Dissertations / Theses on the topic 'Opioid Receptors Signaling'

To see the other types of publications on this topic, follow the link: Opioid Receptors Signaling.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 28 dissertations / theses for your research on the topic 'Opioid Receptors Signaling.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse dissertations / theses on a wide variety of disciplines and organise your bibliography correctly.

1

ZHANG, SHENGWEN. "THE OPIOID RECEPTOR-LIKE RECEPTOR ORL1: SIGNALING AND INTERACTION WITH OPIOID RECEPTORS." University of Cincinnati / OhioLINK, 2002. http://rave.ohiolink.edu/etdc/view?acc_num=ucin1029419843.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Zhang, Shengwen. "The opioid receptor-like receptor ORL1 signaling and interaction with opioid receptors /." Cincinnati, Ohio : University of Cincinnati, 2002. http://rave.ohiolink.edu/etdc/view?acc%5Fnum=ucin1029419843.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Basti, Vida. "Ligand biased signaling of opioid agonists forphosphorylation and regulation of μ -opioid receptors." Thesis, Uppsala universitet, Farmakologi, 2013. http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-192584.

Full text
Abstract:
Abstract Ligand biased signaling of opioid agonists for phosphorylation andregulation of μ -opioid receptors Student: Vida Basti. Supervisor: Prof Macdonald Christie. Departmement of Neuropharmacology, The University of Sydney. Examiner: Prof Ingrid Nylander. Departement of Phamacology, University of Uppsala. Opioid drugs are of great use in the medical practise. The drugs are commonly prescribed formany types of illnesses, mostly in cases of pain management. Although opioids come withmany benefits they are causing a lot of problems as well. The side effects are many andamongst these is tolerance development which may lead to abuse and addiction. Because ofthe fast tolerance development in patients, higher doses up to 10 times the therapeutic doseare being prescribed. This is a major issue in today’s society and must be addressed.Scientists are trying to figure out the mechanism behind tolerance by comparing differenttypes opioid drugs. Some opioids causes tolerance in a much faster rate than others but it isstill uncertain why and what is causing this. Two of the most commonly prescribed opioidsare oxycodone and morphine and so in this rapport these opioids are compared with respectto their capability to cause internalization in neurons. In the experiments a positive control,DAMGO, is being used as well as a negative control. The method being used is an indirectmethod of immunohistochemistry on AtT20 transfected cell culture. The results show thatOxycodone seems to cause no internalization at all in comparison to the control.
APA, Harvard, Vancouver, ISO, and other styles
4

Xiang, Guoqing. "Signaling Through Homomeric and Heteromeric Cannabinoid CB1 receptors." VCU Scholars Compass, 2018. https://scholarscompass.vcu.edu/etd/5683.

Full text
Abstract:
Cannabis (Marijuana) has multiple effects on the human body, such as analgesia, euphoria and memory impairment. Delta-9 tetrahydrocannabinol (D9-THC), the active ingredient in cannabis, binds to cannabinoid receptors, seven-transmembrane G protein-coupled receptors (GPCRs) that mediate a variety of physiological functions. GPCRs were believed to function only in homomeric forms, however, recent findings show that different GPCRs can also form heteromeric complexes that may expand their signaling properties. In this study, we focused on Cannabinoid CB1 receptor (CB1R) heteromers with the mu-opioid receptor (MOR) and the Dopamine type 2 receptor (D2R), respectively. We utilized a variety of techniques, such as the calcium mobilization assay, a luciferase complementation assay and an electrophysiology assay to study the pharmacology of the CB1R-MOR and CB1R-D2R heteromers. Our data demonstrate that co-expression of CB1R enhances the Gi signaling through MOR and inhibits the beta-arrestin recruitment to MOR. We also show that co-application of CB1R ligands can further accentuate the MOR signaling modulation. Co-expression of a CB1R transmembrane domain 5 (TM5), but not a TM1, mini-gene abrogated the signaling change suggesting that it is likely due to heteromerization of MOR and CB1R. Utilizing this herteromeric signaling could provide a novel therapeutic approach that may yield potent analgesic effects with reduced side effects. We have also found that CB1R switched its signaling specificity from Gi to Gs upon its heteromerizaiton with D2R. In conclusion, our data show that CB1R expands its signaling repertory and modulates the partner receptor signaling upon heteromerization.
APA, Harvard, Vancouver, ISO, and other styles
5

Harrison, Charlotte. "Activation of multiple signaling pathways in cells expressing recombinant opioid receptors." Thesis, University of Leicester, 2000. http://hdl.handle.net/2381/29356.

Full text
Abstract:
Although opioids have been used for centuries in the management of pain, the opioid receptor family has only recently been cloned, thus allowing detailed studies on opioid receptor mediated signal transduction pathways to be performed. In addition, two novel opioid peptides, endomorphin-1 and -2, were identified in 1997, and it is believed that these represent new endogenous u-opioid receptor ligands. This thesis represents a study into opioid receptor mediated increases in Ca2+ j and provides an investigation into the cellular signaling pathways of endomorphin-1 and -2. Activation of the recombinant 6-opioid receptor expressed in CHO cells (CH05) by D- Pen2'5 -enkephalin produced a concentration dependent, pertussis toxin and thapsigargin sensitive increase in Ca2+ i in whole cell suspensions. Truncation of this receptor by the 37 C-terminal amino acids produced a rightward shift in the concentration response curve for Ca j release. In single adherent CHOu, CHOk (CHO cells expressing recombinant u- or K-opioid receptors respectively) or CH05 cells, application of fentanyl, spiradoline or D-Pen2' -enkephalin respectively produced an increase in Ca i in some cells. The putative u.-opioid receptor endogenous ligands endomorphin- 1 and endomorphin-2 bound with high affinity and selectivity to u-opioid receptors from CHOu, and SH-SY5Y cells. Endomorphin-1 and -2 concentration-dependently inhibited forskolin stimulated cAMP formation in CHOp. and SH-SY5Y cells and endomorphin-1 and -2 produced an increase in Ca2+ j in CHOji cells. Prolonged endomorphin-1 pretreatment desensitized the -opioid receptor in CHO cells, characterized by a reduction in maximal endomorphin-1 mediated cAMP inhibition, an up-regulation of cAMP formation and was due to receptor - G protein uncoupling. Endomorphin-1 pre-treatment produced a rapid loss of cell surface receptors from CHOp. cells, which was possibly accompanied by receptor degradation. Collectively these data add to our understanding of opioid receptor-mediated signal transduction pathways.
APA, Harvard, Vancouver, ISO, and other styles
6

Yu, Xiaochun, and 喻曉春. "Negative modulation of B-adrenoceptor by K-opioid receptor in the heart: signaling mechanisms and clinicalsignificance." Thesis, The University of Hong Kong (Pokfulam, Hong Kong), 1999. http://hub.hku.hk/bib/B31241323.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Velazquez-Marrero, Cristina M. "Modulation of Neuropeptide Release via Voltage-Dependent and -Independent Signaling in Isolated Neurohypophysial Terminals: a Dissertation." eScholarship@UMMS, 2008. https://escholarship.umassmed.edu/gsbs_diss/367.

