Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Mol Pharmacol ; 100(2): 170-180, 2021 08.
Article in English | MEDLINE | ID: mdl-34031190

ABSTRACT

Opioids play an important role in pain relief, but repeated exposure results in tolerance and dependence. To make opioids more effective and useful, research in the field has focused on reducing the tolerance and dependence for chronic pain relief. Here, we showed the effect of A20-binding inhibitor of nuclear factor-κB (ABIN-1) in modulating morphine function. We used hot-plate tests and conditioned place preference (CPP) tests to show that overexpression of ABIN-1 in the mouse brain attenuated morphine dependence. These effects of ABIN-1 are most likely mediated through the formation of ABIN-1-ß-arrestin2 complexes, which accelerate ß-arrestin2 degradation by ubiquitination. With the degradation of ß-arrestin2, ABIN-1 overexpression also decreased µ opioid receptor (MOR) phosphorylation and internalization after opioid treatment, affecting the ß-arrestin2-dependent signaling pathway to regulate morphine tolerance. Importantly, the effect of ABIN-1 on morphine tolerance was abolished in ß-arrestin2-knockout mice. Taken together, these results suggest that the interaction between ABIN-1 and ß-arrestin2 inhibits MOR internalization to attenuate morphine tolerance, revealing a novel mechanism for MOR regulation. Hence, ABIN-1 may be a therapeutic target to regulate MOR internalization, thus providing a foundation for a novel treatment strategy for alleviating morphine tolerance and dependence. SIGNIFICANCE STATEMENT: A20-binding inhibitor of nuclear factor-κB (ABIN-1) overexpression in the mouse brain attenuated morphine tolerance and dependence. The likely mechanism for this finding is that ABIN-1-ß-arrestin2 complex formation facilitated ß-arrestin2 degradation by ubiquitination. ABIN-1 targeted ß-arrestin2 to regulate morphine tolerance. Therefore, the enhancement of ABIN-1 is an important strategy to prevent morphine tolerance and dependence.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Brain/metabolism , Opioid-Related Disorders/genetics , Receptors, Opioid, mu/metabolism , beta-Arrestin 2/metabolism , Animals , Animals, Genetically Modified , CHO Cells , Cells , Cricetulus , Disease Models, Animal , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Mice , Opioid-Related Disorders/metabolism , Phosphorylation , Proteolysis , Ubiquitination , beta-Arrestin 2/chemistry , beta-Arrestin 2/genetics
2.
Brain Res ; 1748: 147083, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32871137

ABSTRACT

Thienorphine hydrochloride is a new anti-relapse drug for opioid abusers that is currently in phase II clinical trial. In the present study, the antinociception, dependence, and signal transduction induced by thienorphine were examined. Thienorphine showed a potent antinociception effect in acetic acid-induced writhing test and formalin test. In the hot plate test and tail-flick test, thienorphine presented the typical partial opioid agonist character with a ceiling dose-response curve in addition to a bell-shaped curve. The hot plate test revealed that thienorphine induced approximately 50% of antinociception in µ receptor knockout (µ-KO) mice compared to wild-type controls (P < 0.05). The κ, δ selective antagonist nor-binaltorphimine (nor-BNI), and naltrindole decreased approximately 50-60% of theinorphine antinociception in µ-KO mice, respectively. The ORL1 receptor-selective antagonist J113397 did not affect theinorphine antinociception in µ-KO mice. Chronic treatment with thienorphine (1.5 mg/kg) induced some tolerance that was lower compared to buprenorphine or morphine addition. In contrast to buprenorphine or morphine, thienorphine did not lead to psychological dependence by conditioned place preference (CPP). The maximum inhibition of thienorphine on protein kinase A (PKA) activity was about 36%, 100%, 100%, and 12% in CHO-µ/κ/δ/ORL1-PKAcatEGFP cells, respectively. Similar results were observed in cyclic adenosine monophosphate (cAMP) accumulation inhibited by thienorphine in cells. Thienorphine significantly increased pERK1/2 in CHO-κ/δ-PKAcatEGFP cells. These results indicated that thienorphine induced analgesia through activation of κ- and δ-, partial activation of µ- opioid receptor without a bias between G-protein- and ß-arrestin-mediated pathways. Thienorphine might be used for antinociception with minimal adverse effects.


