• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 12
  • 4
  • 2
  • 2
  • Tagged with
  • 29
  • 29
  • 29
  • 6
  • 6
  • 5
  • 5
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
21

EVALUATION OF NATURALLY OCCURRING OPIOIDS AND SYNTHETIC DERIVATIVES FOR THERAPEUTIC APPLICATION IN ALCOHOL ABUSE AND PAIN

Anna M Gutridge (11819636) 19 December 2021 (has links)
<div> <p>Historically, natural products from plants, fungi, bacteria and animals have played an important role in the discovery of new drugs. In fact, it has been found that 34% of new FDA-approved drugs over the last 30 years were derived from natural products or their derivatives. Because of the chemical and structural diversity of natural products, they continue to be one of the best options for discovering novel compounds and scaffolds; this is especially true for compounds targeting the µ-, δ-, and κ- opioid receptors. However, traditional opioids such as morphine cause many therapeutically limiting side effects. Therefore, there have been immense efforts to develop opioids that avoid these side effects, with “signal-biased” compounds being an intense area of interest. The research presented here investigates of the biased mechanisms of compounds found in and derived from <i>Mitragyna speciosa</i>, also known as kratom, and <i>Picralima nitida</i>, also known as akuamma. Kratom and akuamma compounds are examined for their therapeutic potential in treating alcohol abuse and pain, respectively, two prevalent conditions with extreme societal and economic costs.</p> </div> <br>
22

Uncovering the Functional Implications of Mu- and Delta-opioid Receptor Heteromerization in the Brain

Kabli, Noufissa 20 June 2014 (has links)
Opioid Receptors (ORs) are involved in the pathophysiology of several neuropsychiatric conditions yet remain an untapped therapeutic resource. Although only mu-, delta-, and kappa-OR types have been cloned, additional subtypes result from complexes generated by direct receptor-receptor interactions. Mu- and delta-ORs form a heteromeric receptor complex with unique pharmacological and signalling properties distinct from those of mu- and delta-OR homomers. In these studies, we sought to characterize the ligand binding pocket and agonist-induced internalization profile of the mu-delta heteromer, to investigate mu-delta heteromer-specific signalling in brain, and to interrogate the contribution of this receptor complex to opioid-mediated behavioural effects. In competition radioligand binding studies, delta-agonists displaced high affinity mu-agonist binding from the mu-delta heteromer but not the muOR homomer, suggestive of delta-agonists occupying or allosterically modulating the muOR ligand binding pocket within the heteromer. Delta-agonists induced internalization of the mu-delta heteromer in a dose-dependent, pertussis toxin resistant, and muOR- and deltaOR-dependent manner from the cell surface via the clathrin and dynamin endocytic machinery. Agonist-induced internalization of the mu-delta heteromer persisted following chronic morphine treatment conditions which desensitized the muOR homomer. Using Galpha-specific GTPgammaS binding assays, we demonstrated that mu-delta heteromer signalling previously characterized in cell lines was present in the striatum and hippocampus, and did not desensitize following prolonged morphine treatment conditions which desensitized muOR homomer-mediated signalling. Since delta-agonists which also target the mu-delta heteromer possess antidepressant-like and anxiolytic-like properties, we investigated the role of this receptor complex in mood regulation. We devised a strategy to selectively analyze the effects of the mu-delta heteromer by dissociating it using a specific interfering peptide aimed at a sequence implicated in mu-delta heteromerization. The interfering peptide abolished the unique pharmacological and trafficking properties of delta-agonists at the mu-delta heteromer and dissociated this receptor complex in vitro. Intra-accumbens administration of the interfering peptide disrupted the mu-delta interaction in vivo and allowed for isolation of the mu-delta heteromer contribution to the mood-regulatory effects of a delta-agonist with activity at the heteromer. Activation of the mu-delta heteromer in the nucleus accumbens produced antidepressant-like and anxiolytic-like actions in animal models of depression and anxiety.
23

Uncovering the Functional Implications of Mu- and Delta-opioid Receptor Heteromerization in the Brain

