• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 44
  • 19
  • 7
  • 4
  • 4
  • 3
  • 2
  • 1
  • 1
  • Tagged with
  • 104
  • 104
  • 104
  • 104
  • 30
  • 26
  • 26
  • 25
  • 25
  • 21
  • 18
  • 15
  • 14
  • 14
  • 13
  • 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.
61

Modulation endogène des récepteurs métabotropiques du glutamate : bases moléculaires et implications fonctionnelles de la sensibilité au chlore extracellulaire / Endogenous modulation of metabotropic glutamate receptors : molecular basis and functional implications of extracellular chloride sensitivity

Tora, Amélie 20 October 2015 (has links)
Les récepteurs métabotropiques du glutamate (mGluRs) sont des récepteurs couplés aux protéines G (RCPGs) modulant la transmission synaptique au sein du système nerveux central. D'un point de vue structural, ils ont la particularité de posséder un large domaine extracellulaire, le Venus Flytrap (VFT), où se lie leur ligand endogène, le glutamate. Leur domaine transmembranaire à 7 hélices, commun à tous les RCPGs, est connu pour être la cible d'une nouvelle classe de molécules à visée thérapeutique, les modulateurs allostériques. Au contraire, le VFT est le siège du développement de ligands compétitifs du glutamate et peu de choses sont connues quant à l'existence de modulateurs allostériques du VFT. Des études récentes ont mis en évidence une sensibilité des mGluRs aux ions extracellulaires et en particulier au chlore (Cl-), sans que son site de liaison ne soit identifié. Dans ce contexte, ce travail de thèse explore la possibilité d'une modulation allostérique endogène des mGluRs par les ions Cl-, en identifiant leur(s) site(s) de liaison(s) et leur effet sur la dynamique conformationnelle et la fonction des récepteurs. En combinant une approche pharmacologique, biophysique basée sur la technique de FRET, et la modélisation, nous avons tout d'abord confirmé que le Cl- potentialise l'action du glutamate sur tous les mGluRs et qu'il favorise la conformation active des récepteurs en se liant au niveau du VFT. Les mGluRs présentent également une sensibilité différente au Cl-, mGlu4 étant le plus sensible et mGlu2 le moins. Ceci s'explique notamment par le nombre de sites fonctionnels, tous les mGluRs dont mGlu4 possédant 2 sites par monomère à l'exception de mGlu2 qui n'en possède qu'un, en raison d'une mutation « clé » d'une sérine en aspartate dans le lobe 1 du VFT. D'autre part, le récepteur mGlu3 est apparu comme un cas particulier ayant une sensibilité accrue au Cl-, son domaine VFT cumulant la présence et l'orientation adéquate d'acides aminés formant un « verrou » Cl-, qui favorise de manière drastique la conformation active et une activité basale élevée de ce récepteur. Enfin, la modélisation de la variation de la concentration extracellulaire en Cl- lors d'une activité synaptique GABAergique est compatible avec une modulation des mGluRs les plus sensibles. En conclusion, le Cl- est un modulateur allostérique endogène des mGluRs et l'exploitation de ses sites de liaison au sein du VFT pourrait permettre le développement de nouveaux agents thérapeutiques. / Metabotropic glutamate receptors (mGluRs) are G coupled-protein receptors (GPCRs) playing key roles in synaptic transmission in the central nervous system. They display a large extracellular domain, the Venus Flytrap (VFT) where the endogenous ligand, glutamate, binds. Their 7 transmembrane helices spanning domain, common to all GPCRs, is known to be the target of new therapeutic compounds, called allosteric modulators. In contrast, VFT domain is used to develop glutamate competitive ligands and there are only few data about allosteric modulators targeting the VFT. Recent studies have shown mGluRs are sensitive to extracellular ions, particularly to chloride (Cl-), although its binding site has not been elucidated. This thesis work explores the possibility of an endogenous allosteric modulation of mGluRs by Cl-, aiming to delineate its binding site(s) and its effect on receptor conformational dynamics and function. Using pharmacological, FRET based biophysical approaches and modelling, we have first confirmed that Cl- potentiates glutamate action in all mGluRs and that this ion favors agonist induced active conformation by binding to the VFT. mGluRs are also differently sensitive to Cl-, mGlu4 being the most and mGlu2 the least. This difference is notably explained by the number of Cl- functional sites within the VFT, all mGluRs including mGlu4 displaying 2 sites per monomer whereas mGlu2 has only 1 site due to a serine-aspartate “key” mutation in VFT lobe 1. Besides, mGlu3 receptor appears to be a “special case”, as this receptor is highly sensitive to Cl- because its VFT domain is carrying amino acids creating a “Cl- lock”, which dramatically favors active conformation and a high level of basal activity. Finally, modelling of extracellular Cl- concentration variations in a GABAergic synapse is compatible with a modulation of the most sensitive mGluRs. In conclusion, Cl- is an endogenous allosteric modulator of mGluRs and exploiting its binding sites may yield to the development of innovative therapeutic tools.
62

