981 resultados para Histone Deacetylase Inhibitors


Relevância:

90.00% 90.00%

Publicador:

Resumo:

Short chain fatty acids (SCFAs) are fermentation products of anaerobic bacteria. More than just being an important energy source for intestinal epithelial cells, these compounds are modulators of leukocyte function and potential targets for the development of new drugs. The aim of this study was to evaluate the effects of SCFAs (acetate, propionate and butyrate) on production of nitric oxide (NO) and proinflammatory cytokines [tumor necrosis factor alpha (TNF-alpha) and cytokine-induced neutrophil chemoattractant-2 (CINC-2 alpha beta)] by rat neutrophils. The involvement of nuclear factor kappa B (NF-kappa B) and histone deacetylase (HDAC) was examined. The effect of butyrate was also investigated in vivo after oral administration of tributyrin (a pro-drug of butyrate). Propionate and butyrate diminished TNF-alpha, CINC-2 alpha beta and NO production by LPS-stimulated neutrophils. We also observed that these fatty acids inhibit HDAC activity and NF-kappa B activation, which might be involved in the attenuation of the LPS response. Products of cyclooxygenase and 5-lipoxygenase are not involved in the effects of SCFAs as indicated by the results obtained with the inhibitors of these enzymes. The recruitment of neutrophils to the peritonium after intraperitoneal administration of a glycogen solution (1%) and the ex vivo production of cytokines and NO by neutrophils were attenuated in rats that previously received tributyrin. These results argue that this triglyceride may be effective in the treatment of inflammatory conditions. Crown Copyright (C) 2011 Published by Elsevier Inc. All rights reserved.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP)

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:

90.00% 90.00%

Publicador:

Resumo:

