4,063 results on '"µ-Opioid receptor"'
Search Results
2. Positive allosteric modulation of µ-opioid receptor – A new possible approach in the pain management?
- Author
-
Król, Wojciech, Machelak, Weronika, and Zielińska, Marta
- Published
- 2025
- Full Text
- View/download PDF
3. Signal profiles and spatial regulation of β-arrestin recruitment through Gβ5 and GRK3 at the μ-opioid receptor
- Author
-
Carino, Carlo Marion C., Hiratsuka, Suzune, Kise, Ryoji, Nakamura, Gaku, Kawakami, Kouki, Yanagawa, Masataka, and Inoue, Asuka
- Published
- 2025
- Full Text
- View/download PDF
4. CtBP1 is essential for epigenetic silencing of μ-opioid receptor genes in the dorsal root ganglion in spinal nerve ligation-induced neuropathic pain
- Author
-
Lai, Cheng-Yuan, Hsieh, Ming-Chun, Yeh, Chou-Ming, Lin, Tzer-Bin, Chou, Dylan, Wang, Hsueh-Hsiao, Lin, Kuan-Hung, Cheng, Jen-Kun, Yang, Po-Sheng, and Peng, Hsien-Yu
- Published
- 2025
- Full Text
- View/download PDF
5. Effects of Dual Inhibition at Dopamine Transporter and σ Receptors in the Discriminative-Stimulus Effects of Cocaine in Male Rats
- Author
-
Hiranita, Takato, Li, Su-Min, and Katz, Jonathan L.
- Published
- 2024
- Full Text
- View/download PDF
6. Alternative Reinforcers Enhance the Effects of Opioid Antagonists, but Not Agonists, on Oxycodone Choice Self-Administration in Nonhuman Primates
- Author
-
de Moura, Fernando B. and Kohut, Stephen J.
- Published
- 2024
- Full Text
- View/download PDF
7. Identification and quantification of kukoamine A and kukoamine B as novel μ-opioid receptor agonists in potato and other solanaceous plants
- Author
-
Saller, Julia, List, Carina, Hübner, Harald, Gmeiner, Peter, Clark, Timothy, and Pischetsrieder, Monika
- Published
- 2023
- Full Text
- View/download PDF
8. The activated caveolin‐3/μ‐opioid receptor complex drives morphine‐induced rescue therapy in failing hearts.
- Author
-
Guo, Chengxiao, Pan, Xinxin, Dou, Mengyun, Wu, Juan, Chen, Xinyu, Wang, Baoli, Zhu, Rui, Xu, Shijin, Peng, Wenyi, Wu, Chao, He, Shufang, Zhang, Sihe, Zhang, Ye, and Jin, Shiyun
- Subjects
- *
HEART failure , *PROTEIN receptors , *REPERFUSION injury , *OPIOID receptors , *TREATMENT failure , *CAVEOLAE , *OPIOID analgesics - Abstract
Background and Purpose: Opioid analgesics can alleviate ischaemia/reperfusion (I/R) injury in chronic heart failure. However, the underlying mechanisms and targets remain unknown. Here, we investigate if caveolin‐3 (Cav3) interacts with μ opioid receptors and if Cav3–μ receptor interactions play a role in morphine‐induced cardioprotection in failing hearts. Experimental Approach: Cav3 and μ receptor proteins in human and rat heart tissue were determined by western blot, immunofluorescence and co‐immunoprecipitation. Methyl‐β‐cyclodextrin (MβCD), a destroyer of caveolae, and AAV‐Cav3 shRNA were used to reduce Cav3 expression in failing rat hearts. CTOP, a specific μ antagonist, was administrated before morphine preconditioning in perfused failing heart models of myocardial I/R injury. Key Results: Levels of Cav3 and μ receptor proteins were significantly higher in human and rat myocardial tissues with heart failure than in control tissues. Cav3 and μ receptor expression levels were positively correlated with disease severity. The signal of the cardiac Cav3 protein was colocalized with μ receptor in both the human and rat heart sections. Disruption of caveolae in the failing heart by either MβCD or AAV‐Cav3 shRNA significantly inhibits morphine‐induced phosphorylation of ERK1/2 and cardioprotection. Administration of CTOP substantially reduced Cav3 expression and morphine‐induced cardioprotective effect in heart failure. Conclusion and Implications: Our data suggest that up‐regulation of the Cav3/μ receptor complex is critical for morphine protection of the failing heart against I/R injury by regulating the ERK1/2 pathway. The activated Cav3/μ receptor complex is an understudied therapeutic target for opioid treatment of heart failure and ischaemic insult. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
9. Striosome Circuitry Stimulation Inhibits Striatal Dopamine Release and Locomotion.
- Author
-
Taro Okunomiya, Dai Watanabe, Haruhiko Banno, Takayuki Kondo, Keiko Imamura, Ryosuke Takahashi, and Haruhisa Inoue
- Subjects
- *
DESIGNER drugs , *DOPAMINERGIC neurons , *ANIMAL locomotion , *ADENO-associated virus , *DOPAMINE , *DOPAMINE receptors , *OPIOID receptors - Abstract
The mammalian striatum is divided into two types of anatomical structures: the island-like, µ-opioid receptor (MOR)-rich striosome compartment and the surrounding matrix compartment. Both compartments have two types of spiny projection neurons (SPNs), dopamine receptor D1 (D1R)-expressing direct pathway SPNs (dSPNs) and dopamine receptor D2 (D2R)-expressing indirect pathway SPNs. These compartmentalized structures have distinct roles in the development of movement disorders, although the functional significance of the striosome compartment for motor control and dopamine regulation remains to be elucidated. The aim of this study was to explore the roles of striosome in locomotion and dopamine dynamics in freely moving mice. We targeted striosomal MOR-expressing neurons with male MOR-CreER mice, which express tamoxifen-inducible Cre recombinase under MOR promoter, and Cre-dependent adeno-associated virus vector. The targeted neuronal population consisted mainly of dSPNs. We found that the Gq-coupled designer receptor exclusively activated by designer drugs (DREADD)-based chemogenetic stimulation of striatal MOR-expressing neurons caused a decrease in the number of contralateral rotations and total distance traveled. Wireless fiber photometry with a genetically encoded dopamine sensor revealed that chemogenetic stimulation of striatal MOR-expressing neurons suppressed dopamine signals in the dorsal striatum of freely moving mice. Furthermore, the decrease in mean dopamine signal and the reduction of transients were associated with ipsilateral rotational shift and decrease of average speed, respectively. Thus, a subset of striosomal dSPNs inhibits contralateral rotation, locomotion, and dopamine release in contrast to the role of pan-dSPNs. Our results suggest that striatal MOR-expressing neurons have distinct roles in motor control and dopamine regulation. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
10. An OPRM1-SNAP-tag/CMC method to directly identify drug components in sewage: An OPRM1-SNAP-tag/CMC method to directly identify drug components in sewage: C. Li et al.
- Author
-
Li, Chenjia, Liao, Qi, Wang, Rui, Zhang, Xinping, Ma, Mengyang, Liu, Yonghong, Xiao, Lei, Jiao, Ying, and Wang, Nan
- Subjects
- *
SEWAGE , *TELEMATICS , *DRUG monitoring , *PHARMACEUTICAL chemistry , *AFFINITY chromatography - Abstract
The scourge of drug addiction and abuse poses a significant challenge to society. Opioid drugs acting on μ-opioid receptor (OPRM1) make it one of the pivotal targets for drug addiction. In the past decade, sewage analysis has become a prevalent method of drug monitoring. However, traditional methods of detecting drugs in sewage are cumbersome, and rapid detection methods are relatively lacking. To address this, an innovative OPRM1-SNAP-tag/CMC method to directly identify drug components in sewage was established. Cell membrane chromatography (CMC) is an affinity chromatography technique which effectively detects receptor affinity substances. Cells constructed with high expression of specific receptor could be used to screen for compounds acting on the receptor. CMC based on OPRM1 provides a potentially convenient and effective tool for the detection of targeted drug components in sewage. In this study, the selectivity, reproducibility, column lifetime, and carryover of the CMC column had been assessed. Initially, we eluted the collected domestic sewage with methanol and acetonitrile, and the retention peaks were observed on the CMC system. Subsequently, without any preliminary sample preparation, we directly injected filtered samples of suspicious sewage into the OPRM1-SNAP-tag/CMC system, where we observed retention peaks as well. The retained components were further identified as morphine by using UPLC-MS/MS. In conclusion, the OPRM1-SNAP-tag/CMC method stands out as a reliable and robust model for the detection of drug components in sewage. It provides a valuable analytical tool for frontline drug control efforts, enhancing our capacity to monitor and mitigate the impact of drug abuse on society. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. Postsynaptic dopamine D3 receptors selectively modulate μ-opioid receptor-expressing GABAergic inputs onto CA1 pyramidal cells in the rat ventral hippocampus.
- Author
-
Brown, Kyle A., Stramiello, Michael, Clark, Jason K., and Wagner, John J.
