaine intravenously under a chained schedule in daily 2-h sessions for 2 weeks, and then, the cocaine demand function was determined with a modified within-session threshold procedure. Following the test, the rats began to self-administer a higher dose of cocaine (0.25 mg) to increase the cocaine intake. The demand function was then similarly determined in the same rats after 2 weeks of cocaine self-administration of the higher dose. No sex differences were found in either demand intensity or elasticity. Neither did the level of cocaine intake have an impact on demand. The demand elasticity, but not intensity, was significantly lower during proestrus/estrus compared with diestrus. These data suggest that the faster transition to cocaine addiction in women cannot be explained by sex differences in the demand for cocaine and such a demand may change during different phases of estrus cycle.


Subject(s)
Behavior, Addictive/physiopathology , Cocaine-Related Disorders/physiopathology , Cocaine/administration & dosage , Self Administration , Animals , Behavior, Animal , Dose-Response Relationship, Drug , Economics, Behavioral , Estrous Cycle/physiology , Female , Male , Rats , Rats, Wistar , Sex Factors , Time Factors
2.
Neuropharmacology ; 164: 107847, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31758947

ABSTRACT

Tolerance to the antinociceptive effects of cannabinoids represents a significant limitation to their clinical use in managing chronic pain. Tolerance likely results from desensitization and down-regulation of the cannabinoid type 1 receptor (CB1R), with CB1R desensitization occurring via phosphorylation of CB1Rs by a G protein-coupled receptor kinase and subsequent association with an arrestin protein. Previous studies have shown that (1) desensitization-resistant S426A/S430A mice exhibit a modest delay in tolerance for Δ9-THC and (-)-CP55,940 but a more pronounced disruption in tolerance for WIN 55,212-2 and (2) that c-Jun N-terminal kinase (JNK) signaling may selectively mediate antinociceptive tolerance to morphine compared to other opioid analgesics. In the current study, we found that pretreatment with the JNK inhibitor SP600125 (3 mg/kg) attenuates tolerance to the antinociceptive in the formalin test and to the anti-allodynic effects of Δ9-THC (6 mg/kg) in cisplatin-evoked neuropathic pain using wild-type mice. We also find that SP600125 causes an especially robust reduction in tolerance to the antinociceptive effects of Δ9-THC (30 mg/kg), but not WIN 55,212-2 (10 mg/kg) in the tail-flick assay using S426A/S430A mice. Interestingly, SP600125 pretreatment accelerated tolerance to the antinociceptive and anti-allodynic effects of (-)-CP55,940 (0.3 mg/kg) in mice with acute and neuropathic pain. These results demonstrate that inhibition of JNK signaling pathways delay tolerance to Δ9-THC, but not to CP55,940 or WIN55,212-2, demonstrating that the mechanisms of cannabinoid tolerance are agonist-specific.


Subject(s)
Analgesics/pharmacology , Cannabinoids/pharmacology , JNK Mitogen-Activated Protein Kinases/drug effects , Signal Transduction/drug effects , Animals , Anthracenes/pharmacology , Benzoxazines/pharmacology , Cisplatin , Dronabinol/pharmacology , Drug Tolerance , Hyperalgesia/drug therapy , Male , Mice , Morpholines/pharmacology , Naphthalenes/pharmacology , Neuralgia/chemically induced , Neuralgia/drug therapy , Pain Measurement
3.
Mol Pain ; 13: 1744806917728227, 2017.
Article in English | MEDLINE | ID: mdl-28879802