Die am häufigsten auftretende altersassoziierte neurodegenerative Krankheit ist die Alzheimer Demenz. Ein mit entscheidender Schritt bei der Entstehung der Alzheimer Erkrankung ist wahrscheinlich die Produktion des Aβ-Peptids durch proteolytische Spaltung das Amyloid-Vorläuferproteins APP. In der vorliegenden Arbeit wurde die altersabhängige Prozessierung des Amyloid-Vorläuferproteins (APP) in Fibroblasten von Hautbiopsien von Familiärer Alzheimer-, Trisomie21 und Niemann-Pick Typ C-Krankheit untersucht. Die in dieser Arbeit verwendeten Fibroblasten wurden bis zum Erreichen des zellulären Wachstumsstopps (replikative Seneszenz) seriell passagiert und die Untersuchungen erfolgten an Zellen aufsteigender PDL. Dabei zeigte sich, dass, unabhängig von dem durch die Krankheit vorliegenden genetischen biochemischen Hintergrund, die APP-Prozessierung im Laufe der Zellalterung progressiv verringert wird. Die altersabhängig ansteigenden Cholesterinspiegel führten zu einer Reduktion der APP-Reifung und infolge dessen nahmen sowohl die intrazellulären APP-Spaltfragmente (C99, C83 und AICD) als auch die extrazellulären APP-Fragmente (sAPPα, sAPP) ab. Ebenso konnte gezeigt werden, dass die γ-Sekretase-Aktivität abnimmt. Dies war verbunden mit einem Rückgang der Proteinspiegel von Nicastrin und Presenilin, beides Komponenten des γ-Sekretase-Komplexes. Obwohl die Proteinexpression der α-Sekretase ADAM10 altersassoziiert konstant blieb, nahm die α-Sekretase-Aktivität mit steigendem Lebensalter ab. Erste Untersuchungen zeigten, dass die NAD+-abhängige Histon-Deacetylase SIRT1 eine wichtige Rolle im Bezug auf die α-Sekretase-Aktivität spielen könnte. Im Gegensatz zu den Abnahmen der α- und γ-Sekretase-Aktivitäten konnte eine erhöhte Aktivität der β-Sekretase in seneszenten Zellen beobachtet werden. Die mRNA-Menge und Proteinspiegel der ß-Sekretase BACE1 blieben dabei unverändert. Des Weiteren zeigte sich eine Zunahme der β-Sekretase-Aktivität bei Behandlung von jungen Zellen mit konditioniertem Medium seneszenter Zellen. Da sensezente Zellen einem Proliferationsstopp in der G1-Phase unterliegen, wurde der Einfluss des Zellzyklus-Inhibitors Aphidicolin auf die β-Sekretase untersucht. Hier wurde sowohl in IMR90 Fibroblasten als auch in Neuroblastoma-Zellen N2a eine Zunahme der β-Sekretase-Aktivität nach Zugabe der Inhibitoren beobachtet. Auch kommt es im Zuge der Alterung zu einer verstärkten Expression inflammatorischer Zytokine, die mit der Entstehung von Aβ-Peptiden in Verbindung gebracht werden. Deshalb wurde der Einfluss von Zytokinen auf die β-Sekretase-Aktivität untersucht. Die Zugabe von Interferon-γ und Interleukin 6 führte bei jungen IMR90-Zellen zu einem Anstieg der β-Sekretase-Aktivität, während bei alten Zellen keine Änderung zu verzeichnen war.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Nox4 is a member of the NADPH oxidase family, which represents a major source of reactive oxygen species (ROS) in the vascular wall. Nox4-mediated ROS production mainly depends on the expression levels of the enzyme. The aim of my study was to investigate the mechanisms of Nox4 transcription regulation by histone deacetylases (HDAC). Treatment of human umbilical vein endothelial cells (HUVEC) and HUVEC-derived EA.hy926 cells with the pan-HDAC inhibitor scriptaid led to a marked decrease in Nox4 mRNA expression. A similar down-regulation of Nox4 mRNA expression was observed by siRNA-mediated knockdown of HDAC3. HDAC inhibition in endothelial cells was associated with enhanced histone acetylation, increased chromatin accessibility in the human Nox4 promoter region, with no significant changes in DNA methylation. In addition, the present study provided evidence that c-Jun played an important role in controlling Nox4 transcription. Knockdown of c-Jun with siRNA led to a down-regulation of Nox4 mRNA expression. In response to scriptaid treatment, the binding of c-Jun to the Nox4 promoter region was reduced despite the open chromatin structure. In parallel, the binding of RNA polymerase IIa to the Nox4 promoter was significantly inhibited as well, which may explain the reduction in Nox4 transcription. In conclusion, HDAC inhibition decreases Nox4 transcription in human endothelial cells by preventing the binding of transcription factor(s) and polymerase(s) to the Nox4 promoter, most likely because of a hyperacetylation-mediated steric inhibition. In addition, HDAC inhibition-induced Nox4 downregulation may also involves microRNA-mediated mRNA destabilization, because the effect of the scriptaid could be partially blocked by DICER1 knockdown or by transcription inhibition.