Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 164: 107869, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31785260

ABSTRACT

Vesicular glutamate transporter 2 (VGLUT2)-which uptakes glutamate into presynaptic vesicles-is a fundamental component of the glutamate neurotransmitter system. Although several lines of evidence from genetically modified mice suggest a possible association of VGLUT2 with neuropathic pain, the specific role of VGLUT2 in the spinal cord during neuropathic pain, and its regulatory mechanism remain elusive. In this study, we report that spared nerve injury induced an upregulation of VGLUT2 in the spinal cord, and intrathecal administration of small hairpin RNAs (shRNA) against VGLUT2 before or after surgery attenuated mechanical allodynia, and pathologically-enhanced glutamate release. Meanwhile, nerve injury activated the Wnt1/ß-catenin signaling pathway in a quick-onset and sustained manner, and blocking the Wnt1 signaling with a Wnt1 targeting antibody attenuated neuropathic pain. In naïve mice, administration of a Wnt agonist or Wnt1 increased spinal VGLUT2 protein levels. Moreover, intrathecal administration of the Wnt/ß-catenin inhibitor, XAV939 attenuated mechanical allodynia, and this effect was concurrent with that of VGLUT2 downregulation. Pretreatment with VGLUT2 shRNAs abolished the allodynia induced by the Wnt agonist or Wnt1. These findings reveal a novel mechanism wherein there is Wnt1/ß-catenin-dependent VGLUT2 upregulation in neuropathic pain, thus potentiating the development of new therapeutic strategies in pain management.


Subject(s)
Neuralgia/physiopathology , Vesicular Glutamate Transport Protein 2/biosynthesis , Wnt Signaling Pathway/drug effects , Animals , Glutamic Acid/metabolism , Hyperalgesia/drug therapy , Immunohistochemistry , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Spinal Cord/drug effects , Spinal Cord/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Up-Regulation , beta Catenin/metabolism
2.
Eur J Pharmacol ; 828: 103-109, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29605418

ABSTRACT

Vesicular glutamate transporters (VGLUTs) transport glutamate into synaptic vesicles prior to exocytotic release. The expression pattern of VGLUT2 and studies of genetically modified mice have revealed that VGLUT2 contributes to neuropathic pain. We previously showed that VGLUT2 is upregulated in supraspinal regions including the thalamus in mice following spared nerve injury (SNI), and blocking VGLUTs using the VGLUT inhibitor CSB6B attenuated mechanical allodynia. To further evaluate the role of VGLUT2 in neuropathic pain, in this study, we developed a lentiviral vector expressing small hairpin RNAs (shRNAs) against mouse VGLUT2, which was injected into the ventral posterolateral (VPL) nucleus of the thalamus in the presence or absence of SNI. The administration of VGLUT2 shRNAs result in downregulation of VGLUT2 mRNA and protein expression, and decreased extracellular glutamate release in primary cultured neurons. We also showed that VGLUT2 shRNAs attenuated SNI-induced mechanical allodynia, in accordance with knockdown of VGLUT2 in the VPL nucleus in mice. Accordingly, our study supports the essential role of supraspinal VGLUT2 in neuropathic pain in adult mice and, thereby, validates VGLUT2 as a potential target for neuropathic pain therapy.


Subject(s)
Down-Regulation , Hyperalgesia/genetics , Neuralgia/genetics , Ventral Thalamic Nuclei/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Animals , Glutamic Acid/metabolism , Hyperalgesia/metabolism , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Neuralgia/metabolism , Neuralgia/pathology , Neurons/metabolism , RNA, Small Interfering/genetics , Ventral Thalamic Nuclei/pathology
3.
Acta Pharmacol Sin ; 38(7): 977-989, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28502978

ABSTRACT

Opioid analgesics remain the first choice for the treatment of moderate to severe pain, but they are also notorious for their respiratory depression and addictive effects. This study focused on the pharmacology of a novel opioid receptor mixed agonist DPI-125 and attempted to elucidate the relationship between the δ-, µ- and κ-receptor potency ratio and respiratory depression and abuse liability. Five diarylmethylpiperazine compounds (DPI-125, DPI-3290, DPI-130, KUST202 and KUST13T02) were selected for this study. PKA fluorescence redistribution assays in CHO cells individually expressing δ-, µ- or κ-receptors were used to measure the agonist potency. The respiratory safety profiles were estimated in rats by the ratio of ED50 (pCO2 increase)/ED50 (antinociception). The abuse liability of DPI-125 was evaluated with a self-administration model in rhesus monkeys. The observed agonist potencies of DPI-125 for δ-, µ- and κ-opioid receptors were 4.29±0.36, 11.10±3.04, and 16.57±4.14 nmol/L, respectively. The other four compounds were also mixed agonists with varying potencies. DPI-125 exhibited a high respiratory safety profile, clearly related to its high δ-receptor potency. The ratio of the EC50 potencies for the µ- and δ-receptors was found to be positively correlated with the respiratory safety ratio. DPI-125 has similar potencies for µ- and κ-receptors, which is likely the reason for its reduced abuse potential. Our results demonstrate that the opioid receptor mixed agonist DPI-125 is safer and less addictive than traditional µ-agonist analgesics. These findings suggest that the development of δ>µâˆ¼κ opioid receptor mixed agonists is feasible, and such compounds could represent a promising class of potent analgesics with wider therapeutic windows.