Full text
Abstract:
This thesis details my examination of several mechanisms for modulation of neuropeptide release via voltage-dependent and voltage-independent intraterminal signaling in isolated neurohypophysial terminals. The first part of this work characterizes depolarization-induced neuropeptide release in the absence of extracellular calcium. The goal of this project was to examine the relationship between depolarization-induced release of intracellular calcium stores and depolarization-secretion coupling of neuropeptides. We demonstrate that depolarization in the absence of extracellular calcium induced by either High K+ or electrical stimulation induces a rise in [Ca2+]i and subsequent neuropeptide release from Hypothalamic Neurohypophysial System (HNS) terminals. A portion of extracellular calcium-independent neuropeptide release is due to intraterminal calcium, but the remaining depolarization-induced release may be due to calcium-independent voltage-dependent (CIVD) release (Zhang and Zhou, 2002; Zhang et al., 2004; Yang et al., 2005). Nevertheless, our results clearly show that extracellular calcium is notnecessary for depolarization-induced neuropeptide secretion from these CNS terminals. In addition, I investigated the role of internal calcium stores in mediating μ-opioid inhibition of voltage-gated calcium channels (VGCCs). Inhibition of VGCCs via μ-opioid agonists has been shown to reduce neuropeptide release in response to High K+ stimulation of isolated terminals (Bicknell et al., 1985b; Russell et al., 1993; van Wimersma Greidanus and van de Heijning, 1993; Munro et al., 1994; Ortiz-Miranda et al., 2003; Russell et al., 2003; Ortiz-Miranda et al., 2005). My findings show μ-opioid inhibition, of VGCC and High K+-mediated rise in [Ca2+]i, are via a voltage-independent diffusible second-messenger targeting release of calcium from ryanodine-sensitive stores, possibly mediated via the cyclic ADP ribose signaling pathway. Furthermore, I detail a different intracellular messenger pathway mediating the κ-opioid inhibition of VGCC and High K+-mediated rise in [Ca2+]ii. In contrast to the μ-opioid inhibition, κ-receptor activation is coupled to a voltage-dependent membrane-delimited pathway. Inhibition of neuropeptide release via both endogenous and exogenous κ-opioid agonists has been extensively studied (Bicknell et al., 1985a; Nordmann et al., 1986a; Wammack and Racke, 1988; Munro et al., 1994; Ingram et al., 1996; Rusin et al., 1997a). My investigation shows that the κ-inhibition of VGCC is voltage-dependent and is furthermore, relieved within the context of a physiological burst of action potentials (APs). This physiologically-evoked, activity-dependent modulation of VGCC and subsequent release, represents an important mechanism for short-term synaptic plasticity at the level of the terminals. Given the ubiquitous nature of voltage-dependent G-protein signaling in the CNS, our results may prove important in understanding modulatory effects of specific bursting patterns throughout the CNS. In the last 30 years the neurohypophysial system has proven to be an excellent system to study the complexities of depolarization-secretion coupling (DSC). There have been many advances in our understanding of the underlying mechanisms involved and their physiological implications. The current work focuses on two important features of DSC; voltage and calcium. Although in many ways these two are intrinsically linked through VGCC activation, we have found that in isolated HNS terminals that is not always the case. We have further found that when voltage and calcium influx are linked during DSC, modulation by opioids may or may not be linked to activity-dependent relief depending on the opioid receptor activated. This finding has important implications in neuropeptide release during patterned stimulation in vivo. As I will discuss further, many factors play into the complexities of the regulatory mechanisms involving release. As investigations into this remarkable field continue, I hope to have contributed a valuable piece to the puzzle.
APA, Harvard, Vancouver, ISO, and other styles
8

Tumati, Suneeta. "Functional regulation of opioid receptor signaling." Diss., The University of Arizona, 2009. http://hdl.handle.net/10150/194989.

Full text
Abstract:
Studies have shown that long-term opioid agonist (such as morphine) treatment produces antinociceptive tolerance and increased pain sensitivity (hyperalgesia and/or allodynia), limiting the clinical efficacy of morphine. Prolonged opiate administration also upregulates spinal pain neurotransmitter (such as calcitonin gene-related peptide (CGRP)) levels and enhances evoked CGRP release in the dorsal horn of rats. It was suggested that augmented spinal pain neurotransmission may contribute to paradoxical pain sensitization and antinociceptive tolerance. The cellular signal transduction pathways involved in sustained opioid mediated augmentation of spinal pain neurotransmitter are not fully clarified.Sustained morphine treatment was shown to augment the concentrations of inflammatory mediators, such as PGE2 in the spinal cord. Studies have shown that PGE2 stimulates cAMP formation and CGRP release by activation of Gs protein-coupled prostaglandin receptor types in primary sensory neurons. Interestingly, it was found earlier that sustained opioid agonist treatment leads to a Raf-1-dependent sensitization of adenylyl cyclase(s) (AC superactivation), augmenting forskolin-stimulated cAMP formation upon opioid withdrawal (cAMP overshoot). It is well demonstrated that cAMP activates cAMP-dependent protein kinase (PKA), which plays an important role in the modulation of presynaptic neurotransmitter release. Therefore, in this study, we investigate the physiological role of Raf-1 mediated AC superactivation and subsequent PKA activation in A. sustained morphine-mediated augmentation of basal or evoked pain neurotransmitter release in vitro, in cultured primary sensory neurons, and B. in vivo, in sustained morphine mediated paradoxical pain sensitization and antinociceptive tolerance in rats.Our data demonstrates that A. sustained morphine treatment augments both basal and capsaicin-evoked CGRP release from isolated primary sensory neurons in a PKA- and Raf-1- dependent manner. B. sustained morphine treatment- augments of PGE2-evoked CGRP release from these cells. C. selective knockdown of spinal PKA or Raf-1 protein levels by intrathecal PKA- or Raf-1-specific siRNA pretreatment completely attenuates sustained morphine-mediated thermal hyperalgesia, tactile allodynia and greatly reduces antinociceptive tolerance in rats.In conclusion, we suggest that Raf-1-mediated AC superactivation may have a crucial trigger role in sustained morphine-mediated compensatory adaptations in the nervous system. Thus, we expect that pharmacological attenuation of Raf-1-mediated AC superactivation may improve the clinical treatment of chronic and neuropathic pain.
APA, Harvard, Vancouver, ISO, and other styles
9

Olson, Keith Mathew, and Keith Mathew Olson. "Atypical Opioid Interactions – Development of Selective Mu-Delta Heterodimer Antagonists, Clinical Opioids at Non-Mu Pain Targets and Endogenous Biased Signaling." Diss., The University of Arizona, 2017. http://hdl.handle.net/10150/626669.

Full text
Abstract:
Most clinical opioids produce analgesia through the Mu Opioid Receptor (MOR) providing the only effective treatment for chronic pain patients. These studies explore three pre-clinical strategies to improve MOR analgesia and minimize side effects: 1) compounds that target G-protein Coupled Receptors (GPCRs) heterodimers, such as heterodimerization between the Delta Opioid Receptor (DOR) and MOR (MDOR); 2) multi-functional compounds that target multiple receptor systems for synergistic effects, such as a MOR agonist and a the serotonin reuptake transporter (SERT) inhibitor; or 3) biased agonists that preferentially activate one signaling pathway associated with analgesia over another associated with side effects at the same receptor. First, several indirect lines of evidence indicate the MOR-DOR heterodimer (MDOR) can regulate MOR opioid tolerance and withdrawal. However, studying MDOR remains difficult because no selective MDOR antagonists are available. To address this need, we created a novel series of bivalent MDOR antagonists by connecting a low affinity MOR antagonist (H-Tyr-Pro-Phe-D1Nal-NH2) to a moderate affinity DOR (H- Tyr-Tic-OH) antagonist with variable length polyamide spacers (15-41 atoms). In vitro radioligand binding and [35S]-GTPγS coupling assays in MOR, DOR, and MDOR expressing cell lines show bivalent ligands produce a clear length dependence in MDOR but not MOR or DOR cell lines. The lead compound – D24M with a 24-atom spacer – displayed high potency (IC50MDOR = 0.84 nM) with 91-fold selectivity for MDOR:DOR and 1,000-fold MDOR:MOR selectivity. Second, clinicians have long appreciated subtle but distinct differences in analgesia and side effects of MOR opioids. A variety of non-MOR targets including DOR, Kappa Opioid Receptor (KOR), the Cannabinoid Receptor-1 (CB1), the Sigma-1 Receptor (σ1R), the Dopamine- (DAT), Serotonin- (SERT) and Norepinephrine- Reuptake Transporters (NET) induce analgesia and/or modulate MOR mediated side effects. To determine if different opioid profiles arise from non-MOR interactions, we evaluated the binding and function of nine clinical analgesics at the nine aforementioned targets revealing several clinical opioids contain previously unidentified affinity’s or activity’s. Hydrocodone displayed low affinity at the MOR (KI = 1800 nM) and only ~2 fold less affinity at the σ1R (KI = 4000 nM). Second buprenorphine promoted monoamine influx at DAT, SERT and NET with EC50 > 1,000 nM. These novel interactions suggest the nuanced differences of clinical opioids may arise from previously unappreciated off-target effects. Future studies will assess whether these in vitro results predict hydrocodone and buprenorphine activity in vivo. Finally, the unique function of the numerous endogenous opioid peptides at a given receptor remains unclear. How endogenous ligands interact with ORs produces obvious drug design consequences. These studies show two endogenous Dynorphin analogues – Dynorphin A and Dynorphin B – differentially regulate two ubiquitous signaling modules – βarrestin2 and Gαi/o– at the DOR. Dynorphin A and Dynorphin B swap potency rank orders for β-arrestin2 recruitment and [35S]-GTPγS signaling, indicating two distinct signaling platforms are formed. Dynorphin A but not Dynorphin B treatment simulated AC super activation, while Dynoprhin B internalized DOR better than Dynorphin A. These in vitro assays suggest endogenous Dynorphin analogues differentially regulate signals at the DOR in vitro. Future work includes further characterizing signaling differences in vitro and testing these changes in vivo.
APA, Harvard, Vancouver, ISO, and other styles
10

Yu, Y. Joy. "Regulation of mu opioid receptor trafficking, signaling, and recycling in neurons." Diss., Search in ProQuest Dissertations & Theses. UC Only, 2009. http://gateway.proquest.com/openurl?url_ver=Z39.88-2004&rft_val_fmt=info:ofi/fmt:kev:mtx:dissertation&res_dat=xri:pqdiss&rft_dat=xri:pqdiss:3378518.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Cooper, Andrew H. "The postnatal development of nociceptive and opioid receptor signalling pathways." Thesis, University of Nottingham, 2017. http://eprints.nottingham.ac.uk/40839/.

