5 resultados para Gene by environment

em ArchiMeD - Elektronische Publikationen der Universität Mainz - Alemanha


Relevância:

90.00% 90.00%

Publicador:

Resumo:

Cytochrome P450 1A1 (CYP1A1) monooxygenase plays an important role in the metabolism of environmental pollutants such as polycyclic aromatic hydrocarbons (PAHs) and halogenated polycyclic aromatic hydrocarbons (HAHs). Oxidation of these compounds converts them to the metabolites that subsequently can be conjugated to hydrophilic endogenous entities e.g. glutathione. Derivates generated in this way are water soluble and can be excreted in bile or urine, which is a defense mechanism. Besides detoxification, metabolism by CYP1A1 may lead to deleterious effects since the highly reactive intermediate metabolites are able to react with DNA and thus cause mutagenic effects, as it is in the case of benzo(a) pyrene (B[a]P). CYP1A1 is normally not expressed or expressed at a very low level in the cells but it is inducible by many PAHs and HAHs e.g. by B[a]P or 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Transcriptional activation of the CYP1A1 gene is mediated by aryl hydrocarbon receptor (AHR), a basic-helix-loop-helix (bHLH) transcription factor. In the absence of a ligand AHR stays predominantly in the cytoplasm. Ligand binding causes translocation of AHR to the nuclear compartment, its heterodimerization with another bHLH protein, the aryl hydrocarbon nuclear translocator (ARNT) and binding of the AHR/ARNT heterodimer to a DNA motif designated dioxin responsive element (DRE). This process leads to the transcriptional activation of the responsive genes containing DREs in their regulatory regions, e.g. that coding for CYP1A1. TCDD is the most potent known agonist of AHR. Since it is not metabolized by the activated enzymes, exposure to this compound leads to a persisting activation of AHR resulting in diverse toxic effects in the organism. To enlighten the molecular mechanisms that mediate the toxicity of xenobiotics like TCDD and related compounds, the AHR-dependent regulation of the CYP1A1 gene was investigated in two cell lines: human cervix carcinoma (HeLa) and mouse hepatoma (Hepa). Study of AHR activation and its consequence concerning expression of the CYP1A1 enzyme confirmed the TCDD-dependent formation of the AHR/ARNT complex on DRE leading to an increase of the CYP1A1 transcription in Hepa cells. In contrast, in HeLa cells formation of the AHR/ARNT heterodimer and binding of a protein complex containing AHR and ARNT to DRE occurred naturally in the absence of TCDD. Moreover, treatment with TCDD did not affect the AHR/ARNT dimer formation and binding of these proteins to DRE in these cells. Even though the constitutive complex on DRE exists in HeLa, transcription of the CYP1A1 gene was not increased. Furthermore, the CYP1A1 level in HeLa cells remained unchanged in the presence of TCDD suggesting repressional mechanism of the AHR complex function which may hinder the TCDD-dependent mechanisms in these cells. Similar to the native, the mouse CYP1A1-driven reporter constructs containing different regulatory elements were not inducible by TCDD in HeLa cells, which supported a presence of cell type specific trans-acting factor in HeLa cells able to repress both the native CYP1A1 and CYP1A1-driven reporter genes rather than species specific differences between CYP1A1 genes of human and rodent origin. The different regulation of the AHR-mediated transcription of CYP1A1 gene in Hepa and HeLa cells was further explored in order to elucidate two aspects of the AHR function: (I) mechanism involved in the activation of AHR in the absence of exogenous ligand and (II) factor that repress function of the exogenous ligand-independent AHR/ARNT complex. Since preliminary studies revealed that the activation of PKA causes an activation of AHR in Hepa cells in the absence of TCDD, the PKA-dependent signalling pathway was the proposed endogenous mechanism leading to the TCDD-independent activation of AHR in HeLa cells. Activation of PKA by forskolin or db-cAMP