- Subjects
- *
PYRAMIDAL neurons , *CELL morphology , *DOPAMINE receptors , *NEUROPLASTICITY , *NEURONS - Abstract
Although the actions of dopamine throughout the brain are clearly linked to motivation and cognition, the specific role(s) of dopamine in the CA1 subfield of the ventral hippocampus (vH) is unresolved. Prior preclinical studies suggest that dopamine D3 receptors (D3Rs) expressed on CA1 pyramidal cells exhibit a unique capacity to modulate mechanisms of long-term synaptic plasticity, but less is known about how interneuronal inputs modulate these cells. We hypothesized that inputs from μ-opioid receptor (MOR)-expressing inhibitory interneurons selectively modulate the activity of postsynaptic D3Rs expressed on CA1 principal cells to shape neurotransmission in the rat vH. We used the whole cell voltage-clamp technique to test this hypothesis by measuring evoked inhibitory postsynaptic currents (eIPSCs) from CA1 principal cells in vH slices or GABAA currents from acutely dissociated vH neurons. The eIPSC response recorded from CA1 neurons in vH slices was inhibited by either the MOR agonist DAMGO or the D3R agonist PD128907, but pretreatment with DAMGO occluded any further inhibition by PD128907. GABAA currents measured in acutely dissociated vH CA1 neurons were inhibited by D3R activation via PD128907, consistent with postsynaptic localization of D3 receptors. Kinetic alterations induced by the neuromodulatory agonists are consistent with selective targeting of postsynaptic D3Rs expressed on CA1 principal cells by MOR-expressing GABAergic inputs. Our findings suggest that postsynaptic D3R-mediated modulation of MOR-expressing GABAergic inputs is a site at which dopaminergic and opioidergic activity may contribute to disinhibition of vH excitatory neurotransmission and, thus, influence critical physiological processes such as synaptic plasticity and network oscillations. NEW & NOTEWORTHY: We report that the activity of an inhibitory synapse on CA1 pyramidal cells in the rat ventral hippocampus is shaped by heterogeneous neuromodulators. Specifically, postsynaptic dopamine D3 receptors on ventral hippocampal CA1 pyramidal neurons are selectively targeted by an inhibitory input from µ-opioid receptor-expressing GABAergic terminals. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Modulation of Nicotine-Associated Behaviour in Rats By μ-Opioid Signals from the Medial Prefrontal Cortex to the Nucleus Accumbens Shell: F. Zhu et al.: MORs in the mPFC Modulate Nicotine Cravings.
- Author
-
Zhu, Feng, Kanda, Hirosato, Neyama, Hiroyuki, Wu, Yuping, Kato, Shigeki, Hu, Di, Duan, Shaoqi, Noguchi, Koichi, Watanabe, Yasuyoshi, Kobayashi, Kazuto, Dai, Yi, and Cui, Yilong
- Abstract
Nicotine addiction is a concern worldwide. Most mechanistic investigations are on nicotine substance dependence properties based on its pharmacological effects. However, no effective therapeutic treatment has been established. Nicotine addiction is reinforced by environments or habits. We demonstrate the neurobiological basis of the behavioural aspect of nicotine addiction. We utilized the conditioned place preference to establish nicotine-associated behavioural preferences (NABP) in rats. Brain-wide neuroimaging analysis revealed that the medial prefrontal cortex (mPFC) was activated and contributed to NABP. Chemogenetic manipulation of µ-opioid receptor positive (MOR
+ ) neurons in the mPFC or the excitatory outflow to the nucleus accumbens shell (NAcShell) modulated the NABP. Electrophysiological recording confirmed that the MOR+ neurons directly regulate the mPFC-NAcShell circuit via GABAA receptors. Thus, the MOR+ neurons in the mPFC modulate the formation of behavioural aspects of nicotine addiction via direct excitatory innervation to the NAcShell, which may provide new insight for the development of effective therapeutic strategies. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
13. Morphine promotes non-small cell lung cancer progression by downregulating E-cadherin via the PI3K/AKT/mTOR pathway
- Author
-
Fulei Gu, Yuxuan Zhou, Lili Tian, Jinyan Chen, Can Zhang, Zhangxiang Huang, Weifeng Yu, and Kangjie Xie
- Subjects
Morphine ,Non-small cell lung cancer ,µ-Opioid receptor ,Malignant biological behaviour ,E-cadherin ,Medicine ,Science - Abstract
Abstract Morphine has been suggested to affect cancer cell dynamics and decrease survival rates in lung cancer patients at specific doses, but the precise mechanisms poorly understood. In this study, we aimed to investigate the molecular mechanisms by which morphine modulates the malignant characteristics of non-small cell lung cancer. Cell proliferation was assessed via the Cell Counting Kit-8 assay, and cell migration and invasion were examined via wound healing and Transwell assays. We employed immunofluorescence staining to evaluate E-cadherin expression in A549 and Lewis lung cancer (LLC) cell lines and immunohistochemistry to evaluate E-cadherin expression in nude mice tumours. Additionally, the in vivo effects of morphine on lung cancer progression were explored in a xenograft tumour experiments, in which naloxone was used as a morphine antagonist. Western blot analysis was performed to detect E-cadherin, phosphorylated mTOR (p-mTOR), mTOR, phosphorylated AKT (p-AKT), AKT, phosphorylated PI3K (p-PI3K), and PI3K protein levels in A549 and LLC cells as well as in tumour samples. Morphine (10 µM) significantly increased the proliferation of A549 and LLC cells in vitro (p
- Published
- 2024
- Full Text
- View/download PDF
14. μ-Opioid Receptor Modulation of the Glutamatergic/GABAergic Midbrain Inputs to the Mouse Dorsal Hippocampus.
- Author
-
Kim, Haram R., Dey, Soumil, Sekerkova, Gabriella, and Martina, Marco
- Subjects
- *
GRANULE cells , *DENTATE gyrus , *GABA receptors , *CALCIUM channels , *CELL nuclei , *OPIOID receptors , *GLUTAMATE receptors - Abstract
We used virus-mediated anterograde and retrograde tracing, optogenetic modulation, immunostaining, in situ hybridization, and patch-clamp recordings in acute brain slices to study the release mechanism and µ-opioid modulation of the dual glutamatergic/GABAergic inputs from the ventral tegmental area and supramammillary nucleus to the granule cells of the dorsal hippocampus of male and female mice. In keeping with previous reports showing that the two transmitters are released by separate active zones within the same terminals, we found that the short-term plasticity and pharmacological modulation of the glutamatergic and GABAergic currents are indistinguishable. We further found that glutamate and GABA release at these synapses are both virtually completely mediated by N- and P/Q-type calcium channels. We then investigated µ-opioid modulation of these synapses and found that activation of µ-opioid receptors (MORs) strongly inhibits the glutamate and GABA release, mostly through inhibition of presynaptic N-type channels. However, the modulation by MORs of these dual synapses is complex, as it likely includes also a disinhibition due to downmodulation of local GABAergic interneurons which make direct axo-axonic contacts with the dual glutamatergic/GABAergic terminals. We discuss how this opioid modulation may enhance LTP at the perforant path inputs, potentially contributing to reinforce memories of drug-associated contexts. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Slow dissociation kinetics of fentanyls and nitazenes correlates with reduced sensitivity to naloxone reversal at the μ‐opioid receptor.
- Author
-
Alhosan, Norah, Cavallo, Damiana, Santiago, Marina, Kelly, Eamonn, and Henderson, Graeme
- Subjects
- *
ENDORPHIN receptors , *MEMBRANE potential , *NALOXONE , *G proteins , *ARRESTINS , *DRUG overdose , *OPIOID receptors , *G protein coupled receptors - Abstract
Background and Purpose Experimental Approach Key Results Conclusions and Implications Fentanyls and nitazenes are μ‐opioid receptor agonists responsible for a large number of opioid overdose deaths. Here, we determined the potency, dissociation kinetics and antagonism by naloxone at the μ receptor of several fentanyl and nitazene analogues, compared to morphine and DAMGO.In vitro assays of G protein activation and signalling and arrestin recruitment were performed. AtT20 cells expressing μ receptors were loaded with a membrane potential dye and changes in fluorescence used to determine agonist potency, dissociation kinetics and susceptibility to antagonism by naloxone. BRET experiments were undertaken in HEK293T cells expressing μ receptors to assess Gi protein activation and β‐arrestin 2 recruitment.The apparent rate of agonist dissociation from the μ receptor varied: morphine, DAMGO, alfentanil and fentanyl dissociated rapidly, whereas isotonitazene, etonitazene, ohmefentanyl and carfentanil dissociated slowly. Slowly dissociating agonists were more resistant to antagonism by naloxone. For carfentanil, the slow apparent rate of dissociation was not because of G protein receptor kinase‐mediated arrestin recruitment as its apparent rate of dissociation was not increased by inhibition of G protein‐coupled receptor kinases (GRKs) with Compound 101. The in vitro relative potencies of fentanyls and nitazenes compared to morphine were much lower than that previously observed in in vivo experiments.With fentanyls and nitazenes that slowly dissociate from the μ receptor, antagonism by naloxone is pseudo‐competitive. In overdoses involving fentanyls and nitazenes, higher doses of naloxone may be required for reversal than those normally used to reverse heroin overdose. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Non‐linear blood–brain barrier transport and dosing strategies influence receptor occupancy ratios of morphine and its metabolites in pain matrix.