Full text
Abstract:
The physiology and function of nociceptive and opioid signalling pathways undergo substantial postnatal maturation, and supraspinal µ-opioid receptor (MOR)-mediated control of nociceptive signalling is significantly different in juveniles and adults. Here the, mechanisms responsible for these changes were investigated. [35S]GTPγS assays utilising the MOR-selective agonist DAMGO demonstrated concentration-dependent G protein activation within rostral ventromedial medulla (RVM) tissue which was significantly different between rats aged postnatal day (P) 21 and adults (>P40) (adult: EC50 = 351.1 nM; Emax = 179% basal response; P21: EC50 = 129.3 nM; Emax = 150.9% basal response; Extra sum-of-squares F-test comparing concentration-response curves; P < 0.001). However in adult tissue, co-application of DAMGO with the ganglioside GM1 (1 µM), which has previously been shown to alter MOR G protein coupling from Gi to Gs, revealed that modulation of MOR by GM1 was not responsible for this difference (P > 0.05). Aside from expression on neurones, MOR are expressed by microglia, though the extent and functional consequences are subject to some controversy. Intracellular imaging of changes in calcium concentration using the fluorescent indicator dye Fura-2 showed that ATP-induced concentration-dependent increases in intracellular calcium were significantly increased in primary microglial cultures isolated from adult (EC50 = 2.91 µM; Emax = 0.37 Fmax - F0, the increase in 340/380 nm fluorescence ratio from baseline) versus neonatal (P1) (EC50 = 3.03 µM; Emax = 0.20 Fmax - F0; Extra sum-of-squares F-test comparing concentration-response curves; P < 0.01) brain tissue, and that co-application of DAMGO (1 µM) potentiated responses in adult microglia (EC50 = 1.56 µM; Emax = 0.57 Fmax - F0) but inhibited responses in neonatal microglia (EC50 = 2.65 µM; Emax = 0.14 Fmax - F0), suggesting postnatal alterations in microglial modulation of nociceptive signalling. Additionally, the expression of opioid receptors and their endogenous ligands human mid-brain were investigated using TaqMan RT-PCR. This did not reveal any age-related alterations in mRNA transcript levels of these genes (P > 0.05 for all), confirming previous findings in rats. Previously it has been demonstrated that painful experience during the neonatal period can have a lasting influence on pain processing and adult sensory thresholds. Investigations into opioidergic signalling as one of the mechanisms responsible were conducted with hindpaw injection of complete Freund’s adjuvant (CFA; 10 µl), which induced oedema and erythema in rats aged P1, P10 and P21. As expected this failed to induce decreases in mechanical paw withdrawal threshold (PWT) in P1 rats, in contrast to all other ages. Examining infiltrating macrophages during the acute inflammatory process revealed significant alterations between adults and neonates in cells expressing ED1 and mannose receptor, suggesting altered peripheral inflammatory processes in the neonate. Despite the aforementioned postnatal alterations in MOR-mediated control of nociception, administration of the opioid receptor antagonist naltrexone (3 mg/kg) unmasked resolved inflammatory hyperalgesia in rats injured as early as P10, showing that constitutive MOR activity is able to suppress latent pain sensitisation from an early age but not from birth, highlighting the functional immaturity of this system in early postnatal life. The impact of neonatal inflammation on hyperalgesic priming, a model of the transition from acute to chronic pain, was also investigated. CFA at P1 failed to alter adult responses to hindpaw injection of carrageenan (5 µl; 1%)-induced inflammation, did not alter the development of hyperalgesic priming (increased duration of response to hindpaw injection of PGE2; 1 µg/5 µl) and did not itself induce priming. These results suggest that previously observed life-long effects of neonatal injury are dependent on nociceptive signalling to the spinal cord rather than the presence of inflammation itself.
APA, Harvard, Vancouver, ISO, and other styles
12

Blgacim, Nuria. "Molecular Control of the δ-opioid Receptor Signaling and Functional Selectivity by Sodium." Thesis, Université d'Ottawa / University of Ottawa, 2018. http://hdl.handle.net/10393/37806.

Full text
Abstract:
Accumulating evidence suggests a prominent role of the arrestin-dependent signaling pathway in triggering most of the deleterious side effects observed using δ-OR targeting drugs. Numerous small molecules targeting the δ-OR receptors have been developed but their pharmacological properties, including their functional selectivity, have been poorly characterized. The absence of functionally selective opioid drugs, and the lack of knowledge of the pharmacological profile and signaling properties of the δ-OR receptor, limits its therapeutic exploitation. The development of functionally selective modulator toward the canonical G protein pathway could importantly increase the therapeutic potential of this receptor while decreasing its deleterious effects. An approach to fine-tune the functional selectivity of a GPCR is by using allosteric modulators. These allosteric modulators would reduce problems associated with drugs targeting the orthosteric site by not chronically activating the receptor. The overall goal of the proposed research is to study the molecular mechanism by which sodium-channel inhibitors allosterically regulates the delta opioid receptor (δ-OR) signaling and functional selectivity. Additionally, the signaling features of the δ-OR signal transduction triggered by biased receptor activation have been investigated. A combination of approaches, including functional studies, molecular modeling and mutagenesis, were used to study the general mechanism underlying the activation and tuning of the δ-OR signal transduction behavior. Thus, this work suggests the druggability of the allosteric sodium pocket by using sodium channel inhibitors. The current research represent discovery of two different allosteric profiles for the β-arrestin recruitment and one allosteric profile for the G-protein pathway at activated DOR and would serve as scaffold for further refinement of modulators with the desired pharmacological profile.
APA, Harvard, Vancouver, ISO, and other styles
13

Asghar, Muhammad Junaid. "Studies on the biased signalling of some novel delta opioid receptor agonists." Thesis, University of Nottingham, 2017. http://eprints.nottingham.ac.uk/40485/.

Full text
Abstract:
The delta opioid receptor (DOR) is a G protein-coupled receptor (GPCR) which is important in the regulation of neuronal function, predominantly via coupling to heterotrimeric Gi/0 proteins. The receptor has been shown to be a potential target for the treatment of chronic pain and affective disorders. Although a large number of opioid agonists exist, their properties vary widely, at least partly due to their differential coupling to post-receptor signalling systems, a phenomenon referred to as ligand-biased signalling or functional selectivity. The aim of the current project was to examine the signalling properties of a set of established and novel DOR agonists in an attempt to identify compounds that have biased signalling profiles. It was hypothesized that DOR agonists with partial efficacy regarding β-arrestin recruitment would be less liable to induce receptor internalization and desensitization of G protein-mediated signalling than full agonists. Chinese hamster ovary (CHO) cells, stably transfected with GFP-tagged human (h)DOR and CHO-K1 and U2OS cells over-expressing hDOR were exposed to a number of novel selective DOR agonists compared with the commercially available agonists, SNC80, ADL5859 and DADLE. The compounds’ potencies and efficacies were measured in four different assay systems; 1. Inhibition of forskolin-stimulated cyclic AMP accumulation, 2. Extracellular signal-regulated kinase (ERK1/2) phosphorylation using an immunocytochemical In-cell Western (I-CW) assay, 3. β-arrestin 2 recruitment and 4. hDOR internalization. The latter two assays employed DiscoverX Enzyme Fragment Complementation technology. An attempt to develop a secreted placental alkaline phosphatase (SPAP) reporter gene assay to measure DOR-mediated cyclic AMP inhibition was not successful. All of the ligands were nearly full agonists in relation to cyclic AMP inhibition although some were less efficacious than the standard SNC80 regarding ERK1/2 activation. Their absolute potencies and rank orders of potency in inhibiting cyclic AMP and activating ERK1/2 were quite different, although both signalling systems were apparently Gi/o- protein mediated. In contrast, the agonists exhibited a full range of efficacies and potencies in both β-arrestin 2 recruitment and hDOR internalization assays and there was a significant correlation between the maximum efficacies of the compounds in the two assays. A potential relationship between β-arrestin 2 recruitment/ hDOR internalization and desensitization of agonist-induced cyclic AMP accumulation was explored. Responses to the highly arrestin-recruiting agonists SNC80 and DADLE desensitized fully after extended exposure, whereas the novel partial agonists PN6047 and OPD00003 resisted desensitization. Bias factors were calculated for the agonist set and both PN6047 and OPD00003 were found to be significantly biased towards G protein-mediated cyclic AMP inhibition. In conclusion, this study reports, for the first time, a detailed characterization of signalling bias for a set of selective DOR agonists in cells over-expressing human DORs. The findings suggest that it is potentially possible to predict wanted and unwanted properties of agonists by determining post-receptor signalling profiles in vitro which will facilitate the discovery and development of novel therapeutics based on the DOR.
APA, Harvard, Vancouver, ISO, and other styles
14

Schmid, Benedikt [Verfasser], and Carsten [Gutachter] Hoffmann. "Molecular Signalling Mechanisms at the µ-Opioid Receptor / Benedikt Schmid ; Gutachter: Carsten Hoffmann." Würzburg : Universität Würzburg, 2020. http://d-nb.info/1204831513/34.