Subject(s)
Analgesia , Analgesics, Opioid/pharmacology , Pain/drug therapy , Piperazines/pharmacology , Respiratory Insufficiency/drug therapy , Thiophenes/pharmacology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Animals , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Humans , Male , Molecular Conformation , Pain Measurement , Piperazines/administration & dosage , Piperazines/chemistry , Rats , Rats, Wistar , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Structure-Activity Relationship , Thiophenes/administration & dosage , Thiophenes/chemistry
4.
Front Pharmacol ; 7: 242, 2016.
Article in English | MEDLINE | ID: mdl-27555820

ABSTRACT

Thienorphine (TNP) is a novel partial opioid agonist that has completed phase II clinical evaluation as a promising drug candidate for the treatment of opioid dependence. Previous studies have shown that TNP and its glucuronide conjugate (TNP-G) undergo significant bile excretion. The purpose of this study was to investigate the roles of efflux transporters in regulating biliary excretion and plasma exposure of TNP and TNP-G. An ATPase assay suggested that TNP and TNP-G were substrates of P-gp and MRP2, respectively. The in vitro data from rat hepatocytes showed that bile excretion of TNP and TNP-G was regulated by the P-gp and MRP2 modulators. The accumulation of TNP and TNP-G in HepG2 cells significantly increased by the treatment of mdr1a or MRP2 siRNA for P-gp or MRP2 modulation. In intact rats, the bile excretion, and pharmacokinetic profiles of TNP and TNP-G were remarkably changed with tariquidar and probenecid pretreatment, respectively. Tariquidar increased the Cmax and AUC0-t and decreased MRT and T1/2 of TNP, whereas probenecid decreased the plasma exposure of TNP-G and increased its T1/2. Knockdown P-gp and MRP2 function using siRNA significantly increased the plasma exposure of TNP and TNP-G and reduced their mean retention time in mice. These results indicated the important roles of P-gp and MRP2 in hepatobiliary excretion and plasma exposure of TNP and TNP-G. Inhibition of the efflux transporters may affect the pharmacokinetics of TNP and result in a drug-drug interaction between TNP and the concomitant transporter inhibitor or inducer in clinic.

5.
Neurochem Int ; 99: 9-15, 2016 10.
Article in English | MEDLINE | ID: mdl-27210824

ABSTRACT

Disturbance of glutamate homeostasis is a well-characterized mechanism of neuropathic pain. Vesicular glutamate transporters (VGLUTs) determine glutamate accumulation in synaptic vesicles and their roles in neuropathic pain have been suggested by gene-knockout studies. Here, we investigated the spatio-temporal changes in VGLUT expression during the development of neuropathic pain in wild-type rats. Spared nerve injury (SNI) induced mechanical allodynia from postoperative day 1 to at least day 14. Expression of VGLUT1 and VGLUT2 in dorsal root ganglia and spinal cord was examined by western blot analyses on different postoperative days. We observed that VGLUT2 were selectively upregulated in crude vesicle fractions from the ipsilateral lumbar enlargement on postoperative days 7 and 14, while VGLUT1 was transiently downregulated in ipsilateral DRG (day 4) and contralateral lumbar enlargement (day 1). Upregulation of VGLUT2 was not accompanied by alterations in vesicular expression of synaptotagmin or glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Thus, VGLUTs expression, especially VGLUT2, is regulated following peripheral nerve injury. Temporal regulation of VGLUT2 expression in spinal cord may represent a novel presynaptic mechanism contributing to injury-induced glutamate imbalance and associated neuropathic pain.


Subject(s)
Ganglia, Spinal/metabolism , Neuralgia/metabolism , Sciatic Neuropathy/metabolism , Spinal Cord/metabolism , Vesicular Glutamate Transport Protein 1/biosynthesis , Vesicular Glutamate Transport Protein 2/biosynthesis , Animals , Gene Expression , Male , Neuralgia/genetics , Peroneal Nerve/injuries , Peroneal Nerve/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/genetics , Sural Nerve/injuries , Sural Nerve/metabolism , Tibial Nerve/injuries , Tibial Nerve/metabolism , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 2/genetics
6.
Sci Rep ; 5: 16107, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26527075

