• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 9
  • 4
  • 4
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 23
  • 23
  • 23
  • 10
  • 10
  • 10
  • 9
  • 8
  • 8
  • 8
  • 8
  • 8
  • 7
  • 7
  • 7
  • 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.
11

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.
12

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.
13

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.
14

Participação dos receptores delta e kappa -opioides centrais no controle do apetite por sódio em ratos estimulados a ingerir solução salina hipertônica

Nascimento, Ana Isabel Reis January 2015 (has links)
Submitted by Ana Maria Fiscina Sampaio (fiscina@bahia.fiocruz.br) on 2015-10-13T12:34:40Z No. of bitstreams: 1 Ana Isabel Reis Nascimento. Participação...2015.pdf: 1631129 bytes, checksum: d14aa063c882bb96ee29f85751abf2e6 (MD5) / Approved for entry into archive by Ana Maria Fiscina Sampaio (fiscina@bahia.fiocruz.br) on 2015-10-13T12:34:59Z (GMT) No. of bitstreams: 1 Ana Isabel Reis Nascimento. Participação...2015.pdf: 1631129 bytes, checksum: d14aa063c882bb96ee29f85751abf2e6 (MD5) / Made available in DSpace on 2015-10-13T12:34:59Z (GMT). No. of bitstreams: 1 Ana Isabel Reis Nascimento. Participação...2015.pdf: 1631129 bytes, checksum: d14aa063c882bb96ee29f85751abf2e6 (MD5) Previous issue date: 2015 / Fundação Oswaldo Cruz. Centro de Pesquisas Gonçalo Moniz. Salvador, BA, Brasil / Alguns estudos sugerem que as vias opioidérgicas centrais parecem desempenhar um papel regulatório no controle da ingestão de água e sal em mamíferos. As ações dos opioides centrais sobre a regulação do controle hidroeletrolítico são mediadas por vários dos subtipos de receptores opioides. O papel dos receptores delta e kappa-opioides centrais neste processo não está adequadamente elucidado sendo necessário mais estudos que o esclareçam. Objetivo: Este estudo investigou o envolvimento dos receptores delta e kappa-opioides centrais no apetite por sódio em ratos depletados deste íon e em rato ativados centralmente com angiotensina. Material e Métodos: Foram utilizados ratos Wistar (270 ± 20 g), submetidos à cirurgia estereotáxica para implante de cânula guia no ventrículo lateral esquerdo (VL), no órgão subfornical (OSF), no núcleo preóptico mediano (MnPO) e no núcleo basolateral da amígdala (BLA). No protocolo de depleção de sódio os animais foram submetidos à injeção subcutânea de furosemida combinada com dieta hipossódica quatro dias após a cirurgia. Neste modelo de estudo os animais receberam injeção intracerebroventricular (i.c.v.) do antagonista delta-opioide naltrindole no quinto dia pós-cirúrgico, nas doses de 5, 10 e 20 nmol/2 μL e do antagonista kappa-opioide, norbinaltorfimina, injetado no OSF, MnPO e BLA, nas doses de 0,5, 1,0 e 2,0 nmol/0,2 μL.. O agonista específico para os receptores delta-opioides, deltorfina II (2,5, 5,0, 10 e 20 nmol/2 μL), foi injetado i.c.v. em animais depletados de sódio pré-tratados com naltrindole na dose de 20 nmol/2 μL e em animais repletos de sódio na dose de 20 nmol/2 μL. O agonista kappa-opioide, ICI199,441 (2,0 nmol/0,2 μL) foi injetado no OSF, MnPO e BLA em animais depletados de sódio pré-tratados com norbinaltorfimina 2,0 nmol/0,2 μL e em animais repletos de sódio na dose de 2,0 nmol/0,2 μL. Bebedouros de água destilada (H2Od) e de salina foram introduzidos nas caixas15 minutos após a injeção central e tiveram seus volumes monitorados nos tempos 5, 10, 15, 30, 45, 60, 90 e 120 minutos, após a colocação dos bebedouros. No protocolo de ativação angiotensinérgica central, quarto dia após a cirurgia os animais sofreram administração i.c.v. de naltrindole (5, 10 e 20 nmol/2 μL) 15 minutos antes de receberem injeções de angiotensina II na dose de 10 ng/2 μL. Os bebedouros de H2Od e de solução salina foram introduzidos nas caixas logo após a segunda injeção e tiveram seus volumes monitorados nos tempos 5, 10, 15, 30, 45, 60, 90 e 120 minutos, após a colocação dos bebedouros. Para verificar a especificidade de ação dos antagonistas opioides os animais foram submetidos aos testes de sobremesa, campo aberto e medida da pressão arterial. A análise estatística utilizada foi ANOVA modelo misto para medidas repetidas seguida do pós-teste de Bonferroni para múltiplas comparações dos volumes ingeridos e teste “t” de Student não pareado