Full text
APA, Harvard, Vancouver, ISO, and other styles
15

Xiaochun, Yu. "Negative modulation of B-adrenoceptor by K-opioid receptor in the heart : signaling mechanisms and clinical significance /." Hong Kong : University of Hong Kong, 1999. http://sunzi.lib.hku.hk/hkuto/record.jsp?B21415377.

Full text
APA, Harvard, Vancouver, ISO, and other styles
16

Madishetti, Sreedhar [Verfasser], Reinhard [Akademischer Betreuer] Wetzker, and Stefan [Akademischer Betreuer] Schulz. "PI3Kγ as a nodal point in μ-opioid receptor signaling / Sreedhar Madishetti. Gutachter: Reinhard Wetzker ; Stefan Schulz." Jena : Thüringer Universitäts- und Landesbibliothek Jena, 2012. http://d-nb.info/1028236689/34.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

Softah, Abrar. "Multiple Cell Signaling Pathways Modulate the Cocaine-Induced Increase in Mu Opioid Receptor Protein Expression in PC12 Cells." Thesis, Abrar Ameen Softah, 2013. http://hdl.handle.net/10214/7189.

Full text
Abstract:
Cocaine is interrelated with the opioid system on many levels, especially via the mu opioid receptor (MOR). Also, cocaine has been involved in modulating nitric oxide (NO) actions within the cell. The effect of cocaine was first assessed on the MOR, and then on transcription by the use of 1 µg/ mL actinomycin D inhibitor. Several signaling pathways that cocaine may exert its action in modulating the MOR up-regulation in protein expression were also explored. Two dosage regimens were used in cocaine treatment, single continuous treatment (SCT), and repeated intermittent treatment (RIT). Different pathway inhibitors were used on PC12 cells, as follows: the PLC-PKC inhibitors 5 µM U-73122 and 10 µM BIS-1 used to investigate the involvement of the PKC signaling pathways in MOR expression levels, the evaluation of MAPK pathway by the use of 50 µM U0126 inhibitor, and the 10 µM LY94002 inhibitor was used to investigate the PI3K/Akt pathway. Moreover, the effect of NO on these signaling pathways was investigated by the use of 20 mM nonselective L-NAME inhibitor and qualitatively by DAF-2 florescence. Western blot analysis indicated that cocaine up-regulated MOR protein expression. Also, RIT cocaine treatment increased MOR protein levels via transcription. All three signaling pathways, MAPK, Akt and PKC modulated cocaine-induced increase of MOR following SCT cocaine treatment (post-transcriptional). Both MAPK and Akt have been found to modulate the cocaine-induced transcription of MOR via the two dosage regimens of cocaine, SCT and RIT. Also, inhibition of both PLC and PKC did not prevent cocaine-induced increase in MOR transcription, according to RIT of cocaine. Furthermore, Akt and PKC appeared to modulate cocaine-induced NO production while MAPK did not. NO seemed to be involved with the PKC and Akt pathways in up-regulating MOR in RIT of cocaine directly by the Akt pathway, and indirectly by the PKC pathway. On the other hand, NO and MAPK modulated the MOR up-regulation expression simultaneously, but in an individual/parallel manner. Furthermore, signaling pathway activation levels were tested using L-NAME which concluded that NO modulated cocaine-induced increase in total Akt protein levels, but did not appear to have an effect on phosphorylated MAPK activation levels. In conclusion, different treatment regimens of cocaine activate different pathways; SCT of cocaine activated all three signaling pathways, however, RIT of cocaine activated only the MAPK and Akt pathways.
Saudi Bureau in Canada
APA, Harvard, Vancouver, ISO, and other styles
18

Tsisanova, Elena. "Characterization of extracellular signal-regulated Kinase1/2 (ERK1/2) signalling pathway at the μ-opioid receptor (MOPr)." Thesis, University of Bristol, 2015. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.688107.

Full text
Abstract:
Opioid drugs and their receptors are one of the most extensively studied areas of pharmacology, and in relation to biased agonism in particular. Previous research indicated that biased agonist at the Il-opioid receptor (MOPr) can be detected. Biased agonism theory, when applied to MOPr, offers the possibility to synthesize opioid ligands, lacking such lifethreatening side effects as addiction, tolerance, hyperalgesia and respiratory depression. In the current project the ability of MOPr ligands to activate Extracellular signal-Regulated Kinases 1/2 (ERKl/2), a key mediator of GPCR actions in cells, was studied. The MOPr ligands (DAMGO, morphine, buprenorphine, methadone, oxycodone, etorphine, pentazocine and endomorphin-2) were used that display different degrees of bias between G-protein and arrestin pathways as well as different efficacies for G protein activation. The model system used was HEK293 cells stably expressing MOPr.
APA, Harvard, Vancouver, ISO, and other styles
19

Okunomiya, Taro. "Generation of a MOR-CreER knock-in mouse line to study cells and neural circuits involved in mu opioid receptor signaling." Kyoto University, 2020. http://hdl.handle.net/2433/253202.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Gamaleldin, Mariam. "Non-canonical Signaling of μ-opioid Receptors." Master's thesis, 2021. https://doi.org/10.25972/OPUS-24032.

Full text
Abstract:
According to the “canonical” paradigm of GPCR signaling, agonist-bound GPCRs only signal to the downstream adenylyl cyclase enzyme when they are seated at the plasma membrane. Upon prolonged binding of an agonist, receptor internalization usually takes place, leading to the termination of this downstream signaling pathway and activation of alternative ones. However, a set of recent studies have shown that at least some GPCRs (e.g. thyroid stimulating hormone receptor) continue signaling to adenylyl cyclase after internalization. In this study, I aimed to investigate canonical signaling by internalized μ opioid receptors (MORs), which are Gi-coupled receptors, using a fluorescence resonance energy transfer (FRET) sensor for cyclic AMP (cAMP) known as Epac1-camps. My results show that the cyclic AMP inhibition signal induced by the binding of DAMGO, a MOR agonist, persists after agonist washout. We hypothesized that this persistent signal might come from internalized DAMGO-bound receptors located in the endosomal compartment. To test this hypothesis, I used dynasore and Dyngo 4a, two dynamin inhibitors that are known to prevent clathrin-mediated endocytosis. Interestingly, dynasore but not Dyngo 4a pretreatment largely blunted the response to MOR activation as well as to adenylyl cyclase activation with Forskolin (FSK). In addition, DAMGO-induced cAMP signal remained persistent even in the presence of 30 M Dyngo 4a. These results might point to a complex interplay between clathrin-mediated internalization and MOR signaling. Further experiments are required to elucidate the mechanisms underlying the persistent MOR signaling and to fully clarify whether MORs are capable of Gi signaling in the endosomal compartment
Nach dem „kanonischem“ Paradigma der Signaltransduktion akktivieren agonistbindende GPCR's nur dann die Adenylylcyclase, wenn sie sich in der Zellmembran befinden. Ist der Agonist länger gebunden führt dies meist zur Internalisierung des Rezeptors. Dies führt dazu, dass die Signaltransduktion beendet wird und andere Signalwege aktiviert werden. Jedoch haben einige neuere Studien gezeigt, dass zumindest einige GPCR's (z.B. der Thyreotropinrezeptor) auch nach Internalisierung weiter die Adenylylcyclase aktivieren. Das Ziel der vorliegenden Arbeit ist es kanonische Signaltransduktion von internalisierten μ Opioidrezeptoren (MORs) zu untersuchen, welche zu den Gi gekoppelten Rezeptoren gehören. Dazu wird ein Förster Resonanz Energie Transfer (FRET) Sensor für Cyclisches Adenosinmonophosphat (cAMP) benutzt, bekannt als Epac1-camps. Meine Resultate zeigen, dass die Inhibierung des cAMP Signal durch das Binden von DAMGO, einem MOR Agonisten, bestehen bleibt auch nachdem der Agonist ausgewaschen wurde. Unsere Hypothese ist, dass internalisierte Rezeptoren im endosomalen Kompartment, die DAMGO gebunden haben, die Ursache für das fortbestehende Signal verantwortlich sind. Um dies zu überprüfen habe ich Dynasore und Dyngo 4a benutzt. Beides sind Dynamin Inhibitoren von welchen man weiß, dass sie die Clathrin gesteuerte Endocytose unterbinden. Interessanterweise hat nur die Vorbehandlung mit Dynasore die Reaktion auf die MOR und die Adenylylcyclase Aktivierung mit Forskolin (FSK) verringert, jedoch nicht Dyngo 4a. Desweiteren hielt das durch DAMGO induzierte cAMP Signal selbst nach Zugabe von 30 M Dyngo 4a an. Diese Ergebnisse können ein Hinweis für einen komplexen Zusammenhang zwischen Clathrin gesteuerter Internalisierung und MOR Signaltransduktion sein. Jedoch braucht es weitere Experimente um den zugrundeliegenden Mechanismus der anhaltenden MOR Signaltransduktion zu beleuchten und um vollständig zu erklären ob MORs in der Lage für Gi Signaltransduktion im endosomalen Kompartment sind. (Übersetzt von Kerstin Seier)
APA, Harvard, Vancouver, ISO, and other styles
21