ABSTRACT

Incarvillea sinensis is a Bignoniaceae plant used to treat rheumatism and relieve pain in traditional Chinese medicine. As a major component of I. sinensis, incarvillateine has shown analgesic activity in mice formalin tests. Using a series of animal models, this study further evaluated the effects of incarvillateine against acute, inflammatory, and neuropathic pain. Incarvillateine (10 or 20 mg/kg, i.p.) dose-dependently attenuated acetic acid-induced writhing, but did not affect thermal threshold in the hot plate test. In a Complete Freund's Adjuvant model, incarvillateine inhibited both thermal hyperalgesia and paw edema, and increased interleukin-1ß levels. Additionally, incarvillateine attenuated mechanical allodynia induced by spared nerve injury or paclitaxel, whereas normal mechanical sensation was not affected. Incarvillateine did not affect locomotor activity and time on the rotarod at analgesic doses, and no tolerance was observed after 7 consecutive daily doses. Moreover, incarvillateine-induced antinociception was attenuated by theophylline, 1,3-dipropyl-8-cyclopentylxanthine, and 3,7-dimethyl-1-propargylxanthine, but not naloxone, indicating that the effects of incarvillateine on chronic pain were related to the adenosine system, but not opioid system. These results indicate that incarvillateine is a novel analgesic compound that is effective against inflammatory and neuropathic pain, and that its effects are associated with activation of the adenosine system.


Subject(s)
Adenosine/metabolism , Alkaloids/pharmacology , Analgesics/pharmacology , Bignoniaceae/chemistry , Monoterpenes/pharmacology , Alkaloids/chemistry , Alkaloids/therapeutic use , Analgesics/chemistry , Analgesics/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/toxicity , Bignoniaceae/metabolism , Disease Models, Animal , Edema/chemically induced , Edema/prevention & control , Freund's Adjuvant/chemistry , Hyperalgesia/etiology , Hyperalgesia/prevention & control , Interleukin-1beta/metabolism , Medicine, Chinese Traditional , Mice , Monoterpenes/chemistry , Monoterpenes/therapeutic use , Motor Activity/drug effects , Paclitaxel/toxicity , Pain Measurement/drug effects , Theobromine/analogs & derivatives , Theobromine/pharmacology , Theophylline/pharmacology , Xanthines/pharmacology
7.
Brain Res ; 1624: 515-524, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26300222

ABSTRACT

Vesicular glutamate transporters (VGLUTs) control the storage and release of glutamate, which plays a critical role in pain processing. The VGLUT2 isoform has been found to be densely distributed in the nociceptive pathways in supraspinal regions, and VGLUT2-deficient mice exhibit an attenuation of neuropathic pain; these results suggest a possible involvement of VGLUT2 in neuropathic pain. To further examine this, we investigated the temporal changes in VGLUT2 expression in different brain regions as well as changes in glutamate release from thalamic synaptosomes in spared nerve injury (SNI) mice. We also investigated the effects of a VGLUT inhibitor, Chicago Sky Blue 6B (CSB6B), on pain behavior, c-Fos expression, and depolarization-evoked glutamate release in SNI mice. Our results showed a significant elevation of VGLUT2 expression up to postoperative day 1 in the thalamus, periaqueductal gray, and amygdala, followed by a return to control levels. Consistent with the changes in VGLUT2 expression, SNI enhanced depolarization-induced glutamate release from thalamic synaptosomes, while CSB6B treatment produced a concentration-dependent inhibition of glutamate release. Moreover, intracerebroventricular administration of CSB6B, at a dose that did not affect motor function, attenuated mechanical allodynia and c-Fos up-regulation in pain-related brain areas during the early stages of neuropathic pain development. These results demonstrate that changes in the expression of supraspinal VGLUT2 may be a new mechanism relevant to the induction of neuropathic pain after nerve injury that acts through an aggravation of glutamate imbalance.


Subject(s)
Brain/metabolism , Brain/pathology , Neuralgia/pathology , Vesicular Glutamate Transport Protein 2/metabolism , Animals , Coloring Agents/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glutamic Acid/metabolism , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neuralgia/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Statistics, Nonparametric , Synaptosomes/metabolism , Synaptosomes/pathology , Tibial Nerve/injuries , Tibial Nerve/physiopathology , Trypan Blue/pharmacology , Vesicular Glutamate Transport Protein 2/antagonists & inhibitors
8.
J Psychiatr Res ; 59: 161-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25193460

ABSTRACT

Improved utilization of continuous or intermittent opioid administration in pain treatment necessitates a comparison of the antinociceptive effect and tolerance of these two treatment methods. More importantly, the effect of treatment method on subsequent opioid consumption has not been directly compared, although it is widely assumed that continuous opioid treatment may produce lower addictive liability relative to intermittent opioid treatment. In this study, we compared the antinociceptive effect and tolerance of morphine in rats that received repeated injection (10 mg/kg twice daily for 7 days) or continuous infusion (20 mg/kg daily for 7 days) subcutaneously and the self-administration of intravenous morphine in these rats after 7 days of withdrawal. Both intermittent and continuous morphine treatment produced antinociceptive tolerance, but the exhibition of tolerance differed. Moreover, intermittent morphine pretreatment facilitated subsequent morphine self-administration, whereas continuous morphine pretreatment produced minimal effects, as shown by comparable levels of active responses and morphine consumption between continuous morphine and saline-treated rats. These results suggest that the administration method of opioid should be selected according to the specific pain situation and that continuous opioid administration or long-acting therapy may be advantageous, producing less influence on drug-taking behavior than intermittent administration of short-acting drugs.


