Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Biochem Biophys Res Commun ; 614: 219-224, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35636221

ABSTRACT

Cepharanthine is an alkaloid that isolated from Stephania cepharantha Hayata, however,its analgesic properties are unclear and the molecular targets that mediating Cepharanthine-induced analgesia are not explored yet. In the current study, mice pain models including hot plate, acetic acid-induced writhing and formalin tests were conducted to evaluate the antinociceptive actions of Cepharanthine. [3H]-ligand competitive binding assay was applied to determine the binding affinity and selectivity of Cepharanthine at κ, µ and δ opioid receptors. Cepharanthine-induced constipation was investigated using the small intestinal transit test. The results showed that intraperitoneal injection of Cepharanthine produced potent antinociception with an ED50 value of 24.5 mg/kg in the acetic acid-induced writhing test. In the formalin test, Cepharanthine produced moderate antinociception with the maximum analgesic activity of 42.6 ± 11.3% in phase I and 60.1 ± 7.7% in phase Ⅱ, respectively. Cepharanthine had no effects in the hot plate test. In vitro radioligand binding assay, Cepharanthine exhibited a high affinity for µ opioid receptors with a Ki value of 80 nM, without binding to κ and δ opioid receptors. Correspondingly, Cepharanthine-mediated antinociceptive effects were antagonized by pretreatment with opioid receptor antagonist naloxone. Cepharanthine also decreased the small intestine propulsion rates in the small intestinal transit test. Together, this study firstly demonstrates that Cepharanthine produces potent antinociception in acetic acid-induced visceral pain and moderate antinociception in formalin-induced inflammatory pain, and its mechanism of action may be through activation of µ opioid receptors.


Subject(s)
Receptors, Opioid, delta , Receptors, Opioid, mu , Acetates , Analgesics/pharmacology , Analgesics/therapeutic use , Analgesics, Opioid/pharmacology , Animals , Benzylisoquinolines , Disease Models, Animal , Mice , Pain/drug therapy , Receptors, Opioid, kappa/metabolism
2.
Cell Rep ; 37(5): 109913, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731618

ABSTRACT

Opiates produce a strong rewarding effect, but abstinence from opiate use emerges with severe negative emotions. Depression is one of the most frequent emotion disorders associated with opiate abstinence, which is thought to be a main cause for relapse. However, neurobiological bases of such an aversive emotion processing are poorly understood. Here, we find that morphine abstinence activates κ-opioid receptors (KORs) by increasing endogenous KOR ligand dynorphin expression in the amygdala, which in turn facilitates glutamate transporter 1 (GLT1) expression by activation of p38 mitogen-activated protein kinase (MAPK). Upregulation of GLT1 expression contributes to opiate-abstinence-elicited depressive-like behaviors through modulating amygdalar glutamatergic inputs to the nucleus accumbens (NAc). Intra-amygdala injection of GLT1 inhibitor DHK or knockdown of GLT1 expression in the amygdala significantly suppresses morphine-abstinence-induced depressive-like behaviors. Pharmacological and pharmacogenetic activation of amygdala-NAc projections prevents morphine-abstinence-induced behaviors. Overall, our study provides key molecular and circuit insights into the mechanisms of depression associated with opiate abstinence.


Subject(s)
Amygdala/metabolism , Behavior, Animal , Depression/metabolism , Glucose Transporter Type 1/metabolism , Glutamic Acid/metabolism , Morphine , Nucleus Accumbens/metabolism , Receptors, Opioid, kappa/metabolism , Substance Withdrawal Syndrome/metabolism , Amygdala/physiopathology , Animals , Depression/chemically induced , Depression/physiopathology , Depression/psychology , Disease Models, Animal , Dynorphins/metabolism , Excitatory Postsynaptic Potentials , Glucose Transporter Type 1/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/metabolism , Neural Pathways/physiopathology , Nucleus Accumbens/physiopathology , Receptors, Opioid, kappa/genetics , Signal Transduction , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/psychology , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Mol Psychiatry ; 26(11): 6218-6236, 2021 11.
Article in English | MEDLINE | ID: mdl-33963280

ABSTRACT

Exposure to drugs of abuse induces alterations of dendritic spine morphology and density that has been proposed to be a cellular basis of long-lasting addictive memory and heavily depend on remodeling of its underlying actin cytoskeleton by the actin cytoskeleton regulators. However, the actin cytoskeleton regulators involved and the specific mechanisms whereby drugs of abuse alter their expression or function are largely unknown. Twinfilin (Twf1) is a highly conserved actin-depolymerizing factor that regulates actin dynamics in organisms from yeast to mammals. Despite abundant expression of Twf1 in mammalian brain, little is known about its importance for brain functions such as experience-dependent synaptic and behavioral plasticity. Here we show that conditioned morphine withdrawal (CMW)-induced synaptic structure and behavior plasticity depends on downregulation of Twf1 in the amygdala of rats. Genetically manipulating Twf1 expression in the amygdala bidirectionally regulates CMW-induced changes in actin polymerization, spine density and behavior. We further demonstrate that downregulation of Twf1 is due to upregulation of miR101a expression via a previously unrecognized mechanism involving CMW-induced increases in miR101a nuclear processing via phosphorylation of MeCP2 at Ser421. Our findings establish the importance of Twf1 in regulating opioid-induced synaptic and behavioral plasticity and demonstrate its value as a potential therapeutic target for the treatment of opioid addiction.


