• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 16
  • 4
  • 4
  • 2
  • Tagged with
  • 36
  • 36
  • 29
  • 7
  • 6
  • 6
  • 6
  • 6
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 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

DESIGN, SYNTHESES, AND BIOLOGICAL EVALUATION OF 14-N-SUBSTITUTED NALTREXONE DERIVATIVES AS OPIOID RECEPTOR LIGANDS

Elbegdorj, Orgil 29 January 2013 (has links)
Opium, the dried resin obtained from the unripe seedpods of the poppy flower, has been used for medicinal and euphoric purposes since ancient times. Morphine, the main active ingredient of opium, and other clinically useful opioid analgesics all mediate their effects through activating the mu opioid receptor. Studies involving the mu opioid receptor knockout mice showed that the interaction with the mu opioid receptor is also responsible for many notorious side effects associated with these drugs including dependence and addiction. Therefore, selective antagonists for the mu opioid receptor are needed to study its function in drug abuse and addiction. Previously, based on molecular modeling studies and the “message-address” concept, a series of 14-O-substituted naltrexone derivatives were designed and synthesized. These compounds carried an ester-linked heteroaromatic substitution at the 14-position of naltrexone which was designed to interact with the putative “address” site, that was identified in the mu opioid receptor through molecular modeling studies. The lead compound of this series was determined to have a high affinity and selectivity for the mu opioid receptor. Because the 14-O-substituted naltrexone derivatives were not very stable, the ester linkage in these compounds was replaced by an amide one and a series of 14-N-substituted naltrexone derivatives were synthesized. The affinity and selectivity of these novel naltrexone derivatives were determined in a competitive radioligand binding assay. Interestingly, the 14-N-substituted naltrexone derivatives did not maintain the high selectivity of the 14-O-substituted series. It was hypothesized that the conformational constraint introduced by the amide linker was detrimental to the mu opioid receptor selectivity. Therefore, three 14-N-substituted naltrexone derivatives which carried more flexible linkages were synthesized and evaluated. The mu opioid receptor selectivity was not recovered by introducing rotational freedom into the linker. Some of these 14-N-substitued naltrexone derivatives were determined to be mu-kappa opioid receptor dual selective antagonists. Since the mu opioid receptor antagonists are effective at treating drug addiction, while growing evidence suggests that the kappa opioid receptor antagonists may be beneficial in lowering drug cravings, these novel mu-kappa opioid receptor dual selective antagonists may find unique clinical utility in the treatment of opioid dependence.
22

Design, Synthesis and Pharmacological Characterization of Potential Mu Opioid Receptor Selective Ligands

Kulkarni, Abhishek S 01 January 2019 (has links)
Selective Mu Opioid Receptor (MOR) antagonists possess immense potential in the treatment of opioid abuse/addiction. Utilizing the “message-address” concept, our laboratory reported a novel, reversible, non-peptide MOR selective antagonist 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-[(4՛-pyridyl)carboxamido]morphinan (NAP). Molecular modeling studies revealed that the selectivity of NAP for the MOR is because of a π-π stacking interaction of its pyridine ring with the Trp318residue in theMOR. Pharmacological characterization showed that NAP is a P-glycoprotein substrate, thereby limiting its use in the treatment of opioid abuse/addiction. Thus, to modify NAP, we replaced the pyridine ring with its isosteric counterpart thiophene. Isosteric replacement could lead to development of compounds with different pharmacologic properties. Additionally, exploring other ring systems would diversify and enrich our library of compounds and aid in establishing a comprehensive structure-activity relationship. Therefore, newly synthesized compounds included thiophene derivatives of 6α/β-naltrexamine with potential to be used in the treatment of opioid abuse/addiction. Preliminary in vivo screening revealed that compounds 8 and 11 could be acting as antagonists. To aid in the design and synthesis of newer generation of MOR selective analogs, a 3-Dimensional Quantitative Structure-Activity Relationship (3D-QSAR) Comparative Molecular Field Analysis (CoMFA) on 6β-N-heterocyclic substituted naltrexamine derivatives was conducted. After rigorous optimizations, the best CoMFA model possessed low predictive power. Results obtained suggested that small structural changes could lead to significant change in binding modes of these ligands. To further validate this observation, molecular docking studies were performed which revealed that these ligands indeed possessed multiple distinct binding modes thereby offering rationale for the CoMFA results. Thus, overall this study furnished useful information about the complexity of protein-ligand interactions which will aid in designing more potent and selective MOR ligands.
23