Subject(s)
Analgesics, Opioid/administration & dosage , Morphine/administration & dosage , Pain Measurement/drug effects , Pain/drug therapy , Administration, Intravenous , Analysis of Variance , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Delivery Systems , Drug Tolerance , Hot Temperature/adverse effects , Male , Pain/etiology , Rats , Rats, Sprague-Dawley , Self Administration , Time Factors
9.
Dig Dis Sci ; 59(10): 2417-22, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24801687

ABSTRACT

BACKGROUND AND AIMS: Ilaprazole is a novel proton pump inhibitor that has been marketed as an oral therapy for acid-related diseases in China and Korea. This study aimed to compare the gastroprotective effects of intravenous and enteral ilaprazole in rat models. METHODS: The rats were divided into 7-8 groups receiving vehicle, esomeprazole, and different doses of intravenous and enteral ilaprazole. The rats were then exposed to indomethacin (30 mg/kg, i.g.), or water-immersion stress and gastric lesions were examined. The effects of different treatments on histamine (10 µmol/kg/h)-induced acid secretion were also observed. RESULTS: Intravenous ilaprazole exhibited high antiulcer activity in a dose-dependent manner. Ilaprazole at a dose of 3 mg/kg decreased ulcer number and index to the same extent as 20 mg/kg esomeprazole. Moreover, the potency of intravenous ilaprazole is superior to that of intragastric ilaprazole. In anesthetized rats, the inhibitory effect of intravenous ilaprazole on histamine-induced acid secretion is faster and longer-lasting than that of intraduodenal ilaprazole. CONCLUSION: Intravenous ilaprazole is more potent than oral ilaprazole against indomethacin- or stress-induced gastric lesions, with faster and longer inhibition of acid secretion.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/administration & dosage , 2-Pyridinylmethylsulfinylbenzimidazoles/therapeutic use , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/therapeutic use , Stomach Ulcer/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Histamine/pharmacology , Indomethacin/toxicity , Male , Rats , Rats, Sprague-Dawley , Stomach/drug effects , Stomach/pathology , Stress, Physiological
10.
CNS Neurosci Ther ; 20(3): 282-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24330593

ABSTRACT

AIMS: It is considered that a long-acting therapy would be advantageous in the treatment of addiction. In a search for novel buprenorphine analogues, thienorphine was demonstrated to be an extremely long-acting orally active partial opioid agonist. This study explored the mechanisms underlying the long-lasting effects of thienorphine. METHODS: The binding kinetics of [(3) H]thienorphine were measured in membrane preparations expressing cloned rat opioid receptors. Flow cytometric analysis was used to determine the effect of thienorphine on the surface opioid receptor number. The long-lasting effects of thienorphine were also confirmed at the tissue level and in vivo. RESULTS: At 37°C, [(3) H]thienorphine showed rapid association with µ- and κ-opioid receptors, while its dissociation was sluggish and biphasic (K-1 = 0.21 min(-1) , K-2 = 0.0078 min(-1) for the µ-receptor; K-1 = 0.17 min(-1) , K-2 = 0.0042 min(-1) for the κ-receptor). Treatment with thienorphine for 24, 48, and 72 h downregulated surface µ-receptor in a dose- and time-dependent manner. The inhibitory effect of thienorphine on guinea pig ileum persisted for more than 120 min after prolonged washing. In vivo, thienorphine exhibited significant antagonism of morphine-induced antinociception for more than 7 days. CONCLUSIONS: These results indicate that multiple factors, including persistent receptor occupation and enhanced receptor downregulation, may contribute to the long-lasting effects of thienorphine that would be beneficial for its application in addiction treatment.