Subject(s)
Analgesics, Opioid/therapeutic use , Brain/drug effects , Buprenorphine/analogs & derivatives , Nociception/drug effects , Pain/drug therapy , Analgesics, Opioid/pharmacology , Animals , Brain/metabolism , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Cell Line, Tumor , Cyclic AMP/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Pain/genetics , Pain/metabolism , Rats , Rats, Wistar , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism
3.
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
4.
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
5.
Mol Pharmacol ; 93(2): 36-48, 2018 02.
Article in English | MEDLINE | ID: mdl-29237725

ABSTRACT

The µ-opioid receptor (MOR) is a Gi/o protein-coupled receptor that mediates analgesic, euphoric, and reward effects. Using a bacterial two-hybrid screen, we reported that the carboxyl tail of the rat MOR associates with A20-binding inhibitor of nuclear factor κB (ABIN-1). This interaction was confirmed by direct protein-protein binding and coimmunoprecipitation of MOR and ABIN-1 proteins in cell lysates. Saturation binding studies showed that ABIN-1 had no effect on MOR binding. However, the interaction of ABIN-1 and MOR inhibited the activation of G proteins induced by DAMGO ([d-Ala2,N-Me-Phe4,Gly5-ol]-Enkephalin). MOR phosphorylation, ubiquitination, and internalization induced by DAMGO were decreased in Chinese hamster ovary cells that coexpressed MOR and ABIN-1. The suppression of forskolin-stimulated adenylyl cyclase by DAMGO was also inhibited by the interaction of ABIN-1 with MOR. In addition, extracellular signal-regulated kinase activation was also negatively regulated by overexpression of ABIN-1. These data suggest that ABIN-1 is a negative coregulator of MOR activation, phosphorylation, and internalization in vitro. ABIN-1 also inhibited morphine-induced hyperlocomotion in zebrafish larvae (AB strain). By utilization of an antisense morpholino oligonucleotide (MO) gene knockdown technology, the ABIN-1 MO-injected zebrafish larvae showed a significant increase (approximately 60%) in distance moved compared with control MO-injected larvae after acute morphine treatment (P < 0.01). Taken together, ABIN-1 negatively regulates MOR function in vitro and in vivo.


Subject(s)
DNA-Binding Proteins/physiology , Receptors, Opioid, mu/physiology , Analgesics, Opioid/pharmacology , Animals , Binding Sites , CHO Cells , Cell Line, Tumor , Cricetulus , Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Endocytosis , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Larva , Ligands , Luminescent Proteins , Phosphorylation , Rats , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Signal Transduction , Sulfur Radioisotopes/metabolism , Ubiquitination , Zebrafish/growth & development
6.
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
7.
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.

8.
Eur J Pharmacol ; 789: 144-151, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27445237

ABSTRACT

Opioid analgesics are widely used in moderate to severe pain including renal colic. Morphine is believed to cause spasm of ureter and affect the bladder contractions. Thienorphine is a partial opioid agonist that is a good candidate for the treatment of opioid dependence and pain. This study examined the effects of thienorphine on the guinea pig isolated ureter and bladder. The contractile amplitude of isolated ureter induced by KCl (40mM) was not influenced by thienorphine or buprenorphine, whereas morphine increased the amplitude of the isolated ureter. Thienorphine, buprenorphine or naloxone concentration-dependently antagonized the isolated ureter contraction induced by morphine. Thienorphine (1.0-32.0µM) or buprenorphine (1.0-32.0µM) had no effects on the spontaneous or acetylcholine (Ach) induced contractions of isolated bladder, but decreased the amplitude of the contractions of isolated bladder at 100µM concentration. Morphine (0.1-3.2mM) concentration dependently increased the spontaneous movement and Ach (1µM) induced contractions of isolated bladder. The mRNA levels of µ receptor in the ureter and bladder was as the same as that in the frontal cortex. In comparison, the mRNA levels of κ receptor, δ receptor and N/OFQ receptor was fewer than that in the frontal cortex. In summary, thienorphine has little influence on the guinea pig isolated ureter and bladder compared with morphine, which may result in a lack of adverse renal colic effects.