Kabli, Noufissa 20 June 2014 (has links)
Opioid Receptors (ORs) are involved in the pathophysiology of several neuropsychiatric conditions yet remain an untapped therapeutic resource. Although only mu-, delta-, and kappa-OR types have been cloned, additional subtypes result from complexes generated by direct receptor-receptor interactions. Mu- and delta-ORs form a heteromeric receptor complex with unique pharmacological and signalling properties distinct from those of mu- and delta-OR homomers. In these studies, we sought to characterize the ligand binding pocket and agonist-induced internalization profile of the mu-delta heteromer, to investigate mu-delta heteromer-specific signalling in brain, and to interrogate the contribution of this receptor complex to opioid-mediated behavioural effects. In competition radioligand binding studies, delta-agonists displaced high affinity mu-agonist binding from the mu-delta heteromer but not the muOR homomer, suggestive of delta-agonists occupying or allosterically modulating the muOR ligand binding pocket within the heteromer. Delta-agonists induced internalization of the mu-delta heteromer in a dose-dependent, pertussis toxin resistant, and muOR- and deltaOR-dependent manner from the cell surface via the clathrin and dynamin endocytic machinery. Agonist-induced internalization of the mu-delta heteromer persisted following chronic morphine treatment conditions which desensitized the muOR homomer. Using Galpha-specific GTPgammaS binding assays, we demonstrated that mu-delta heteromer signalling previously characterized in cell lines was present in the striatum and hippocampus, and did not desensitize following prolonged morphine treatment conditions which desensitized muOR homomer-mediated signalling. Since delta-agonists which also target the mu-delta heteromer possess antidepressant-like and anxiolytic-like properties, we investigated the role of this receptor complex in mood regulation. We devised a strategy to selectively analyze the effects of the mu-delta heteromer by dissociating it using a specific interfering peptide aimed at a sequence implicated in mu-delta heteromerization. The interfering peptide abolished the unique pharmacological and trafficking properties of delta-agonists at the mu-delta heteromer and dissociated this receptor complex in vitro. Intra-accumbens administration of the interfering peptide disrupted the mu-delta interaction in vivo and allowed for isolation of the mu-delta heteromer contribution to the mood-regulatory effects of a delta-agonist with activity at the heteromer. Activation of the mu-delta heteromer in the nucleus accumbens produced antidepressant-like and anxiolytic-like actions in animal models of depression and anxiety.
24

La comorbidité entre dépendance aux opiacés et dépression : mécanismes sérotoninergiques dans un modèle murin / Comorbidity between opiate addiction and depression : serotonergic mechanisms in a mouse model

Lutz, Pierre-Eric 03 September 2012 (has links)
L’addiction ou dépendance aux substances psychoactives est une affection chronique, fréquente et grave, émaillée de rechutes et de périodes d’abstinence. Les études épidémiologiques montrent que l’abstinence aux opiacés est fortement associée à une prévalence accrue de la dépression. Nous résumons ici les principaux aspects cliniques de la dépendance aux opiacés et de la dépression, en détaillant leurs mécanismes physiopathologiques. Puis, nous présentons notre modèle d’abstinence aux opiacés chez la souris. Suite à un traitement morphinique chronique et au cours de l’abstinence apparaissent progressivement des comportements apparentés à la dépression. Ce traitement morphinique modifie profondément le fonctionnement du système sérotoninergique, notamment dans le noyau du raphé dorsal. De plus, les déficits comportementaux observés peuvent être prévenus par un traitement chronique par la fluoxétine, un antidépresseur ciblant ce système. Nous avons généralisé ce modèle à l’héroïne, un autre opiacé illicite. Nous avons révélé par des approches génétiques de délétion constitutive et conditionnelle les rôles distincts des 3 récepteurs opioïdes (mu, delta et kappa) lors de l’abstinence à l’héroïne. Enfin, nous avons initié une étude de caractérisation, à l’échelle de l’ensemble du génome, des adaptations transcriptomiques (ARN messagers et micro-ARN) dans le noyau du raphé dorsal au cours de l’abstinence à l’héroïne et du traitement antidépresseur. Ce travail devrait permettre d’améliorer notre compréhension des mécanismes neurobiologiques à l’œuvre dans la comorbidité entre dépendance aux opiacés et dépression et pourrait suggérer de nouvelles pistes thérapeutiques. / Addiction is a chronic, frequent and serious brain disease, with relapse alternating with abstinence periods. Epidemiological studies show that abstinence, notably from opiates, is strongly associated with depression.Here we present the main clinical aspects of opiate addiction and depression, and most recent advances in molecular pathophysiology of both disorders. Then, we present our mouse model of opiate abstinence. Following chronic morphine exposure, depressive-like behaviours progressively emerge. Morphine treatment profoundly disrupts serotonergic signalling, notably in the dorsal raphe nucleus. In addition, behavioural deficits can be prevented by chronic treatment with fluoxetine, an antidepressant targeting serotonergic neurons. We then generalized our mouse model to heroin, another major illicit opiate. Using constitutive and conditional knockout strategies, we documented distinct roles for all 3 opioid receptors (mu, delta and kappa) in heroin abstinence. Finally, we initiated a large-scale analysis of transcriptomic regulations (mRNA and micro-RNA) occurring in our model as a function of heroin abstinence and fluoxetine treatment.These studies should reveal an unforeseen contribution of the dorsal raphe nucleus to addiction. They should uncover new molecular mechanisms underlying depressive-like behaviors in mice during opiate abstinence and thus put forward new therapeutic targets in humans.
25