Engineering the angiotensin II type 1 receptor for structural studies

Thomas, Jennifer Ann January 2015 (has links)
G protein-coupled receptors (GPCRs) are eukaryotic integral membrane proteins that perform transmembrane signal transduction. Due to their pivotal role in a wide range of essential physiological functions GPCRs represent a high proportion of all drug targets. High resolution X-ray structures of GPCRs are however underrepresented in the Protein Data Bank. This is due to their instability in detergent, low expression levels and the presence of misfolded receptors in many heterologous expression systems. The objective of this project was to engineer the angiotensin II type 1 receptor (AT1R), a human GPCR, to make it suitable for structural studies. It was determined that detergentsolubilised AT1R was thermostable with antagonist bound with an apparent Tm of ~45°C, which was sufficiently stable for purification without further thermostabilisation by rational mutagenesis. Two expression systems were then evaluated for large-scale production of AT1R, namely baculovirus-mediated expression in insect cells and mammalian expression in HEK293 cells. Radioligand binding assays showed that only the mammalian system produced sufficient quantities of active AT1R for structural studies. Expression in the mammalian system was further optimised to approximately 6 mg/L. An AT1R-GFP fusion was created to examine membrane localisation using confocal laser scanning microscopy, to assay expression levels, to select highly expressing monoclonal cell lines using fluorescence activated flow cytometry and to develop a fluorescence size-exclusion chromatographybased assay to examine the suitability of 12 different ligands for co-crystallization. AT1R was also engineered to facilitate crystallisation, including C-terminal truncations to remove predicted disordered regions and bacteriophage T4-lysozyme being added to the third intracellular loop to provide additional points of contact for crystallisation, which increased the apparent Tm by approximately 10°C. All modified versions of AT1R were assessed for expression, stability and monodispersity. Additionally a rapid western blotting based assay was developed for the detection of unfolded membrane proteins, which will have wide applicability in the field.
63

Activation Of Glycoprotein Hormone Receptors : Role Of Different Receptor Domains In Hormone Binding And Signaling