Subject(s)
Analgesics/pharmacology , Buprenorphine/analogs & derivatives , Muscle Contraction/drug effects , Ureter/drug effects , Ureter/physiology , Urinary Bladder/drug effects , Urinary Bladder/physiology , Animals , Buprenorphine/pharmacology , Female , Gene Expression Regulation/drug effects , Guinea Pigs , Male , Potassium Chloride/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Opioid/genetics , Ureter/metabolism , Urinary Bladder/metabolism , Nociceptin Receptor
9.
Bioorg Med Chem Lett ; 26(15): 3598-602, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27311893

ABSTRACT

Novel water-soluble paclitaxel prodrugs were designed and synthesized by introducing morpholino groups through different linkers. These derivatives showed 400-20,000-times greater water solubility than paclitaxel as well as comparable activity in MCF-7 and HeLa cell lines. The prodrug PM4 was tested in the S-180 tumor mouse model, with paclitaxel as the positive control. The results showed that PM4 had comparable antitumor activity as paclitaxel, with tumor inhibition of 54% versus 56%, and remarkably decreased toxicity. The survival rate of treated mice was 8/8 in the PM4 group, compared to 3/8 in the paclitaxel group.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Design , Morpholinos/pharmacology , Paclitaxel/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HeLa Cells , Humans , MCF-7 Cells , Mice , Molecular Structure , Morpholinos/chemistry , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Paclitaxel/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Solubility , Structure-Activity Relationship , Water/chemistry
10.
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
11.
J Med Chem ; 59(8): 3953-63, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27022999

ABSTRACT

(-)-Incarvillateine (INCA) is the major antinociceptive component of Incarvillea sinensis, which has been used to treat rheumatism and relieve pain in traditional Chinese medicine. We have developed a concise and general synthetic approach for INCA, which enabled gram-scale asymmetric syntheses of (-)-INCA, (-)-incarvilline, (-)-isoincarvilline, and six other INCA analogues. The synthesis of isoincarvilline was reported for the first time. Three structurally simplified analogues of INCA were also synthesized. In vivo screening found that INCA and two structurally optimized analogues were efficacious in preventing the acetic acid-induced writhing response. Moreover, their analgesic efficacy was demonstrated in formalin induced pain model. More importantly, administration of 20 or 40 mg/kg INCA and two structurally optimized analogues showed strong analgesic effects in spared nerve injury (SNI) model, and their effective doses were lower than the current gold standard, gabapentin (100 mg/kg in this model).


Subject(s)
Alkaloids/chemical synthesis , Alkaloids/pharmacology , Monoterpenes/chemical synthesis , Monoterpenes/pharmacology , Neuralgia/drug therapy , Alkaloids/therapeutic use , Animals , Drug Discovery , Male , Mice , Mice, Inbred ICR , Monoterpenes/therapeutic use , Structure-Activity Relationship
12.
Behav Brain Res ; 298(Pt A): 97-104, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26433144

ABSTRACT

Methamphetamine (METH), a substance with a high potential for abuse and addiction, is a serious worldwide public health problem. METH addicts often show extreme paranoia, anxiety, and depression. Thus, there is no effective medication for the treatment of METH-induced abnormalities. In the present study, we investigated the effects of l-Scoulerine (l-SLR), a tetrahydroprotoberberine (THPBS) alkaloid, on METH-induced anxiety-like behaviour in zebrafish and METH-induced addictive behavior in mice. In the novel tank test, acute administration of METH (2 mg/L) induced a significant decrease in the number of total vertical transitions and time spent in the upper zone. Moreover, METH produced significant avoidance behaviour showing increased swimming time in the central area and high speed movement in the mirror area in the mirror stimulation test; these anxiety-like changes were attenuated by l-SLR. Chronic administration of METH (2 mg/kg) produced a steady increase in locomotor activity and conditioned place preference in mice. l-SLR (5 mg/kg) failed to reduce acute METH-induced hyperlocomotion, but attenuated chronic METH-induced behavioural sensitization and significantly blocked the expression of conditioned place preference induced by METH in mice. The present study suggests that l-SLR may be a promising agent for the treatment of addiction and anxiety induced by METH.