para análise dos testes de comportamento, através do programa GraphPad Prism 6.0. Resultados: Os grupos de ratos que receberam injeções i.c.v. de naltrindole após depleção de sódio e ativação angiotensinérgica central, apresentaram redução estatisticamente significante na ingestão de salina quando comparados ao grupo de animais controles. Os ratos que receberam injeção de norbinaltorfimina no OSF, MnPO e BLA após depleção de sódio apresentaram redução estatisticamente significante na ingestão de salina quando comparados ao grupo de animais controles. A estimulação dos receptores delta-opioides em animais repletos de sódio aumentou a ingestão de salina hipertônica. Conclusões: Os dados presentes sugerem que os receptores delta-opioides centrais e os receptores kappa-opioides localizados no OSF, MnPO e BLA parecem desempenhar papel fundamental na expressão do comportamento de aquisição de sal em ratos que sofreram depleção de sódio e ativação central do apetite por sódio induzido pela via angiotensinérgica. / Central opioid pathways seem to have an important role on the control of water and salt intake in mammals, and brain opioid peptides may influence hydroelectrolyte balance through a myriad of actions mediated by distinct opioid receptors. The specific role of central delta and kappa-opioid receptors (DOR and KOR) in this process is far from being fully understood. In the present work, we investigated the role of those receptors in the control of water and salt intake, in sodium-depleted rats and rats with activation central angiotensinergic. Method: Wistar male rats (250 ± 20 g) were used in the experiment after stereotaxic cannulation of the VL left, SFO, MnPO and BLA. To study the effect of the blockade of central DOR and KOR on water and salt intake in rats were sodium depleted by the concomitant use of s.c. injections of furosemide and were kept in hypossodic diet, five days after surgery. In the sixth day, they received i.c.v. injections of a selective delta-opioid receptor antagonist (naltrindole) at the doses of 5, 10 and 20 nmol/2 μL and injections in the SFO, MnPO and BLA of a selective kappa-opioid receptor antagonist (norbinaltorphimine) at the doses of 0.5, 1.0 and 2.0 nmol/0.2 μL. The specific agonist for delta-opioid receptor deltorphin II (2.5, 5.0, 10 and 20 nmol / 2 !L) was injected i.c.v. in animals depleted pretreated with sodium naltrindole at the dose 20 nmol /2 !L . The kappa-opioid agonist, ICI199,441 (2 nmol /0.2 !L) was injected into the SFO, MnPO and BLA in animals depleted pretreated with sodium norbinaltorphimine 2.0 nmol / 0.2 !L. Bottles containing water or hypertonic saline solution were introduced into the cages 15 min after the central administration. To study the effect of the blockade of central DOR and KOR on water and salt intake in animals after central angiotensinergic stimulation, the animals received intracerebroventricular injections of naltrindole at the doses of 5, 10 and 20 nmol/2 μL 30 min before receiving central injections of angiotensin II at the dose of 10 ng/2 μL. In this case, bottles containing water or hypertonic saline solution were introduced into the cages immediately after the central administration of angiotensin II. Water and salt intake were recorded for the next 2 hours after the introduction of the bottles into the cages. To verify the specificity of action of opioid antagonists animals were submitted to the dessert test, open field and measurement of blood pressure. Data were analyzed by Two-Way ANOVA mixed model followed by Bonferroni as post-hoc test. Results: The groups of rats that received i.c.v. injections naltrindole after sodium depletion and central angiotensinergic activation, showed a statistically significant reduction in salt intake when compared to control animals group. Rats receiving norbialtorphimine injection in the SFO, MnPO and BLA after sodium depletion showed a statistically significant reduction in salt intake when compared to control animals group. The stimulation of delta-opioid receptors in animals full of sodium increased intake of hypertonic saline. Conclusions: The present data suggest that the delta-opioid receptors central, and the kappa-opioid receptors located in the SFO, MnPO and BLA appear to play a key role in the expression of the salt acquisition behavior in rats with sodium appetite.
15

