• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 212
  • 56
  • 26
  • 17
  • 16
  • 14
  • 13
  • 9
  • 6
  • 4
  • 4
  • 4
  • 4
  • 4
  • 4
  • Tagged with
  • 457
  • 457
  • 302
  • 214
  • 159
  • 81
  • 62
  • 55
  • 53
  • 51
  • 49
  • 46
  • 45
  • 44
  • 43
  • 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.
321

Functional Analysis of Adapter Protein c-Abl Src Homology 3 Domain-binding Protein 2

Chen, Grace Yi-Ying 23 September 2009 (has links)
3BP2 is a pleckstrin homology (PH) domain- and Src homology 2 (SH2) domain-containing adapter protein that has been linked through genetic evidence to a rare human disease called cherubism 146. 3BP2 was originally cloned in a screen to identify c-Abl SH3 binding proteins 23,24. In overexpression studies, 3BP2 has been implicated as a positive regulatory adapter molecule coupled to immunoreceptor on T cells 67,69,70, B cells 68, NK cells 71-73 and mast cells 74,75. It was also evident that 3BP2 forms complexes with a number of signaling molecules, such as Zap-70, LAT, phospholipase C-γ1 (PLC-γ1), Grb2, Cbl, and Fyn in Jurkat cells 67 and Vav1, Vav2, PLC-γ, and Syk in Daudi B cells 68. Despite the growing body of biochemical data to support the importance of 3BP2 in cells of the hematopoietic lineage, a clear picture of the biological function of 3BP2 has yet to emerge. To elucidate the in vivo function of 3BP2, our laboratory has generated 3BP2 gene-deficient mice through homologous recombination 452. The 3BP2-deficient (3BP2-/-) mice were born at the expected Mendelian frequency and were fertile and viable. 3BP2-/- mice accumulate splenic marginal-zone (MZ) B cells, possess a reduced frequency of peritoneal B-1 B cells, and have a diminished thymus-independent type 2 (TI-2) antigen response. 3BP2-/- B cells demonstrate diminished proliferation and cell survival following cross-linking of the B-cell receptor (BCR). Following BCR ligation, 3BP2 might be recruited to BCR complex through its inducible interaction with BCR costimulatory molecule CD19. In the absence of 3BP2, the activation of BCR downstream effectors such as MAPK Erk1/2, JNK, and c-Abl is normal; however, 3BP2 deficiency leads to defects in Syk phosphorylation and calcium flux. In addition to defects in peripheral B cell activities, 3BP2 deficiency contributes to defects in neutrophil activities. In response to the chemotactic peptide, fMLF, 3BP2-/- neutrophils fail to establish directional migration in vitro. There is a defect in the accumulation of filamentous actin at the leading edge of migrating 3BP2-/- neutrophils which might be responsible for the random movement of these cells under shallow gradient of fMLF. In vivo, there is a delay in the recruitment of circulating neutrophils to the site of chemically induced inflammation in 3BP2-/- mice. Compared to wildtype neutrophils, 3BP2-/- neutrophils fail to properly produce superoxide anion (O2-) following fMLF stimulation. Defects in both directional migration and superoxide production of 3BP2-/- neutrophils might contribute to the reduction in bacteria clearance and the increased mortality in 3BP2-/- mice post Listeria Monocytogenes infection. In Chapter 1 of this thesis, I have reviewed basic structures and functions of the domain modules found in adapter proteins. In addition, I have reviewed the findings from numerous reports on the function of 3BP2 in different cell types. A discussion of the physical appearance and some of the initial characterization of 3BP2-deficient mice (3BP2-/-) we have generated in our laboratory are included in Chapter 1. The second part of Chapter 1 consists of an introduction on B cell receptor signaling pathway and B-cell development and activation. A discussion of G protein-coupled receptor-mediated neutrophil functions can also be found in Chapter 1. Chapter 2 contains all the methods and materials used in my study. Chapter 3 includes the characterization of peripheral B cell compartment of 3BP2-/- mice as well as the role of 3BP2 downstream of B-cell antigen receptor and in T-independent immune response. In chapter 4, I present data from experiments designed to examine the role of 3BP2 downstream of a G protein-coupled receptor, fMLF receptor, of neutrophils. I also show the requirement of 3BP2 in the clearance of Listeria Monocytogenes. In chapter 5, I propose two models for 3BP2 action based on the findings in B cells and neutrophils and discuss future areas for investigation.
322