Mu opioid receptors and neuronal circuits of addiction : genetic approaches in mice / Récepteurs opioïdes mu et circuits neuronaux de l'addiction : approches génétiques chez la souris

Charbogne, Pauline 09 July 2015 (has links)
Le récepteur opioïde mu est responsable des propriétés analgésiques et addictives puissantes de la morphine et de l’héroïne, mais son mode d’action à l’échelle des circuits neuronaux est mal connu et a été peu étudié par des approches génétiques. Le récepteur mu est largement exprimé dans le système nerveux, essentiellement dans des neurones GABAergiques. Le premier objectif de mon projet a été d’inactiver le gène codant pour le récepteur mu dans les neurones GABAergiques du cerveau antérieur et d’en étudier les conséquences comportementales. Notre étude montre que ces récepteurs ne sont pas impliqués dans l’analgésie et la dépendance physique à la morphine, mais qu’ils sont essentiels à l’effet hyperlocomoteur de l’héroïne. De plus, nos résultats indiquent que ces récepteurs limitent la motivation à consommer de l’héroïne et du chocolat, révélant un rôle entièrement nouveau pour cette population particulière de récepteurs (Manuscrit 1 : Mu opioid receptors in GABAergic forebrain neurons are necessary for heroin hyperlocomotion and reduce motivation for heroin and palatable food). Aussi, cette population de récepteurs mu n’est pas responsable du syndrome autistique décrit chez les souris knockout totales (Manuscrit 2 : Mu opioid receptors in GABAergic forebrain neurons are not involved in autistic-like symptoms). Enfin, nous avons développé un nouveau modèle transgénique visant l’inactivation génétique du récepteur mu dans les neurones glutamatergiques, mais qui n’a pas abouti à un knockout conditionnel détectable. Nous avons aussi initié la création d’une lignée transgénique Cre pour l’inactivation de gènes d’intérêt dans l’amygdale étendue, qui permettra notamment d’étudier le rôle du récepteur mu dans ce microcircuit. / Mu opioid receptors mediate the strong analgesic and addictive properties of morphine and heroin;however mu receptor function at circuit levels is not well understood and has been poorly studied by genetic approaches. These receptors are widely expressed throughout the nervous system, essentially in GABAergic neurons. The first aim of my project was to genetically inactivate the mu receptor gene in GABAergic forebrain neurons and study the behavioral consequences. Our study shows that these mu receptors are not implicated in morphine-induced analgesia and physical dependence, but are essential for locomotor effects of heroin. Moreover, our data show that these receptors inhibit motivation to consume heroin and chocolate, revealing an entirely new role for this particular population of mu receptors (Manuscript 1: Mu opioid receptors in GABAergic forebrain neurons are necessary for heroin hyperlocomotion and reduce motivation for heroin and palatable food). Also, mu receptors expressed in forebrain GABAergic neurons are not responsible for the autistic syndrome described in total mu receptor knockout mice (Manuscript 2: Mu opioid receptors in GABAergic forebrain neurons are not involved in autistic-like symptoms). Finally, we developed a new transgenic model targeting the mu receptor gene in glutamatergic neurons, but receptor deletion was not detectable in conditional mice. We also initiated the creation of a transgenic Cre driver line to knockout genes of interest in the extended amygdala, and this tool will enable us to study mu receptor function within this microcircuit.
26