Regulation of the endogenous opioid system by acute nicotine and nicotine withdrawal

McCarthy, Michael J. 27 April 2004 (has links)
No description available.
16

Régulation du complexe constitutif formé par le récepteur opioïde delta et le canal potassique de la famille Kir3

Nagi, Karim 01 1900 (has links)
Les opioïdes sont les analgésiques les plus efficaces dans le traitement des douleurs sévères. Ils produisent leurs effets en ciblant spécifiquement les récepteurs opioïdes localisés tout le long de la voie de perception de la douleur où ils modulent la transmission de l'information douloureuse. La plupart des études dans ce domaine essaient de caractériser les récepteurs opioïdes à l'état isolé de tout partenaire de signalisation. Cette thèse, par contre, montre que le récepteur opioïde delta (DOR) peut former un complexe avec sa protéine G et l'un de ses effecteurs impliqués dans la production de l'effet analgésique, le canal potassique à rectification entrante activée par les protéines G (Kir3 ou GIRK). Après avoir établi la présence de ce complexe constitutif, on a ensuite caractérisé sa stabilité, modulation et régulation suite à une stimulation avec des agonistes opioïdes. En premier lieu, on a caractérisé la transmission de l'information du récepteur DOR, suite à son activation par un agoniste, vers le canal Kir3. On a remarqué que cette transmission ne suit pas le modèle de collision, généralement accepté, mais nécessite plutôt un simple changement dans la conformation du complexe préformé. Ensuite, on a déterminé que même suite à l'activation prolongée du récepteur DOR par un agoniste complet, le complexe DOR/Kir3 maintenait son intégrité et a été reconnu par la βarrestine (βarr) comme une seule unité signalétique provoquant ainsi l'internalisation de DOR et Kir3 par un mécanisme clathrine et dynamine-dépendant. Ainsi, prises ensemble, ces données montrent que l'activation du récepteur DOR déclenche non seulement l'activation de l'effecteur Kir3 mais également un mécanisme de régulation qui élimine cet effecteur de la membrane plasmique. / Opioids are the most effective analgesics in the treatment of severe pain. They produce their effects by specifically targeting opioid receptors located all along the pain perception pathway where they modulate the transmission of pain information. Most studies in this area try to characterize the opioid receptor in isolation from any signaling partner. This thesis, on the other hand, shows that the delta opioid receptor (DOR) can form a complex with its G protein and one of its effectors involved in the production of the analgesic effect, the G protein coupled inward rectifying potassium channel (Kir3 or GIRK). Having established the presence of this constitutive complex, we then characterized its stability, modulation and regulation following stimulation with opioid agonists. First, we characterized the transmission of information from DOR, following its activation by an agonist, to the Kir3 channel. We have noticed that this transmission does not follow the collision model, generally accepted, but rather requires a simple change in the conformation within the preformed complex. Then, we have determined that even following prolonged DOR activation by a full agonist, the DOR/Kir3 complex maintained its integrity and was recognized by βarrestin (βarr) as a single signaling unit producing the internalization of DOR and Kir3 by a clathrin and dynamin-dependent mechanism. Thus, taken together, these data show that DOR activation triggers not only activation of the Kir3 effector but also a regulatory mechanism that removes this effector from the plasma membrane.
17