ABSTRACT

The cannabinoid 1 receptor and cannabinoid 2 receptor can both be targeted in the treatment of pain; yet, they have some important differences. Cannabinoid 1 receptor is expressed at high levels in the central nervous system, whereas cannabinoid 2 receptor is found predominantly, although not exclusively, outside the central nervous system. The objective of this study was to investigate potential interactions between cannabinoid 2 receptor and the mu-opioid receptor in pathological pain. The low level of adverse side effects and lack of tolerance for cannabinoid 2 receptor agonists are attractive pharmacotherapeutic traits. This study assessed the anti-nociceptive effects of a selective cannabinoid 2 receptor agonist (JWH-133) in pathological pain using mice subjected to inflammatory pain using the formalin test. Furthermore, we examined several ways in which JWH-133 may interact with morphine. JWH-133 produces dose-dependent anti-nociception during both the acute and inflammatory phases of the formalin test. This was observed in both male and female mice. However, a maximally efficacious dose of JWH-133 (1 mg/kg) was not associated with somatic withdrawal symptoms, motor impairment, or hypothermia. After eleven once-daily injections of 1 mg/JWH-133, no tolerance was observed in the formalin test. Cross-tolerance for the anti-nociceptive effects of JWH-133 and morphine were assessed to gain insight into physiologically relevant cannabinoid 2 receptor and mu-opioid receptor interaction. Mice made tolerant to the effects of morphine exhibited a lower JWH-133 response in both phases of the formalin test compared to vehicle-treated morphine-naïve animals. However, repeated daily JWH-133 administration did not cause cross-tolerance for morphine, suggesting opioid and cannabinoid 2 receptor cross-tolerance is unidirectional. However, preliminary data suggest co-administration of JWH-133 with morphine modestly attenuates morphine tolerance. Isobolographic analysis revealed that co-administration of JWH-133 and morphine has an additive effect on anti-nociception in the formalin test. Overall these findings show that cannabinoid 2 receptor may functionally interact with mu-opioid receptor to modulate anti-nociception in the formalin test.


Subject(s)
Analgesics, Opioid/therapeutic use , Inflammation/complications , Inflammation/drug therapy , Nociception/drug effects , Pain/complications , Pain/drug therapy , Receptor, Cannabinoid, CB2/agonists , Analgesics, Opioid/pharmacology , Animals , Cannabinoids/administration & dosage , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Drug Interactions , Drug Tolerance , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Morphine/administration & dosage , Morphine/pharmacology , Morphine/therapeutic use , Pain Measurement , Receptors, Opioid, mu/metabolism
4.
Neuroreport ; 27(6): 392-6, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-26914092

ABSTRACT

The abuse and overdose of opioid drugs are growing public health problems worldwide. Although progress has been made toward understanding the mechanisms governing tolerance to opioids, the exact cellular machinery involved remains unclear. However, there is growing evidence to suggest that c-Jun N-terminal kinases (JNKs) play a major role in mu-opioid receptor regulation and morphine tolerance. In this study, we aimed to determine the potential roles of different JNK isoforms in the development of tolerance to the antinociceptive and hypothermic effects of morphine. We used the hot-plate and tail-flick tests for thermal pain to measure tolerance to the antinociceptive effects of once-daily subcutaneous injections with 10 mg/kg morphine. Body temperature was also measured to determine tolerance to the hypothermic effects of morphine. Tolerance to morphine was assessed in wild-type mice and compared with single knockout mice each lacking the JNK isoforms (JNK1, JNK2, or JNK3). We found that loss of each individual JNK isoform causes impairment in tolerance for the antinociceptive and hypothermic effects of daily morphine. However, disruption of JNK2 seems to have the most profound effect on morphine tolerance. These results indicate a clear role for JNK signaling pathways in morphine tolerance. This complements previous studies suggesting that the JNK2 isoform is required for morphine tolerance, but additionally presents novel data suggesting that additional JNK isoforms also contribute toward this process.


Subject(s)
Analgesics, Opioid/therapeutic use , Hypothermia, Induced/methods , JNK Mitogen-Activated Protein Kinases/metabolism , Morphine/therapeutic use , Pain Management/methods , Pain/drug therapy , Animals , Body Temperature/drug effects , Drug Tolerance , JNK Mitogen-Activated Protein Kinases/classification , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Pain/etiology , Pain Measurement , Protein Isoforms/genetics , Protein Isoforms/metabolism
5.
Brain Res Bull ; 123: 5-12, 2016 05.
Article in English | MEDLINE | ID: mdl-26521067