Tsai, Hsin-Ju, and 蔡欣儒. "The Role of Opioid Receptors Signaling in Electroacupuncture-induced Cardioprotection in Rats." Thesis, 2014. http://ndltd.ncl.edu.tw/handle/53382433111524419769.

Full text
Abstract:
碩士
國立陽明大學
傳統醫藥研究所
102
Rationale: Our previous studies demonstrated that electroacupuncture (EA) increased myocardial GSK3 phosphorylation. Recent studies showed that GSK3 and opioid receptors activation can attenuate ischemia/ reperfusion (I/R) injury in rat hearts. However, the role of various opioid receptors (OR) subtypes in preconditioned EA-induced myocardial protection and the cross reaction with GSK3 remain unknown. Objective: We investigated the role of OR subtype signaling in EA-induced cardioprotection against I/R injury of the rat heart. Methods & Results: Male Spraque-Dawley rats were preconditioned by EA with/without various receptors antagonists such as opioid receptor (OR) subtype receptors (kappa OR, delta OR, and mu OR). The expressions of Akt, GSK3, and PKC-ε expression were analyzed by Western blotting. Our results showed that GSK3 and PKC-ε expression levels were significantly increased in the EA group compared to the sham group, which were blocked by pretreatment with specific antagonists targeting KOR and DOR, but not MOR subtype. In I/R model, rats randomly divided to sham EA (I/R alone), EA (I/R pretreatment with EA), EA+KOR left (EA with delta and mu OR antagonists), EA+KOR blocked (EA with kappa OR antagonist), EA+DOR left (EA with kappa and mu OR antagonist) and EA+DOR blocked (EA with delta OR antagonist). During ischemia and reperfusion, the rat's hemodynamic index, arrhythmia, mortality, infarction size, and serum CK-MB and troponin-I were evaluated. In our results, EA with/without OR antagonists did not change the mean blood pressure and heart rate. The incidence and duration of ventricular tachycardia and mortality during myocardial ischemia were significantly reduced by EA compared to sham EA. Moreover, the protection effects were complete reversed in EA+KOR blocked. Similarly, the infarction size was significantly reduced by EA and increased in EA+KOR blocked than sham EA. In the myocardial infarction marker, EA with/without did not significantly alter troponin-I. In RT-PCR analysis, the mRNA expression of opioid receptor subtypes were delta>kappa>mu. Moreover, the location of kappa OR in rat heart was in myocyte not vein analysis by immunofluorescence stain. Conclusion: The mechanism of EA-induced myocardial protection against I/R injury seems to involve multiple target pathways such as Akt, KOR and/or DOR signaling.
APA, Harvard, Vancouver, ISO, and other styles
22

Moutelíková, Karolína. "Vliv chronického působení morfinu na přežití buněk po působení oxidativního stresu u neuroblastomové linie SH-SY5Y buněk." Master's thesis, 2018. http://www.nusl.cz/ntk/nusl-388352.

Full text
Abstract:
Morphine is a natural opioid which is used in medicine due to his potent analgesic and sedative effects. In the forefront of scientific interest is a chronic usage of opioids which can lead to a development of drug addiction. Morphine role in oxidative stress was described in last years. It was revealed its protective potencial by many studies. However, some studies described its pro-oxidative effect. The aim of this study was to determinate effect of chronic morphine on cell survival after oxidative stress caused by H202 analog - tBHP in the SH-SY5Y neuroblastoma cell line. The results verified morphine protective effect against oxidative stress. The highest protective effect of morphine was achieved in a concetration of 10 µM. It was desribed that morphine can induce activation of mu-opioid (MOR) and Toll-like 4 (TLR4) receptors signalling pathway on molecular level. The aim of this thesis was to evaluate the role of MOR a TLR4 in protective effect of morphine against oxidative stress by two methods. Firstly, it was used tests of oxidative stress on cell viability. The obtained results demonstrated majority role of TLR4 and minory role of MOR. Afterwards, we assesed changes in the expression of MOR a TLR4 after chronic morphine by SDS-PAGE electrophoresis. Results of these experiments did not...
APA, Harvard, Vancouver, ISO, and other styles
23

Παπακωνσταντίνου, Μαρία-Παγώνα. "Αλληλεπιδράσεις των επταελικοειδών υποδοχέων με διάφορες πρωτεΐνες. Χαρακτηρισμός νέων σηματοδοτικών μονοπατιών." Thesis, 2014. http://hdl.handle.net/10889/8440.