Store-Operated Calcium Channels in the Function of Intracardiac Neurons

Bonds, Timetria 01 January 2012 (has links)
Proper autonomic regulation of mammalian cardiac function is dependent upon very complex and precise communication among the intracardiac ganglia and individual neurons within the ganglia. An array of neuromodulators is found within the ganglia that direct neuronal activity by modulating the movement of calcium. The current study determines that opioidergic agonists, which have been found to contribute to severe cardiac disease states and intracellular calcium mobilization, are also responsible for changes in the function of the intracardiac neuron via their effects on store-operated calcium channels (SOCs). Previous studies suggest that phosphorylation plays a role in SOC regulation. Using Fura-2 calcium fluorometry, we determined that protein kinase A (PKA), protein kinase C (PKC), and cyclic adenosine monophosphate (cAMP) had no effect on store-operated calcium entry in the presence of antagonists, phorbol 12, 13 dibutyrate (PDBu), forskolin, and 8-Br cAMP, respectively. We also found pharmacologically that using both electrophysiology and calcium imaging that μ-opioid agonists, met-enkephalin (ME) and endomorphin (EM) depress SOC activity in intracardiac neurons. Arachidonic acid (AA), which has been found to depress SOC function in rat liver cells and μ-opioid receptor activation (MOR), blocked both store-operated calcium entry (SOCE) and the calcium release-activated current (ICRAC) significantly. Contrastingly, AA metabolites, prostaglandin E2)(PGE2) and prostaglandin D2 (PGD2), do not significantly influence SOCE which suggests that the effects of AA may be direct. The block elicited by EM was partially reversed by pertussis toxin (PTX), indicative of activation of a PTX-sensitive G-protein following MOR activation. Similarly, PLA2 inhibitors, OBAA and AACOCF3, decreased the percent block of SOCE due to opioid agonist-induced inhibition. Using the perforated-patch method of I-clamp electrophysiology, we demonstrated that gadolinium, at low micromolar concentrations, reversibly reduced action potential firing. Importantly, these results suggest that SOCs may influence action potential firing in mammalian intracardiac neurons. Similarly, AA and EM depressed action potential firing. Taken together, these experiments suggest that a pathway involving EM and AA influences repetitive firing through SOC inhibition. The importance of SOCs in the maintenance of action potential firing and more specifically, the expression and biophysical functionality of the individual pore-forming subunits (Orai1, 2, and 3) in any neuronal cell type has previously not been explored. Quantitative RT-PCR along with I-clamp electrophysiology revealed that Orai3 was exclusive to repetitively firing neurons. As a result, we hypothesize that robust Ca2+-dependent fast inactivation, also associated Orai3, is a factor in the maintenance of repetitive action potential firing. Using Fura-2 calcium fluorometry and patch-clamp electrophysiology, we determined pharmacologically that μ-opioid receptor activation precedes an intracellular cascade that is dependent on a PTX-sensitive G-protein and AA but independent of prostaglandin and protein kinase activity. Finally, we used RT-PCR to determine the Orai subunits expressed in the intracardiac neurons and their influence on neuronal firing patterns. This study is the first to determine the role expressed subunits has in the maintenance of the electrical activity of the neuron.
24