Modulation of N-methyl-D-aspartate receptors by Gαs- and Gαi/o-coupled receptors

Trepanier, Catherine Helene 07 January 2013 (has links)
The induction of synaptic plasticity at CA1 synapses requires NMDAR activation. Modulation of NMDAR function by various GPCRs can shift the thresholds for LTP and LTD induction and contribute to metaplasticity. Here we showed that the activity of GluN2A- and GluN2B-containing NMDARs is differentially regulated by Gαi/o-coupled, Gαq- and Gαs-coupled receptors. Furthermore, enhancing the relative function of GluN2A-to-GluNB NMDAR activity by GPCRs can alter the balance of LTP and LTD induction and contribute to metaplasticity. In CA1 neurons, activation of the Gαs-coupled D1/D5R selectively recruited Fyn kinase and enhanced GluN2B-mediated NMDAR currents. Biochemical experiments confirmed that D1/D5R stimulation activates Fyn kinase and enhances the tyrosine phosphorylation of GluN2B subunits. In contrast, activation of the Gαq-coupled PAC1R selectively recruited Src kinase to enhance the function of GluN2A-containing NMDARs. Enhancing the functional ratio of GluN2A-to-GluN2B subunits by PAC1R activation lowered the threshold for LTP induction whereas enhancing the functional ratio of GluN2B-to-GluN2A subunits by D1/D5R activation increased the threshold for LTP induction. Unexpectedly, activation of the Gαi/o-coupled mGluR2/3 enhanced NMDAR-mediated function via a previously unidentified mechanism. Inhibition of the cAMP-PKA pathway via mGluR2/3 activation resulted in activation of Src via decreased phosphorylation of its C-terminal Tyr527 by Csk. Stimulation of mGluR2/3 selectively potentiated the function of GluN2A-containing NMDARs but whether it shifted the modification threshold θm to the left requires further investigation.
323

Functional Analysis of Adapter Protein c-Abl Src Homology 3 Domain-binding Protein 2

Chen, Grace Yi-Ying 23 September 2009 (has links)
3BP2 is a pleckstrin homology (PH) domain- and Src homology 2 (SH2) domain-containing adapter protein that has been linked through genetic evidence to a rare human disease called cherubism 146. 3BP2 was originally cloned in a screen to identify c-Abl SH3 binding proteins 23,24. In overexpression studies, 3BP2 has been implicated as a positive regulatory adapter molecule coupled to immunoreceptor on T cells 67,69,70, B cells 68, NK cells 71-73 and mast cells 74,75. It was also evident that 3BP2 forms complexes with a number of signaling molecules, such as Zap-70, LAT, phospholipase C-γ1 (PLC-γ1), Grb2, Cbl, and Fyn in Jurkat cells 67 and Vav1, Vav2, PLC-γ, and Syk in Daudi B cells 68. Despite the growing body of biochemical data to support the importance of 3BP2 in cells of the hematopoietic lineage, a clear picture of the biological function of 3BP2 has yet to emerge. To elucidate the in vivo function of 3BP2, our laboratory has generated 3BP2 gene-deficient mice through homologous recombination 452. The 3BP2-deficient (3BP2-/-) mice were born at the expected Mendelian frequency and were fertile and viable. 3BP2-/- mice accumulate splenic marginal-zone (MZ) B cells, possess a reduced frequency of peritoneal B-1 B cells, and have a diminished thymus-independent type 2 (TI-2) antigen response. 3BP2-/- B cells demonstrate diminished proliferation and cell survival following cross-linking of the B-cell receptor (BCR). Following BCR ligation, 3BP2 might be recruited to BCR complex through its inducible interaction with BCR costimulatory molecule CD19. In the absence of 3BP2, the activation of BCR downstream effectors such as MAPK Erk1/2, JNK, and c-Abl is normal; however, 3BP2 deficiency leads to defects in Syk phosphorylation and calcium flux. In addition to defects in peripheral B cell activities, 3BP2 deficiency contributes to defects in neutrophil activities. In response to the chemotactic peptide, fMLF, 3BP2-/- neutrophils fail to establish directional migration in vitro. There is a defect in the accumulation of filamentous actin at the leading edge of migrating 3BP2-/- neutrophils which might be responsible for the random movement of these cells under shallow gradient of fMLF. In vivo, there is a delay in the recruitment of circulating neutrophils to the site of chemically induced inflammation in 3BP2-/- mice. Compared to wildtype neutrophils, 3BP2-/- neutrophils fail to properly produce superoxide anion (O2-) following fMLF stimulation. Defects in both directional migration and superoxide production of 3BP2-/- neutrophils might contribute to the reduction in bacteria clearance and the increased mortality in 3BP2-/- mice post Listeria Monocytogenes infection. In Chapter 1 of this thesis, I have reviewed basic structures and functions of the domain modules found in adapter proteins. In addition, I have reviewed the findings from numerous reports on the function of 3BP2 in different cell types. A discussion of the physical appearance and some of the initial characterization of 3BP2-deficient mice (3BP2-/-) we have generated in our laboratory are included in Chapter 1. The second part of Chapter 1 consists of an introduction on B cell receptor signaling pathway and B-cell development and activation. A discussion of G protein-coupled receptor-mediated neutrophil functions can also be found in Chapter 1. Chapter 2 contains all the methods and materials used in my study. Chapter 3 includes the characterization of peripheral B cell compartment of 3BP2-/- mice as well as the role of 3BP2 downstream of B-cell antigen receptor and in T-independent immune response. In chapter 4, I present data from experiments designed to examine the role of 3BP2 downstream of a G protein-coupled receptor, fMLF receptor, of neutrophils. I also show the requirement of 3BP2 in the clearance of Listeria Monocytogenes. In chapter 5, I propose two models for 3BP2 action based on the findings in B cells and neutrophils and discuss future areas for investigation.
324