- Author
-
Budda, Divakar, Gülave, Berfin, van Hasselt, J. G. Coen, and de Lange, Elizabeth C. M.
- Subjects
- *
ANIMAL experimentation , *CHRONIC pain , *MORPHINE , *ANALGESIA , *PHARMACOKINETICS - Abstract
Background and Purpose: Morphine is important for treatment of acute and chronic pain. However, there is high interpatient variability and often inadequate pain relief and adverse effects. To better understand variability in the dose‐effect relationships of morphine, we investigated the effects of its non‐linear blood–brain barrier (BBB) transport on μ‐receptor occupancy in different CNS locations, in conjunction with its main metabolites that bind to the same receptor. Experimental Approach: CNS exposure profiles for morphine, M3G and M6G for clinically relevant dosing regimens based on intravenous, oral immediate‐ and extended‐release formulations were generated using a physiology‐based pharmacokinetic model of the CNS, with non‐linear BBB transport of morphine. The simulated CNS exposure profiles were then used to derive corresponding μ‐receptor occupancies at multiple CNS pain matrix locations. Key Results: Simulated CNS exposure profiles for morphine, M3G and M6G, associated with non‐linear BBB transport of morphine resulted in varying μ‐receptor occupancies between different dose regimens, formulations and CNS locations. At lower doses, the μ‐receptor occupancy of morphine was relatively higher than at higher doses of morphine, due to the relative contribution of M3G and M6G. At such higher doses, M6G showed higher occupancy than morphine, whereas M3G occupancy was low throughout the dose ranges. Conclusion and Implications: Non‐linear BBB transport of morphine affects the μ‐receptor occupancy ratios of morphine with its metabolites, depending on dose and route of administration, and CNS location. These predictions need validation in animal or clinical experiments, to understand the clinical implications. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Opioids and the Gastrointestinal Tract: The Role of Peripherally Active μ-Opioid Receptor Antagonists in Modulating Intestinal Permeability.
- Author
-
Lacy, Brian E. and Cangemi, David J.
- Subjects
- *
INTESTINAL barrier function , *OPIOID receptors , *BACTERIAL toxins , *TOLL-like receptors , *LARGE intestine - Abstract
Opioid receptors are found throughout the gastrointestinal tract, including the large intestine. Many patients treated with opioids experience opioid-induced constipation (OIC). Laxatives are not effective in most patients, and in those who do initially respond, the efficacy of laxatives generally diminishes over time. In addition, OIC does not spontaneously resolve for most patients. However, complications of opioids extend far beyond simply slowing gastrointestinal transit. Opioid use can affect intestinal permeability through a variety of mechanisms. Toll-like receptors are a crucial component of innate immunity and are tightly regulated within the gut epithelium. Pathologic m-opioid receptor (MOR) and toll-like receptor signaling, resulting from chronic opioid exposure, disrupts intestinal permeability leading to potentially harmful bacterial translocation, elevated levels of bacterial toxins, immune activation, and increased cytokine production. Peripherally active MOR antagonists, including methylnaltrexone, are effective at treating OIC. Benefits extend beyond simply blocking the MOR; these agents also act to ameliorate opioid-induced disrupted intestinal permeability. In this review, we briefly describe the physiology of the gastrointestinal epithelial border and discuss the impact of opioids on gastrointestinal function. Finally, we consider the use of peripherally activeMORantagonists to treat disrupted intestinal permeability resulting from opioid use and discuss the potential for improved morbidity and mortality in patients treated with methylnaltrexone for opioid-induced bowel disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Morphine promotes non-small cell lung cancer progression by downregulating E-cadherin via the PI3K/AKT/mTOR pathway.
- Author
-
Gu, Fulei, Zhou, Yuxuan, Tian, Lili, Chen, Jinyan, Zhang, Can, Huang, Zhangxiang, Yu, Weifeng, and Xie, Kangjie
- Subjects
NON-small-cell lung carcinoma ,CELL migration ,LUNG cancer ,CADHERINS ,CANCER invasiveness - Abstract
Morphine has been suggested to affect cancer cell dynamics and decrease survival rates in lung cancer patients at specific doses, but the precise mechanisms poorly understood. In this study, we aimed to investigate the molecular mechanisms by which morphine modulates the malignant characteristics of non-small cell lung cancer. Cell proliferation was assessed via the Cell Counting Kit-8 assay, and cell migration and invasion were examined via wound healing and Transwell assays. We employed immunofluorescence staining to evaluate E-cadherin expression in A549 and Lewis lung cancer (LLC) cell lines and immunohistochemistry to evaluate E-cadherin expression in nude mice tumours. Additionally, the in vivo effects of morphine on lung cancer progression were explored in a xenograft tumour experiments, in which naloxone was used as a morphine antagonist. Western blot analysis was performed to detect E-cadherin, phosphorylated mTOR (p-mTOR), mTOR, phosphorylated AKT (p-AKT), AKT, phosphorylated PI3K (p-PI3K), and PI3K protein levels in A549 and LLC cells as well as in tumour samples. Morphine (10 µM) significantly increased the proliferation of A549 and LLC cells in vitro (p < 0.05). It also enhanced the migratory and invasive capacities of these cell lines (p < 0.01). Mechanistically, morphine treatment (10 µM) led to a reduction in the expression of E-cadherin, and an increase in the phosphorylation of PI3K, AKT, and mTOR in A549 and LLC cells (p < 0.01). Morphine treatment (1.5 mg/kg) also reduced E-cadherin expression in xenograft tumours and promoted tumour growth in vivo (p < 0.05). This effect was reversed by naloxone (0.1 mg/kg). The results demonstrated that morphine stimulates the malignant proliferation of A549 and LLC cell lines and promotes xenograft tumour growth. Perhaps by specifically targeting MOR, morphine triggers a signalling cascade that activates the PI3K/AKT/mTOR pathway while inhibiting the EMT marker E-cadherin, which may consequently promote the progression of lung cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Characterization of novel nitazene recreational drugs: Insights into their risk potential from in vitro µ-opioid receptor assays and in vivo behavioral studies in mice
- Author
-
Marthe M. Vandeputte, Grant C. Glatfelter, Donna Walther, Nathan K. Layle, Danielle M. St. Germaine, István Ujváry, Donna M. Iula, Michael H. Baumann, and Christophe P. Stove
- Subjects
Nitazene ,New synthetic opioids ,In vitro and in vivo pharmacology ,Structure-activity relationships ,2-Benzylbenzimidazoles ,µ-Opioid receptor ,Therapeutics. Pharmacology ,RM1-950 - Abstract
2-Benzylbenzimidazole derivatives or ‘nitazenes’ are increasingly present on the recreational drug market. Here, we report the synthesis and pharmacological characterization of 15 structurally diverse nitazenes that might be predicted to emerge or grow in popularity. This work expands the existing knowledge about 2-benzylbenzimidazole structure-activity relationships (SARs), while also helping stakeholders (e.g., forensic toxicologists, clinicians, policymakers) in their risk assessment and preparedness for the potential next generation of nitazenes. In vitro µ-opioid receptor (MOR) affinity was determined via competition radioligand (3[H]DAMGO) binding assays in rat brain tissue. MOR activation (potency and efficacy) was studied by means of a cell-based β-arrestin 2 recruitment assay. For seven nitazenes, including etonitazene, opioid-like pharmacodynamic effects (antinociception, locomotor activity, body temperature changes) were evaluated after subcutaneous administration in male C57BL/6 J mice. The results showed that all nitazenes bound to MOR with nanomolar affinities, and the functional potency of several of them was comparable to or exceeded that of fentanyl. In vivo, dose-dependent effects were observed for antinociception, locomotor activity, and body temperature changes in mice. SAR insights included the high opioid-like activity of methionitazene, iso-butonitazene, sec-butonitazene, and the etonitazene analogues 1-ethyl-pyrrolidinylmethyl N-desalkyl etonitazene and ethylene etonitazene. The most potent analogue of the panel across all functional assays was α’-methyl etonitazene. Taken together, through critical pharmacological evaluation, this work provides a framework for strengthened preparedness and risk assessments of current and future nitazenes that have the potential to cause harm to users.