as well as inhibition of the kinase by H89 in both HeLa and Hepa cells did not lead to alterations in the AHR interaction with ARNT in the absence of TCDD and had no effect on binding of these proteins to DRE. Moreover, the modulators of PKA did not influence the CYP1A1 activity in these cells in the presence and in the absence of TCDD. Thus, an involvement of PKA in the regulation of the CYP1A1 Gen in HeLa cells was not evaluated in the course of this study. Repression of genes by transcription factors bound to their responsive elements in the absence of ligands has been described for nuclear receptors. These receptors interact with protein complex containing histone deacetylase (HDAC), enzyme responsible for the repressional effect. Thus, a participation of histone deacetylase in the transcriptional modulation of CYP1A1 gene by the constitutively DNA-bound AHR/ARNT complex was supposed. Inhibition of the HDAC activity by trichostatin A (TSA) or sodium butyrate (NaBu) led to an increase of the CYP1A1 transcription in the presence but not in the absence of TCDD in Hepa and HeLa cells. Since amount of the AHR and ARNT proteins remained unchanged upon treatment of the cells with TSA or NaBu, the transcriptional upregulation of CYP1A1 gene was not due to an increased expression of the regulatory proteins. These findings strongly suggest an involvement of HDAC in the repression of the CYP1A1 gene. Similar to the native human CYP1A1 also the mouse CYP1A1-driven reporter gene transfected into HeLa cells was repressed by histone deacetylase since the presence of TSA or NaBu led to an increase in the reporter activity. Induction of reporter gene did not require a presence of the promoter or negative regulatory regions of the CYP1A1 gene. A promoter-distal fragment containing three DREs together with surrounding sequences was sufficient to mediate the effects of the HDAC inhibitors suggesting that the AHR/ARNT binding to its specific DNA recognition site may be important for the CYP1A1 repression. Histone deacetylase is recruited to the specific genes by corepressors, proteins that bind to the transcription factors and interact with other members of the HDAC complex. Western blot analyses revealed a presence of HDAC1 and the corepressors mSin3A (mammalian homolog of yeast Sin3) and SMRT (silencing mediator for retinoid and thyroid hormone receptor) in both cell types, while the corepressor NCoR (nuclear receptor corepressor) was expressed exclusively in HeLa cells. Thus the high inducibility of CYP1A1 in Hepa cells may be due to the absence of NCoR in these cells in contrast to the non-responsive HeLa cells, where the presence of NCoR would support repression of the gene by histone deacetylase. This hypothesis was verified in reporter gene experiments where expression constructs coding for the particular members of the HDAC complex were cotransfected in Hepa cells together with the TCDD-inducible reporter constructs containing the CYP1A1 regulatory sequences. An overexpression of NCoR however did not decrease but instead led to a slight increase of the reporter gene activity in the cells. The expected inhibition was observed solely in the case of SMRT that slightly reduced constitutive and TCDD-induced reporter gene activity. A simultaneous expression of NCoR and SMRT shown no further effects and coexpression of HDAC1 with the two corepressors did not alter this situation. Thus, additional factors that are likely involved in the repression of CYP1A1 gene by HDAC complex remained to be identified. Taking together, characterisation of an exogenous ligand independent AHR/ARNT complex on DRE in HeLa cells that repress transcription of the CYP1A1 gene creates a model system enabling investigation of endogenous processes involved in the regulation of AHR function. This study implicates HDAC-mediated repression of CYP1A1 gene that contributes to the xenobiotic-induced expression in a tissue specific manner. Elucidation of these processes gains an insight into mechanisms leading to deleterious effects of TCDD and related compounds.