Full text
Abstract:
Οι οπιοειδείς υποδοχείς (OR), μ, δ, κ και NOP, είναι μέλη των επταελικοειδών υποδοχέων που συζεύγνυνται με G πρωτεΐνες (7ΤΜ ή GPCR), οι οποίοι αποτελούν τη μεγαλύτερη υπεροικογένεια υποδοχέων και έναν από τους κύριους φαρμακολογικούς στόχους λόγω της υψηλής φυσιολογικής τους σημασίας. Οι OR ρυθμίζουν μια ποικιλία φυσιολογικών αποκρίσεων στο νευρικό σύστημα, με κυριότερη την αναλγησία. Τα οπιοειδή φάρμακα είναι τα πιο ισχυρά και αποτελεσματικά αναλγητικά έναντι στον οξύ πόνο, όμως η παρατεταμένη χρήση τους οδηγεί σε φαινόμενα ανοχής και εξάρτησης. Γι’ αυτό υπάρχει έντονο ενδιαφέρον στην αποσαφήνιση των μηχανισμών που εμπλέκονται στα φαινόμενα αυτά προκειμένου να σχεδιαστούν πιο αποτελεσματικά φάρμακα χωρίς τέτοιες παρενέργειες. Η σηματοδότηση των οπιοειδών υποδοχέων γίνεται κυρίως μέσω της ενεργοποίησης των Gi/o πρωτεϊνών που με τη σειρά τους ρυθμίζουν κατάλληλους τελεστές. Πέρα όμως από αυτούς τους κλασσικούς αλληλεπιδρώντες εταίρους οι OR έχουν την ικανότητα να αλληλεπιδρούν και με πολλές άλλες πρωτεΐνες κυρίως μέσω των περιοχών της τρίτης ενδοκυτταρικής τους θηλιάς (i3L) και του καρβοξυτελικού τους άκρου (CT) (Georgoussi et al., 2006- Georgoussi, 2008- Georgoussi et al., 2012). Οι αλληλεπιδράσεις αυτές επηρεάζουν όχι μόνο την σηματοδότηση των OR αλλά και την εν γένει εύρυθμη λειτουργία τους. Μια σημαντική πρωτεϊνική οικογένεια που ελέγχει τη μεταγωγή σήματος από τις G πρωτεΐνες βρέθηκε να είναι οι πρωτεΐνες Ρυθμιστές της κυτταρικής Σηματοδότησης μέσω G πρωτεϊνών ή RGS πρωτεΐνες (Regulators of G protein signaling, RGS). Ο πρωταρχικός τους ρόλος είναι η αλληλεπίδραση τους με τις Gα υπομονάδες των G πρωτεϊνών και η επιτάχυνση της υδρόλυσης του GTP από τις τελευταίες οδηγώντας στη μείωση της σηματοδότησης των GPCR. Μέλη της οικογένειας των RGS πρωτεϊνών είχε δειχθεί ότι πέρα από τις Gα πρωτεΐνες αλληλεπιδρούν επίσης με υποδοχείς GPCR, τελεστές αλλά και με άλλες ρυθμιστικές πρωτεΐνες, προσδίδοντας τους έναν ιδιαίτερο οργανωτικό ρόλο στη λειτουργία του κυττάρου και καθιστώντας τις RGS πρωτεΐνες μόρια υψηλού φαρμακολογικού ενδιαφέροντος. Παρελθόντα πειράματα in vitro συγκατακρήμνισης, του εργαστηρίου Κυτταρικής Σηματοδότησης και Μοριακής Φαρμακολογίας, με τη χρήση GST-χιμαιρικών πεπτιδίων των καρβοξυτελικών άκρων των μ-OR και δ-OR (μ-CT και δ-CT αντίστοιχα) και της τρίτης ενδοκυτταρικής θηλιάς του δ-OR (δ-i3L), έδειξαν ότι η RGS4, ένα μέλος της B/R4 υποοικογένειας, αλληλεπιδρά και με τους δυο υποδοχείς στις περιοχές αυτές (Georgoussi et al., 2006- Leontiadis et al., 2009). Η αλληλεπίδραση της RGS4 στα καρβοξυτελικά άκρα των υποδοχέων αυτών γίνεται στην περιοχή που σχηματίζει μια 8η αμφιπαθική α-έλικα (έλικα VIII), σημείο επαφής των OR και για άλλες πρωτεϊνικές αλληλεπιδράσεις όπως αυτή των STAT5A/B ((Mazarakou and Georgoussi, 2005- Georganta et al., 2010), της σπινοφιλίνης (Fourla et al., 2012) και άλλων πρωτεϊνών (Georgoussi et al., 2012). Βρέθηκε επίσης ότι η RGS4 είναι αρνητικός ρυθμιστής της κυτταρικής σηματοδότησης των μ-OR και δ-OR (Georgoussi et al., 2006- Leontiadis et al., 2009). Τέλος, αποδείχθηκε για πρώτη φορά ότι η RGS4 παίξει το ρόλο «μοριακού φίλτρου» καθοδηγώντας τους μ-OR και δ-OR να αλληλεπιδράσουν με συγκεκριμένο διαφορετικό υποπληθυσμό Gα υπομονάδων των G πρωτεϊνών (Leontiadis et al., 2009). Καμία πληροφορία για τον ρόλο των RGS πρωτεϊνών δεν υπάρχει για τον κ-OR. Για τον λόγο αυτό σκοπός της παρούσας διατριβής ήταν να ελέγξουμε αν οι RGS πρωτεΐνες της Β/R4 υποοικογένειας αλληλεπιδρούν με τον κ-OR και αν ναι, ποιος είναι ο ρόλος τους στη σηματοδότηση του κ-OR και των G πρωτεϊνών με τις οποίες ο τελευταίος συζεύγνυται. Τα αποτελέσματά μας έδειξαν ότι ο κ-OR μπορεί να αλληλεπιδράσει και με την RGS4 και με την RGS2 τόσο in vitro όσο και in vivo. Η δημιουργία GST-χιμαιρικών πεπτιδίων του καρβοξυτελικού άκρου του κ-OR (κ-CT) έδειξε ότι η RGS4 αλληλεπιδρά επίσης εντός της έλικας VIII ενώ η RGS2 αλληλεπιδρά με το τελικό μη συντηρημένο άκρο του κ-CT όσο και του δ-CT. Επιπλέον η συνέκφραση της RGS4 ή της RGS2 σε κύτταρα 293F που εκφράζουν τον κ-OR έδειξε ότι και οι δυο RGS πρωτεΐνες προάγουν την επιλεκτική και διαφορική σύζευξη του κ-OR με συγκεκριμένο υποπληθυσμό των Gαi/o υπομονάδων. Σε ότι αφορά τον φυσιολογικό ρόλο των RGS4 και RGS2 στις ελεγχόμενες από τον κ-OR κυτταρικές αποκρίσεις βρήκαμε ότι τόσο η RGS4 όσο και η RGS2 ανέστειλαν την καταστολή της αδενυλικής κυκλάσης που ελέγχει ο κ-OR, αλλά όχι ο δ-OR, με την RGS2 να έχει ισχυρότερη επίδραση στο μονοπάτι αυτό. Επίσης οι RGS4 και RGS2 μείωσαν την ενεργοποίηση των ERK1,2 κινασών που σηματοδοτούσε ο κ-OR. Τέλος, βρήκαμε ότι παρόλο που καμία από τις δυο RGS δεν επηρεάζει την εσωτερίκευση του κ-OR, η RGS4 επιταχύνει την εσωτερίκευση του δ-OR. Τα ευρήματά μας καταδεικνύουν ότι οι RGS4 και RGS2 πρωτεΐνες είναι δυο νέοι αρνητικοί ρυθμιστές στην σηματοδότηση των κ-OR και δ-OR. Εμφανίζουν διαφορικό ρυθμιστικό ρόλο στα σηματοδοτικά μονοπάτια καθενός OR, με ρόλο κλειδί στην καθοδήγηση της σύζευξής τους με τις Gα υπομονάδες και μπορούν να αποτελέσουν ενδιαφέροντες φαρμακολογικούς στόχους για τον έλεγχο της δράσης των οπιοειδών.
Οpioid receptors (OR) (subtypes μ, δ, κ and NOP) belong to the superfamily of the Heptahelical G protein-coupled receptors (7TM or GPCRs), the largest class of receptors in the human genome and common targets for therapeutics. ORs mediate their responses in the nervous system via coupling to members of the Gi/Go proteins to regulate the activity of various effector systems. Opioids are the most potent analgesics but prolonged administration leads to phenomena of tolerance and dependence thus there is a great interest towards understanding of OR signalling in an effort to develop new drugs devoid of adverse effects. Extended observations have demonstrated that the cytoplasmic face of the ORs is critical in mediating their signal through interactions not only with G proteins but also with multiple other proteins. These regulatory proteins play distinct roles in the regulation of the OR signalling, and in the fine tuning of these receptors. Regulators of G protein signalling (RGS) proteins is a class of proteins that modulate G protein signalling events by directly interacting with Gα subunits and accelerating the GTP hydrolysis, thus reducing GPCR signalling towards their effectors. RGS can also interact with many GPCRs, effectors and auxiliary proteins thus playing a key role in the cell functions, making them highly attractive as pharmacological targets (Abramow-Newerly et al., 2006). Our previous in vitro studies have shown that a member of the B/R4 subfamily of RGS proteins such as RGS4 interacts directly with μ-OR and δ-OR within a conserved region in their C-termini (μ-CT and δ-CT), forming a helix VIII, as well as within the δ-third intracellular loop (δ-i3L). RGS4 associates with μ-OR and δ-OR in living cells and forms selective complexes with Gαi/o proteins in a receptor dependent manner. Expression of RGS4 in HEK293 cells attenuated adenylyl cyclase inhibition mediated by μ-OR and agonist-mediated ERK1,2 phosphorylation for both receptors (Georgoussi et al., 2006- Leontiadis et al., 2009), suggesting for the first time that RGS4 is a negative modulator of μ-OR and δ-OR signalling. To deduce whether similar effects also occur for the κ-opioid receptor (κ-ΟR) and define the ability of other members of the B/R4 subfamily of RGS proteins, such as RGS2, to interact with OR we generated fusion peptides encompassing the C-terminus of κ-OR (κ-CT). Results from pull down experiments indicated that RGS2 interacts with the κ-CT, the δ-CT and the δ-i3L but fails to interact with the μ-CT. RGS4-N-terminal domain is responsible for OR interaction. Mapping the sites of RGS2 interaction indicated that RGS2 interacts with the non conserved portion of the C-termini of ORs exhibiting a different docking site as compared to that of RGS4. Co-precipitation studies in living cells indicated that RGS2 and RGS4 associate with κ-ΟR constitutively and upon receptor activation and confer selectivity for coupling with a specific subset of G proteins in an RGS protein dependent manner. Expression of both RGS2 and/or RGS4, in 293F cells attenuated agonist mediated-adenylyl cyclase inhibition for κ-ΟR, but not δ-OR, with RGS2 exhibiting a more robust effect. RGS4 and RGS2 reduced κ-ΟR-mediated ERK1,2 phosphorylation whereas, RGS4 accelerated agonist-induced internalization of the δ-OR but not of the κ-OR. Collectively, our observations demonstrate that RGS2 and RGS4 are novel interacting partners and negative modulators of κ-ΟR and δ-OR signalling. These two RGS proteins display a differential modulatory effect in each signalling pathway tested and play a key functional role by conferring selectivity for both κ-OR and δ-OR coupling with a specific subset of G proteins. Therefore they can be considered as attractive new pharmacological targets to manipulate opioid receptors signalling.
APA, Harvard, Vancouver, ISO, and other styles
24

Schmid, Benedikt. "Molecular Signaling Mechanisms at the µ-Opioid Receptor." Doctoral thesis, 2020. https://nbn-resolving.org/urn:nbn:de:bvb:20-opus-176850.