慢性疼痛或壓力情境對於類鴉片delta受體的調節與其抗憂鬱功能的改變 / Effects of chronic pain or stress on the modulation of delta opioid receptor and its mediated antidepressant-like effect

陳昶名 Unknown Date (has links)
憂鬱症是盛行的精神疾病之一。慢性疼痛或是處在長期壓力情境的患者常與憂鬱症產生共病。在動物研究中,類鴉片delta受體制效劑能產生抗憂鬱效果,並且在發炎性疼痛的研究也指出類鴉片delta受體制效劑能展現抗痛覺過敏的效果。本研究主要利用大白鼠腦室內給予類鴉片delta受體制效劑SNC80以及三環抗憂鬱劑amitriptyline,來探討並比較其所產生的抗憂鬱效果在發炎性疼痛或長期壓力情境下與正常情境下的異同。大白鼠強迫游泳試驗被用來比較測試藥物的抗憂鬱效果;佛氏完全佐劑經由皮下注射至大白鼠右後腳掌底板來產生發炎性疼痛;腎上腺皮質酮經由皮下注射且持續21天來產生長期性壓力;西方墨點法用來檢驗在發炎性疼痛或長期壓力下,類鴉片delta受體蛋白質在大白鼠海馬迴的細胞膜上的改變。另外,拮抗劑實驗則用來確認類鴉片delta受體所產生的抗憂鬱效果。實驗結果顯示,大白鼠在正常情境下,SNC80及amitriptyline皆能產生抗憂鬱效果;然而在發炎性疼痛下,SNC80所產生的抗憂鬱效果有提高的表現,並且類鴉片delta受體蛋白質的數量在海馬迴的細胞膜上也隨著疼痛的時間增長而增加,amitriptyline則跟正常情境下的效果相似。另外,大白鼠在長期性壓力下,SNC80的抗憂鬱效果則沒有提高的表現,並且類鴉片delta受體蛋白質的數量在海馬迴的細胞膜上也未受到改變。本研究透過行為實驗提出類鴉片delta受體制效劑的藥理特性,並用分子生物學的方法來對應行為實驗的結果。本研究可做為未來類鴉片delta受體制效劑在治療慢性疼痛的憂鬱症患者上,可能發展為抗憂鬱藥的一個證據。 / Depression is one of the most prevalent mental illnesses all over the world. Patients with chronic pain or stress often have depression. Previous studies have shown that delta opioid receptor (DOR) agonists produced antidepressant-like effects in animal models and that antihyperalgesic effects of DOR agonists can be enhanced in rats under inflammatory pain. The aim of the study was to investigate and compare the antidepressant-like effects of a DOR agonist, SNC80, and a tricyclic antidepressant, amitriptyline, following intracerebroventricular (i.c.v.) administration in rats under different states. The forced swim test was used to determine the antidepressant-like effects of test compounds. Complete Freund’s adjuvant was injected subcutaneously into the right hind paw of rats to elicit inflammatory pain. Corticosterone was injected subcutaneously once per day for 21 days to induce chronic stress. The western blot was used to quantify the levels of DOR protein on plasma membrane in the hippocampus of rats under inflammatory pain or chronic stress. In addition, antagonist experiment was conducted to verify the receptor mechanism underlying the antidepressant-like effects of DOR agonist. Results indicated that i.c.v. SNC80 and amitriptyline dose-dependently produced antidepressant-like effects in rats under normal state. More importantly, the potency of SNC80-induced antidepressant-like effects, but not amitriptyline, was enhanced in rats under inflammatory pain. In addition, up-regulation of supraspinal DORs was time-dependently associated with enhanced antidepressant-like effects of SNC80 in rats under inflammatory pain. On the other hand, SNC80 did not produce enhanced antidepressant-like effects, and DOR density was not changed in rats under chronic stress. This study provides evidence of the DOR agonist’s state-dependent effects and suggests that DOR agonists may be more effective as potential antidepressants for patients with depression comorbid with chronic pain.
18