Subject(s)
Buprenorphine/analogs & derivatives , Morphine Dependence/drug therapy , Narcotic Antagonists/therapeutic use , Acetylcholine/pharmacology , Animals , Buprenorphine/pharmacokinetics , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Cell Line, Transformed , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Guinea Pigs , Ileum/drug effects , Ileum/physiology , Male , Mice , Mice, Inbred Strains , Muscle Contraction/drug effects , Narcotic Antagonists/pharmacokinetics , Protein Binding/drug effects , Rats , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Time Factors , Tritium/pharmacokinetics
11.
J Recept Signal Transduct Res ; 34(1): 44-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24219160

ABSTRACT

CONTEXT: Our previous study demonstrated that acute and repeated morphine treatment differentially regulated κ-opioid receptor mRNA levels in the rat mesocorticolimbic system. Here, we further investigated the effects of morphine on protein levels of κ-opioid receptor in this reward-related circuitry. METHODS: Three groups of rats received intraperitoneal injection of saline, acute morphine (8.0 mg/kg) and repeated morphine (8.0 mg/kg, once daily for 5 consecutive days) and the κ-receptor protein expression was examined by Western blot analysis. RESULTS: We found that acute morphine treatment did not affect the κ-receptor protein levels in medial prefrontal cortex (mPFC), nucleus accumbens (NAc) and ventral tegmental area (VTA). However, repeated morphine treatment downregulated the κ-receptor protein levels in mPFC and VTA, while there was no significant change in NAc. CONCLUSIONS: These results, along with those reported previously, suggested that morphine dependence may be associated with regionally specific changes in κ-opioid receptor expression in mesocorticolimbic system.


Subject(s)
Morphine/administration & dosage , RNA, Messenger/drug effects , Receptors, Opioid, kappa/biosynthesis , Animals , Gene Expression Regulation/drug effects , Humans , Prefrontal Cortex/drug effects , Rats , Ventral Tegmental Area/drug effects
12.
Yao Xue Xue Bao ; 48(4): 484-8, 2013 Apr.
Article in Chinese | MEDLINE | ID: mdl-23833933

ABSTRACT

Schizophrenia, described as the worst disease affecting mankind, is a severe and disabling mental disorder. Schizophrenia is characterized by complicated symptoms and still lacks a diagnostic neuropathology, so developing schizophrenia animal models which have quantifiable measures tested in a similar fashion in both humans and animals will play a key role in new therapeutic approaches. According to the symptoms of cognitive impairment and emotional disorder, the N-methyl-d-aspartate (NMDA)-receptor antagonist MK-801 was applied to induce schizophrenia-like behavior in mice. Locomotor activity and prepulse inhibition (PPI) were selected as indices and the effect of clozapine was also investigated in this model. The results showed that compared with the normal group, MK-801-treated mice exhibited significantly increased locomotor activity and impaired PPI, and pre-exposure to clozapine could ameliorate the abnormality and make it back to normal level. These findings suggest that the model we established could be a useful tool for antipsychotic drug screening.


Subject(s)
Clozapine/pharmacology , Disease Models, Animal , Inhibition, Psychological , Motor Activity/drug effects , Schizophrenia/chemically induced , Animals , Antipsychotic Agents/pharmacology , Dizocilpine Maleate , Male , Mice , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Schizophrenia/physiopathology
13.
World J Gastroenterol ; 19(9): 1444-50, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23539497

ABSTRACT

AIM: To evaluate the effect of thienorphine on small intestinal transit in vivo and on guinea-pig ileum (GPI) contraction in vitro. METHODS: The effects of thienorphine on intestinal transit were examined in mice and in isolated GPI. Buprenorphine and morphine served as controls. The distance traveled by the head of the charchol and the total length of the intestine were measured in vivo. Gastrointestinal transit was expressed as a percentage of the distance traveled by the head of the marker relative to the total length of the small intestine. The isolated GPI preparations were connected to an isotonic force transducer and equilibrated for at least 1 h before exposure to drugs. Acetylcholine was used for muscle stimulation. RESULTS: Thienorphine (0.005-1.0 mg/kg, ig) or buprenorphine (0.005-1.0 mg/kg, sc) dose-dependently significantly inhibited gut transit compared with saline. Thienorphine inhibited gut transit less than buprenorphine. The maximum inhibition by thienorphine on the intestinal transit was 50%-60%, whereas the maximum inhibition by morphine on gut transit was about 100%. Thienorphine also exhibited less inhibition on acetylcholine-induced contraction of GPI, with a maximum inhibition of 65%, compared with 93% inhibition by buprenorphine and 100% inhibition by morphine. Thienorphine induced a concentration-dependent decrease in the basal tonus of spontaneous movement of the GPI, the effect of which was weaker than that with buprenorphine. The duration of the effect of thienorphine on the GPI was longer than that with buprenorphine. CONCLUSION: Thienorphine had less influence, but a longer duration of action on GPI contraction and moderately inhibited intestinal transit.