Majumdar, Ritankar 04 1900 (has links) (PDF)
The glycoprotein hormones, Luteinizing Hormone (LH), human Chorionic Gonadotropin (hCG), Follicle Stimulating Hormone (FSH) and Thyroid Stimulating Hormone (TSH) are heterodimeric proteins with an identical α-subunit associated non-covalently with the hormone specific β-subunit and play important roles in reproduction and overall physiology of the organism [1]. The receptors of these hormones belong to the family of G-protein coupled receptors (GPCR) and have a large extracellular domain (ECD) comprising of 9-10 leucine rich repeats (LRR) followed by a flexible hinge region, a seven helical transmembrane domain (TMD) and a C terminal cytoplasmic tail [2]. Despite significant sequence and structural homologies observed between the ECDs of the receptors and the specific β-subunits of the hormones, the hormone-receptor pairs exhibit exquisite specificity with very low cross-reactivity with other members of the family. The TSH receptor (TSHR) is an especially interesting member of this family as it not only recognizes is cognate ligand, i.e. TSH, but also binds to the non-cognate ligands such as autoantibodies. TSHR autoantibodies come in different flavors; inhibitory antibodies that compete with the hormone for receptor binding and block its action, stimulatory antibodies that activate the receptor in a hormone independent manner and neutral antibodies that bind to the receptor but do not directly influence its functions. The inhibitory autoantibodies cause hypothyroidism and are responsible for Hashimoto’s Thyroiditis, whereas the stimulatory autoantibodies cause Graves’ thyrotoxicosis characterized by hyperthyroid condition [3]. The exact epitopes of these autoantibodies are not well delineated although it has been hypothesized that the blocking type- and the stimulatory type- autoantibodies have predominant epitopes in the TSHR ECD that overlap with hormone binding regions [4]. Insights into the mode of hormone or autoantibody binding to the receptor was primarily derived from the crystal structure of FSHR leucine rich repeat domain (LRRD) bound to single chain analog of FSH, and the crystal structures of TSHR LRRD bound to the stimulatory type human monoclonal antibody M22 [5] and the inhibitory type- monoclonal antibody K1-70 [6]. Both these crystal structures propose LRRDs as the primary ligand binding site which interacts with the hormone through its determinant loop in a hand-clasp fashion [7] while the autoantibodies mimics the hormone binding to a large extent [8] . These structures, while providing detailed understanding of the molecular interactions of the LRRs with the hormone, shed little light on the mechanism by which the signal generated at the LRRD are transduced to the downstream effector regions at the distally situated TMD. Hence, while one understands the ligand binding to a large extent, the activation process is not well understood, one of the central objective of the present study. Ligand-receptor interactions are typically studied by perturbing ligand/receptor structure by mutagenesis or by mapping conformational changes by biophysical or computational approaches. In addition to the above-mentioned approaches, the present work also uses highly specific antibodies against different domains of the receptor as molecular probes due to the ability of antibodies to distinguish between conformations likely to arise during the activation process. Use of antibodies to understand the receptor activation process is especially apt for TSHR due to the presence of physiologically relevant TSHR autoantibodies and their ability to influence hormone binding and receptor activation [9, 10]. Chapter 2 attempts to provide a comparison between the interactions of the hormone and the autoantibodies with TSHR. For this purpose, two assays were developed for identification of TSHR autoantibodies in the sera of patients suffering from autoimmune thyroid diseases (AITD), the first assay is based on the ability of TSHR autoantibodies to compete for radiolabeled hormone (The TSH binding inhibition (TBI), assay) and the second based on the capability of stimulatory antibody to produce cAMP in cells expressing TSHR (TSHR stimulatory immunoglobin (TSI) assay). A stable cell line expressing TSHR capable of recognizing both TSH and TSHR autoantibodies was thus created and used for prospective and retrospective analysis of AITD patients. Based on the TBI and TSI profiles of IgGs, purified from AITD patient's sera, it was recognized that TSHR stimulatory and TSH binding inhibitory effects of these antibodies correlated well, indicating overlap between hormone binding and IgG binding epitopes. It was also recognized that stimulatory IgGs are not affected by negative regulatory mechanism that governs TSH secretion substantiating the persistence of these antibodies in circulation. Kinetics of cAMP production by Graves’ stimulatory IgG was found to be fundamentally distinct, where the autoantibodies displayed pronounce hysteresis during the onset of the activation process when compared to the hormone. This could possibly be explained by the oligoclonality of the autoantibody population, a different mechanism of receptor activation or dissimilarity in autoantibody and hormone epitopes. To gain additional insights into the epitopes of TSHR autoantibodies and the regions that might be critical in the activation process, different overlapping fragments encompassing the entire TSH receptor ECD were cloned, expressed in E.coli as GST fusion proteins and purified: 1] the first three LRRs (TLRR 1-3, amino acid (aa) 21-127), 2] the first six LRRs (TLRR 1-6, aa 21-200), 3] the putative major hormone binding domain (TLRR 4-6, aa 128-200), and 4] the hinge region of TSH receptor along with LRR 7 to 9, (TLRR 7-HinR, aa 201-413). The receptor fragment TLRR 7-HinR was further subdivided into LRR 7-9 (TLRR 7-9, aa 201-161) and the hinge region (TSHR HinR, aa 261-413), expressed as N-terminal His-Tagged protein and purified using IMAC chromatography. Simultaneously, the full-length TSHR ECD was cloned, expressed and purified using the Pichia pastoris expression system. ELISA or immunoblot analysis of autoantibodies with the TSHR exodomain fragments suggested that Graves’ stimulatory antibody epitopes were distributed throughout the ECD with LRR 4-9 being the predominant site of binding. Interestingly, experiments involving neutralization of Graves’ IgG stimulated cAMP response by different receptor fragment indicated that fragments corresponding to the TSHR hinge region were better inhibitors of autoantibody stimulated receptor response than corresponding LRR fragments, suggesting that the hinge region might be an important component of the receptor activation process. This was in contrast to prevalent beliefs that considered the hinge region to be an inert linker connecting the LRRs to the TMD, a structural entity without any known functional significance. Mutagenesis in TSHR hinge region and agonistic antibodies against FSHR and LHR hinge regions, reported by the laboratory, recognized the importance of the hinge regions as critical for receptor activation and may not simply be a scaffold [11-13]. Unfortunately, the mechanism by which the hinge region regulates binding or response or both have not been well understood partially due to unavailability of structural information about this region. In addition poor sequence similarity within the GpHR family and within proteins of known structure, make this region difficult to model structurally. In chapter 3, effort is made to model the hinge regions of the three GpHR based on the knowledge driven and Ab initio protocols. An assembled structure comprising of the LRR domain (derived from the known structures of FSHR and TSHR LRR domains) and the modeled hinge region and transmembrane domain presents interesting differences between the three receptors, especially in the manner the hormone bound LRRD is oriented towards the TMD. These models also suggested that the α-subunit interactions in these three receptors are fundamentally different and this was verified by investigating the effects of two α-subunit specific MAbs C10/2A6 on hCG-LHR and hTSH-TSHR