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Krebs stellt eine der häufigsten Todesursachen in Europa dar. Grundlage für eine langfristige Verbesserung des Behandlungserfolgs ist ein molekulares Verständnis der Mechanismen, welche zur Krankheitsentstehung beitragen. In diesem Zusammenhang spielen Proteasen nicht nur eine wichtige Rolle, sondern stellen auch bei vielerlei Erkrankungen bereits anerkannte Zielstrukturen derzeitiger Behandlungsstrategien dar. Die Protease Threonin Aspartase 1 (Taspase1) spielt eine entscheidende Rolle bei der Aktivierung von Mixed Lineage Leukemia (MLL)-Fusionsproteinen und somit bei der Entstehung aggressiver Leukämien. Aktuelle Arbeiten unterstreichen zudem die onkologische Relevanz von Taspase1 auch für solide Tumore. Die Kenntnisse über die molekularen Mechanismen und Signalnetzwerke, welche für die (patho)biologischen Funktionen von Taspase1 verantwortlich sind, stellen sich allerdings noch immer als bruchstückhaft dar. Um diese bestehenden Wissenslücken zu schließen, sollten im Rahmen der Arbeit neue Strategien zur Inhibition von Taspase1 erarbeitet und bewertet werden. Zusätzlich sollten neue Einsichten in evolutionären Funktionsmechanismen sowie eine weitergehende Feinregulation von Taspase1 erlangt werden. Zum einen erlaubte die Etablierung und Anwendung eines zellbasierten Taspase1-Testsystem, chemische Verbindungen auf deren inhibitorische Aktivität zu testen. Überraschenderweise belegten solch zelluläre Analysen in Kombination mit in silico-Modellierungen eindeutig, dass ein in der Literatur postulierter Inhibitor in lebenden Tumorzellen keine spezifische Wirksamkeit gegenüber Taspase1 zeigte. Als mögliche Alternative wurden darüber hinaus Ansätze zur genetischen Inhibition evaluiert. Obwohl publizierte Studien Taspase1 als ααββ-Heterodimer beschreiben, konnte durch Überexpression katalytisch inaktiver Mutanten kein trans-dominant negativer Effekt und damit auch keine Inhibition des wildtypischen Enzyms beobachtet werden. Weiterführende zellbiologische und biochemische Analysen belegten erstmalig, dass Taspase1 in lebenden Zellen in der Tat hauptsächlich als Monomer und nicht als Dimer vorliegt. Die Identifizierung evolutionär konservierter bzw. divergenter Funktionsmechanismen lieferte bereits in der Vergangenheit wichtige Hinweise zur Inhibition verschiedenster krebsrelevanter Proteine. Da in Drosophila melanogaster die Existenz und funktionelle Konservierung eines Taspase1-Homologs postuliert wurde, wurde in einem weiteren Teil der vorliegenden Arbeit die evolutionäre Entwicklung der Drosophila Taspase1 (dTaspase1) untersucht. Obwohl Taspase1 als eine evolutionär stark konservierte Protease gilt, konnten wichtige Unterschiede zwischen beiden Orthologen festgestellt werden. Neben einem konservierten autokatalytischen Aktivierungsmechanismus besitzt dTaspase1 verglichen mit dem humanen Enzym eine flexiblere Substraterkennungs-sequenz, was zu einer Vergrößerung des Drosophila-spezifischen Degradoms führt. Diese Ergebnisse zeigen des Weiteren, dass zur Definition und Vorhersage des Degradoms nicht nur proteomische sondern auch zellbiologische und bioinformatische Untersuchungen geeignet und notwendig sind. Interessanterweise ist die differentielle Regulation der dTaspase1-Aktivität zudem auf eine veränderte intrazelluläre Lokalisation zurückzuführen. Das Fehlen von in Vertebraten hochkonservierten aktiven Kernimport- und nukleolären Lokalisationssignalen erklärt, weshalb dTaspase1 weniger effizient nukleäre Substrate prozessiert. Somit scheint die für die humane Taspase1 beschriebene Regulation von Lokalisation und Aktivität über eine Importin-α/NPM1-Achse erst im Laufe der Entwicklung der Vertebraten entstanden zu sein. Es konnte also ein bislang unbekanntes evolutionäres Prinzip identifiziert werden, über welches eine Protease einen Transport- bzw. Lokalisations-basierten Mechanismus zur Feinregulation ihrer Aktivität „von der Fliege zum Menschen“ nutzt. Eine weitere Möglichkeit zur dynamischen Funktionsmodulation bieten post-translationale Modifikationen (PTMs) der Proteinsequenz, zu welcher Phosphorylierung und Acetylierung zählen. Interessanterweise konnte für die humane Taspase1 über den Einsatz unabhängiger Methoden einschließlich massenspektrometrischer Analysen eine Acetylierung durch verschiedene Histon-Acetyltransferasen (HATs) nachgewiesen werden. Diese Modifikation erfolgt reversibel, wobei vor allem die Histon-Deacetylase HDAC1 durch Interaktion mit Taspase1 die Deacetylierung der Protease katalysiert. Während Taspase1 in ihrer aktiven Konformation acetyliert vorliegt, kommt es nach Deacetylierung zu einer Reduktion ihrer enzymatischen Aktivität. Somit scheint die Modulation der Taspase1-Aktivität nicht allein über intra-proteolytische Autoaktivierung, Transport- und Interaktionsmechanismen, sondern zudem durch post-translationale Modifikationen gesteuert zu werden. Zusammenfassend konnten im Rahmen dieser Arbeit entscheidende neue Einblicke in die (patho)biologische Funktion und Feinregulation der Taspase1 gewonnen werden. Diese Ergebnisse stellen nicht nur einen wichtigen Schritt in Richtung eines verbesserten Verständnis der „Taspase1-Biologie“, sondern auch zur erfolgreichen Inhibition und Bewertung der krebsrelevanten Funktion dieser Protease dar.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