PERIPHERALLY RESTRICTED OPIOID CONJUGATES AND ITS USE AS PHARMACOLOGICAL PROBES AND POTENTIAL THERAPEUTICS

Tuhin, Md Tariqul Haque 01 January 2022 (has links)
Opioid-induced constipation (OIC) is one of the major adverse effects of opioid analgesics used by millions of patients each year. While progress has been made, there remains a significant unmet medical need in the treatment of OIC. Major gaps remain in our understanding of the role of the gastrointestinal tract and central nervous system (CNS) in precipitating OIC. For the last four decades, numerous investigations to study the sites of action of opioid analgesics have utilized peripherally acting mu-opioid receptor antagonists (PAMORAs), which have been incorrectly believed to have limited penetration across the blood-brain barrier (BBB). Several preclinical and clinical reports indicate that significant amounts of PAMORAs penetrate the BBB quite readily. As a result, the usage of current PAMORAs have resulted in misunderstandings of the role of the CNS and gastrointestinal tract in causing side effects such as opioid-induced constipation (OIC). We have developed a transthyretin-based novel drug delivery approach for restricting the passage of small molecules across the BBB. Our approach involves endowing the opioid agonist/antagonist with the selective transthyretin ligand, AG10. The newly synthesized naloxone- and oxycodone-based conjugates have demonstrated superior peripheral selectivity, improved pharmacokinetics, and efficacy in rats compared to other clinically used PAMORAs. Here we present chemical synthesis, in vitro binding and stability studies, as well as pharmacokinetic and pharmacodynamic evaluations of the AG10-opioid conjugates in rats. Our AG10-based PAMORA allowed us to obtain new insights into the important role of mu-opioid receptors in the central nervous system (CNS) in causing constipation. Additionally, our results demonstrate for the first time that synergy between mu-opioid receptors in the central nervous system and the gastrointestinal tract is crucial to the understanding of OIC and the development of effective treatment regimens. These findings contradict prior ideas that OIC was caused by a mechanism that involves primarily the gastrointestinal mu-opioid receptors. Moreover, we confirmed our findings by a AG10-oxycodone conjugate, a peripherally restricted opioid agonist. This molecule demonstrated the predominant role of CNS in OIC precipitation. The newly synthesized AG10-opioid conjugates represent a novel class of pharmacological probes that will aid in our understanding of OIC and other undesirable adverse effects of opioids. In addition, these conjugates have been evaluated for their potential therapeutic value in the preclinical studies. Collectively our approach to limit the BBB penetration of opioids will contribute to develop safer and more effective opioid medications.
27

Long-Term Opiate-Induced Adaptations in Lateral Paracapsular Neurons of the Basolateral Amygdala

Werner, Sara Jane 09 April 2020 (has links)
Increases in basolateral amygdala (BLA) activity drive avoidance-seeking behavior that may be associated with stress induced drug seeking. Activity of BLA pyramidal neurons is regulated by local and paracapsular gamma aminobutyric acid (GABA) interneurons. The lateral paracapsular interneurons (LPCs) border the external capsule, receive dense cortical/thalamic input and provide feed-forward inhibition onto BLA principle neurons. The GABAergic LPCs also express high concentrations of g-protein coupled µ-opioid receptors (MORs). Therefore, the effects of opiates on LPC activity and local GABA release were examined. Fluorescently double labeled LPCs were observed in glutamate decarboxylase (GAD) 65-mcherry/GAD67-green fluorescent protein (GFP) transgenic mice. Whole-cell electrophysiology experiments demonstrated that acute exposure to [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO; a synthetic selective MOR agonist), reduced LPC firing and spontaneous inhibitory postsynaptic current (sIPSC) frequency in LPCs, with no apparent effect on spontaneous excitatory currents (sEPSCs). Current injection induced firing in LPC neurons, but less effectively than in saline controls. Morphine-exposed mice (10mg/kg/day, across 5 days, 1-2 days off) had increased sIPSCs compared to saline-injected controls, as well as enhanced adenylyl cyclase (AC) activity. Together these data show that LPC neurons are a highly sensitive targets for opiate-induced inhibition, and that long-term opiate exposure results in impaired LPC excitability, possibly contributing to anxiety observed during opiate withdrawal.
28