- Published
- 2024
- Full Text
- View/download PDF
20. ANALYSIS OF THE DOCKING RESULTS OF SOME SELECTIVE MOR LIGANDS.
- Author
-
Dzimbova, Tatyana, Sapundzhi, Fatima, and Milanov, Peter
- Subjects
- *
OPIOID receptors , *RECEPTOR-ligand complexes , *OPIOID peptides , *AMINO acid residues , *BINDING sites - Abstract
Endogenous opioids produce the same effects as the chemicals known as classic alkaloid opiates, which include morphine and heroin. Endogenous opioid peptides function both as hormones and as neuromodulators. The aim of the present study was to analyze the results of docking of ligands with MOR to identify the key elements required for selectivity. Many of the ligands have been synthesized and biologically tested by our colleagues. The other part is compounds known in the literature. The analysis of the obtained ligand-receptor complexes makes it possible to determine the key structural elements associated with the manifestation of specificity with respect to the receptor. These results will assist in the design of new compounds with potential MOR agonistic or antagonistic effects. In order to be active and effective, a ligand must have certain properties. First, it must be stable in a biological environment so that it can reach the place where it will manifest its action. Second, be of a suitable structure to allow it to reach and interact with the receptor's binding site. Third, upon binding, the resulting ligand-receptor complex should be stable, i.e. its energy to be small. Fourth, the ligand induces the appropriate conformations in the receptor molecule upon interaction, i.e. to bind to precisely defined amino acid residues. Therefore, the present study aims to analyze the docking results of dalargine derivatives with MOR and determine the necessary conditions for the manifestation of the biological effect. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Covalent and Visible‐Light Photoswitchable Derivatives of the Potent Synthetic Opioid Isotonitazene and Other Nitazenes.
- Author
-
Lahmy, Ranit, Hübner, Harald, Gmeiner, Peter, and König, Burkhard
- Subjects
- *
VISIBLE spectra , *OPIOIDS - Abstract
Isotonitazene belongs to a potent class of μ‐opioid receptor (μOR) ligands, known as nitazenes. The lack of knowledge surrounding this agonist and others in its class has sparked thorough re‐investigations. To aid in these investigations, the purportedly covalent yet underexplored nitazene BIT was biochemically re‐evaluated in this work, along with a newly synthesized analogue, Iso‐BIT. Moreover, in the pursuit of understanding the mechanism, function and interactions of the μOR, this study involved developing photoswitchable nitazene derivatives as potential probe molecules. Converting known ligands into azo‐containing photoswitchable derivatives offers the opportunity to modulate ligand structure with light, allowing for photocontrol of compound activity. While photocontrol of μOR activity could not be entirely achieved, photophysical evaluation of these 2‐benzimidazole azo‐arenes revealed a novel photoswitch scaffold that responds to visible light. Furthermore, azo‐containing 2 e and 3 e emerged as promising nitazene derivatives that were able to form an exceptionally high fraction of covalent‐ligand receptor complexes with wild‐type μOR at physiological pH. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Spinal-Supraspinal and Intrinsic µ-Opioid Receptor Agonist-Norepinephrine Reuptake Inhibitor (MOR-NRI) Synergy of Tapentadol in Diabetic Heat Hyperalgesia in Mice
- Author
-
Christoph, Thomas, Schröder, Wolfgang, Tallarida, Ronald J., De Vry, Jean, and Tzschentke, Thomas M.
- Published
- 2013
- Full Text
- View/download PDF
23. Novel Screening Assay for the Selective Detection of G-Protein-Coupled Receptor Heteromer Signaling
- Author
-
van Rijn, Richard M., Harvey, Jessica H., Brissett, Daniela I., DeFriel, Julia N., and Whistler, Jennifer L.
- Published
- 2013
- Full Text
- View/download PDF
24. Stereoselective recognition of morphine enantiomers by μ-opioid receptor.
- Author
-
Wang, Yibo, Ngo, Van A, and Wang, Xiaohui
- Subjects
- *
OPIOID receptors , *ENANTIOMERS , *MOLECULAR dynamics , *MORPHINE , *MOLECULAR recognition , *CHIRAL recognition - Abstract
Stereospecific recognition of chiral molecules plays a crucial role in biological systems. The μ-opioid receptor (MOR) exhibits binding affinity towards (−)-morphine, a well-established gold standard in pain management, while it shows minimal binding affinity for the (+)-morphine enantiomer, resulting in a lack of analgesic activity. Understanding how MOR stereoselectively recognizes morphine enantiomers has remained a puzzle in neuroscience and pharmacology for over half-a-century due to the lack of direct observation techniques. To unravel this mystery, we constructed the binding and unbinding processes of morphine enantiomers with MOR via molecular dynamics simulations to investigate the thermodynamics and kinetics governing MOR's stereoselective recognition of morphine enantiomers. Our findings reveal that the binding of (−)-morphine stabilizes MOR in its activated state, exhibiting a deep energy well and a prolonged residence time. In contrast, (+)-morphine fails to sustain the activation state of MOR. Furthermore, the results suggest that specific residues, namely D1142.50 and D1473.32, are deprotonated in the active state of MOR bound to (−)-morphine. This work highlights that the selectivity in molecular recognition goes beyond binding affinities, extending into the realm of residence time. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Ubiquitination of G Protein-Coupled Receptors: Functional Implications and Drug Discovery
- Author
-
Dores, Michael R. and Trejo, JoAnn
- Published
- 2012
- Full Text
- View/download PDF
26. Prolonged Stimulation of μ-Opioid Receptors Produces β-Arrestin-2-Mediated Heterologous Desensitization of α2-Adrenoceptor Function in Locus Ceruleus Neurons
- Author
-
Dang, Vu C., Chieng, Billy C., and Christie, MacDonald J.
- Published
- 2012
- Full Text
- View/download PDF
27. Signaling Cascades for δ-Opioid Receptor-Mediated Inhibition of GABA Synaptic Transmission and Behavioral Antinociception
- Author
-
Zhang, Zhi and Pan, Zhizhong Z.
- Published
- 2012
- Full Text
- View/download PDF
28. Desensitization and Trafficking of μ-Opioid Receptors in Locus Ceruleus Neurons: Modulation by Kinases
- Author
-
Arttamangkul, Seksiri, Lau, Elaine K., Lu, Hsin-Wei, and Williams, John T.
- Published
- 2012
- Full Text
- View/download PDF
29. The role of endogenous opioid neuropeptides in neurostimulation-driven analgesia.
- Author
-
Lubejko, Susan, Graham, Robert, Schaefer, Robert, Creed, Meaghan, Banghart, Matthew, and Livrizzi, Giulia
- Subjects
analgesia ,deep brain stimulation (DBS) ,neuromodulation ,neurostimulation ,opioid ,pain ,spinal cord stimulation (SCS) ,μ-opioid receptor - Abstract
Due to the prevalence of chronic pain worldwide, there is an urgent need to improve pain management strategies. While opioid drugs have long been used to treat chronic pain, their use is severely limited by adverse effects and abuse liability. Neurostimulation techniques have emerged as a promising option for chronic pain that is refractory to other treatments. While different neurostimulation strategies have been applied to many neural structures implicated in pain processing, there is variability in efficacy between patients, underscoring the need to optimize neurostimulation techniques for use in pain management. This optimization requires a deeper understanding of the mechanisms underlying neurostimulation-induced pain relief. Here, we discuss the most commonly used neurostimulation techniques for treating chronic pain. We present evidence that neurostimulation-induced analgesia is in part driven by the release of endogenous opioids and that this endogenous opioid release is a common endpoint between different methods of neurostimulation. Finally, we introduce technological and clinical innovations that are being explored to optimize neurostimulation techniques for the treatment of pain, including multidisciplinary efforts between neuroscience research and clinical treatment that may refine the efficacy of neurostimulation based on its underlying mechanisms.
- Published
- 2022
30. Phosphorylation of the μ-Opioid Receptor at Tyrosine 166 (Tyr3.51) in the DRY Motif Reduces Agonist Efficacy
- Author
-
Clayton, Cecilea C., Bruchas, Michael R., Lee, Michael L., and Chavkin, Charles
- Published
- 2010
- Full Text
- View/download PDF
31. Opioid-Induced Hyperalgesia and Tolerance Are Driven by HCN Ion Channels.
- Author
-
Xue Han, Pinto, Larissa Garcia, Vilar, Bruno, and McNaughton, Peter A.