Subject(s)
Analgesics, Opioid , Microfilament Proteins/metabolism , Substance Withdrawal Syndrome , Actin Cytoskeleton/metabolism , Actins , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Dendritic Spines/metabolism , Rats , Substance Withdrawal Syndrome/metabolism , Synapses/metabolism
4.
ACS Med Chem Lett ; 12(3): 397-403, 2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33738067

ABSTRACT

SIRT1, a member of the sirtuin family, catalyzes the deacetylation of proteins with the transformation of NAD+ into nicotinamide and 2'-O-acetyl-ADP-ribose. Selective SIRT1/2 inhibitors have potential application in the chemotherapy of colorectal carcinoma, prostate cancer, and myelogenous leukemia. Here we identified novel SIRT1 inhibitors with the scaffold of 5-benzylidene-2-phenyl-1,3-dioxane-4,6-dione. The most potent inhibitor 12n displayed an IC50 of 460 nM and a selectivity for SIRT1 over SIRT2, SIRT3, and SIRT5 of 113.5-, 254.3-, and 10.83-fold, respectively. It did not affect the activity of SIRT6. To elucidate the inhibitory mechanism, we determined the inhibition type of the inhibitor by enzyme kinetic analysis, showing that the inhibitor was competitive to the acetyl peptide and noncompetitive to NAD+. Further, the interaction of the inhibitor in SIRT1 was studied by using molecular docking, which was validated by the structure-activity relationship analysis of the inhibitors and the site-directed mutagenesis of SIRT1. Consistent with the in vitro assays, the inhibitors increased the acetylation level of p53 in a concentration-dependent manner in cells.

5.
Sci Rep ; 8(1): 14087, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30237513

ABSTRACT

Dezocine is becoming dominated in China market for relieving moderate to severe pain. It is believed that Dezocine's clinical efficacy and little chance to provoke adverse events during the therapeutic process are mainly attributed to its partial agonist activity at the µ opioid receptor. In the present work, we comprehensively studied the pharmacological characterization of Dezocine and identified that the analgesic effect of Dezocine was a result of action at both the κ and µ opioid receptors. We firstly found that Dezocine displayed preferential binding to µ opioid receptor over κ and δ opioid receptors. Dezocine, on its own, weakly stimulated G protein activation in cells expressing κ and µ receptors, but in the presence of full κ agonist U50,488 H and µ agonist DAMGO, Dezocine inhibited U50,488H- and DAMGO-mediated G protein activation, indicating that Dezocine was a κ partial agonist and µ partial agonist. Then the in intro results were verified by in vivo studies in mice. We observed that Dezocine-produced antinociception was significantly inhibited by κ antagonist nor-BNI and µ antagonist ß-FNA pretreatment, indicating that Dezocine-mediated antinociception was via both the κ and µ opioid receptors. When co-administrating of Dezocine with U50,488 H or morphine, Dezocine was capable of inhibiting U50,488H- or morphine-induced antinociception. Finally, κ receptor activation-associated side effect sedation was investigated. We found that Dezocine displayed limited sedative effect with a ceiling effecting at a moderate dose. Thus, our work led to a better understanding of the analgesic mechanism of action of Dezocine in vivo.


Subject(s)
Analgesics, Opioid/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Nociception/drug effects , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Tetrahydronaphthalenes/pharmacology , Animals , CHO Cells , Cricetulus , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , Male , Mice , Morphine/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain Measurement , Protein Binding , Signal Transduction/drug effects
6.
Front Mol Neurosci ; 10: 313, 2017.
Article in English | MEDLINE | ID: mdl-29051727

ABSTRACT

Abuse of methamphetamine (METH), a powerful addictive amphetamine-type stimulants (ATS), is becoming a global public health problem. The gamma-aminobutyric acid (GABA)ergic system plays a critical role in METH use disorders. By using rat METH conditioned place preference (CPP) model, we previously demonstrated that METH-associated rewarding memory formation was associated with the reduction of GABAAα1 expression in the dorsal straitum (Dstr), however, the underlying mechanism was unclear. In the present study, we found that METH-induced CPP formation was accompanied by a significant increase in the expression of Synovial apoptosis inhibitor 1 (SYVN1), an endoplasmic reticulum (ER)-associated degradation (ERAD) E3 ubiquitin ligase, in the Dstr. The siRNA knockdown of SYVN1 significantly increased GABAAα1 protein levels in both primary cultured neurons and rodent Dstr. Inhibition of proteasomal activity by MG132 and Lactacystin significantly increased GABAAα1 protein levels. We further found that SYVN1 knockdown increased GABAAα1 in the intra-ER, but not in the extra-ER. Accordingly, endoplasmic reticulum stress (ERS)-associated Glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP) increased. Thus, this study revealed that SYVN1, as the ERAD E3 ubiquitin ligase, was associated with Dstr GABAAα1 degradation induced by METH conditioned pairing.