Structural studies of Gαq signaling and regulation

Shankaranarayanan, Aruna 07 November 2012 (has links)
Gαq signaling is implicated in a number of physiological processes that include platelet activation, cardiovascular development and smooth muscle function. Historically, Gαq is known to function by activating its effector, phospholipase Cβ. Desensitization of Gαq signaling is mediated by G-protein coupled receptor kinases (GRK) such as GRK2 that phosphorylates the activated receptor and also sequesters activated Gαq and Gβγ subunits. Our crystal structure of Gαq-GRK2-Gβγ complex shows that Gαq forms effector-like interactions with the regulator of G-protein signaling (RGS) homology domain of GRK2 involving the classic effector-binding site of Gα subunits, raising the question if GRK2 can itself be a Gáq effector and initiate its own signaling cascade. In the structure, Gα and Gβγ subunits are completely dissociated from one another and the orientation of activated Gαq with respect to the predicted cell membrane is drastically different from its position in the inactive Gαβγ heterotrimer. Recent studies have identified a novel Gαq effector, p63RhoGEF that activates RhoA. Our crystal structure of the Gαq-p63RhoGEF-RhoA complex reveals that Gαq interacts with both the Dbl homology (DH) and pleckstrin homology (PH) domains of p63RhoGEF with its C-terminal helix and its effector-binding site, respectively. The structure predicts that Gαq relieves auto-inhibition of the catalytic DH domain by the PH domain. We show that Gαq activates p63RhoGEF-related family members, Trio and Kalirin, revealing several conduits by which RhoA is activated in response to Gq-coupled receptors. The Gαq effector-site interaction with p63RhoGEF/GRK2 does not overlap with the Gαq-binding site of RGS2/RGS4 that function as GTPase activating proteins (GAPs). This suggests that activated G proteins, effectors, RGS proteins, and activated receptors can form high-order complexes at the cell membrane. We confirmed the formation of RGS-Gαq-effector complexes and our results suggest that signaling pathways initiated by GRK2 and p63RhoGEF are regulated by RGS proteins via both allosteric and GAP mechanisms. Our structural studies of Gαq signaling provide insight into protein-protein interactions that induce profound physiological changes. Understanding such protein interfaces is a key step towards structure-based drug design that can be targeted to treat diseases concerned with impaired Gαq signaling. / text
325

Αλληλεπιδράσεις των επταελικοειδών υποδοχέων με διάφορες πρωτεΐνες. Χαρακτηρισμός νέων σηματοδοτικών μονοπατιών / Protein-protein interactions of the heptahelical receptors. Identification of new signaling pathways

Παπακωνσταντίνου, Μαρία-Παγώνα 07 April 2015 (has links)
Οι οπιοειδείς υποδοχείς (OR), μ, δ, κ και NOP, είναι μέλη των επταελικοειδών υποδοχέων που συζεύγνυνται με G πρωτεΐνες (7ΤΜ ή GPCR), οι οποίοι αποτελούν τη μεγαλύτερη υπεροικογένεια υποδοχέων και έναν από τους κύριους φαρμακολογικούς στόχους λόγω της υψηλής φυσιολογικής τους σημασίας. Οι OR ρυθμίζουν μια ποικιλία φυσιολογικών αποκρίσεων στο νευρικό σύστημα, με κυριότερη την αναλγησία. Τα οπιοειδή φάρμακα είναι τα πιο ισχυρά και αποτελεσματικά αναλγητικά έναντι στον οξύ πόνο, όμως η παρατεταμένη χρήση τους οδηγεί σε φαινόμενα ανοχής και εξάρτησης. Γι’ αυτό υπάρχει έντονο ενδιαφέρον στην αποσαφήνιση των μηχανισμών που εμπλέκονται στα φαινόμενα αυτά προκειμένου να σχεδιαστούν πιο αποτελεσματικά φάρμακα χωρίς τέτοιες παρενέργειες. Η σηματοδότηση των οπιοειδών υποδοχέων γίνεται κυρίως μέσω της ενεργοποίησης των Gi/o πρωτεϊνών που με τη σειρά τους ρυθμίζουν κατάλληλους τελεστές. Πέρα όμως από αυτούς τους κλασσικούς αλληλεπιδρώντες εταίρους οι OR έχουν την ικανότητα να αλληλεπιδρούν και με πολλές άλλες πρωτεΐνες κυρίως μέσω των περιοχών της τρίτης ενδοκυτταρικής τους θηλιάς (i3L) και του καρβοξυτελικού τους άκρου (CT) (Georgoussi et al., 2006- Georgoussi, 2008- Georgoussi et al., 2012). Οι αλληλεπιδράσεις αυτές επηρεάζουν όχι μόνο την σηματοδότηση των OR αλλά και την εν γένει εύρυθμη λειτουργία τους. Μια σημαντική πρωτεϊνική οικογένεια που ελέγχει τη μεταγωγή σήματος από τις G πρωτεΐνες βρέθηκε να είναι οι πρωτεΐνες Ρυθμιστές της κυτταρικής Σηματοδότησης μέσω G πρωτεϊνών ή RGS πρωτεΐνες (Regulators of G protein signaling, RGS). Ο πρωταρχικός τους ρόλος είναι η αλληλεπίδραση τους με τις Gα υπομονάδες των G πρωτεϊνών και η επιτάχυνση της υδρόλυσης του GTP από τις τελευταίες οδηγώντας στη μείωση της σηματοδότησης των GPCR. Μέλη της οικογένειας των RGS πρωτεϊνών είχε δειχθεί ότι πέρα από τις Gα πρωτεΐνες αλληλεπιδρούν επίσης με υποδοχείς GPCR, τελεστές αλλά και με άλλες ρυθμιστικές πρωτεΐνες, προσδίδοντας τους έναν ιδιαίτερο οργανωτικό ρόλο στη λειτουργία του κυττάρου και καθιστώντας τις RGS πρωτεΐνες μόρια υψηλού φαρμακολογικού ενδιαφέροντος. Παρελθόντα πειράματα in vitro συγκατακρήμνισης, του εργαστηρίου Κυτταρικής Σηματοδότησης και Μοριακής Φαρμακολογίας, με τη χρήση GST-χιμαιρικών πεπτιδίων των καρβοξυτελικών άκρων των μ-OR και δ-OR (μ-CT και δ-CT αντίστοιχα) και της τρίτης ενδοκυτταρικής θηλιάς του δ-OR (δ-i3L), έδειξαν ότι η RGS4, ένα μέλος της B/R4 υποοικογένειας, αλληλεπιδρά και με τους δυο υποδοχείς στις περιοχές αυτές (Georgoussi et al., 2006- Leontiadis et al., 2009). Η αλληλεπίδραση της RGS4 στα καρβοξυτελικά άκρα των υποδοχέων αυτών γίνεται στην περιοχή που σχηματίζει μια 8η αμφιπαθική α-έλικα (έλικα VIII), σημείο επαφής των OR και για άλλες πρωτεϊνικές αλληλεπιδράσεις όπως αυτή των STAT5A/B ((Mazarakou and Georgoussi, 2005- Georganta et al., 2010), της σπινοφιλίνης (Fourla et al., 2012) και άλλων πρωτεϊνών (Georgoussi et al., 2012). Βρέθηκε επίσης ότι η RGS4 είναι αρνητικός ρυθμιστής της κυτταρικής σηματοδότησης των μ-OR και δ-OR (Georgoussi et al., 2006- Leontiadis et al., 2009). Τέλος, αποδείχθηκε για πρώτη φορά ότι η RGS4 παίξει το ρόλο «μοριακού φίλτρου» καθοδηγώντας τους μ-OR και δ-OR να αλληλεπιδράσουν με συγκεκριμένο διαφορετικό υποπληθυσμό Gα υπομονάδων των G πρωτεϊνών (Leontiadis et al., 2009). Καμία πληροφορία για τον ρόλο των RGS πρωτεϊνών δεν υπάρχει για τον κ-OR. Για τον λόγο αυτό σκοπός της παρούσας διατριβής ήταν να ελέγξουμε αν οι RGS πρωτεΐνες της Β/R4 υποοικογένειας αλληλεπιδρούν με τον κ-OR και αν ναι, ποιος είναι ο ρόλος τους στη σηματοδότηση του κ-OR και των G πρωτεϊνών με τις οποίες ο τελευταίος συζεύγνυται. Τα αποτελέσματά μας έδειξαν ότι ο κ-OR μπορεί να αλληλεπιδράσει και με την RGS4 και με την RGS2 τόσο in vitro όσο και in vivo. Η δημιουργία GST-χιμαιρικών πεπτιδίων του καρβοξυτελικού άκρου του κ-OR (κ-CT) έδειξε ότι η RGS4 αλληλεπιδρά επίσης εντός της έλικας VIII ενώ η RGS2 αλληλεπιδρά με το τελικό μη συντηρημένο άκρο του κ-CT όσο και του δ-CT. Επιπλέον η συνέκφραση της RGS4 ή της RGS2 σε κύτταρα 293F που εκφράζουν τον κ-OR έδειξε ότι και οι δυο RGS πρωτεΐνες προάγουν την επιλεκτική και διαφορική σύζευξη του κ-OR με συγκεκριμένο υποπληθυσμό των Gαi/o υπομονάδων. Σε ότι αφορά τον φυσιολογικό ρόλο των RGS4 και RGS2 στις ελεγχόμενες από τον κ-OR κυτταρικές αποκρίσεις βρήκαμε ότι τόσο η RGS4 όσο και η RGS2 ανέστειλαν την καταστολή της αδενυλικής κυκλάσης που ελέγχει ο κ-OR, αλλά όχι ο δ-OR, με την RGS2 να έχει ισχυρότερη επίδραση στο μονοπάτι αυτό. Επίσης οι RGS4 και RGS2 μείωσαν την ενεργοποίηση των ERK1,2 κινασών που σηματοδοτούσε ο κ-OR. Τέλος, βρήκαμε ότι παρόλο που καμία από τις δυο RGS δεν επηρεάζει την εσωτερίκευση του κ-OR, η RGS4 επιταχύνει την εσωτερίκευση του δ-OR. Τα ευρήματά μας καταδεικνύουν ότι οι RGS4 και RGS2 πρωτεΐνες είναι δυο νέοι αρνητικοί ρυθμιστές στην σηματοδότηση των κ-OR και δ-OR. Εμφανίζουν διαφορικό ρυθμιστικό ρόλο στα σηματοδοτικά μονοπάτια καθενός OR, με ρόλο κλειδί στην καθοδήγηση της σύζευξής τους με τις Gα υπομονάδες και μπορούν να αποτελέσουν ενδιαφέροντες φαρμακολογικούς στόχους για τον έλεγχο της δράσης των οπιοειδών. / Οpioid receptors (OR) (subtypes μ, δ, κ and NOP) belong to the superfamily of the Heptahelical G protein-coupled receptors (7TM or GPCRs), the largest class of receptors in the human genome and common targets for therapeutics. ORs mediate their responses in the nervous system via coupling to members of the Gi/Go proteins to regulate the activity of various effector systems. Opioids are the most potent analgesics but prolonged administration leads to phenomena of tolerance and dependence thus there is a great interest towards understanding of OR signalling in an effort to develop new drugs devoid of adverse effects. Extended observations have demonstrated that the cytoplasmic face of the ORs is critical in mediating their signal through interactions not only with G proteins but also with multiple other proteins. These regulatory proteins play distinct roles in the regulation of the OR signalling, and in the fine tuning of these receptors. Regulators of G protein signalling (RGS) proteins is a class of proteins that modulate G protein signalling events by directly interacting with Gα subunits and accelerating the GTP hydrolysis, thus reducing GPCR signalling towards their effectors. RGS can also interact with many GPCRs, effectors and auxiliary proteins thus playing a key role in the cell functions, making them highly attractive as pharmacological targets (Abramow-Newerly et al., 2006). Our previous in vitro studies have shown that a member of the B/R4 subfamily of RGS proteins such as RGS4 interacts directly with μ-OR and δ-OR within a conserved region in their C-termini (μ-CT and δ-CT), forming a helix VIII, as well as within the δ-third intracellular loop (δ-i3L). RGS4 associates with μ-OR and δ-OR in living cells and forms selective complexes with Gαi/o proteins in a receptor dependent manner. Expression of RGS4 in HEK293 cells attenuated adenylyl cyclase inhibition mediated by μ-OR and agonist-mediated ERK1,2 phosphorylation for both receptors (Georgoussi et al., 2006- Leontiadis et al., 2009), suggesting for the first time that RGS4 is a negative modulator of μ-OR and δ-OR signalling. To deduce whether similar effects also occur for the κ-opioid receptor (κ-ΟR) and define the ability of other members of the B/R4 subfamily of RGS proteins, such as RGS2, to interact with OR we generated fusion peptides encompassing the C-terminus of κ-OR (κ-CT). Results from pull down experiments indicated that RGS2 interacts with the κ-CT, the δ-CT and the δ-i3L but fails to interact with the μ-CT. RGS4-N-terminal domain is responsible for OR interaction. Mapping the sites of RGS2 interaction indicated that RGS2 interacts with the non conserved portion of the C-termini of ORs exhibiting a different docking site as compared to that of RGS4. Co-precipitation studies in living cells indicated that RGS2 and RGS4 associate with κ-ΟR constitutively and upon receptor activation and confer selectivity for coupling with a specific subset of G proteins in an RGS protein dependent manner. Expression of both RGS2 and/or RGS4, in 293F cells attenuated agonist mediated-adenylyl cyclase inhibition for κ-ΟR, but not δ-OR, with RGS2 exhibiting a more robust effect. RGS4 and RGS2 reduced κ-ΟR-mediated ERK1,2 phosphorylation whereas, RGS4 accelerated agonist-induced internalization of the δ-OR but not of the κ-OR. Collectively, our observations demonstrate that RGS2 and RGS4 are novel interacting partners and negative modulators of κ-ΟR and δ-OR signalling. These two RGS proteins display a differential modulatory effect in each signalling pathway tested and play a key functional role by conferring selectivity for both κ-OR and δ-OR coupling with a specific subset of G proteins. Therefore they can be considered as attractive new pharmacological targets to manipulate opioid receptors signalling.
326

Uncovering the mechanisms of trans-arachidonic acids : function and implications for cerebral ischemia and beyond

Kooli, Amna. January 2008 (has links)
Cerebral ischemia is the principal cause of morbidity and mortality worldwide. In addition to neuronal loss associated with hypoxic-ischemic damage, cerebral ischemia is characterized by a neuromicrovascular injury. Nitrative stress and lipid peroxidation increase in hypoxic-ischemic damages and play an essential role in neuromicrovascular injury leading to cerebral ischemia. We hypothesized that newly described lipid peroxidation products, termed trans-arachidonic acids (TAA), could be implicated in the pathogenesis of hypoxia-ischemia by affecting the cerebral vasomotricity and microvascular integrity. / The effects of TAA on neuromicrovascular tone were tested ex vivo by monitoring the changes in vascular diameter of rat cerebral pial microvessels. Four isomers of TAA, namely 5 E-AA, 8E-AA, IIE-AA and 14 E-AA induced an endothelium-dependent vasorelaxation. Possible mechanisms involved in TAA-induced vasorelaxation were thoroughly investigated. Collectively, data enclosed revealed that TAA induce cerebral vasorelaxation through the interactive activation of BKCa channels with heme oxygenase-2. This interaction leads to generation of carbon monoxide which in turn activates soluble guanylate cyclase and triggers vasorelaxation. / Chronic effects of TAA on microvascular integrity were examined by generating a unilateral hypoxic-ischemic (HI) model of cerebral ischemia on newborn rat pups. Our HI model showed microvascular degeneration as early as 24h post-HI, preceded by an increase in cerebral TAA levels. HI-induced microvascular lesions were dependent on nitric oxide synthase activation and ensued TAA formation. Although the molecular mechanisms leading to TAA-induced microvascular degeneration were, in part uncovered for the retina, the primary site of action of TAA remains unknown. We demonstrated that TAA binds and activates GPR40 receptor, a newly described free fatty acid receptor. Importantly, GPR40 receptor knock-out prevents TAA-induced reduction in cerebral microvascular density and limits HI-induced brain infarct.
327

Étude des mécanismes moléculaires menant à la migration cellulaire associée à Rac1 et ARF6.

Cotton, Mathieu 12 1900 (has links)
Le facteur de l’ADP-ribosylation 6 (ARF6) et Rac1 sont des petites protéines liant le GTP qui régulent plusieurs voies de signalisation comprenant le trafic de vésicules, la modification des lipides membranaires et la réorganisation du cytosquelette d’actine. Cependant, les mécanismes moléculaires par lesquels ARF6 et Rac1 agissent de concert afin de contrôler ces différents processus cellulaires restent méconnus. Dans cette étude, nous montrons que, dans les cellules HEK293, ARF6 et Rac1 sont retrouvées en complexe suite à la stimulation du récepteur à l’angiotensine. Des expériences réalisées in vitro nous indiquent que ces deux GTPases interagissent ensemble directement, et que ARF6 s’associe préférentiellement avec la forme inactive de Rac1. L’inhibition de l’expression de ARF6 par interférence à l’ARN entraîne une activation marquée en cellule de Rac1 via le facteur PIX, indépendamment de la stimulation d’un récepteur, ce qui provoque la migration non contrôlée des cellules. Les arrestines, protéines de régulation de la désensibilisation des récepteurs couplés aux protéines G, servent de protéines d’échafaudage pour Rac1 et ARF6, en interagissant directement avec les GTPases et en augmentant leur association stimulée par l’angiotensine. De plus, les arrestines permettent l’activation, en s’en dissociant, de la MAP Kinase p38 qui régule l’activité de ARF6 et son interaction précoce avec les arrestines. Mis ensemble, ces résultats montrent que les arrestines contrôlent l’activité de ARF6, en influençant p38. ARF6 joue un rôle inhibiteur sur l’activation basale de Rac1 pour permettre ensuite son recrutement et son activation dépendante de l’angiotensine. Cette étude nous a permis de préciser le mode de régulation mis en jeu dans l’initiation de la migration cellulaire, suite à l’activation d’un récepteur couplé aux protéines G. Par le fait même, nous avons identifié certains des acteurs impliqués dans ce processus, offrant ainsi de nouvelles cibles pour le traitement des déséquilibres pathophysiologiques de la migration cellulaire. / The ADP-ribosylation factor 6 (ARF6) and Rac1 are small GTP-binding proteins that regulate several signaling events ranging from vesicle trafficking, to modification of membrane lipids and reorganization of the actin cytoskeleton. However, the molecular mechanisms by which ARF6 and Rac1 act in concert to control these different cellular processes remain unclear. Here, we show that in HEK 293 cells, ARF6 and Rac1 can be found in complex upon stimulation of the angiotensin receptor (ATR). In vitro experiments indicate that these two small G proteins can directly interact together, and that ARF6 preferentially interacts with the GDP-bound form of Rac1. Depletion of ARF6 by RNA interference leads to a marked PIX-dependent Rac1 activation in cells, independently of receptor stimulation, leading to uncontrolled cell migration. Arrestins, which are known for their role in G protein-coupled receptor desensitization, act as scaffold proteins toward Rac1 and ARF6, by directly interacting with the GTPases and by increasing their agonist-promoted association. Besides, arrestins allow p38 MAP Kinase activation, by releasing it, which regulates ARF6 activity and early association occurring between arrestins and ARF6. Taken together, this study shows that arrestins control ARF6 activity, by managing p38. ARF6 is an inhibitor of basal Rac1 activation to further allow the protein to be recruited and activated following angiotensin treatment. This study allowed us to precise how cell migration induction is regulated following G protein-coupled receptor activation. As a result, we identified some of the key players implicated in this process, providing new targets in the treatment of patho-physiological inbalance in cell migration.
328

Rôle du dimère Gbetagamma dans l’organisation des systèmes de signalisation cellulaire

Robitaille, Mélanie 11 1900 (has links)
Selon le modèle classique, le signal reçu par les récepteurs couplés aux protéines G (RCPG) se propage suite à des interactions transitoires et aléatoires entre les RCPGs, les protéines G et leurs effecteurs. Par les techniques de transfert d’énergie de résonance de bioluminescence (BRET), de complémentation bimoléculaire de protéines fluorescentes (BiFC) et de co-immunoprécipitation, nous avons observé que les récepteurs, les protéines G et les effecteurs forment un complexe stable, avant et après l’activation des récepteurs. L’interaction entre l’effecteur Kir3 et le dimère Gbetagamma se produit initialement au réticulum endoplasmique et est sensible à un agoniste liposoluble des récepteurs beta2-adrénergiques. Bien que peu de spécificité pour les nombreux isoformes des sous-unités Gbetagamma ait été observée pour l’activation du canal Kir3, les interactions précoces au RE sont plus sensibles aux différentes combinaisons de Gbetagamma présentes. En plus de son rôle dans la régulation des effecteurs, le dimère Gbetagamma peut interagir avec de nombreuses protéines possédant des localisations cellulaires autres que la membrane plasmique. Nous avons identifié une nouvelle classe de protéines interagissant avec la sous-unité Gbeta, autant en système de surexpression que dans des extraits de cerveaux de rats, soit les protéines FosB et cFos, qui forment le complexe de transcription AP-1, suite à leur dimérisation avec les protéines de la famille des Jun. La coexpression du dimère Gbetagamma réduit l’activité transcriptionnelle du complexe AP-1 induit par le phorbol 12-,myristate 13-acetate (PMA), sans toutefois interférer avec la formation du complexe Fos/Jun ou son interaction avec l’ADN. Toutefois, le dimère Gbetagamma colocalise au noyau avec le complexe AP-1 et recrute les protéines histones déacétylases (HDAC) afin d’inhiber l’activité transcriptionnelle du complexe AP-1. / Based on the classical model of G protein activation, signal transduction occurs by transient and random interactions between the receptor, the G protein and the effectors. Bioluminescence resonance energy transfer (BRET), bimolecular fluorescence complementation assay (BiFC) and co-immunoprecipitation experiments revealed that receptor, heterotrimeric G proteins and effectors were found in stable complexes that persisted during signal transduction. Kir3 channel and Gbetagamma dimer interacts first in the endoplasmic reticulum (ER) and this interaction can be modulated by the membrane-permeable beta2-adrenergic agonist cimaterol. Little specificity has been reported for several isoforms of the Gbetagamma dimer in the activation of the Kir3 channel. However, we found that the “precocious” interaction in the ER is sensitive to the presence of different combination of Gbeta and Ggamma subunits. Recently, a number of new proteins, which are not classical effectors at the plasma membrane have been shown to interact with GbetagammaThese include histone deacetylases 4 and 5 (HDAC)[1, 2] and the glucocorticoid receptor. We identified a novel interaction between Gbetagamma subunit and the Fos proteins, which form the transcription factor AP-1 following their dimerization with Jun proteins. Gbetagamma and Fos interactions can be detected in HEK 293 cells overexpressing the two proteins as well as in brains from rats pre-treated with amphetamine. Gbetagamma/Fos interaction favours the nuclear translocation of Gbetagamma dimer and inhibits AP-1 transcriptional activity. Gbetagamma did not block Fos/Jun dimerization or the interaction of AP-1 with DNA but recruited HDACs to the AP-1 complex.
329

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.
330

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.

Page generated in 0.0344 seconds