- Subjects
- *
ION channels , *NOCICEPTORS , *HYPERALGESIA , *GENE expression , *SPINAL cord , *TREATMENT of addictions - Abstract
Prolonged exposure to opioids causes an enhanced sensitivity to painful stimuli (opioid-induced hyperalgesia, OIH) and a need for increased opioid doses to maintain analgesia (opioid-induced tolerance, OIT), but the mechanisms underlying both processes remain obscure. We found that pharmacological block or genetic deletion of HCN2 ion channels in primary nociceptive neurons of male mice completely abolished OIH but had no effect on OIT. Conversely, pharmacological inhibition of central HCN channels alleviated OIT but had no effect on OIH. Expression of C-FOS, a marker of neuronal activity, was increased in second-order neurons of the dorsal spinal cord by induction of OIH, and the increase was prevented by peripheral block or genetic deletion of HCN2, but block of OIT by spinal block of HCN channels had no impact on C-FOS expression in dorsal horn neurons. Collectively, these observations show that OIH is driven by HCN2 ion channels in peripheral nociceptors, while OIT is driven by a member of the HCN family located in the CNS. Induction of OIH increased cAMP in nociceptive neurons, and a consequent shift in the activation curve of HCN2 caused an increase in nociceptor firing. The shift in HCN2 was caused by expression of a constitutively active μ-opioid receptor (MOR) and was reversed by MOR antagonists. We identified the opioid-induced MOR as a six-transmembrane splice variant, and we show that it increases cAMP by coupling constitutively to Gs. HCN2 ion channels therefore drive OIH, and likely OIT, and may be a novel therapeutic target for the treatment of addiction. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Neural basis of opioid-induced respiratory depression and its rescue
- Author
-
Liu, Shijia, Kim, Dong-Il, Oh, Tae Gyu, Pao, Gerald M, Kim, Jong-Hyun, Palmiter, Richard D, Banghart, Matthew R, Lee, Kuo-Fen, Evans, Ronald M, and Han, Sung
- Subjects
Neurosciences ,Lung ,Neurological ,Good Health and Well Being ,Analgesics ,Opioid ,Animals ,Mice ,Mice ,Transgenic ,Morphine ,Neurons ,Receptors ,Opioid ,mu ,Respiratory Insufficiency ,parabrachial nucleus ,mu-opioid receptor ,OIRD ,μ-opioid receptor - Abstract
Opioid-induced respiratory depression (OIRD) causes death following an opioid overdose, yet the neurobiological mechanisms of this process are not well understood. Here, we show that neurons within the lateral parabrachial nucleus that express the µ-opioid receptor (PBL Oprm1 neurons) are involved in OIRD pathogenesis. PBL Oprm1 neuronal activity is tightly correlated with respiratory rate, and this correlation is abolished following morphine injection. Chemogenetic inactivation of PBL Oprm1 neurons mimics OIRD in mice, whereas their chemogenetic activation following morphine injection rescues respiratory rhythms to baseline levels. We identified several excitatory G protein-coupled receptors expressed by PBL Oprm1 neurons and show that agonists for these receptors restore breathing rates in mice experiencing OIRD. Thus, PBL Oprm1 neurons are critical for OIRD pathogenesis, providing a promising therapeutic target for treating OIRD in patients.
- Published
- 2021
33. Repurposing EGFR Inhibitors for Oral Cancer Pain and Opioid Tolerance.
- Author
-
Santi, Maria Daniela, Zhang, Morgan, Liu, Naijiang, Viet, Chi T., Xie, Tongxin, Jensen, Dane D., Amit, Moran, Pan, Huilin, and Ye, Yi
- Subjects
- *
CANCER pain , *PAIN tolerance , *ORAL cancer , *EPIDERMAL growth factor receptors , *METHYL aspartate receptors , *PAIN threshold - Abstract
Oral cancer pain remains a significant public health concern. Despite the development of improved treatments, pain continues to be a debilitating clinical feature of the disease, leading to reduced oral mobility and diminished quality of life. Opioids are the gold standard treatment for moderate-to-severe oral cancer pain; however, chronic opioid administration leads to hyperalgesia, tolerance, and dependence. The aim of this review is to present accumulating evidence that epidermal growth factor receptor (EGFR) signaling, often dysregulated in cancer, is also an emerging signaling pathway critically involved in pain and opioid tolerance. We presented preclinical and clinical data to demonstrate how repurposing EGFR inhibitors typically used for cancer treatment could be an effective pharmacological strategy to treat oral cancer pain and to prevent or delay the development of opioid tolerance. We also propose that EGFR interaction with the µ-opioid receptor and glutamate N-methyl-D-aspartate receptor could be two novel downstream mechanisms contributing to pain and morphine tolerance. Most data presented here support that repurposing EGFR inhibitors as non-opioid analgesics in oral cancer pain is promising and warrants further research. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
34. Mu opioid receptors on distinct neuronal populations mediate different aspects of opioid reward-related behaviors
- Author
-
Severino, Amie L, Mittal, Nitish, Hakimian, Joshua K, Velarde, Nathanial, Minasyan, Ani, Albert, Ralph, Torres, Carlos, Romaneschi, Nicole, Johnston, Camille, Tiwari, Suchi, Lee, Alex S, Taylor, Anna M, Gavériaux-Ruff, Claire, Kieffer, Brigitte L, Evans, Christopher J, Cahill, Catherine M, and Walwyn, Wendy M
- Subjects
Behavioral and Social Science ,Neurosciences ,Basic Behavioral and Social Science ,Brain Disorders ,Substance Misuse ,Drug Abuse (NIDA only) ,Neurological ,Good Health and Well Being ,Analgesics ,Opioid ,Animals ,Mice ,Morphine ,Neurons ,Receptors ,Opioid ,mu ,Reward ,floxed MOR ,hyperlocomotion ,intravenous self-administration ,morphine ,mu-opioid receptor ,oxycodone ,μ-opioid receptor - Abstract
μ-Opioid receptors (MORs) are densely expressed in different brain regions known to mediate reward. One such region is the striatum where MORs are densely expressed, yet the role of these MOR populations in modulating reward is relatively unknown. We have begun to address this question by using a series of genetically engineered mice based on the Cre recombinase/loxP system to selectively delete MORs from specific neurons enriched in the striatum: dopamine 1 (D1) receptors, D2 receptors, adenosine 2a (A2a) receptors, and choline acetyltransferase (ChAT). We first determined the effects of each deletion on opioid-induced locomotion, a striatal and dopamine-dependent behavior. We show that MOR deletion from D1 neurons reduced opioid (morphine and oxycodone)-induced hyperlocomotion, whereas deleting MORs from A2a neurons resulted in enhanced opioid-induced locomotion, and deleting MORs from D2 or ChAT neurons had no effect. We also present the effect of each deletion on opioid intravenous self-administration. We first assessed the acquisition of this behavior using remifentanil as the reinforcing opioid and found no effect of genotype. Mice were then transitioned to oxycodone as the reinforcer and maintained here for 9 d. Again, no genotype effect was found. However, when mice underwent 3 d of extinction training, during which the drug was not delivered, but all cues remained as during the maintenance phase, drug-seeking behavior was enhanced when MORs were deleted from A2a or ChAT neurons. These findings show that these selective MOR populations play specific roles in reward-associated behaviors.
- Published
- 2020
35. Coarse-Grained MD Simulations of Opioid Interactions with the μ-Opioid Receptor and the Surrounding Lipid Membrane
- Author
-
Sourav Ray, Konstantin Fackeldey, Christoph Stein, and Marcus Weber
- Subjects
coarse-grain ,opioid ,μ-opioid receptor ,membrane bilayer ,molecular dynamics ,SqrA ,Biology (General) ,QH301-705.5 - Abstract
In our previous studies, a new opioid (NFEPP) was developed to only selectively bind to the μ-opoid receptor (MOR) in inflamed tissue and thus avoid the severe side effects of fentanyl. We know that NFEPP has a reduced binding affinity to MOR in healthy tissue. Inspired by the modelling and simulations performed by Sutcliffe et al., we present our own results of coarse-grained molecular dynamics simulations of fentanyl and NFEPP with regards to their interaction with the μ-opioid receptor embedded within the lipid cell membrane. For technical reasons, we have slightly modified Sutcliffe’s parametrisation of opioids. The pH-dependent opioid simulations are of interest because while fentanyl is protonated at the physiological pH, NFEPP is deprotonated due to its lower pKa value than that of fentanyl. Here, we analyse for the first time whether pH changes have an effect on the dynamical behaviour of NFEPP when it is inside the cell membrane. Besides these changes, our analysis shows a possible alternative interaction of NFEPP at pH 7.4 outside the binding region of the MOR. The interaction potential of NFEPP with MOR is also depicted by analysing the provided statistical molecular dynamics simulations with the aid of an eigenvector analysis of a transition rate matrix. In our modelling, we see differences in the XY-diffusion profiles of NFEPP compared with fentanyl in the cell membrane.