Subject(s)
Amphetamine-Related Disorders/drug therapy , Anxiety Disorders/drug therapy , Berberine Alkaloids/pharmacology , Central Nervous System Agents/pharmacology , Amphetamine-Related Disorders/physiopathology , Amphetamine-Related Disorders/psychology , Animals , Anxiety Disorders/physiopathology , Berberine Alkaloids/chemistry , Central Nervous System Agents/chemistry , Central Nervous System Stimulants/adverse effects , Central Nervous System Stimulants/pharmacology , Disease Models, Animal , Female , Male , Methamphetamine/adverse effects , Methamphetamine/pharmacology , Mice , Motor Activity/drug effects , Random Allocation , Spatial Behavior/drug effects , Zebrafish
13.
J Mater Chem B ; 4(23): 4147-4155, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-32264617

ABSTRACT

Addition polymerization usually results in polymers with long carbon-carbon main chains. Cyanoacrylate (CA) is arguably an important example of such polymerization and has gained widespread acceptance as an all-purpose adhesive. However, CA-based medical adhesives have never been approved by the U.S. Federal Drug Administration for use below the skin, mainly due to the low biodegradability and biocompatibility of their solid glue after polymerization. In this research, a cross-linking strategy involving the combination of alkyl-CA and the cross-linking agent poly(ethylene glycol)-di(cyanoacrylate) (CA-PEG-CA) to form a copolymeric network was used to synthesize a new generation of biodegradable CA medical adhesives. The degradability could be modulated by adjusting the ratio of CA-PEG-CA to alkyl-CA and the length of PEG. An optimal composite adhesive, LKJ11, was shown to have excellent biodegradability, adhesive capability, and biocompatibility. Importantly, the molecular weight of polycyanoacrylate chains in the polymerized LKJ11 was greatly reduced compared to those polymerized from pure butyl-CA. Thus, the degradation product could be readily extracted. The results showed that LKJ11 represents a new generation of CA-based biodegradable medical adhesives. This advance also provides a general strategy to facilitate the conversion of other polymers with long carbon-carbon main chains to a biodegradable form, thereby expanding the novel applications available for traditional polymeric materials.

14.
Life Sci ; 143: 156-67, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26522050

ABSTRACT

AIMS: Opioid addiction is associated with long-term adaptive changes in the brain that involve protein expression. The carboxyl-terminal of the µ opioid receptor (MOR-C) is important for receptor signal transduction under opioid treatment. However, the proteins that interact with MOR-C after chronic morphine exposure remain unknown. The brain cDNA library of chronic morphine treatment rats was screened using rat MOR-C to investigate the regulator of opioids dependence in the present study. MAIN METHODS: The brain cDNA library from chronic morphine-dependent rats was constructed using the SMART (Switching Mechanism At 5' end of RNA Transcript) technique. Bacterial two-hybrid system was used to screening the rat MOR-C interacting proteins from the cDNA library. RT-qPCR and immunoblotting were used to determine the variation of MOR-C interacting proteins in rat brain after chronic morphine treatment. Column overlay assays, immunocytochemistry and coimmunoprecipitation were used to demonstrate the interaction of MOR-C and p75NTR-associated cell death executor (NADE). KEY FINDINGS: 21 positive proteins, including 19 known proteins were screened to interact with rat MOR-C. Expression of several of these proteins was altered in specific rat brain regions after chronic morphine treatment. Among these proteins, NADE was confirmed to interact with rat MOR-C by in vitro protein-protein binding and coimmunoprecipitation in Chinese hamster ovary (CHO) cells and rat brain with or without chronic morphine treatment. SIGNIFICANCE: Understanding the rat MOR-C interacting proteins and the proteins variation under chronic morphine treatment may be critical for determining the pathophysiological basis of opioid tolerance and addiction.