Vliv hypotermie na úspěch resuscitace a neurologické postižení po dlouhodobé srdeční zástavě léčené metodou Emergency Preservation and Resuscitation / The effect of hypothermia on outcome and neurologic injury after prolonged cardiac arrest treated by emergency preservation and delayed resuscitation

Drábek, Tomáš January 2013 (has links)
5 Summary: Currently, the outcomes from traumatic exsanguination cardiac arrest (CA) show that over 50% of deaths due to trauma occur at the scene, where medical care is limited. Less than 10% of patients who become pulseless from trauma survive. However, in an appropriate setting, some of those traumatic injuries could be surgically repairable. Emergency preservation and resuscitation (EPR) is a novel approach for resuscitation of exsanguination CA victims. EPR uses deep hypothermic preservation for prolonged CA to buy time for transport, damage control surgery, and delayed resuscitation with cardiopulmonary bypass (CPB). Initially, we used a dog model to maximize clinical relevance. We showed that the efficacy of EPR is related to the depth of hypothermia and duration of CA. Pharmacologic adjuncts tested to augment hypothermia generally failed. Extended hemorrhagic shock did not prevent the success of EPR vs. conventional resuscitation if extended post-resuscitative hypothermia was provided. Oxygenation of the flush allowed extending of survivable duration of deep hypothermic CA. Because of the lack of molecular tools available for use in dogs, we developed a rat EPR model to study the cellular and molecular mechanisms underlying deep hypothermic neuroprotection to allow us to define specific targets for...
19

CELLULAR AND BEHAVIORAL CHARACTARIZATION OF δ-OPIOID RECEPTOR MEDIATED ß-ARRESTIN SIGNALING

Arryn T Blaine (13154670) 26 July 2022 (has links)
<p>The following thesis will focus on understanding the downstream behavioral effects of δORmediated β-arrestinsignaling. δORagonists have been implicated as effective targets for a variety of diseases, however detrimental side effects of opioid-targeting agonists limit their clinical use. δORagonists specifically can induce seizures, however the underlying mechanism contributing to this  behavior  is  unknown.  We  review  this  phenomenon  in  more  detail,  highlighting  current agonists known to induce seizures and potential circuits and pathways involved. Our work suggests β-arrestinsignaling  is  involved,  specifically β-arrestin2  mediated  signaling  may  be  largely contributing  to δORagonist-induced  seizure  behavior.  As  it  is  possible  the β-arrestinisoforms have unique roles in seizure behavior, we also analyzed methods in which to provoke β-arrestinisoform bias of δORtargeting compounds. Though the full mechanism relating δORagonists with seizures remains unknown, our work provides foundational detail of this behavior, implicating the importance of β-arrestinisoform signaling through δOR; allowing for future studies to full define this seizure pathway and develop δORsafer agonists.  </p>
20

À la recherche de meilleurs traitements analgésiques : interactions entre le récepteur opioïde δ et ses différents agonistes