interactions. These two α-subunit MAbs had inverse effects on binding of hormone to the receptor. MAb C10 inhibited TSH binding to TSHR but not that of hCG, whereas MAb 2A6 inhibited binding of hCG to LHR but not of hTSH. Investigation into the accessibility of their epitopes in a preformed hormone receptor complex indicated that the α-subunit may become buried or undergo conformational change during the activation process and interaction may be different for LHR and TSHR. Fundamental differences in TSHR and LHR were further investigated in the next chapter (Chapter 4), especially with regards to the ligand independent receptor activation. Polyclonal antibodies were developed against LRR 1-6, TLRR 7-HinR and the TSHR HinR receptor fragments. The LRR 1-6 antibodies were potent inhibitor of receptor binding as well as response, similar to that observed with antibodies against the corresponding regions of LHR. Interestingly, the antibodies against the hinge region of TSHR were unable to inhibit hTSH binding, but were effective inhibitors of cAMP production suggesting that this region may be involved in a later stage of a multi-step activation process. This was also verified by studying the mechanism of inhibition of receptor response and their effect on ligand-receptor association and dissociation kinetics. Hinge region-specific antibodies immunopurified from TLRR 7-HinR antibodies behaved akin to those of the pure hinge region antibodies providing independent validation of the above results. This result was, however, in contrast to those observed with a similar antibody against LHR hinge region. As compared to the TSHR antibody, the LHR antibody inhibited both hormone binding and response. In addition, this antibody could dissociate a preformed hormone-receptor complex which was not observed for TSHR hinge region antibodies. Although unable to dissociate preformed hormone-receptor complex by itself, the TSHR HinR antibodies augmented hormone induced dissociation of the hormone-receptor complex suggesting that this region may be involved in modulation of negative cooperativity associated with TSHR. Molecular dissection of the role of hinge region of TSHR was further carried out by using monoclonal antibodies against LRR 1-3 (MAb 413.1.F7), LRR 7-9 (MAb 311.87), TSHR hinge region (MAb 311.62 and MAb PD1.37). MAb 311.62 which identifies the LRR/Cb-2 junction (aa 265-275), increased the affinity of TSHR for the hormone while concomitantly decreasing its efficacy, whereas MAb 311.87 recognizing LRR 7-9 (aa 201-259) acted as a non-competitive inhibitor of TSH binding. MAb 413.1.F7 did not affect hormone binding or response and was used as the control antibody for different experiments. Binding of MAbs was sensitive to the conformational changes caused by the activating and inactivating mutations and exhibited differential effects on hormone binding and response of these mutants. By studying the effects of these MAbs on truncation and chimeric mutants of thyroid stimulating hormone receptor (TSHR), this study confirms the tethered inverse agonistic role played by the hinge region and maps the interactions between TSHR hinge region [14] and exoloops responsible for maintenance of the receptor in its basal state. Mechanistic studies on the antibody-receptor interactions suggest that MAb 311.87 is an allosteric insurmountable antagonist and inhibits initiation of the hormone induced conformational changes in the hinge region, whereas MAb 311.62 acts as a partial agonist that recognizes a conformational epitope critical for coupling of hormone binding to receptor activation. Estimation of apparent affinities of the antibody to the receptor and the cooperativity factor suggests that epitope of MAb 311.87 (LRR 7-9) may act as a pivot involved in the initial events immediate to hormone binding at the LRRs. The anatgonsitic effect of MAB 311.62 on binding and response also suggested that binding of hormone is conformationally selective rather than an induced event. The hinge region, probably in close proximity with the α-subunit in the hormone-receptor complex, acts as a tunable switch between hormone binding and receptor activation. In contrast to the stimulatory nature of Cb-2 antibody such as MAb 311.62, MAb PD1.37, which identified residues aa 366–384 near Cb-3, was found to be inverse agonistic. Unlike other known inverse agonistic MAbs such as CS-17 [15] and 5C9 [16], MAb PD1.37 did not compete for TSH binding to TSHR, although it could inhibit hormone stimulated response. Moreover, unlike CS-17, MAb PD1.37 was able to decrease elevated basal cAMP of hinge region constitutively activated mutations only but not those in the extracellular loops. This is particularly important as interaction of hinge region residues with those of ECLs had been thought to be critical in maintenance of the basal level of receptor activation and are responsible for attenuating the constitutive basal activity of the mutant and wild-type receptors in the absence of the hormone. This was demonstrated by a marked increase in the basal constitutive activity of the receptor upon the complete removal of its extracellular domain, which returned to the wild-type levels upon reintroduction of the hinge region. However, careful comparison of the activities of the mutants (receptors harboring deletions and gain-of-function mutations) with maximally stimulated wild-type TSHR indicated that these mutations of the receptor resulted primarily in partial activation of the serpentine domain suggesting that only the ECD in complex with the hormone is the full agonist of the receptor. Confirmation of the above proposition has been difficult to verify primarily due to a highly transient conformational change in the tripartite interaction of the hinge region/hormone and the ECLs. The current approaches of using antibodies to probe the ECLs are difficult due to the conformational nature of the antigen as well as difficulty in obtaining a soluble protein. In chapter 5, the ligand induced conformational alterations in the hinge regions and inter-helical loops of LHR/FSHR/TSHR were mapped using the exoloop specific antibodies generated against a mini-Transmembrane domain (mini-TMD) protein. This mini-TMD protein, designed to mimic the native exoloop conformations, was created by joining the TSHR exoloops, constrained through the helical tethers and library derived linkers. The antibody against mini-TMD specifically recognized all three GpHRs and inhibited the basal and hormone stimulated cAMP production without affecting hormone binding. Interestingly, binding of the antibody to all three receptors was abolished by prior incubation of the receptors with the respective hormones suggesting that the exoloops are buried in the hormone-receptor complexes. The antibody also suppressed the high basal activities of gain-of-function mutations in the hinge regions, exoloops and TMDs such as those involved precocious puberty and thyroid toxic adenomas. Using the antibody and point/deletion/chimeric receptor mutants, dynamic changes in hinge region-exoloop interactions were mapped. The computational analysis suggests that mini-TMD antibodies act by conformationally locking the transmembrane helices by restraining the exoloops and juxta-membrane regions. This computational approach of generating synthetic TMDs bears promise in development of interesting antibodies with therapeutic potential, as well as, explains the role of exoloops during receptor activation. In conclusion (Chapter 6), the study provides a comprehensive outlook on the highly dynamic interaction of ligand and different subdomains of the TSHR (and to a certain extent of LHR and FSHR) and proposes a model of receptor activation where the receptor is in a dynamic equilibrium between the low affinities constrained state and the high affinity unconstrained state and bind to the hormone through the LRR 4-6. Upon binding the βL2 loop of the hormone contact LRR 8-10 that triggers a conformational change in the hinge region driving the α-subunit to contact the ECLs. Upon contact, the ECLs cooperatively causes helix movement in the TMH and ultimately in ICLs causing the inbuilt GTP-exchange function of a GPCR.
64