Subject(s)
Brain/metabolism , Gene Library , Morphine Dependence/metabolism , Receptors, Opioid, mu/metabolism , Two-Hybrid System Techniques , Animals , Brain/drug effects , CHO Cells , Cricetinae , Cricetulus , Male , Morphine/administration & dosage , Morphine/metabolism , Protein Binding/physiology , Rats , Rats, Wistar , Receptors, Opioid, mu/agonists
15.
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
16.
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
17.
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
18.
Eur J Pharmacol ; 738: 153-7, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-24886878

ABSTRACT

Reversing the respiratory depression induced by carfentanil involves intravenous administration of naloxone or naltrexone, but this treatment has disadvantages. Hence, finding a more appropriate treatment to counter the depressive actions of carfentanil is needed. In the present study, with the naloxone as a control, we investigated the efficacy of nalmefene for countering the depressive actions of carfentanil. Rats were treated successively with carfentanil (10 µg/kg, i.v.) and nalmefene (9.4-150.0 µg/kg, i.m.), and the duration of loss of righting reflex (LORR) recorded. Respiratory parameters were measured in free-moving rats using a whole-body plethysmograph after rats were administered carfentanil (20 µg/kg, i.v.) and nalmefene (9.4-150.0 µg/kg, i.m.) sequentially. The parameters of arterial blood gases were also examined. Nalmefene (9.4-150.0 µg/kg, i.m.) treatment dose-dependently decreased the duration of carfentanil-induced LORR. The respiratory rate after 60 min of nalmefene (150.0 µg/kg, i.m.) treatment increased from 34.3 ± 5.3 bursts/min to 117.8 ± 18.9 bursts/min, and enhanced pause decreased from 1.1 ± 0.1 to 0.4 ± 0.1, and was close to those of normal rats. Furthermore, nalmefene (37.5-150.0 µg/kg) treatment could enable the PaO2, SaO2 and PaCO2 to approach normal levels 10 min (15 min after carfentanil injection) or 30 min (25 min after carfentanil injection) after injection. While, a single injection of naloxone (150.0 µg/kg, i.m.) only achieved partial remission of respiratory depression. These data suggest that nalmefene more effectively counters the depressive actions induced by carfentanil and is a more appropriate treatment to antagonize carfentanil toxicity compared with naloxone.


Subject(s)
Analgesics, Opioid/adverse effects , Fentanyl/analogs & derivatives , Naltrexone/analogs & derivatives , Reflex, Righting/drug effects , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Analgesics, Opioid/antagonists & inhibitors , Animals , Blood Gas Analysis , Dose-Response Relationship, Drug , Fentanyl/adverse effects , Fentanyl/antagonists & inhibitors , Male , Naltrexone/pharmacology , Naltrexone/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors
19.
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
20.
Eur J Pharmacol ; 737: 22-8, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24830319

ABSTRACT

Opioid analgesics are widely believed to cause spasm of the bile duct sphincter and so impede bile flow. Thienorphine is a partial opioid agonist that is a good candidate for the treatment of opioid dependence; however, to date, no studies have reported the effects of thienorphine on the function of the biliary tract. This study examined the in vivo effects of thienorphine on the guinea pig isolated sphincter of Oddi, choledochus and gall bladder and on bile flow. The area under the curve (AUC) of isolated sphincter of Oddi was not influenced by thienorphine or buprenorphine, whereas morphine increased the AUC of the isolated sphincter of Oddi in a concentration-dependent manner. Thienorphine and buprenorphine concentration-dependently decreased the AUC of isolated choledochus, while morphine increased the AUC of isolated choledochus. Thienorphine had no effect on the contractile amplitude or basal tension of isolated gall bladder muscle strips. In contrast, buprenorphine and morphine increased the contractile basal tension of isolated gall bladder muscle strips in a concentration-dependent manner. Thienorphine (0.01-1.0mg/kg) had no significant inhibitory effect on bile flow. However, morphine (1.0-10mg/kg) and buprenorphine (1.0mg/kg) significantly inhibited bile flow. The maximum inhibition of bile flow by buprenorphine was 63.9±12.9% and by morphine was 74.1±11.3%. In summary, thienorphine has little influence on the guinea pig isolated sphincter of Oddi, choledochus and gall bladder or on bile flow, which may result in a lack of adverse biliary colic effects.


Subject(s)
Buprenorphine/analogs & derivatives , Common Bile Duct/drug effects , Gallbladder/drug effects , Muscle Contraction/drug effects , Sphincter of Oddi/drug effects , Animals , Bile/metabolism , Buprenorphine/pharmacology , Common Bile Duct/physiology , Female , Gallbladder/physiology , Guinea Pigs , Male , Sphincter of Oddi/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...