- Published
- 2023
- Full Text
- View/download PDF
36. Role for μ-opioid receptor in antidepressant effects of δ-opioid receptor agonist KNT-127
- Author
-
Yuki Moriya, Yoshiyuki Kasahara, Masafumi Shimada, Yasufumi Sakakibara, Hideaki Fujii, Hiroshi Nagase, Soichiro Ide, Kazutaka Ikeda, F. Scott Hall, George R. Uhl, and Ichiro Sora
- Subjects
Antidepressant ,μ-opioid receptor ,δ-opioid receptor agonist ,Anxiolytic ,Lack of motivated behavior ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Previous pharmacological data have shown the possible existence of functional interactions between μ- (MOP), κ- (KOP), and δ-opioid receptors (DOP) in pain and mood disorders. We previously reported that MOP knockout (KO) mice exhibit a lower stress response compared with wildtype (WT) mice. Moreover, DOP agonists have been shown to exert antidepressant-like effects in numerous animal models. In the present study, the tail suspension test (TST) and forced swim test (FST) were used to examine the roles of MOP and DOP in behavioral despair. MOP-KO mice and WT mice were treated with KNT-127 (10 mg/kg), a selective DOP agonist. The results indicated a significant decrease in immobility time in the KNT-127 group compared with the saline group in all genotypes in both tests. In the saline groups, immobility time significantly decreased in MOP-KO mice compared with WT mice in both tests. In female MOP-KO mice, KNT-127 significantly decreased immobility time in the TST compared with WT mice. In male MOP-KO mice, however, no genotypic differences were found in the TST after either KNT-127 or saline treatment. Thus, at least in the FST and TST, the activation of DOP and absence of MOP had additive effects in reducing measures of behavioral despair, suggesting that effects on this behavior by DOP activation occur independently of MOP.
- Published
- 2023
- Full Text
- View/download PDF
37. Targeting Peripheral μ-opioid Receptors or μ-opioid Receptor-Expressing Neurons Does not Prevent Morphine-induced Mechanical Allodynia and Anti-allodynic Tolerance.
- Author
-
Du, Feng, Yin, Guangjuan, Han, Lei, Liu, Xi, Dong, Dong, Duan, Kaifang, Huo, Jiantao, Sun, Yanyan, and Cheng, Longzhen
- Abstract
The chronic use of morphine and other opioids is associated with opioid-induced hypersensitivity (OIH) and analgesic tolerance. Among the different forms of OIH and tolerance, the opioid receptors and cell types mediating opioid-induced mechanical allodynia and anti-allodynic tolerance remain unresolved. Here we demonstrated that the loss of peripheral μ-opioid receptors (MORs) or MOR-expressing neurons attenuated thermal tolerance, but did not affect the expression and maintenance of morphine-induced mechanical allodynia and anti-allodynic tolerance. To confirm this result, we made dorsal root ganglia-dorsal roots-sagittal spinal cord slice preparations and recorded low-threshold Aβ-fiber stimulation-evoked inputs and outputs in superficial dorsal horn neurons. Consistent with the behavioral results, peripheral MOR loss did not prevent the opening of Aβ mechanical allodynia pathways in the spinal dorsal horn. Therefore, the peripheral MOR signaling pathway may not be an optimal target for preventing mechanical OIH and analgesic tolerance. Future studies should focus more on central mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
38. μ-阿片受体基因多态性与带状疱疹后神经痛患者 疼痛敏感性及阿片类药物用量的关系.
- Author
-
朱琨
- Abstract
Objective To explore the associations of the polymorphism of μ - opioid receptor gene (OPRM1) with pain sensitivity and opioid dosage in patients with postherpetic neuralgia (PHN). Methods The clinical data of 170 herpes zoster (HZ) patients who visited the pain clinic of our hospital from January 2019 to July 2022 were reviewed, 90 of them put in the PHN group and 80 put in the non-PHN group according to the presence or absence of postherpetic neuralgia 3 months after HZ onset. The factors influencing the occurrence of PHN were analyzed by Logistic regression analysis. The two groups were compared in terms of 48h opioid dosage, sleep quality score and state trait anxiety score among different genotypes of PHN patients with moderate and severe pains. #The independent risk factors for PHN were found to include initial treatment > 72 hours, severe pain in acute stage and genotype GG. The frequency of GG genotype distribution was significantly increased across the PHN mild pain, moderate pain and severe pain groups. As compared with the PHN moderate group, the 48 h dosage of opioids in the patients with different genotypes in the PHN moderate and severe pain group were significantly increased (P < 0.05), with the highest in the GG genotype patients. Conclusion The independent risk factors for PHN are initial treatment >72 h, acute severe pain and genotype GG. The polymorphism of OPRM1 gene is the genetic factor that causes individual differences of opioid pharmacodynamics in PHN patients. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. 3a-(4-Chlorophenyl)-1-methyl-3a,4-dihydroimidazo[1,5- a ]quinazolin-5(3 H)-one: Synthesis and In Silico Evaluation as a Ligand in the µ-Opioid Receptor.
- Author
-
Defant, Andrea, Innocenti, Nicole, and Mancini, Ines
- Subjects
- *
OPIOID receptors , *BLOOD-brain barrier , *BIOMOLECULES , *MORPHINE , *ENANTIOMERS , *FENTANYL , *SYNTHETIC biology - Abstract
In the search for fused heterocycle molecules with potential biological activities, the new title compound was produced in racemic form via a four step-synthetic sequence with an overall yield of 60%. It was structurally characterised via 1H-, 13C-NMR and IR analyses, and the molecular composition was confirmed through a high-resolution MS experiment. After predicting its analgesic activity using PASS online software, wherein a good overlap between its enantiomers and the structure of the natural opioid morphine was observed, the compound was evaluated through docking calculations as a ligand of the µ-opioid receptor. The resulting energy values and interactions were comparable to the data obtained for morphine and its synthetic derivative fentanyl, which is used in the therapeutic treatment of severe forms of pain. Moreover, the title compound displayed favourable predicted blood–brain barrier permeation and drug-likeness. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Effect of Exercise on β-Endorphin and Its Receptors in Myasthenia Gravis Patients.
- Author
-
Tripathi, Gyanesh M., Misra, Usha K., Kalita, Jayantee, Singh, Varun K., and Tripathi, Abhilasha
- Abstract
This is a prospective observational study evaluating the change in β-endorphin (BE) and its receptors following exercise in patients with myasthenia gravis (MG) and their association with clinical improvement. Fifteen patients with mild to moderate MG, aged 16–70 years, who were able to do 6-Minute Walk Test (6-MWT) and had MG Quality of Life-15 (MGQoL-15) ≤ 45 without any contraindication for exercise were included. The patients walked 30 min daily for 3 months. The primary outcome at 3 months was > 50% improvement in MGQoL-15 from the baseline, and the secondary outcomes were MG Activities of Daily Living (MGADL), Hospital Anxiety and Depression Scale (HADS), number of steps, and distance covered on 6-MWT and adverse events. Plasma BE level, μ-opioid receptor (MOR), and δ-opioid receptor (DOR) were measured on admission and at 1 and 3 months. Twelve age- and gender-matched healthy controls who were not on regular exercise were included for comparison of BE, MOR, and DOR levels. Plasma BE level (P = 0.007) and DOR expression (P = 0.001) were lower in MG patients compared to the healthy controls. After 3 months of exercise, 6 patients improved. Plasma BE, MOR, and DOR levels increased in the first and decreased in the third month. MGQoL-15 (P < 0.001), HADS (P < 0.0001), number of steps (P < 0.007), distance (P = 0.030), and MGADL (P < 0.001) significantly improved compared to baseline. At 3 months, MGQoL-15 was associated with HADS score (P = 0.001), reduced depression (P = 0.013), MGADL (P = 0.035), and distance travelled on the 6-WMW test (P = 0.050). The improvement in depression was associated with higher BE level. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