The role of human cytomegalovirus encoded viral G protein-coupled receptors in onco-modulatory signalling

Subramoney, Preya 22 June 2011 (has links)
Human cytomegalovirus (HCMV) is a ubiquitous virus of the herpes type that infects a high percentage of some populations. One of the most researched genes expressed by HCMV with close homology to human chemokine receptors is the US28 G protein-coupled receptor. Study design: This study was initiated to elucidate the intracellular signalling pathways of an inflammatory factor (IL-6) and an angiogenic factor (STAT3) triggered by the viral US28 oncogene and the presence of US28 in the HCMV viral particle. These pathways were observed by introducing the US28 gene into two human cell lines by infection with a HCMV strain that expresses the US28 gene (wild type), and two HCMV strains where the US28 gene was deleted (ÄUS28 and ÄUS28/UL33). Special attention was directed at the expression of IL-6 after promotion of the US28 gene and subsequent phosphorolation of STAT3. A new US28 antibody was validated and a method developed in an attempt to determine US28 on the viral particle. The following techniques were applied: Cell culture work, two mammalian cell lines were used, HFF’s and U373 MG. Virus stock titre determination to determine the multiplicity of infection. Protein quantitation to determine very small quantities of protein for Western blot analysis. ELISA for the quantitative determination of IL-6. Western blotting for phospho- STAT3 determination and validation of the US28 antibody. Immunocytochemistry was used for back titrations of virally infected cells. Immunofluorescence assay and use of confocal microscopic techniques was used for the location of the US28 gene in the virion and for tSTAT3 translocation to the nucleus. Conclusion: A clear increase in IL-6 secretion (495% ± 1%) was seen, and this was after only an hour in HCMV WT infected cells. From the increase in IL-6 secretion a subsequent increase in STAT3 phosphorylation was detected in the same samples. A clear link has been established between IL-6 and STAT3. A method to determine whether US28 was present in the HCMV viral particle was designed and preliminary results obtained. The results were inclusive. / Dissertation (MSc)--University of Pretoria, 2011. / Pharmacology / unrestricted
65

Régulation de l'activité autophagique par les récepteurs chimiotactiques couplés aux protéines G : rôle essentiel dans la migration directionnelle / Inhibition of autophagic activity by chemotactic receptors coupled to G proteins : essential role in cell migration