Relevância:

80.00% 80.00%

Publicador:

Resumo:

Maligne Melanome sind gegenüber Chemotherapeutika relativ resistent. Das methylierende Alkylanz Temozolomid sowie das chlorethylierende und DNA-Interstrand Crosslink (ICL) bildende Alkylanz Fotemustin kommen bei der Behandlung des malignen Melanoms als Mittel erster Wahl zum Einsatz. In der vorliegenden Arbeit konnte das erste Mal nachgewiesen werden, dass die zytotoxische Wirkung von Temozolomid und Fotemustin in Melanomzellen durch Apoptose vermittelt wird. Unter Verwendung klinisch relevanter Dosen der beiden Alkylantien konnte die Induktion von Apoptose durch vier unabhängige Methoden (Bestimmung der SubG1-Fraktion und der Apoptose- / Nekrose-Frequenz, Aktivierung der Effektorcaspasen-3 und -7 sowie Spaltung von PARP-1) nachgewiesen werden. Die Alkylierungen an der O6-Position des Guanins, welche durch beide Agenzien induziert werden, sind auch in Melanomzellen die wichtigsten Zytotoxizität-bewirkenden Läsionen in der DNA, und die O6-Methylguanin-DNA-Methyltransferase (MGMT) ist folglich ein herausragender Resistenzmarker. Eine der verwendeten Zelllinien (D05) exprimierte p53-Wildtypprotein. Diese Zelllinie war resistenter als alle anderen Zelllinien gegenüber Temozolomid und Fotemustin. Dies weist darauf hin, dass p53 nicht die Apoptoseinduktion in Melanomzellen verstärkt. Die Prozessierung des O6MeG erfolgt über die Mismatch-Reparatur (MMR) unter Generierung von DNA-Doppelstrangbrüchen (DSBs). Die Untersuchung der durch Temozolomid induzierten DSBs, nachgewiesen durch gammaH2AX-Induktion, korrelierte direkt mit der apoptotischen Antwort von Melanomzelllinien und DSBs können somit als eine entscheidende apoptoseauslösende Größe angesehen werden. Eine Resistenz gegenüber dem methylierenden Temozolomid in der Zelllinie MZ7 konnte auf einen Defekt in der MMR-Schadenserkennung auf der Ebene des MutSalpha-Komplexes zurückgeführt werden. Dieser Defekt hatte keinen Einfluss auf die Fotemustin-vermittelte Apoptoseinduktion. Neben MGMT konnte somit die MMR als Resistenzfaktor gegenüber methylierenden Agenzien in Melanomen identifiziert werden. Die Fotemustin-induzierte Apoptose wurde in Melanomzelllinien im Detail untersucht. Es konnte erstmals gezeigt werden, dass Fotemustin-bedingte ICLs in Zellen einen G2/M-Arrest im Behandlungszyklus induzieren. Wie anhand G1-arretierter Zellen nachgewiesen werden konnte, war das Durchlaufen der DNA-Replikation vor Erreichen des Arrests für die Induktion der Apoptose notwendig. Die Prozessierung von ICLs ist im Vergleich zu Methylierungen der DNA deutlich komplexer. Dies könnte erklären, warum in Melanomzellen die durch gammaH2AX-Induktion repräsentierten DSBs nicht mit der Sensitivität der einzelnen Zelllinien korreliert. Die Untersuchung unterschiedlich sensitiver Zelllinien zeigte ein vergleichbares Schadensniveau an ICLs und eine ebenso vergleichbare initiale Prozessierung derselben unter Generierung von DSBs. Die Prozessierung dieser sekundären Läsionen, welche anhand der Abnahme von gammaH2AX-Foci untersucht wurde, war hingegen in der sensitiveren Melanomzelllinie deutlich weniger effektiv. Es konnte weiterhin nachgewiesen werden, dass eine uneffektive Prozessierung der sekundären Läsionen einhergeht mit einer verstärkten und länger anhaltenden Aktivierung der in der DSB-Detektion beteiligten Kinase ATM und der Checkpoint Kinase 1. Es wäre daher denkbar, dass eine verstärkte Aktivität dieser Kinasen proapoptotische Signale vermittelt. Unterschiede in der Prozessierung der sekundären Läsionen könnten somit ein wichtiger Marker der ICL-induzierten Apoptose darstellen. Des weitern konnte nachgewiesen werden, dass nach Fotemustingabe die mitochondrial-vermittelte Apoptose einen effektiven Exekutionsweg in Melanomen darstellt. Während Cytochrom C-Freigabe, Bcl-2-Abnahme an den Mitochondrien, Bax-Rekrutierung und Caspase-9 Aktivität nachgewiesen werden konnten, wurden keine Hinweise auf eine Fas-Rezeptor-vermittelte Apoptose gefunden. Die Unfähigkeit, Rezeptor-vermittelte Apoptose zu unterlaufen, könnte die Bedeutungslosigkeit des p53-Gens in Melanomen begründen, da gerade dieser Weg in der Alkylantien-induzierten Apoptose in anderen Zellsystemen eine große Relevanz besitzt. Bei der Suche nach einem alternativen proapoptotischen Signalweg konnten Hinweise für die Beteiligung des Rb/E2F-1-Wegs, welcher über p73 agiert, in einer p53-mutierten Melanomzelllinie gefunden werden. Einen Einfluss der Proteine Survivin und XIAP als Resistenzfaktoren auf die Fotemustin-induzierte Apoptose wurde hingegen nicht nachgewiesen.