41. Coarse-Grained MD Simulations of Opioid Interactions with the μ -Opioid Receptor and the Surrounding Lipid Membrane.
- Author
-
Ray, Sourav, Fackeldey, Konstantin, Stein, Christoph, and Weber, Marcus
- Subjects
BILAYER lipid membranes ,OPIOID receptors ,FENTANYL ,MOLECULAR dynamics ,CELL membranes - Abstract
In our previous studies, a new opioid (NFEPP) was developed to only selectively bind to the μ -opoid receptor (MOR) in inflamed tissue and thus avoid the severe side effects of fentanyl. We know that NFEPP has a reduced binding affinity to MOR in healthy tissue. Inspired by the modelling and simulations performed by Sutcliffe et al., we present our own results of coarse-grained molecular dynamics simulations of fentanyl and NFEPP with regards to their interaction with the μ -opioid receptor embedded within the lipid cell membrane. For technical reasons, we have slightly modified Sutcliffe's parametrisation of opioids. The pH-dependent opioid simulations are of interest because while fentanyl is protonated at the physiological pH, NFEPP is deprotonated due to its lower pK
a value than that of fentanyl. Here, we analyse for the first time whether pH changes have an effect on the dynamical behaviour of NFEPP when it is inside the cell membrane. Besides these changes, our analysis shows a possible alternative interaction of NFEPP at pH 7.4 outside the binding region of the MOR. The interaction potential of NFEPP with MOR is also depicted by analysing the provided statistical molecular dynamics simulations with the aid of an eigenvector analysis of a transition rate matrix. In our modelling, we see differences in the XY-diffusion profiles of NFEPP compared with fentanyl in the cell membrane. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
42. Optogenetic activation of β-endorphin terminals in the medial preoptic nucleus regulates sexual receptivity
- Author
-
Johnson, Caroline, Hong, Weizhe, and Micevych, Paul
- Subjects
Biological Psychology ,Biomedical and Clinical Sciences ,Psychology ,Neurosciences ,Estrogen ,Substance Misuse ,Drug Abuse (NIDA only) ,Animals ,Arcuate Nucleus of Hypothalamus ,Estradiol ,Female ,Mice ,Optogenetics ,Posture ,Preoptic Area ,Progesterone ,Rats ,Rats ,Long-Evans ,Sexual Behavior ,Animal ,beta-Endorphin ,beta-endorphin ,estradiol ,lordosis ,mu-opioid receptor ,POMC ,β-endorphin ,μ-opioid receptor - Abstract
Estrogen and progesterone (P4) act in neural circuits to elicit lordosis, the stereotypical female sexual receptivity behavior. Estradiol acts through membrane receptors to rapidly activate a limbic-hypothalamic circuit consisting of the arcuate (ARH), medial preoptic (MPN), and ventromedial (VMH) nuclei of the hypothalamus. This initial activation results in a transient but necessary inhibition of lordosis, which appears to be a result of the release of β-endorphin (β-End) from proopiomelanocortin (POMC) terminals onto cells containing the µ-opioid receptor (MOR) in the MPN. To functionally examine the role of the MOR in the hypothalamic lordosis circuit, we transfected a channelrhodopsin (ChR2) adeno-associated virus into POMC cell bodies in the ARH and photostimulated POMC/β-End axon terminals in the MPN in sexually receptive female Pomc-cre mice. Following estrogen and P4 priming, sexual receptivity was assessed by measuring the lordosis quotient (LQ). Following an initial trial for sexual receptivity, mice were photostimulated during behavioral testing, and brains were processed for MOR immunohistochemistry (IHC). Photostimulation decreased the LQ only in ChR2-expressing Pomc-cre mice. Furthermore, photostimulation of ChR2 in POMC/β-End axon terminals in the MPN resulted in the internalization of MOR, indicating activation of the receptor. Our results suggest that the activation of the MOR in the MPN is sufficient to attenuate lordosis behavior in a hormone-primed, sexually receptive female mouse. These data support a central role of MOR in female sexual behavior, and provide further insight into the hypothalamus control of sexual receptivity.
- Published
- 2020
43. Lactobacillus rhamnosus GG and butyrate supplementation in rats with bone cancer reduces mechanical allodynia and increases expression of μ-opioid receptor in the spinal cord
- Author
-
Wenxi Yuan, Jie Xiao, Huabao Liao, Zhiyuan Xie, Yiran Zhao, Cheng Li, Keying Zhou, and Xue-Jun Song
- Subjects
Lactobacillus rhamnosus GG ,cancer pain ,mechanical allodynia ,μ-opioid receptor ,gut microbiota ,butyrate ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
IntroductionChronic cancer pain is one of the most unbearable symptoms for the patients with advanced cancer. The treatment of cancer pain continues to possess a major challenge. Here, we report that adjusting gut microbiota via probiotics can reduce bone cancer pain (BCP) in rats.MethodsThe model of BCP was produced by tumor cell implantation (TCI) to the tibia in rats. Continuous feeding of Lactobacillus rhamnosus GG (LGG) was used to modulate the gut microbiota. Mechanical allodynia, bone destruction, fecal microbiota, and neurochemical changes in the primary dorsal root ganglion (DRG) and the spinal dorsal horn (DH) were assessed.ResultsLGG supplementation (109 CFU/rat/day) delayed the production of BCP for 3–4 days and significantly alleviated mechanical allodynia within the first 2 weeks after TCI. TCI-induced proinflammatory cytokines TNF-α and IL-β in the DH, and TCI-induced bone destruction in the tibia were both significantly reduced following LGG supplementation examined on day 8 after TCI. Meanwhile, we found that LGG supplementation, in addition to inhibiting TCI-induced pain, resulted in a significantly increased expression of the μ-opioid receptor (MOR) in the DH, but not in the DRG. LGG supplementation significantly potentiated the analgesic effect of morphine. Furthermore, LGG supplementation led to an increase in butyrate levels in the feces and serum and a decrease in histone deacetylase 2 (HDAC2) expression in the DH. Feeding TCI-rats with sodium butyrate solution alone, at a dose of 100 mg/kg, resulted in decreased pain, as well as decreased HDAC2 expression and increased MOR expression in the DH. The increased expression of MOR and decreased HDAC2 were also observed in neuro-2a cells when we treated the cells with serum from TCI rats with supplementation of LGG or sodium butyrate.DiscussionThis study provides evidence that reshaping the gut microbiota with probiotics LGG can delay the onset of cancer pain. The butyrate-HDAC2-MOR pathway may be the underlying mechanism for the analgesic effect of LGG. These findings shed light on an effective, safe, and non-invasive approach for cancer pain control and support the clinical implication of probiotics supplementation for patients with BCP.
- Published
- 2023
- Full Text
- View/download PDF
44. The inhibition of enkephalin catabolism by dual enkephalinase inhibitor: A novel possible therapeutic approach for opioid use disorders.
- Author
-
Alvarez‐Perez, Beltran, Poras, Hervé, and Maldonado, Rafael
- Subjects
- *
OPIOID abuse , *OPIOID receptors , *THERAPEUTICS , *OPIOID peptides , *OPIOID epidemic , *ENKEPHALINS - Abstract
Despite the increasing impact of opioid use disorders on society, there is a disturbing lack of effective medications for their clinical management. An interesting innovative strategy to treat these disorders consists in the protection of endogenous opioid peptides to activate opioid receptors, avoiding the classical opioid‐like side effects. Dual enkephalinase inhibitors (DENKIs) physiologically activate the endogenous opioid system by inhibiting the enzymes responsible for the breakdown of enkephalins, protecting endogenous enkephalins and increasing their half‐lives and physiological actions. The activation of opioid receptors by the increased enkephalin levels, and their well‐demonstrated safety, suggests that DENKIs could represent a novel analgesic therapy and a possible effective treatment for acute opioid withdrawal, as well as a promising alternative to opioid substitution therapy minimizing side effects. This new pharmacological class of compounds could bring effective and safe medications avoiding the major limitations of exogenous opioids, representing a novel approach to overcome the problem of opioid use disorders. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
45. Sex‐specific effects of developmental morphine exposure and rearing environments on hippocampal spatial memory.
- Author
-
Sarkaki, Alireza, Mard, Seyed Ali, Bakhtiari, Nima, and Yazdanfar, Neda
- Subjects
- *
SPATIAL memory , *BRAIN-derived neurotrophic factor , *CYCLIC adenylic acid , *HIPPOCAMPUS (Brain) , *MORPHINE - Abstract
Developmental morphine (MOR) exposure (DME) detrimentally affects the cognitive abilities of the next generation. It is shown that postnatal rearing environments and prenatal conditions effectively impact memory. The present study investigated the effects of DME, postweaning rearing, and sex on spatial learning and memory. At molecular level, we evaluated mRNA levels of brain‐derived neurotrophic factor, cyclic AMP response element‐binding protein (CREB), μ‐opioid receptor, and ΔFosB in the hippocampus of male offspring. Female Wistar rats were treated with escalating doses of MOR or saline before mating, gestation, and lactation. On Postnatal Day 22, the male and female pups were divided into 12 groups and raised for 2 months under different conditions: standard, isolated (ISO), or enriched environment. Afterward, the Morris water maze task measured spatial learning and reference memory; rats were then sacrificed to assess hippocampus gene expressions. Results indicated the DME and isolated rearing increased latency to find the hidden platform in male offspring. DME was insignificant in female offspring, whereas rearing environments significantly altered escape latency in both sexes. We also found that the enriched environment upregulated the brain‐derived neurotrophic factor mRNA in both saline and MOR groups, whereas it downregulated the mRNA levels of CREB1, μ‐opioid receptor, and ΔFosB in the MOR group. In addition, the DME enhanced CREB1, μ‐opioid receptor, and ΔFosB gene expression in the MOR + isolated group. Our findings signified the effects of DME, rearing environment, and sex on the spatial learning abilities of offspring. Also, we showed that DME and rearing conditions could manipulate hippocampal neurochemistry. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. Analysis of highly potent synthetic opioid nitazene analogs and their positional isomers.
- Author
-
Kanamori, Tatsuyuki, Okada, Yuki, Segawa, Hiroki, Yamamuro, Tadashi, Kuwayama, Kenji, Tsujikawa, Kenji, and Iwata, Yuko T.