Coly, Pierre-Michaël 02 February 2017 (has links)
L’autophagie est un processus catabolique par lequel certaines protéines cytosoliquessont dirigées vers le compartiment lysosomial, afin d’y être dégradées. Ce processus débutepar la séquestration de constituants cytoplasmiques par une structure multimembranaireappelée phagophore. La fermeture du phagophore donne naissance à une vésicule à doublemembrane nommée autophagosome, qui fusionne avec les lysosomes, ce qui conduit à ladégradation du contenu de sa lumière. Ainsi, la modulation de l’autophagie permet unremodelage dynamique du protéome cellulaire. Bien que des données récentes ont permis dedémontrer la dégradation autophagique de protéines impliquées dans la migration cellulaire,telles que des intégrines, ou encore les protéines RhoA et Src, l'impact fonctionnel del'autophagie sur la migration cellulaire demeure sujet à controverse. Alors que l'autophagie estdécrite comme un processus pro-migratoire et pro-invasif dans certaines études, d'autrestravaux indiquent que l'inactivation des protéines pro-autophagiques stimule l'invasion descellules cancéreuses. De plus, l'effet fonctionnel des RCPG chimiotactiques sur l’activitéautophagique reste totalement inexploré. Sur la base de ces données, les objectifs de mon travail de thèse ont été i) d’évaluer les effets des RCPG chimiotactiques, le CXCR4 et l’UT,sur le processus autophagique et ii) d’étudier l’impact de cette modulation sur la migrationcellulaire. Pour ce faire, nous avons utilisé des cellules HEK-293, transfectées à l’aide deconstruits permettant l’expression des RCPG CXCR4 et UT, ainsi que la lignée deglioblastome humain U87, exprimant ces deux récepteurs de manière endogène.Nous avons dans un premier temps évalué l’activité autophagique à l’aide de laprotéine de fusion EGFP-LC3, marqueur des autophagosomes. Nous avons ainsi démontréque l’activation du CXCR4 et de l’UT provoque une diminution significative de la biogénèsedes autophagosomes. Une étape essentielle de cette biogenèse est le recrutement des protéinesAtg16L1 et Atg5 à la membrane plasmique, conduisant à la formation d'endosomes Atg16L1-Atg5-positifs, appelés « endosomes pré-autophagiques ». Cette population d’endosomesconstitue une source importante de phospholipides nécessaire à l’expansion du phagophore etla formation d’un autophagosome mature. Afin d’évaluer l’impact des RCPG chimiotactiquessur le recrutement de la protéine Atg16L1 à la membrane plasmique, nous avons bloqué leprocessus d’endocytose par l’utilisation d’un inhibiteur de la dynamine, le Dynasore. Cettemolécule provoque une accumulation marquée de la protéine Atg16L1 dans les endosomespré-autophagiques en formation, retenus à la membrane plasmique. / Autophagy is a catabolic process by which certain cytosolic proteins are directed to thelysosomal compartment to be degraded. This process begins with the sequestration ofcytoplasmic components, by a multimembrane structure called the phagophore. The closure ofthe phagophore gives rise to a double membrane vesicle called autophagosome, which thenmerges with lysosomes in order to degrade its luminal content. Autophagy modulation allowsa dynamic remodeling of the cellular proteome. Although recent evidence has demonstratedautophagic degradation of key proteins involved in cell migration, such as integrins, RhoAand the Src kinase, the functional impact of autophagy on cell migration remainscontroversial. While autophagy is described as a pro-migratory and pro-invasive process insome studies, others indicate that the inactivation of pro-autophagic proteins stimulates thecancer cell invasion. In addition, the functional effect of chemotactic GPCR on autophagicactivity remains unexplored. On the basis of these data, the objectives of my thesis were i) toevaluate the effects of the chemotactic GPCRs for SDF-1 (CXCR4) and for the vasoactivepeptide urotensin II (UT), on the autophagic process and ii) to study the impact of thismodulation on cell migration. In order to do this, we used HEK-293 cells, transfected with constructs allowing the expression of CXCR4 and UT, as well as the human glioblastomaline, U87, which endogenously expresses these two receptors. Previous studies have demonstrated a direct interaction of Atg5 with membranes,suggesting that recruitment of Atg16L1 to the plasma membrane may depend on Atg5. This prompted us to evaluate the formation of Atg16L1-positive pre-autophagic endosomes,following depletion of Atg5 levels. Several interfering RNAs, targeting the transcriptencoding Atg5, have been tested and, as expected, these interfering RNAs completely blockedthe recruitment of Atg16L1 to forming pre-autophagic endosomes. We then tested the effectsof chemotactic GPCRs on the subcellular localization of the Atg5 protein. By confocalmicroscopy, we found that a significant fraction of Atg5 localized to the plasma membraneunder basal conditions. The activation of CXCR4 or UT is accompanied by a marked decreaseof the Atg5 pool localized at the plasma membrane. Furthermore, we have demonstrated thatthe anti-autophagic effects of chemotactic GPCRs are completely abrogated byoverexpression of a recombinant Atg5 protein, suggesting that chemotactic GPCRs exert theiranti-autophagic effects by reducing the membrane pool of Atg5, necessary for the productionof pre-autophagic endosomes, and the expansion of the phagophore.
66

Vliv chronického působení morfínu na funkci signálních systémů řízených trimérními G-proteiny v srdci potkana / Effect of chronic morphine treatment of rats on myocardial signaling systems regulated by trimeric G-proteins

Škrabalová, Jitka January 2011 (has links)
It has recently been discovered that the effect of morphine can significantly reduce the tissue damage that occurs during myocardial ischemia. The molecular mechanisms by which morphine acts on the heart are still little understood. The aim of this thesis was to monitor the effect of chronic 27-day and 10-day administration of low (1 mg/kg/day) and high (10 mg/kg/ day) doses of morphine on the expression of selected G-protein-coupled receptors (GPCR) and on the expression and activity of adenylyl cyclase (AC). Chronic (27 days) morphine treatment reduced the expression of к-opioids receptors, but 10-day morphine exposure did not influence the expression of these receptors. Assessment of β1- and β2-AR by immunoblot technique did not show any significant change in the expression, but the more accurate determination of β-AR expression using the saturation binding studies revealed that 27-day treatment with high doses of morphine appreciable increased the total number of these receptors. Administration of high doses of morphine led to marked up-regulation of adenylyl cyclase (AC) isoforms V/VI, and the amount of AC decreased proportionally with the time of discontinuation of morphine administration. Low doses of morphine up- regulated AC only during 27-day administration. Chronic morphine exposure did...
67