7.
Behav Brain Res ; 323: 133-140, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28147236

ABSTRACT

Repeated exposure to methamphetamine (METH) can cause severe neurotoxicity to the cortical neurons. In the present study, we investigated the effect of METH on cognitive function deficits, and determined the neuroprotective effects of memantine (MEM) on memory impairment induced by METH. The protein levels of Bcl-2 and cleaved caspase-3 in prefrontal cortex (PFC) were further examined to exploring the underlying mechanism. We found that repeated METH administration impaired long term (24h) memory retention without affecting short term (5min) memory retention. Co-administration of MEM with METH before training session significantly improved METH-induced cognitive function. METH significantly decreased expression level of Bcl-2 and increased expression level of cleaved caspase-3 in the PFC. The changes can be prevented by MEM pretreatment. Thus, these results demonstrated that MEM pretreatment reversed METH-induced changes of protein levels of apoptotic-related gene, and produced protective effects against METH-induced cognitive deficits, suggesting the effectiveness of MEM may be due to its anti-apoptotic activity.


Subject(s)
Cognition/drug effects , Memantine/administration & dosage , Memory/drug effects , Methamphetamine/toxicity , Neuroprotective Agents/administration & dosage , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Male , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Mice , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
8.
ACS Chem Neurosci ; 8(4): 766-776, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28033462

ABSTRACT

To develop novel analgesics with no side effects or less side effects than traditional opioids is highly demanded to treat opioid receptor mediated pain and addiction issues. Recently, κ-opioid receptor (KOR) has been established as an attractive target, although its selective agonists could bear heterogeneous pharmacological activities. In this study, we designed and synthesized two new series of nepenthone derivatives by inserting a spacer (carbonyl) between 6α,14α-endo-ethenylthebaine and the 7α-phenyl substitution of the skeleton and by substituting the 17-N-methyl group with a cyclopropylmethyl group. We performed in vitro tests (binding and functional assays) and molecular docking operations on our newly designed compounds. The results of wet-experimental measures and modeled binding structures demonstrate that these new compounds are selective KOR agonists with nanomolar level affinities. Compound 4 from these new derivatives showed the highest affinity (Ki = 0.4 ± 0.1 nM) and the highest selectivity (µ/κ = 339, δ/κ = 2034) toward KOR. The in vivo tests revealed that compound 4 is able to induce stronger (ED50 = 2.1 mg/kg) and much longer antinociceptive effect than that of the typical KOR agonist U50488H (ED50 = 4.4 mg/kg). Therefore, compound 4 can be used as a perfect lead compound for future design of potent analgesics acting through KOR.


Subject(s)
Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Morphinans/chemistry , Morphinans/pharmacology , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Analgesics, Opioid/chemical synthesis , Animals , Humans , Molecular Docking Simulation , Morphinans/chemical synthesis , Protein Structure, Quaternary , Rats , Receptors, Opioid, kappa/agonists , Structure-Activity Relationship
9.
Acta Pharmacol Sin ; 36(12): 1437-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26567727

ABSTRACT

AIM: Brain-derived neurotrophic factor (BDNF) plays an important role in learning and memory in multiple brain areas. In the present study, we investigated the roles of BDNF in aversive memories associated with conditioned drug withdrawal in acute morphine-dependent rats. METHODS: Conditioned place aversion (CPA) was induced in male SD rats exposed to a single dose of morphine (10 mg/kg, sc) followed by naloxone (0.3 mg/kg, sc). In some rats, BDNF receptor antagonist K252a (8.5 ng per side) or BDNF scavenger TrkB-FC (0.65 µg per side) was bilaterally microinjected into amygdala before naloxone injection. BDNF mRNA and protein expression levels in amygdala were detected after the behavior testing. RESULTS: CPA behavior was induced in rats by the naloxone-precipitated morphine withdrawal, which was accompanied by significantly increased levels of BDNF mRNA and protein in the amygdala. Bilateral microinjection of TrkB-FC or K252a into the amygdala completely blocked CPA behavior in the rats. CONCLUSION: Formation of aversive memories associated with conditioned drug withdrawal in acute morphine-dependent rats requires BDNF expression in the amygdala.


Subject(s)
Amygdala/drug effects , Morphine Dependence/genetics , Morphine Dependence/therapy , Naloxone/therapeutic use , Narcotic Antagonists/therapeutic use , Receptor, trkB/genetics , Substance Withdrawal Syndrome/genetics , Amygdala/metabolism , Amygdala/physiopathology , Animals , Aversive Therapy , Male , Morphine/adverse effects , Morphine Dependence/physiopathology , Narcotics/adverse effects , RNA, Messenger/genetics , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/physiopathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...