- Abstract
Four nitazenes (metonitazene, etonitazene, protonitazene, and isotonitazene), highly potent benzimidazole synthetic opioids, and their four nitro group positional isomers (isonitazenes) were synthesized and analyzed using infrared (IR) spectroscopy, gas chromatography/mass spectrometry (GC/MS), and liquid chromatography/mass spectrometry (LC/MS). In addition, the agonistic activity of all compounds at the human μ‐opioid receptor was measured using a cell‐based assay system. In the IR spectra, characteristic peaks for nitazenes and isonitazenes were observed. In GC/MS, all compounds were well separated on the chromatogram, although distinguishing nitazenes from the corresponding isonitazenes by electron ionization mass spectra was difficult. In LC/MS, all compounds were detected in both positive and negative modes of electrospray ionization. Characteristic fragment ions were observed in the product ion spectra of isonitazenes, enabling nitazenes to be distinguished from isonitazenes. All nitazenes tested demonstrated higher agonistic activity at the human μ‐opioid receptors than the synthetic opioid fentanyl. The agonistic activities of isonitazenes were 11–35 times lower than those of the corresponding nitazenes. However, iso‐etonitazene and iso‐isotonitazene showed moderate activity similar to that of fentanyl, indicating that these drugs could cause poisoning at a comparable level as fentanyl, if these drugs are abused in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
47. Elucidating the harm potential of brorphine analogues as new synthetic opioids: Synthesis, in vitro, and in vivo characterization.
- Author
-
Vandeputte, Marthe M., Bilel, Sabrine, Tirri, Micaela, Corli, Giorgia, Bassi, Marta, Layle, Nathan K., Fantinati, Anna, Walther, Donna, Iula, Donna M., Baumann, Michael H., Stove, Christophe P., and Marti, Matteo
- Subjects
- *
RADIOLIGAND assay , *RESPIRATORY insufficiency , *CHEMICAL structure , *OPIOIDS , *PIPERIDINE , *OPIOID receptors - Abstract
The emergence of new synthetic opioids (NSOs) has added complexity to recreational opioid markets worldwide. While NSOs with diverse chemical structures have emerged, brorphine currently remains the only NSO with a piperidine benzimidazolone scaffold. However, the emergence of new generations of NSOs, including brorphine analogues, can be anticipated. This study explored the pharmaco-toxicological, opioid-like effect profile of brorphine alongside its non-brominated analogue (orphine) and three other halogenated analogues (fluorphine, chlorphine, iodorphine). In vitro , radioligand binding assays in rat brain tissue indicated that all analogues bind to the μ-opioid receptor (MOR) with nM affinity. While analogues with smaller-sized substituents showed the highest MOR affinity, further in vitro characterization via two cell-based (HEK 293T) MOR activation (β-arrestin 2 and mini-G αi recruitment) assays indicated that chlorphine, brorphine, and iodorphine were generally the most active MOR agonists. None of the compounds showed significant in vitro biased agonism compared to hydromorphone. In vivo , we investigated the effects of intraperitoneal (IP) administration of the benzimidazolones (0.01–15 mg/kg) on mechanical and thermal antinociception in male CD-1 mice. Chlorphine and brorphine overall induced the highest levels of antinociception. Furthermore, the effects on respiratory changes induced by a fixed dose (15 mg/kg IP) of the compounds were investigated using non-invasive plethysmography. Fluorphine-, chlorphine-, and brorphine-induced respiratory depressant effects were the most pronounced. For some compounds, pretreatment with naloxone (6 mg/kg IP) could not reverse respiratory depression. Taken together, brorphine-like piperidine benzimidazolones are opioid agonists that have the potential to cause substantial harm to users should they emerge as NSOs. This article is part of the Special Issue on "Novel Synthetic Opioids (NSOs)". • Analogues of brorphine have the potential to emerge as new synthetic opioids. • Piperidine benzimidazolones are μ-opioid receptor agonists in vitro. • Chlorphine and brorphine induced the highest levels of antinociception in vivo. • Fluorphine, chlorphine, and brorphine induced pronounced respiratory depression. • Non-opioid-mediated effects may contribute to some of the in vivo effects. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Solid-Phase Synthesis of the Bicyclic Peptide OL-CTOP Containing Two Disulfide Bridges, and an Assessment of Its In Vivo μ-Opioid Receptor Antagonism after Nasal Administration.
- Author
-
Rayala, Ramanjaneyulu, Tiller, Annika, Majumder, Shahayra A., Stacy, Heather M., Eans, Shainnel O., Nedovic, Aleksandra, McLaughlin, Jay P., and Cudic, Predrag
- Subjects
- *
SOLID-phase synthesis , *INTRANASAL administration , *OPIOID receptors , *PEPTIDE synthesis , *CYCLIC peptides , *PEPTIDES - Abstract
New strategies facilitate the design of cyclic peptides which can penetrate the brain. We have designed a bicyclic peptide, OL-CTOP, composed of the sequences of a selective μ-opioid receptor antagonist, CTOP (f-cyclo(CYwOTX)T) (X = penicillamine, Pen; O = ornithine) and odorranalectin, OL (YASPK-cyclo(CFRYPNGVLAC)T), optimized its solid-phase synthesis and demonstrated its ability for nose-to-brain delivery and in vivo activity. The differences in reactivity of Cys and Pen thiol groups protected with trityl and/or acetamidomethyl protecting groups toward I2 in different solvents were exploited for selective disulfide bond formation on the solid phase. Both the single step and the sequential strategy applied to macrocyclization reactions generated the desired OL-CTOP, with the sequential strategy yielding a large quantity and better purity of crude OL-CTOP. Importantly, intranasally (i.n.s.) administered OL-CTOP dose-dependently antagonized the analgesic effect of morphine administered to mice through the intracerebroventricular route and prevented morphine-induced respiratory depression. In summary, the results demonstrate the feasibility of our solid-phase synthetic strategy for the preparation of the OL-CTOP bicyclic peptide containing two disulfide bonds and reveal the potential of odorranalectin for further modifications and the targeted delivery to the brain. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. Identification of the Putative Binding Site of a Benzimidazole Opioid (Etazene) and Its Metabolites at µ-Opioid Receptor: A Human Liver Microsomal Assay and Systematic Computational Study.
- Author
-
Chaturvedi, Krishna, Hewamanna, Isuru, Pandey, Pankaj, Khan, Washim, Wang, Yan-Hong, Chittiboyina, Amar G., Doerksen, Robert J., and Godfrey, Murrell
- Subjects
- *
OPIOID receptors , *BINDING sites , *LIQUID chromatography-mass spectrometry , *METABOLITES , *MOLECULAR dynamics , *LIVER microsomes - Abstract
The synthetic benzimidazole opioid etazene (which has a 70-times higher analgesic activity than morphine), a recreational drug, has gained popularity as a novel psychoactive substance (NPS) on the illegal/darknet market; however, no experimental information is available at the molecular level on the binding mechanism and putative binding site of etazene and its metabolites at the µ-opioid receptor (MOR). In the present study, we investigated the metabolism of etazene in human liver microsomes using ultra-high-performance liquid chromatography–mass spectrometry (UHPLC–MS). We also explored the possibilities of MOR activation by etazene and its metabolites by studying their binding mechanisms and interaction profiles at an active-state MOR model via molecular docking, binding free energy calculations, and all-atom molecular dynamics (MD) simulations. The putative metabolites of etazene were also predicted using the ADMET Predictor 10.1. The molecular docking studies and free energy calculations showed that etazene and its metabolites (M1, M2, and M5–M7) exhibited strong predicted binding affinity at MOR and showed overlapped binding orientation with MOR-bound agonist BU72, which was co-crystallized in the MOR X-ray crystal structure (PDB ID: 5C1M). MD also confirmed the stability of the MOR–etazene and MOR–M6 complexes. These results suggest that etazene and its metabolites may act as strong MOR agonists, highlighting the necessity of experimental validation. The insights from this study, such as key interactions between etazene and its metabolites and the MOR, will allow authorities to predict potential analogs and clarify the target–protein interactions associated with this illicit substance, granting advanced or rapid reactions to confiscating or banning potential emerging drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
50. Ubiquitination of the μ-opioid receptor regulates receptor internalization without affecting Gi/o-mediated intracellular signaling or receptor phosphorylation.
- Author
-
Miyoshi, Kentaro, Shimizu, Satoshi, Shiraki, Atsuko, and Egi, Moritoki
- Subjects
- *
OPIOID receptors , *POST-translational modification , *UBIQUITINATION , *PHOSPHORYLATION - Abstract
Opioids are highly potent analgesics but develop tolerance. Previous studies have focused on phosphorylation of the μ-opioid receptor as it is involved in maintaining cellular sensitivity via desensitization, recycling, and degradation of the activated receptor. Recently, ubiquitination, another form of posttranslational modification has attracted attention in terms of triggering intracellular signaling and regulation of the activated receptor. Here, we generated a ubiquitination-deficient mutant of the μ-opioid receptor to investigate whether ubiquitination is involved in driving G i/o -mediated analgesic signaling, receptor desensitization or subsequent receptor internalization. Our study shows that the G i/o pathway and receptor phosphorylation do not require ubiquitination. Instead, ubiquitination regulates the internalization efficiency and might help in promoting internalization of the desensitized MOP. • The activation of Gi/o-mediated analgesic pathway does not require MOP ubiquitination. • MOP phosphorylation does not require ubiquitination. • Ubiquitination of MOP affects receptor internalization. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.