Subject(s)
Analgesics, Opioid/pharmacology , Buprenorphine/analogs & derivatives , Gastrointestinal Transit/drug effects , Ileum/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Animals , Buprenorphine/pharmacology , Cholinergic Agonists/pharmacology , Dose-Response Relationship, Drug , Guinea Pigs , Male , Mice , Morphine/pharmacology , Narcotic Antagonists/pharmacology , Time Factors
14.
J Pain Res ; 5: 491-502, 2012.
Article in English | MEDLINE | ID: mdl-23166448

ABSTRACT

The discovery of the tetrodotoxin-resistant (TTX-R) Na(+) channel in nociceptive neurons has provided a special target for analgesic intervention. In a previous study we found that both morphine tolerance and persistent visceral inflammation resulted in visceral hyperalgesia. It has also been suggested that hyperexcitability of sensory neurons due to altered TTX-R Na(+) channel properties and expression contributes to hyperalgesia; however, we do not know if some TTX-R Na(+) channel property changes can be triggered by visceral hyperalgesia and morphine tolerance, or whether there are similar molecular or channel mechanisms in both situations. To evaluate the effects of morphine tolerance and visceral inflammation on the channel, we investigated the dorsal root ganglia (DRG) neuronal change following these chronic treatments. Using whole-cell patch clamp recording, we recorded TTX-R Na(+) currents in isolated adult rat lumbar and sacral (L6-S2) DRG neurons from normal and pathologic rats with colon inflammatory pain or chronic morphine treatment. We found that the amplitudes of TTX-R Na(+) currents were significantly increased in small-diameter DRG neurons with either morphine tolerance or visceral inflammatory pain. Meanwhile, the result also showed that those treatments altered the kinetics properties of the electrical current (ie, the activating and inactivating speed of the channel was accelerated). Our current results suggested that in both models, visceral chronic inflammatory pain and morphine tolerance causes electrophysiological changes in voltage-gated Na channels due to the chronic administration of these medications. For the first time, the present investigation explored the adaptations of this channel, which may contribute to the hyperexcitability of primary afferent nerves and hyperalgesia during these pathologic conditions. The results also suggest that neurophysiologic mechanisms of morphine tolerance and visceral hyperalgesia are related at the TTX-R Na(+) channel.

15.
Pharmacol Rep ; 64(2): 445-8, 2012.
Article in English | MEDLINE | ID: mdl-22661197

ABSTRACT

BACKGROUND: Morphine induces adaptive changes in gene expression throughout the reward circuitry of the brain. Here, we investigated the acute and chronic effects of morphine on mRNA levels of κ-opioid receptor in the rat mesocorticolimbic system. METHODS: Three groups of rats received ip injection of saline, acute morphine (8.0 mg/kg) and repeated morphine (8.0 mg/kg, once daily for 5 consecutive days) and the κ-receptor mRNA expression was examined using real-time quantitative PCR method. RESULTS: We found that κ-receptor mRNA in medial prefrontal cortex (mPFC) increased after acute and repeated morphine treatment. However, the mRNA levels in nucleus accumbens (NAc) and ventral tegmental area (VTA) were upregulated after acute morphine treatment and returned to basal levels after repeated morphine treatment. CONCLUSIONS: These results suggest that morphine dependence was associated with regionally specific changes in mRNA levels of κ-opioid receptor in mesocorticolimbic system.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , RNA, Messenger/biosynthesis , Receptors, Opioid, kappa/biosynthesis , Ventral Tegmental Area/drug effects , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Morphine/administration & dosage , Morphine Dependence/metabolism , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Up-Regulation , Ventral Tegmental Area/metabolism
16.
Yao Xue Xue Bao ; 46(9): 1078-83, 2011 Sep.
Article in Chinese | MEDLINE | ID: mdl-22121778

ABSTRACT

This study investigates whether kappa-opioid receptor and ORL1 receptor may interact to form a heterodimer. In immunofluorescence and co-immunoprecipitation experiments, differentially epitope-tagged receptors, colocalization and heterodimerization of kappa-opioid receptor and ORL1 receptor were used and examined in primary culturing rat neurons, Chinese hamster ovary (CHO) or human embryonic kidney 293 (HEK293) cells. The results show that fluorescence of both kappa-opioid receptor and ORL1 receptor were overlapping in primary culturing hippocampal and cortical neurons. Similarly in co-expressing CHO or HEK293 cells, HA-KOR and Myc-ORL1 were almost exclusively confined to the membranes, revealing extensive colocalization. When Flag-KOR and Myc-ORL1 were co-expressing in CHO cells, heterodimerization was identified to have the ability to co-immunoprecipitate ORL1-receptors with kappa-opioid receptor and vice versa. In the current study, further evidence was provided for the direct interaction of two subtypes of opioid receptors, kappa-opioid receptor and ORL1-receptor, to form the heterodimerization. The finding represents the novel pharmacological mechanism for modulation of opioid receptor function as well as diversity of G protein-coupled receptors.