ABSTRACT

The rewarding and antinociceptive effects of opioids are mediated through the mu-opioid receptor. The A118G single nucleotide polymorphism in this receptor has been implicated in drug addiction and differences in pain response. Clinical and preclinical studies have found that the G allele is associated with increased heroin reward and self-administration, elevated post-operative pain, and reduced analgesic responsiveness to opioids. Male and female mice homozygous for the "humanized" 118AA or 118GG alleles were evaluated to test the hypothesis that 118GG mice are less sensitive to the rewarding and antinociceptive effects of morphine. We found that 118AA and 118GG mice of both genders developed conditioned place preference for morphine. All mice developed tolerance to the antinociceptive and hypothermic effects of morphine. However, morphine tolerance was not different between AA and GG mice. We also examined sensitivity to the antinociceptive and hypothermic effects of cumulative morphine doses. We found that 118GG mice show reduced hypothermic and antinociceptive responses on the hotplate for 10mg/kg morphine. Finally, we examined basal pain response and morphine-induced antinociception in the formalin test for inflammatory pain. We found no gender or genotype differences in either basal pain response or morphine-induced antinociception in the formalin test. Our data suggests that homozygous expression of the GG allele in mice blunts morphine-induced hypothermia and hotplate antinociception but does not alter morphine CPP, morphine tolerance, or basal inflammatory pain response.


Subject(s)
Pain/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Alleles , Analgesics/metabolism , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Drug Tolerance/genetics , Female , Male , Mice , Mice, Transgenic , Morphine/pharmacology , Pain/drug therapy , Pain Measurement/drug effects , Polymorphism, Single Nucleotide/genetics
6.
Mol Pain ; 11: 34, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-26065412

ABSTRACT

BACKGROUND: Morphine and fentanyl are opioid analgesics in wide clinical use that act through the µ-opioid receptor (MOR). However, one limitation of their long-term effectiveness is the development of tolerance. Receptor desensitization has been proposed as a putative mechanism driving tolerance to G protein-coupled receptor (GPCR) agonists. Recent studies have found that tolerance to morphine is mediated by the c-Jun N-terminal Kinase (JNK) signaling pathway. The goal of the present study was to test the hypotheses that: 1) JNK inhibition will be antinociceptive on its own; 2) JNK inhibition will augment morphine antinociception and; 3) JNK mediates chronic tolerance for the antinociceptive effects of morphine using acute (hotplate and tail-flick), inflammatory (10 µl of formalin 2.5%) and chemotherapy (cisplatin 5 mg/kg ip once weekly)-induced neuropathic pain assays. RESULTS: We found that JNK inhibition by SP600125 (3 mg/kg) produces a greater antinociceptive effect than morphine (6 mg/kg) alone in the formalin test. Moreover, co-administration of morphine (6 mg/kg) with SP600125 (3 mg/kg) produced a sub-additive antinociceptive effect in the formalin test. We also show that pre-treatment with SP600125 (3 or 10 mg/kg), attenuates tolerance to the antinociceptive effects of morphine (10 mg/kg), but not fentanyl (0.3 mg/kg), in the tail-flick and hotplate tests. Pre-treatment with SP600125 also attenuates tolerance to the hypothermic effects of both morphine and fentanyl. We also examined the role of JNK in morphine tolerance in a cisplatin-induced model of neuropathic pain. Interestingly, treatment with SP600125 (3 mg/kg) alone attenuated mechanical and cold allodynia in a chemotherapy-induced pain model using cisplatin. Strikingly, SP600125 (3 mg/kg) pre-treatment prolonged the anti-allodynic effect of morphine by several days (5 and 7 days for mechanical and cold, respectively). CONCLUSIONS: These results demonstrate that JNK signaling plays a crucial role in mediating antinociception as well as chronic tolerance to the antinociceptive effects of morphine in acute, inflammatory, and neuropathic pain states. Thus, inhibition of JNK signaling pathway, via SP600125, represents an efficacious pharmacological approach to delay tolerance to the antinociceptive effects of chronic morphine in diverse pain models.


Subject(s)
Analgesics/pharmacology , Drug Tolerance , JNK Mitogen-Activated Protein Kinases/metabolism , Morphine/pharmacology , Animals , Anthracenes/pharmacology , Cisplatin/pharmacology , Fentanyl/pharmacology , Formaldehyde , Hyperalgesia/pathology , Hypothermia, Induced , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice, Inbred C57BL , Models, Biological , Morphine/administration & dosage , Nociception/drug effects , Protein Kinase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...