Imaging nociceptive signaling in peripheral CGRP terminal fibres

2015 June 1900 (has links)
In this dissertation I introduce a simple experimental approach for studying afferent pain fibre physiology. I developed an en bloc dural-skull preparation that pairs electrophysiological stimulations, pharmacological manipulations, and the UV photolysis of caged compounds in and around selectively identified individual C-fibre nociceptors with microfluorometric imaging of Ca2+ responses. This allows the observation of physiological functioning in individual nociceptive fibre free nerve endings. I show high-resolution functional imaging of single action potential-evoked fluorescent transients, as well as sub- and supra-threshold calcium signaling events within individual nociceptive fibre terminations. Utilizing the dural-skull preparation I was able to identify a peripheral mechanism of action in the terminals of CGRP nociceptive fibres for an effective migraine therapeutic, the selective 5-HT1 receptor agonist, sumatriptan. I found sumatriptan to cause an approximately 40% reduction in the amplitude of action potential-evoked Ca2+ transients in the peripheral terminals of CGRP nociceptive fibres that was mediated selectively through the inhibition of N-type Ca2+ channels. Observations from this study support a peripheral site of action for sumatriptan in inhibiting the activity of dural pain fibres and adds to our understanding of the mechanisms that underlie the clinical effectiveness of 5-HT1 receptor agonists such as sumatriptan. While μ-opioid receptor agonists remain the most powerful analgesics for the treatment of severe pain, their mechanism of action in peripheral primary afferent pain fibres remain to be established. Further exploiting the dural-skull preparation I found activation of μ-opioid receptors in individual CGRP terminals had a dual modulatory effect; inhibition of N-type Ca2+ channel signaling and a frequency dependent, BKCa channel-mediated, suppression of action potential firing. These results establish possible anti-nociceptive mechanisms of μ-opioid receptor activation in the peripheral terminals of CGRP nociceptive fibres and identify new pathways to target for peripherally mediated analgesia. The development and subsequent testing of the dural-skull preparation in this dissertation displays its utility and opens up a new window for studying nociceptive fibre physiology and pathophysiology.
25

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>
26

Mu opioid receptors in the habenula : dissecting reward and aversion in addiction / Récepteurs Mu aux opioïdes dans l'habénula : récompense et/ou aversion dans l'addiction

Boulos, Laura-Joy 19 December 2017 (has links)
Les récepteurs mu aux opioïdes (MORs) jouent un rôle central dans l’addiction et ce majoritairement via le contrôle qu’ils exercent sur les phénomènes de récompense. Les effets des MORs sur le système de récompense sont généralement attribués à leur expression dans le circuit mésocorticolimbique. Les MORs sont toutefois exprimés dans d’autres régions du cerveau, notamment dans une petite structure cérébrale épithalamique qui exprime la plus forte densité de MORs : l’habénula médiane (MHb). Le rôle de cette population spécifique de récepteurs n’a jamais été exploré, malgré l’implication fortement suggérée dans la littérature de l’habénula dans l’addiction et les processus de récompense/aversion sous-jacents. Nous avons donc généré un modèle de souris knockout conditionnel chez lesquelles nous supprimons les MORs uniquement dans l’habénula et nous les avons soumis à des tests comportementaux dans le but d’évaluer les fonctions de ces récepteurs et leur impact sur le comportement avec un focus sur les processus aversifs, cognitifs et récompensant. Nos résultats révèlent que les récepteurs mu que nous supprimons dans l’habénula limitent l’aversion somatique et affective mais ne modulent ni les réponses locomotrices, analgésiques et de récompense à la morphine, ni les fonctions cognitives que nous avons testées. Nous identifions donc pour la première fois une population de MORs dans l’habénula qui freinent l’aversion et nous suggérons par là-même que les récepteurs mu de l’habénula pourraient être cruciaux dans le stage «sevrage aversif» des cycles d’addiction. / Mu opioid receptors (MORs) have been extensively studied for their addictive properties that are thought to operate through the control of reward processes. While the importance of MORs in reward is generally attributed to their presence in the mesocorticolimbic circuitry, their role in the medial habenula (MHb), the structure in which MORs are most densely expressed, remains unexplored to date. This is quite surprising given the increasing literature on the habenula’s role in addiction as well as reward/aversion processes. Here we generated a conditional knockout mouse model that lacks MORs solely in the MH band we investigated the contribution of habenular MORs in brain functions and behavioural out comes with emphasis on reward, aversion and cognition. While the performance of our mutant model did not differ in locomotor, analgesic and reward responses to morphine norincognitive tasks compared to control mice, we uncovered a novel role for MORs in aversive states.This is the first report demonstrating that MORs control both somatic and affective aversion specifically at the level of the MHb. Habenular MORs could thus be crucial to the aversive with drawal stage of addiction cycles that is thought to increase craving and prevent success in quitting.
27

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.
28

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.
29

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.
30

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.

Page generated in 0.0207 seconds