Subject(s)
Dimerization , Receptors, Opioid, kappa/metabolism , Receptors, Opioid/metabolism , Animals , CHO Cells , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cricetinae , Cricetulus , Female , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Immunoprecipitation , Male , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Nociceptin Receptor
17.
Acta Pharmacol Sin ; 32(10): 1215-24, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21863064

ABSTRACT

AIM: To investigate possible pharmacological mechanisms underlying the antinociceptive effect of and tolerance to N-methyl-7α-[(R)-1-hydroxy-1-methyl-3-(thien-3-yl)-propyl]-6,14-endo-ethanotetrahydronororipavine (030418), a derivative of thienorphine. METHODS: The binding affinity and efficacy of 030418 were determined using receptor binding and guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPγS) assays in CHO-µ, CHO-κ, CHO-δ, and CHO-ORL1 cell membranes. The analgesic activity of and tolerance to 030418 were evaluated in thermal nociceptive tests in mice. The effects of 030418 on opioid receptors were further investigated using in vivo pharmacological antagonist blockade and in vitro tissue preparations. RESULTS: The compound 030418 displayed high binding affinity to all subtypes of opioid receptors with K(i) values in the nanomolar range. In [(35)S]GTPγS binding assay, the maximal stimulation of 030418 to µ-, κ-, δ-receptors and the ORL1 receptor was 89%, 86%, 67% and 91%, respectively. In hot-plate test, the antinociceptive effect of 030418 was more potent and longer than morphine. The nonselective opioid receptor antagonist naloxone could completely block 030418-induced antinociception, while both the µ-opioid receptor antagonist ß-FNA and the κ-opioid receptor antagonist nor-BNI attenuated 030418-induced antinociception. In contrast, the ORL1 receptor antagonist J-113397 enhanced the antinociceptive effect of 030418. Additionally, chronic treatment with 030418 resulted in a dramatic development of tolerance that could not be effectively prevented by J-113397. In guinea pig ileum preparation, the existing action of 030418 could be removed with difficulty after prolonged washing. CONCLUSION: The compound 030418 is a novel agonist of opioid receptors with high efficiency, long-lasting effect and liability to tolerance, which may be closely correlated with the methyl group at the N(17) position and the high hydrophobicity of the C(7)-thiophene group in its chemical structure.


Subject(s)
Analgesics/chemistry , Analgesics/therapeutic use , Pain Measurement/drug effects , Thebaine/analogs & derivatives , Analgesics/pharmacology , Animals , Buprenorphine/analogs & derivatives , Buprenorphine/chemistry , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , CHO Cells , Cricetinae , Drug Tolerance , Female , Guinea Pigs , Male , Mice , Nociception/drug effects , Receptors, Opioid/metabolism , Thebaine/chemistry , Thebaine/pharmacology , Thebaine/therapeutic use
18.
Naunyn Schmiedebergs Arch Pharmacol ; 384(2): 125-31, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21594658

ABSTRACT

Numerous efforts have been made on the chemical modification of opioid compounds, with the ultimate goal of developing new opioid analgesics that is highly potent and low/non-addictive. In a search for such compounds, TH-030418 [7α-[(R)-1-hydroxy-1-methyl-3-(thien-3-yl)-propyl]-6,14-endo-ethanotetrahydrooripavine] was synthesized. Here, we evaluated the pharmacological activities of TH-030418, in comparison with morphine, the prototype opioid analgesic. In radioligand binding assays, TH-030418 bound potently and nonselectively to µ-, δ-, κ-, and ORL1 (opioid receptor-like 1) receptors stably expressed in CHO (Chinese hamster ovary) cells with K (i) values of 0.56, 0.73, 0.60, and 1.55 nM, respectively. When administered subcutaneously, TH-030418 was much more potent than morphine in analgesia, with the ED(50) values of 1.37 µg/kg and 1.70 µg/kg in hot plate and acetic acid writhing tests, respectively. The opioid antagonist naloxone blocked the antinociceptive effect of TH-030418, indicating that the action of TH-030418 was mediated by opioid receptors. The antinociceptive effect of s.c. TH-030418 in hot plate test lasted for more than 12 h, which is much longer than those of morphine (2.5 h) and dihydroetorphine (1.5 h). In addition, naloxone did not precipitate withdrawal syndrome in the mice treated with TH-030418 previously. Most importantly, TH-030418 did not induce conditioned place preference in mice after chronic treatment. These results indicate that TH-030418 is a potent long-acting opioid analgesic with low dependence liability and may be of some value in the development of new analgesics.


Subject(s)
Analgesics, Opioid/pharmacology , Etorphine/analogs & derivatives , Opioid-Related Disorders/etiology , Receptors, Opioid/metabolism , Analgesics, Opioid/adverse effects , Analgesics, Opioid/therapeutic use , Animals , Behavior, Animal/drug effects , CHO Cells , Cell Culture Techniques , Conditioning, Classical , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Etorphine/adverse effects , Etorphine/pharmacology , Etorphine/therapeutic use , Female , Injections, Subcutaneous , Ligands , Male , Mice , Mice, Inbred Strains , Molecular Structure , Pain/drug therapy , Protein Binding , Radioligand Assay , Receptors, Opioid/genetics , Time Factors , Transfection , Nociceptin Receptor
19.
J Cardiovasc Pharmacol ; 56(3): 246-54, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20531217

ABSTRACT

Pulmonary hypertension is a kind of disease associated with a very high rate of mortality, and there are not many effective drugs for the treatment. Today, endothelin (ET)-1 receptor antagonists were proved to be effective in the treatment of pulmonary hypertension. Aiming at developing new endothelin-A receptor (ETA) antagonist for treatment of pulmonary hypertension, di-n-butylaminocarbamyl-L-leucyl-D-tryptophanyl-D-4-chloro-Phe, named GF063, was synthesized at base of selective ETA receptor antagonist BQ485 and selected for the further pharmacological characterization. The preliminary pharmacodynamics of GF063 was evaluated by radioligand receptor binding assay and test of antivasoconstriction effects in vitro and in vivo. The integrative pharmacodynamics was evaluated in hypoxia-induced rat pulmonary hypertension. In vitro, GF063 bound to ETA receptor with 100,000-fold higher affinity than to ETB receptor. GF063 concentration dependently inhibited contraction of isolated rat aortic ring induced by ET-1 and shifted the cumulative concentration-contraction response curve to right with no change in the maximal response. In vivo, GF063 inhibited the increase of mean systemic arterial pressure induced by ET-1 in anesthetized rat. In hypoxia-induced rat pulmonary hypertension model, pretreatment with GF063 (40 mg/kg, s.c.) significantly decreased pulmonary artery pressure and right ventricular hypertrophy, also significantly inhibited the increase of ET-1 level in lung, improved hemodynamics, and alleviated the wall thickness of pulmonary vessels. This study indicated that GF063, as a selective ETA receptor antagonist, could inhibit vasoconstriction effects in vivo and in vitro, could prevent pulmonary hypertension induced by hypoxia, and may have great potential to be developed as a new drug of antipulmonary hypertension.


Subject(s)
Blood Pressure/drug effects , Endothelin A Receptor Antagonists , Endothelin-1/antagonists & inhibitors , Hypoxia/physiopathology , Oligopeptides/pharmacology , Pulmonary Artery/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Dose-Response Relationship, Drug , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , In Vitro Techniques , Lung/blood supply , Lung/drug effects , Lung/pathology , Male , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Wistar , Vasoconstriction/drug effects
20.
Eur J Pharmacol ; 636(1-3): 73-81, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20361956

ABSTRACT

SO-3, a novel Omega-superfamily conotoxin derived from Conus striatus, selectively inhibits N-type neuronal voltage-sensitive calcium channels. In current study, antinociception of SO-3 compared with MVIIA or morphine and its effects on morphine analgesia were investigated in rodent chemical stimulus tests after acute or repeated intrathecal administration. In mice acetic acid writhing test, similar to MVIIA, SO-3 caused dose- and time-dependent spinal antinociception with ED(50) of 0.25 microg/kg and t(1/2) of 4h, which was more potent and longer-acting than morphine. In rat formalin test after intrathecal bolus injection, SO-3 produced dose- and time-dependent antinociception by suppressing acute (ED(50), 1.79 microg/kg) and tonic phases (ED(50), 0.41 microg/kg), which was similar to MVIIA and approximately 10-fold potency and twice longer-acting of morphine in blocking tonic phase responses. After repeated intrathecal injections twice daily for 5 consecutive days, SO-3 produced analgesia without loss of potency whereas morphine produced analgesia tolerance in rat formalin test; further, SO-3 still produced potent analgesia in morphine-tolerant rats. SO-3 co-administered with morphine left-shift the dose-response curve of morphine in mice acetic acid writhing test and significantly potentiated morphine analgesia in rat formalin test. No changes in motor function were seen in mice or rats receiving antinociceptive doses of SO-3 whereas MVIIA caused motor dysfunction at doses of 1.0-2.0 microg/kg in rats. This study showed that (1) novel SO-3 produced potent and long-acting spinal antinociception without observable motor dysfunction, (2) SO-3 significantly potentiated morphine analgesia, (3) After repeated intrathecal administration, SO-3 produced neither tolerance nor cross-tolerance to morphine analgesia.


Subject(s)
Calcium Channels, N-Type/metabolism , Morphine/pharmacology , Neurons/drug effects , omega-Conotoxins/administration & dosage , omega-Conotoxins/pharmacology , Acetic Acid/toxicity , Analgesics/administration & dosage , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Drug Interactions , Formaldehyde/toxicity , Injections, Spinal , Locomotion/drug effects , Male , Mice , Neurons/metabolism , Pain/chemically induced , Pain/drug therapy , Pain/physiopathology , Rats , omega-Conotoxins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...