Lagréou, Alexandre 09 1900 (has links)
Les opioïdes restent encore à l’heure actuelle les composés pharmacologiques les plus efficaces pour traiter les différentes formes de douleurs, et donc fournir une analgésie thérapeutique. Cependant, l’administration répétée de ces composés entraîne des effets secondaires majeurs comme la dépression respiratoire, la tolérance, mais également, il a été montré que certains de ces opioïdes pouvaient engendrer des états proépileptiques. D’un point de vue thérapeutique, il existe donc un réel besoin pour de nouveaux et meilleurs traitements analgésiques, n’élicitant pas ces effets secondaires. Notre laboratoire étudie la signalétique des récepteurs couplés aux protéines G comme les récepteurs opioïdes et leur capacité de sélectivité fonctionnelle depuis des années, et en particulier celle du récepteur delta opioïde (DOP). En effet, celui-ci présenterait moins d’effets indésirables que le récepteur mu opioïde (MOP) qui est la cible principale des opioïdes classiques comme la morphine. Cependant, il semblerait que le DOP justement soit à l’origine des états proépileptiques précédemment décrits. Ainsi malgré la promesse initiale des agonistes delta par rapport à la diminution des effets secondaires, les effets proépileptiques de certains ont notamment contribué à une baisse d’intérêt vers le DOP et aucun de ses agonistes n’a pu passer les phases de tests cliniques. Cependant, il a été démontré que certains agonistes delta n’entraînaient pas d’effet proépileptique; tandis que d’autres oui. Comment expliquer un tel phénomène ? Ceci est la question que pose la présente recherche. Ainsi notre objectif sera d’obtenir et de comparer les signatures pharmacologiques des agonistes connus pour être proépileptiques versus ceux qui ne le sont pas ; par rapport à la transduction de signal via le récepteur delta opioïde et sa protéine G hétérotrimérique ; et par rapport à un de ses effecteurs principaux pour l’analgésie, un canal potassique rectifiant entrant. Cette comparaison se fera selon les paramètres du modèle classique de la pharmacologie, comme l’efficacité et la puissance ; mais également avec un outil plus récent appelé modèle opérationnel, utilisant des paramètres comme l’affinité et le coefficient de transduction. Pour se faire, le transfert d'énergie par résonance de bioluminescence ou BRET sera utilisé afin de caractériser les différentes voies signalétiques impliquées. Cette recherche s’inscrit dans un vaste contexte de collaboration entre différents laboratoires, et au sein de chacun d’entre eux, dans l’espoir de pouvoir synthétiser un jour, de meilleurs composés pharmacologiques, capables de cibler uniquement les voies médiatrices des effets thérapeutiques voulus, ici l’analgésie ; sans éliciter celles entraînant les effets secondaires associés, ici, les états proconvulsifs. L’aboutissement de cette recherche permettrait donc d’impacter la vie de millions de gens en souffrance, et c’est pourquoi il nous semble plus qu’important de continuer à l’entreprendre. / Opioids are still nowadays the most efficacious pharmacological compounds available to treat the different types of pain, and therefore provide a therapeutic analgesia. However, repeated administration of those compounds lead to major secondary effects like respiratory depression, tolerance, but also it was shown that some opioid compounds could induce seizures. From a therapeutical point of view, there is a serious need for new and better analgesic treatments that do not elicit such adverse effects. Our lab has been studying for years the signaletics of G-protein coupled receptors like the opioid receptors, and their capacity for functional selectivity, especially more recently the one of the delta opioid receptor (DOP). Indeed, this receptor elicits fewer adverse effects compared to the mu opioid receptor (MOP) that is the main target of all clinically used opioids such as morphine. However, it seems like the DOP itself would be responsible for the pro-epileptic states previously described. Thus, despite initial promises of the delta agonists towards reducing adverse effects whilst providing analgesia, the pro-convulsive effects that some seem to elicit have induced a loss of interest towards the DOP, and so far none of its agonists have gone further than pre-clinical trials. However, it has been shown that not all of those DOP agonists had those pro-convulsive adverse effects. How to explain such a phenomenon? This is the question which the present research will be asking. Thus our goal is to obtain and compare pharmacological signatures of the agonists known for being pro-convulsive versus those that are not ; regarding the transduction of signals through the delta opioid receptor and its heterotrimeric G-Protein ; and also regarding one of its main effectors to induce analgesia, an inwardly rectifying potassium channel. This comparison will be done according to the classical parameters of pharmacology, such as efficacy and potency ; but also according to the newest operational model, with parameters such as affinity and transduction coefficients. In order to do so, bioluminescence resonance energy transfer or BRET, will be used in order to characterize and quantify the signalling pathways there implicated. This research is embedded in a vast collaboration context, in between laboratories around the world, and within those laboratories as well, in hope to be able to one day synthesize, better pharmacological compounds, capable of targeting only the pathways responsible for the desired effects, here analgesia ; without triggering the associated adverse effects, here pro-convulsive states. The culmination of this research could allow to impact the lives of millions of people throughout the world, and this is why it is more than important for us to keep on pursuing it.

Page generated in 0.0597 seconds