Full text
Abstract:
To this day, opioids represent the most effective class of drugs for the treatment of severe pain. On a molecular level, all opioids in use today are agonists at the μ-opioid receptor (μ receptor). The μ receptor is a class A G protein-coupled receptor (GPCR). GPCRs are among the biological structures most frequently targeted by pharmaceuticals. They are membrane bound receptors, which confer their signals into the cell primarily by activating a variety of GTPases called G proteins. In the course of the signaling process, the μ receptor will be phosphorylated by GRKs, increasing its affinity for another entity of signaling proteins called β-arrestins (β-arrs). The binding of a β-arr to the activated μ receptor will end the G protein signal and cause the receptor to be internalized into the cell. Past research showed that the μ receptor’s G protein signal puts into effect the desired pain relieving properties of opioid drugs, whereas β-arr recruitment is more often linked to adverse effects like obstipation, tolerance, and respiratory depression. Recent work in academic and industrial research picked up on these findings and looked into the possibility of enhancing G protein signaling while suppressing β-arr recruitment. The conceptual groundwork of such approaches is the phenomenon of biased agonism. It appreciates the fact that different ligands can change the relative contribution of any given pathway to the overall downstream signaling, thus enabling not only receptor-specific but even pathway-specific signaling. This work examined the ability of a variety of common opioid drugs to specifically activate the different signaling pathways and quantify it by means of resonance energy transfer and protein complementation experiments in living cells. Phosphorylation of the activated receptor is a central step in the canonical GPCR signaling process. Therefore, in a second step, expression levels of the phosphorylating GRKs were enhanced in search for possible effects on receptor signaling and ligand bias. In short, detailed pharmacological profiles of 17 opioid ligands were recorded. Comparison with known clinical properties of the compounds showed robust correlation of G protein activation efficacy and analgesic potency. Ligand bias (i.e. significant preference of any path- way over another by a given agonist) was found for a number of opioids in native HEK293 cells overexpressing μ receptor and β-arrs. Furthermore, overexpression of GRK2 was shown to fundamentally change β-arr pharmacodynamics of nearly all opioids. As a consequence, any ligand bias as detected earlier was abolished with GRK2 overexpression, with the exception of buprenorhin. In summary, the following key findings stand out: (1) Common opioid drugs exert biased agonism at the μ receptor to a small extent. (2) Ligand bias is influenced by expression levels of GRK2, which may vary between individuals, target tissues or even over time. (3) One of the opioids, buprenorhin, did not change its signaling properties with the overexpression of GRK2. This might serve as a starting point for the development of new opioids which could lack the ability of β-arr recruitment altogether and thus might help reduce adverse side effects in the treatment of severe pain
Nach wie vor stellen Opioide die wirkstärkste Gruppe von Medikamenten zu Behandlung starker Schmerzen dar. Auf molekularer Ebene sind alle heute gebräuchlichen Opioide Agonisten am μ-Opioidrezeptor. Der μ-Opioidrezeptor ist ein G-Protein-gekoppelter Rezeptor (GPCR) der Klasse A. GPCR zählen zu den häufigsten Zielstrukturen von Pharmaka. Sie sind membranständige Rezeptoren, die ihr Signal in erster Linie durch die Aktivierung von G-Proteine genannten GTPasen in die Zelle weiterleiten. Im Laufe des Signalprozesses wird der GPCR von GRK phosphoryliert, wodurch seine Affinität zu einer weiteren Gruppe von Signalproteinen, den sog. β-Arrestinen erhöht wird. Bindet ein β-Arrestin an den Rezeptor, beendet dies das G-Proteinsignal und veranlasst die Internalisierung des Rezeptors ins Zellinnere. Bisherige Forschung zeigte, dass das G-Proteinsignal des μ-Opioidrezeptors die erwünschte Schmerzlinderung vermittelt, wohingegen die Rekrutierung von β-Arrestin oftmals mit unerwünschten Wirkungen wie Obstipation, Toleranzentwicklung und Atemdepression in Verbindung gebracht wird. Neuere akademische und industrielle Forschung griff diese Erkenntnisse auf und erkundete die Möglichkeit, das G-Proteinsignal zu verstärken und zur gleichen Zeit die β-Arrestinrekrutierung zu inhibieren. Die theoretische Grundlage solcher Ansätze liegt im Konzept des biased agonism. Dieses berücksichtigt die Tatsache, dass verschiedene Liganden den Anteil eines bestimmten Signalweges am gesamten vom Rezeptor ausgehenden Signals beeinflussen kann und damit nicht nur rezeptor-, sondern sogar signalwegspezifische Signale möglich sein sollten. Die vorliegende Arbeit untersuchte eine Reihe von gängigen Opioiden auf ihre Fähigkeit hin, die einzelnen Signalwege spezifisch zu aktivieren und quantifizierte dies mit Methoden des Resonanzenergietransfers sowie der Proteinkomplementierung in lebenden Zellen. Die Phosphorylierung des Rezeptors ist ein zentrales Ereignis in der anerkannten Abfolge der Signalprozesse an GPCR. Daher wurde in einem weiteren Schritt die Expression der phosphorylierenden GRK erhöht und nach möglichen Auswirkungen auf die Selektivität der Signalwegaktivierung gesucht. Hierbei wurde detaillierte pharmakologische Profile von 17 Opioiden erstellt. Der Abgleich mit bekannten klinischen Wirkeigenschaften der Substanzen zeigte einen robusten Zusammenhang zwischen der Fähigkeit, G-Proteine zu aktivieren und der analgetischen Wirkstärke. Ligand bias, d.h. die signifikante Bevorzugung eines Signalweges gegenüber einem anderen durch einen Liganden, konnte für eine Reihe von Opioiden in lebenden HEK293-Zellen gezeigt werden, die den μ-Opioidrezeptor sowie β-Arrestine überexprimierten. Darüber hinaus konnte gezeigt werden, dass die zusätzliche Überexpression von GRK2 die pharmakodynamischen Eigenschaften nahezu aller Opioide grundlegend veränderte. In der Folge war jeder zuvor gezeigte ligand bias mit Ausnahme von Buprenorphin aufgehoben. Zusammenfassend stehen die folgenden drei Erkenntnisse im Vordergrund: (1) Gängige Opioide zeigen in einem gewissen Maß Selektivität zwischen den Signalwegen. (2) Ligand bias wird beeinflusst von GRK2-Expressionsleveln, welche zwischen Individuen, verschiedenen Gewebetypen oder auch im zeitlichen Verlauf variieren können. (3) Als einziges der untersuchten Opioide änderte Buprenorphin seine Signaleigenschaften durch die Überexpression von GRK2 nicht. Dies könnte als Anknüpfungspunkt in der Entwicklung neuer Opioide dienen, die keinerlei β-Arrestinrekrutierung bewirken und dadurch helfen könnten, unerwünschte Wirkungen in der Behandlung starker Schmerzen zu verhindern
APA, Harvard, Vancouver, ISO, and other styles
25

Brejchová, Jana. "Úloha membránového cholesterolu v signalizaci delta-opioidního receptoru Korelace se strukturou plazmatické membrány." Doctoral thesis, 2014. http://www.nusl.cz/ntk/nusl-338451.

Full text
Abstract:
Study of HEK293 cells stably expressing fusion protein between delta opioid receptor (δ-OR) and pertussis toxin-insensitive mutant of Gi1α protein, δ-OR-Gi1α (Cys351 -Ile351 ), provided the following results. Decrease of plasma membrane cholesterol content (cholesterol depletion) induced by cyclic oligosaccharide β-cyclodextrin did not affect binding of specific δ-OR agonist, [3 H]DADLE. Neither the maximum number of binding sites nor the affinity of [3 H]DADLE binding was changed by cholesterol depletion. However, the ability of δ-OR to activate cognate trimeric G proteins was impaired. EC50 value of agonist-stimulated [35 S]GTPγS binding was an order of magnitude higher. This effect was observed in case of both control and pertussis toxin-treated cells. It means that cholesterol depletion markedly reduced the efficiency of functional coupling of δ-OR to endogenously expressed pertussis toxin-sensitive G proteins of Gi/Go family as well as covalently bound Gi1α (Cys351 -Ile351 ) protein. Unchanged plasma membrane cholesterol content is therefore important requirement for proper δ-OR function. Detection of the effect of cholesterol depletion on the functional activity of δ-OR was supported by the analysis of changes in biophysical state of plasma membrane using fluorescent membrane probes,...
APA, Harvard, Vancouver, ISO, and other styles
26

"Mu-Opioid Receptor - pAKT Signaling in the Ventral Tegmental Area is Critical for the Behavioral and Cellular Consequences of Social Stress." Doctoral diss., 2015. http://hdl.handle.net/2286/R.I.29778.