Neuronal and Molecular Adaptations of GABA Neurons in the Ventral Tegmental Area to Chronic Alcohol

Hales, Kimberly 03 December 2007 (has links) (PDF)
The purpose of this thesis project was to examine the effects of chronic alcohol on the excitability and molecular adaptation of GABA neurons of the ventral tegmental area (VTA). GABA neurons are of interest with regards to ethanol intoxication, reinforcement, and dependence due to their widespread distribution and connectivity to mesocorticolimbic dopamine (DA) neurons implicated in alcohol reward and addiction. Since we have previously shown adaptation of VTA GABA neuron firing rate to chronic ethanol (Gallegos, Criado et al. 1999) and suppression of gap-junction (GJ) mediated coupling between these neurons by acute ethanol (Stobbs, Ohran et al. 2004), we wanted to further characterize the effects of chronic ethanol on VTA GABA neuron excitability, electrical coupling and molecular adaptation. In particular, we analyzed the GJ mediated coupling and protein regulation of VTA GABA neurons following a three week period of continuous ethanol exposure via liquid diet. Although some animals showed tolerance, there was no significant tolerance to ethanol inhibition of GJ-mediated electrical coupling. In addition, we were able to characterize differences in mRNA expression levels for the DA synthesizing enzyme tyrosine hydroxylase (TH), the DA D2 receptor and the NMDAR2B receptor subunit in DA versus GABA neurons, all three of which were expressed at higher levels in DA neurons. We also determined the effects of chronic ethanol on mRNA levels of these same proteins as well as μ-opioid receptors (μORs) and connexin-36 (Cx36) GJs. Most significantly, we found a down-regulation of the DA D2 receptor, confirming that molecular modification occurs in these VTA GABA neurons with chronic alcohol. While we reject our hypothesis that acute ethanol inhibition of VTA GABA neuron electrical coupling would undergo tolerance to chronic ethanol in these non-dependent rats, which was the focus of this thesis, it remains to be determined if tolerance to chronic ethanol might be obtained in ethanol-dependent rats.
29

Development of Pharmacologically Distinct Opioid Analgesics

Patel, Shivani 29 September 2022 (has links)
Opioid analgesics have been a major contribution to pain therapy with opioids being used as an effective treatment for various recalcitrant pain conditions. The drug class has come under increased scrutiny due to the raising concerns about the public health crisis of opioid misuse and addiction, thereby increasing the need for alternative and safer analgesics. The exploration of alternative pharmacotherapy for pain management has led to an increasing paradigm shift towards the development of a single-drug-multiple-target approach that takes inspiration from numerous naturally occurring drugs. The mu-opioid receptor has been the primary target for the management of pain; however, the voltage-gated sodium channel Nav1.7 is gaining attention as a putative antinociceptive target based on human genetic evidence. The proposed research aims to develop multi-target directed ligands (MTDL) that modulates two key targets for pain perception, the MOR, and Nav1.7 to generate analgesics with reduced side effects and enhanced analgesia. This will be achieved by exploiting polypharmacology to develop hybrid analgesia in two ways: (i) performing structure-activity relationship (SAR) studies to design a single drug with two pharmacophores that specifically interacts with both the targets (ii) exploiting in silico techniques by performing structure-based virtual ligand screening (VLS) of a chemical library. In our work, we report that through SAR studies and molecular docking studies that the designed compounds having in combination the pharmacophore of PZM21 and aryl sulfonamide demonstrate significant interactions between the active compounds and both the MOR and Nav1.7 proteins. This study also reports the first ever bifunctional virtual ligand screening where a library consisting of over a million compounds was screened for bifunctional activity at the MOR and the Nav1.7 ion channel. We also report the development of a novel mechanism-specific membrane potential assay to that can be used to screen for subtype selective Nav1.7 inhibitors. The research performed in this thesis will serve as a platform to explore the possibility of MTDL as potential therapeutic solutions to diseases of complex etiologies such as chronic pain. It will also serve as a starting point to exploring bifunctional VLS as a way to screen large chemical libraries for MTDLs.

Page generated in 0.0514 seconds