Discovering Natural Product Chemistries for Vector Control

Lide Bi (15347593) 25 April 2023 (has links)
<p>  </p> <p>Vector-borne diseases (VBDs) represent a significant health burden worldwide, threatening approximately 80% of the global population. Insecticide-based vector control is the most effective method to manage many VBDs, but its efficacy has been declining due to high levels of resistance in vector populations to the main insecticide classes which operate via limited modes of action. Therefore, the discovery of new chemistries from non-conventional chemical classes and with novel modes of action is a priority for the control of vectors and VBDs. Natural products (NPs) are diverse in chemical structures and, potentially, modes of action. They have been used as insecticides for many decades and have inspired the development of multiple synthetic insecticides, suggesting the discovery of novel NPs could lead to the development of highly effective insecticides. </p> <p><br></p> <p>In this thesis, I report two studies with a main goal to identify novel mosquito-active insecticide leads that operate via modes of action distinct from existing insecticides. First, I tested the hypothesis that new mosquito-active insecticide leads with novel chemical structures, possibly operating via novel modes of action, can be identified by high-content larval phenotypic screening against a natural product collection and using novel phenotypic endpoints in addition to mortality endpoints. Here, I performed a high-content larval phenotypic screen using first instar (L1) larvae of <em>Aedes aegypti</em> (Linnaeus, 1762) against 3,680 compounds from the AnalytiCon MEGx Natural Product Libraries and a screening platform developed by Murgia et al., (2022). Compounds were screened in a 384-well plate format using the Perkin Elmer Opera Phenix and larvae were scored for lethal and novel phenotypic endpoints. Screening revealed five chemistries that caused larval mortality, including rotenone and a new NP chemistry, NP-4. The identification of rotenone confirmed the ability of the screen to detect mosquito-active NP chemistries. NP-4 caused high levels of larval mortality in the screen, and toxicity was confirmed in a subsequent concentration-response assay against third instar (L3) larvae of <em>Ae. aegypti</em>. 140 chemistries that caused atypical larval phenotypes, including cuticular pigmentation and morphometric changes relative to negative controls, were also identified by the screen. Some of these chemistries may operate by disruption of pathways regulating melanization, growth and development, and novel targets in the insect nervous systems, thus representing potential leads for further insecticide toxicity and mode of action studies. To facilitate quantitative analyses of atypical phenotypes, an attempt was made to assess the morphometrics of the thorax in larvae exposed to test chemistry, relative to control larvae. However, assessment was limited by the number of larvae images of suitable quality for measurements. </p> <p><br></p> <p>In the second study, I tested the hypothesis that metergoline (Murgia et al., 2022) and NP-4 (this study), two chemistries identified by the HTP phenotypic screen described in this project, operate via disruption of targets in the insect nervous systems that are distinct from the current insecticidal modes of products used in mosquito control programs. Specifically, I explored the hypothesis that metergoline operates via one or more insect orthologs of the mammalian G protein-coupled serotonin and dopamine receptors. An electrophysiology study was performed using the suction electrode technique and ganglia of the German cockroach, <em>Blattella germanica </em>(Linnaeus, 1767). To facilitate the investigation of metergoline agonism/antagonism and disruption of invertebrate GPCR signaling, 5-hydroxytryptamine (5-HT; serotonin) was included as a chemical probe. Electrophysiological recordings showed 5-HT (10µM and 1mM) and metergoline (10µM) caused no significant neurological activity at the tested concentrations in comparison to the saline control. However, a consistent neuro-inhibitory trend was observed, suggesting possible agonism of a 5-HT1-like receptor ortholog and antagonism of a putative 5-HT7-like receptor ortholog in the cockroach, respectively.  NP-4 caused significant neuro-inhibition at the tested concentration of 20µM, in comparison to the negative saline control. Given the demonstration of rapid contact toxicity to <em>Ae. aegypti</em> larvae and neurological inhibition in <em>B. germanica</em>, we propose NP-4 may act at one or more conserved targets in the insect nervous systems, which remain to be elucidated. </p> <p><br></p> <p>The significance of the present study is three-fold. First, this study reports the first high-content phenotypic screen of mosquito larvae against a NP collection and identification of 145 mosquito-active chemistries associated with lethal and phenotypic endpoints. These data confirm that the screening platform provided an innovative and effective system to rapidly identify mosquito-active small molecules with potential novel modes of action. Second, metergoline and NP-4 represent potential novel chemical leads for the development of new insecticides that can be incorporated into vector control programs targeting insecticide-resistant populations. Lastly, the study describes the first electrophysiology study of 5-HT, metergoline, and NP-4 via the suction electrode technique in an insect system and contributes new knowledge to the study of the insect serotonergic system, which represents an expanding area of vector biology research given its roles in feeding regulation.  </p> <p><br></p> <p>Future studies resulting from this thesis might include: (1) development of a set of morphometric criteria for quantitative analyses of atypical larval phenotypes, (2) incorporation of new phenotypic endpoints to expand the capacity of the screen to identify novel mode of action chemistries for insecticide discovery, and (3) identification of chemistry candidates suitable for further development from the 140 chemistries associated with atypical larval phenotypes in the primary screen using chemo-informatic and toxicological studies. In addition, studies using reverse transcription-polymerase chain reaction (RT-PCR), cell-based expression systems, mutant/insecticide resistant strains, and patch clamp electrophysiology could be pursued to further investigate the molecular mode of action of metergoline and NP-4, and potential for vector control.</p>
68