Full text
Abstract:
abstract: Intermittent social defeat stress produces vulnerability to drugs of abuse, a phenomena known as cross-sensitization, which is proceeded by a corresponding upregulation of ventral tegmental area (VTA) mu-opioid receptors (MORs). Since VTA MORs are implicated in the expression of psychostimulant sensitization, they may also mediate social stress-induced vulnerability to drugs of abuse. Social stress and drugs of abuse increase mesolimbic brain-derived neurotrophic factor (BDNF) signaling with its receptor, tropomyosin-related kinase B (TrkB). These studies examined whether VTA MOR signaling is important for the behavioral and cellular consequences of social stress. First, the function of VTA MORs in the behavioral consequences of intermittent social defeat stress was investigated. Lentivirus-mediated knockdown of VTA MORs prevented social stress-induced cross-sensitization, as well as stress-induced social avoidance and weight gain deficits. Next it was examined whether VTA MOR expression is critical for stress-induced alterations in the mesocorticolimbic circuit. At the time cross-sensitization was known to occur, lentivirus-mediated knockdown of VTA MORs prevented stress-induced increases in VTA BDNF and its receptor, TrkB in the nucleus accumbens (NAc), and attenuated NAc expression of delta FosB. There was no effect of either stress or virus on BDNF expression in the prefrontal cortex. Since social stress-induced upregulation of VTA MORs is necessary for consequences of social stress, next activity dependent changes in AKT, a downstream target of MOR stimulation associated with sensitization to psychostimulant drugs, were investigated. Using fluorescent immunohistochemical double labeling for the active form of AKT (pAKT) and markers of either GABA or dopamine neurons in the VTA, it was determined that social stress significantly increased the expression of pAKT in GABA, but not dopamine neurons, and that this effect was dependent on VTA MOR expression. Moreover, intra-VTA inhibition of pAKT during stress prevented stress-induced weight gain deficits, while acute inhibition of VTA pAKT blocked the expression of cross-sensitization in subjects that had previously exhibited sensitized locomotor activity. Together these results suggest that social stress upregulates MORs on VTA GABA neurons, resulting in AKT phosphorylation, and that increased VTA MOR-pAKT signaling may represent a novel therapeutic target for the intervention of substance abuse disorders.
Dissertation/Thesis
Doctoral Dissertation Neuroscience 2015
APA, Harvard, Vancouver, ISO, and other styles
27

Bagheri, Haniyeh. "L'inhibition de la production d'AMPc est modulée différemment à court et long terme par l'internalisation du récepteur opioïde delta." Thèse, 2014. http://hdl.handle.net/1866/11815.

Full text
Abstract:
Les opioïdes sont les analgésiques les plus puissants mais leur utilisation prolongée peut entraîner le développement d’une tolérance analgésique. La tolérance serait en partie associée à l’inhibition prolongée de l’adénosine monophosphate cyclique (AMPc) entraînant des changements compensatoires dans la voie de l’adénylate cyclase. Pour cette étude, nous avons eu recours à un biosenseur basée sur la technologie de Bioluminescence Resonnance Energy Transfer (BRET) et qui fournit des mesures de l’AMPc en fonction du temps réel. Durant les 15 premières minutes de stimulation, la réponse de l’AMPc est bi-phasique. Cette progression de la réponse à l’AMPc n’est pas la même pour tous les ligands. Par exemple, la deltorphine II qui induit l’internalisation du récepteur opioïde delta (DOR) affiche une baisse de l’inhibition de l’AMPc. À l’inverse la morphine qui n’induit pas l’internalisation du DOR affiche une réponse stable à l’inhibition de l’AMPc. Ainsi le profil d’internalisation permet de prédire la progression de l’inhibition de l’AMPc à court terme (15 minutes). Nous avons aussi mesuré la réponse à l’AMPc durant 30, 60 et 120 min, étant donné qu’un traitement chronique aux opioïdes induit une tolérance analgésique. Selon les résultats obtenus, le profil d’internalisation du DOR induits par les ligands ne permet pas d’expliquer l’inhibition persistante de l’AMPc.
Opioids are the most powerful analgesics but their prolonged use can cause the development of analgesic tolerance. The tolerance may be associated with the duration of response to cAMP. For this study, we used a biosensor based on Bioluminescence Resonnance Energy Transfer technology that provides measurements of cAMP levels as a function of real time. The aim of our study was to determine whether there is a correlation between the internalization profile of delta-opioid receptor (DOR) when stimulated by different ligands, with respect to the duration of signaling in the short-term (≤15 min) and long term (120 min). This evolution of the duration of cAMP inhibition is biphasic and is explained in part by the efficiency of ligands to promote Gαi activation and by the profile of internalization for each of the different ligands used in this study. For example, deltorphin II which displayed high efficiency to promote Gαi activation and internalization shows a more pronounced decline in cAMP response, unlike morphine which displayed low efficiency to promote Gαi activation, and a poor sequestration, displaying a minimal response decay of inhibition of cAMP. However, ligand ability to promote internalization of DOR does not explain the kinetic profile of a persistent inhibition of cAMP over a longer period of 120 min.
APA, Harvard, Vancouver, ISO, and other styles
28

Charfi, Iness. "Étude du trafic du récepteur delta-opiacé suite à sa stimulation par différents agonistes." Thèse, 2012. http://hdl.handle.net/1866/8899.

Full text
Abstract:
Les opiacés figurent parmi les analgésiques les plus puissants pour le traitement des douleurs sévères. Les agonistes du DOR (récepteur delta opiacé) induisent moins d'effets secondaires que ceux du mu, ce qui les rend une cible d'intérêt pour le traitement des douleurs chroniques. Cependant, ils induisent la tolérance à l'analgésie. Des hypothèses récentes proposent que le potentiel des drogues à induire la tolérance soit la conséquence de la stabilisation de différentes conformations du récepteur induites par la liaison avec différents ligands, chacune ayant différentes propriétés de trafic. Dans ce contexte, nous avons déterminé si différents ligands du DOR différaient dans leur capacité à induire la signalisation et le trafic du récepteur. Nos résultats indiquent que DPDPE et SNC-80 sont les drogues les plus efficaces à inhiber la production d’AMPc, suivis par UFP-512, morphine et TIPP. DPDPE et SNC-80 induisent à eux seuls l’internalisation du DOR dans les cellules HEK-293 de façon dépendante de la β-arrestine mais pas de la GRK2 ni PKC. Ces deux drogues induisent également l’internalisation du DOR dans les neurones corticaux et c’est seulement le DPDPE qui permet au DOR de regagner la membrane des cellules HEK-293 et des neurones après récupération. Cette capacité de recyclage était suggérée comme un mécanisme protégeant contre la survenue de la tolérance. Ces observations indiquent que le DOR peut subir différentes régulations en fonction du ligand lui étant associé. Cette propriété de sélectivité fonctionnelle des ligands pourrait être utile pour le développement de nouveaux opiacés ayant une activité analgésique plus durable.
Opiates are among the most powerful painkillers to treat severe pain. Delta opioid receptor (DOR) agonists induce fewer side effects than mu opioid receptor agonists, which makes them a target of interest for the treatment of chronic pain. However, they induce tolerance to analgesia. Recent hypotheses suggest that drugs tolerance is the result of stabilization of ligand-specific conformations of the receptor, with distinct traffic properties such as internalization and/or recycling. In this context, we determined whether different DOR ligands differed with respect to their ability to induce signaling and receptor trafficking. Our results indicate that DPDPE and SNC-80 are the most effective drugs to inhibit the production of cAMP, followed by UFP-512, morphine and TIPP. Only DPDPE and SNC-80 manage to induce DOR internalization in HEK-293 cells. This effect is dependent on β-arrestin but not on GRK2 or PKC. Of these two internalizing agonists, only DPDPE allows the DOR to recycle back to the membrane of HEK-293 cells after recovery. DPDPE and SNC-80 also trigger similar DOR internalization in cortical neurons, and as observed in HEK293 cells only DPDPE allowed the receptor to recycle back to the membrane. This recycling capacity was suggested as a mechanism to protect against the onset of tolerance. These observations indicate that the DOR can undergo different regulations depending on the ligand bound to it. This property of functional selectivity of DOR ligands could be useful for the development of new opiates with longer lasting analgesic properties.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography