2 resultados para Cell aggregation

em ArchiMeD - Elektronische Publikationen der Universität Mainz - Alemanha


Relevância:

60.00% 60.00%

Publicador:

Resumo:

Zelladhäsionsphänomene spielen eine wichtige Rolle in vielen biologischen Vorgängen, wie z. B. in der Embryogenese, der Wundheilung, der Immunantwort und Entzündungsprozessen. So wird die inflammatorische Kaskade durch P- und E-Selektin vermittelte Adhäsion der im Blutstrom zirkulierenden Leukozyten an Endothelzellen eingeleitet. Übermäßige Zelladhäsion kann hingegen eine Vielzahl von Krankheiten bewirken. Nachgewiesen ist eine solche Beteiligung der Selektine u. a. bei rheumatoider Arthritis, Erkrankungen der Herzkranzgefäße und dem Reperfusionssyndrom. Ein Ansatz zur Therapie besteht in der Verabreichung löslicher Selektinliganden, die kompetitiv an die Selektine binden und deren Aktivität dadurch mindern. Die wichtigste Leitstruktur für pharmakologisch aktive Selektinliganden stellt hierbei das Tetrasaccharid-Epitop Sialyl-Lewisx (sLex) dar.rnrnIn dieser Arbeit wurde nach einer nicht enzymatischen Strategie ein auf der sLex Struktur basierender Baustein für die Festphasen-Peptidsynthese synthetisiert. Aus diesem sollen nach Schutzgruppen-Manipulationen durch Einsatz in automatisierten Festphasen-Peptidsynthesen, verschiedene Glycopeptide als Zelladhäsionsinhibitoren für Selektine gewonnen werden. Der rasche Abbau der sLex-Struktur im Organismus schränkt die pharmakologische Nutzung von sLex-Derivaten jedoch stark ein. Das synthetisierte Kohlenhydrat Epitop weist daher gegenüber der natürlichen sLex-Struktur mehrere Modifikationen auf, die zu einer erhöhten metabolischen Stabilität führen sollen, ohne die für eine effektive Bindung zum Rezeptor nötigen Wechselwirkungen zu beeinträchtigen. rnrnAus diesem Grund wurde der synthetisch anspruchsvolle Sialinsäure-Baustein durch die L-Cyclohexylmilchsäure substituiert. Die Anbindung des Kohlenhydrat-Epitops an das Peptid wird im Gegensatz zur natürlichen N-glycosidischen Verknüpfung hier über eine zusätzliche Glucosamin-Einheit O-glycosidisch an die Aminosäure L-Threonin bewirkt. Außerdem wird der labile Fucose-Rest durch die D-Arabinose ersetzt, die nicht im menschlichen Organismus vorkommt, jedoch die drei essentiellen pharmakophoren Hydroxylfunktionen in der gleichen dreidimensionalen Orientierung präsentiert wie die L-Fucose.

Relevância:

60.00% 60.00%

Publicador:

Resumo:

The amyloid precursor protein (APP) is a type I transmembrane glycoprotein, which resembles a cell surface receptor, comprising a large ectodomain, a single spanning transmembrane part and a short C-terminal, cytoplasmic domain. It belongs to a conserved gene family, with over 17 members, including also the two mammalian APP homologues proteins APLP1 and APLP2 („amyloid precursor like proteins“). APP is encoded by 19 exons, of which exons 7, 8, and 15 can be alternatively spliced to produce three major protein isoforms APP770, APP751 and APP695, reflecting the number of amino acids. The neuronal APP695 is the only isoform that lacks a Kunitz Protease Inhibitor (KPI) domain in its extracellular portion whereas the two larger, peripheral APP isoforms, contain the 57-amino-acid KPI insert. rnRecently, research effort has suggested that APP metabolism and function is thought to be influenced by homodimerization and that the oligomerization state of APP could also play a role in the pathology of Alzheimer's disease (AD), by regulating its processing and amyloid beta production. Several independent studies have shown that APP can form homodimers within the cell, driven by motifs present in the extracellular domain, as well as in the juxtamembrane (JM) and transmembrane (TM) regions of the molecule, whereby the exact molecular mechanism and the origin of dimer formation remains elusive. Therefore, we focused in our study on the actual subcellular origin of APP homodimerization within the cell, an underlying mechanism, and a possible impact on dimerization properties of its homologue APLP1. Furthermore, we analyzed homodimerization of various APP isoforms, in particular APP695, APP751 and APP770, which differ in the presence of a Kunitz-type protease inhibitor domain (KPI) in the extracellular region. In order to assess the cellular origin of dimerization under different cellular conditions, we established a mammalian cell culture model-system in CHO-K1 (chinese hamster ovary) cells, stably overexpressing human APP, harboring dilysine based organelle sorting motifs at the very C-terminus [KKAA-Endoplasmic Reticulum (ER); KKFF-Golgi]. In this study we show that APP exists as disulfide-bound, SDS-stable dimers, when it was retained in the ER, unlike when it progressed further to the cis-Golgi, due to the KKFF ER exit determinant. These stable APP complexes were isolated from cells, and analyzed by SDS–polyacrylamide gel electrophoresis under non-reducing conditions, whereas strong denaturing and reducing conditions completely converted those dimers to monomers. Our findings suggested that APP homodimer formation starts early in the secretory pathway and that the unique oxidizing environment of the ER likely promotes intermolecular disulfide bond formation between APP molecules. We particularly visualized APP dimerization employing a variety of biochemical experiments and investigated the origin of its generation by using a Bimolecular Fluorescence Complementation (BiFC) approach with split GFP-APP chimeras. Moreover, using N-terminal deletion constructs, we demonstrate that intermolecular disulfide linkage between cysteine residues, exclusively located in the extracellular E1 domain, represents another mechanism of how an APP sub-fraction can dimerize within the cell. Additionally, mutational studies revealed that cysteines at positions 98 and 105, embedded in the conserved loop region within the E1 domain, are critical for interchain disulfide bond formation. Using a pharmacological treatment approach, we show that once generated in the oxidative environment of the ER, APP dimers remain stably associated during transport, reaching the plasma membrane. In addition, we demonstrate that APP isoforms, encompassing the KPI domain, exhibit a strongly reduced ability to form cis-directed dimers in the ER, whereas trans-directed cell aggregation of Drosophila Schneider (S2)-cells was isoform independent, mediating cell-cell contacts. Thus, suggesting that steric properties of KPI-APP might be the cause for weaker cis-interaction in the ER, compared to APP695. Finally, we provide evidence that APP/APLP1 heterointeractions are likewise initiated in the ER, suggesting a similar mechanism for heterodimerization. Therefore, dynamic alterations of APP between monomeric, homodimeric, and possibly heterodimeric status could at least partially explain some of the variety in the physiological functions of APP.rn