Differential regulation of GABAB receptor trafficking by different modes of N-methyl-D-aspartate (NMDA) receptor signaling

Kantamneni, Sriharsha, Gonzàlez-Gonzàlez, I.M., Luo, J., Cimarosti, H., Jacobs, S.C., Jaafari, N., Henley, J.M. 24 December 2013 (has links)
yes / Inhibitory GABAB receptors (GABABRs) can down-regulate most excitatory synapses in the CNS by reducing postsynaptic excitability. Functional GABABRs are heterodimers of GABAB1 and GABAB2 subunits and here we show that the trafficking and surface expression of GABABRs is differentially regulated by synaptic or pathophysiological activation of NMDA receptors (NMDARs). Activation of synaptic NMDARs using a chemLTP protocol increases GABABR recycling and surface expression. In contrast, excitotoxic global activation of synaptic and extrasynaptic NMDARs by bath application of NMDA causes the loss of surface GABABRs. Intriguingly, exposing neurons to extreme metabolic stress using oxygen/glucose deprivation (OGD) increases GABAB1 but decreases GABAB2 surface expression. The increase in surface GABAB1 involves enhanced recycling and is blocked by the NMDAR antagonist AP5. The decrease in surface GABAB2 is also blocked by AP5 and by inhibiting degradation pathways. These results indicate that NMDAR activity is critical in GABABR trafficking and function and that the individual subunits can be separately controlled to regulate neuronal responsiveness and survival. / BBSRC, MRC and the European Research Council
69

Molecular Effects of Auto-Antibodies on Angiotensin II Type 1 Receptor Signaling and Cell Proliferation

Philippe, Aurelie, Kleinau, Gunnar, Gruner, Jason Jannis, Wu, Sumin, Postpieszala, Daniel, Speck, David, Heidecke, Harald, Dowell, Simon J., Riemekasten, Gabriela, Hildebrand, Peter W., Kamhieh-Milz, Julian, Catar, Rusan, Szczepek, Michal, Dragun, Duska, Scheerer, Patrick 17 January 2024 (has links)
The angiotensin II (Ang II) type 1 receptor (AT1R) is involved in the regulation of blood pressure (through vasoconstriction) and water and ion homeostasis (mediated by interaction with the endogenous agonist). AT1R can also be activated by auto-antibodies (AT1R-Abs), which are associated with manifold diseases, such as obliterative vasculopathy, preeclampsia and systemic sclerosis. Knowledge of the molecular mechanisms related to AT1R-Abs binding and associated signaling cascade (dys-)regulation remains fragmentary. The goal of this study was, therefore, to investigate details of the effects of AT1R-Abs on G-protein signaling and subsequent cell proliferation, as well as the putative contribution of the three extracellular receptor loops (ELs) to Abs-AT1R signaling. AT1R-Abs induced nuclear factor of activated T-cells (NFAT) signaling, which reflects Gq/11 and Gi activation. The impact on cell proliferation was tested in different cell systems, as well as activation-triggered receptor internalization. Blockwise alanine substitutions were designed to potentially investigate the role of ELs in AT1R-Abs-mediated effects. First, we demonstrate that Ang II-mediated internalization of AT1R is impeded by binding of AT1R-Abs. Secondly, exclusive AT1RAbs- induced Gq/11 activation is most significant for NFAT stimulation and mediates cell proliferation. Interestingly, our studies also reveal that ligand-independent, baseline AT1R activation of Gi signaling has, in turn, a negative effect on cell proliferation. Indeed, inhibition of Gi basal activity potentiates proliferation triggered by AT1R-Abs. Finally, although AT1R containing EL1 and EL3 blockwise alanine mutations were not expressed on the human embryonic kidney293T (HEK293T) cell surface, we at least confirmed that parts of EL2 are involved in interactions between AT1R and Abs. This current study thus provides extended insights into the molecular action of AT1R-Abs and associated mechanisms of interrelated pathogenesis.
70

cAMP BIOSENSORS AND SPATIOTEMPORAL cAMP SIGNALING IN ADULT CARDIAC MYOCYTES

Warrier, Sunita 06 April 2007 (has links)
No description available.

Page generated in 0.2071 seconds