Relevância:

40.00% 40.00%

Publicador:

Resumo:

Ziel dieser Arbeit war es, die funktionelle Bedeutung des Drosophila melanogaster tumor suppressor Gens lethal(2)tumorous imaginal discs (l(2)tid) durch die Identifikation von molekularen Partnern der vom Gen kodierten Proteine zu etablieren. Mit dem Screening einer Expressionsbibliothek mittels des Hefe-Di-Hybrid-Systems wurde das Protein Patched (Ptc) als ein neues Tid-bindendes Protein identifiziert. Ptc ist ein Zentralregulator der Hedhehog-Signalkette. Diese ist in der Entwicklung konserviert und in manchen humanen Krebsarten verwickelt. Die Tid/Ptc-Interaktion wurde mittels unabhängigen biochemischen Methoden wie dem GST-pulldown-Test oder der Immunopräzipitation überprüft. Außerdem ergaben funktionelle Studien in tumorosen Imaginalscheiben einen möglichen inhibitorischen Effekt von Tid über die Hh Signaltransduktion.Im letzten Teil dieser Arbeit wurde die Interaktion zwischen Tid und dem E-APC-Protein (Adenomatous polyposis coli) bewiesen. Polakis und seine Gruppe zeigten durch Studien mit dem Hefe-Di-Hybrid-System und in vitro, dass das hTid mit dem APC-Protein interagiert. Um dies auch auf Drosophila-Ebene zu überprüfen, wurden Immunopräzipitation-Studien mit den Drosophila-Gegenstücken durchgeführt. Diese Studien zeigen zum ersten Mal eine direkte Interaktion beider Proteine in vivo.

Relevância:

40.00% 40.00%

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

Relevância:

40.00% 40.00%

Publicador:

Resumo:

The cannabinoid type 1 (CB1) receptor is involved in a plethora of physiological functions and heterogeneously expressed on different neuronal populations. Several conditional loss-of-function studies revealed distinct effects of CB1 receptor signaling on glutamatergic and GABAergic neurons, respectively. To gain a comprehensive picture of CB1 receptor-mediated effects, the present study aimed at developing a gain-of-function approach, which complements conditional loss-of-function studies. Therefore, adeno-associated virus (AAV)-mediated gene delivery and Cre-mediated recombination were combined to recreate an innovative method, which ensures region- and cell type-specific transgene expression in the brain. This method was used to overexpress the CB1 receptor in glutamatergic pyramidal neurons of the mouse hippocampus. Enhanced CB1 receptor activity at glutamatergic terminals caused impairment in hippocampus-dependent memory performance. On the other hand, elevated CB1 receptor levels provoked an increased protection against kainic acid-induced seizures and against excitotoxic neuronal cell death. This finding indicates the protective role of CB1 receptor on hippocampal glutamatergic terminals as a molecular stout guard in controlling excessive neuronal network activity. Hence, CB1 receptor on glutamatergic hippocampal neurons may represent a target for novel agents to restrain excitotoxic events and to treat neurodegenerative diseases. Endocannabinoid synthesizing and degrading enzymes tightly regulate endocannabinoid signaling, and thus, represent a promising therapeutic target. To further elucidate the precise function of the 2-AG degrading enzyme monoacylglycerol lipase (MAGL), MAGL was overexpressed specifically in hippocampal pyramidal neurons. This genetic modification resulted in highly increased MAGL activity accompanied by a 50 % decrease in 2-AG levels without affecting the content of arachidonic acid and anandamide. Elevated MAGL protein levels at glutamatergic terminals eliminated depolarization-induced suppression of excitation (DSE), while depolarization-induced suppression of inhibition (DSI) was unchanged. This result indicates that the on-demand availability of the endocannabinoid 2-AG is crucial for short-term plasticity at glutamatergic synapses in the hippocampus. Mice overexpressing MAGL exhibited elevated corticosterone levels under basal conditions and an increase in anxiety-like behavior, but surprisingly, showed no changes in aversive memory formation and in seizure susceptibility. This finding suggests that 2 AG-mediated hippocampal DSE is essential for adapting to aversive situations, but is not required to form aversive memory and to protect against kainic acid-induced seizures. Thus, specific inhibition of MAGL expressed in hippocampal pyramidal neurons may represent a potential treatment strategy for anxiety and stress disorders. Finally, the method of AAV-mediated cell type-specific transgene expression was advanced to allow drug-inducible and reversible transgene expression. Therefore, elements of the tetracycline-controlled gene expression system were incorporated in our “conditional” AAV vector. This approach showed that transgene expression is switched on after drug application and that background activity in the uninduced state was only detectable in scattered cells of the hippocampus. Thus, this AAV vector will proof useful for future research applications and gene therapy approaches.