"Silent mating type information regulation 2 Type" 1 (SIRT1), das humane Homolog der NAD+-abhängigen Histondeacetylase Sir2 aus Hefe, besitzt Schlüsselfunktionen in der Regulation des Metabolismus, der Zellalterung und Apoptose. Letztere wird vor allem durch die Deacetylierung von p53 an Lys382 und der dadurch verringerten Transkription proapoptotischer Zielgene vermittelt. Im Rahmen der vorliegenden Arbeit wurde die SIRT1 Regulation im Zusammenhang mit der DNA-Schadensantwort untersucht.rnIn der Apoptoseregulation übernimmt die Serin/Threonin-Kinase "Homeodomain interacting protein kinase" 2 (HIPK2) eine zentrale Rolle und daher wurde die SIRT1 Modifikation und Regulation durch HIPK2 betrachtet. Durch Phosphorylierung des Tumorsuppressorproteins p53 an Ser46 aktiviert HIPK2 das Zielprotein und induziert die Transkription proapoptotischer Zielgene von p53. Es wurde beschrieben, dass HIPK2 nach DNA-Schädigung über einen bisher unbekannten Mechnismus die Acetylierung von p53 potenzieren kann.rnIn der vorliegenden Arbeit konnte gezeigt werden, dass SIRT1 von HIPK2 in vitro und in Zellen an Serin 27 und 682 phosphoryliert wird. Weiterhin ist die Interaktion von SIRT1 mit HIPK2 sowie die SIRT1 Phosphorylierung an Serin 682 durch DNA-schädigende Adriamycinbehandlung erhöht. Es gibt Hinweise, dass HIPK2 die Expression von SIRT1 reguliert, da HIPK2 RNA-Interferenz zur Erniedrigung der SIRT1 Protein- und mRNA-Mengen führt.rnEin weiterer interessanter Aspekt liegt in der Beobachtung, dass Ko-Expression von PML-IV, welches SIRT1 sowie HIPK2 in PML-Kernkörper rekrutiert, die SIRT1 Phosphorylierung an Serin 682 verstärkt. Phosphorylierung von SIRT1 an Serin 682 interferiert wiederum mit der SUMO-1 Modifikation, welche für die Lokalisation in PML-Kernkörpen wichtig ist.rnBemerkenswerterweise reduziert die DNA-schadendsinduzierte SIRT1 Phosphorylierung die Bindung des SIRT1 Ko-Aktivators AROS, beeinflusst aber nicht diejenige des Inhibitors DBC1. Dies führt zur Reduktion der enzymatischen Aktivität von SIRT1 und der darausfolgenden weniger effizienten Deacetylierung des Zielproteins p53.rnDurch die von mir in der vorliegenden Promotionsarbeit erzielten Ergebnisse konnte ein neuer molekularer Mechanismus entschlüsselt werden, welcher die durch HIPK2 modulierte Acetylierung von p53 und die daran anschließende Induktion der Apoptose beschreibt.rnHIPK2-vermittelte SIRT1 Phosphorylierung resultiert in einer verminderten Deacetylasefunktion von SIRT1 und führt so zu einer verstärkten acetylierungsinduzierten Expression proapoptotischer p53 Zielgene.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

The corepressor complex Tup1-Ssn6 regulates many classes of genes in yeast including cell type specific, glucose repressible, and DNA damage inducible. Tup1 and Ssn6 are recruited to target promoters through their interactions with specific DNA binding proteins such as α2, Mig1, and Crt1. Most promoters that are repressed by this corepressor complex exhibit a high degree of nucleosomal organization. This chromatin domain occludes transcription factor access to the promoter element resulting in gene repression. Previous work indicated that Tup1 interacts with underacetylated isoforms of H3 and H4, and that mutation of these histones synergistically compromises repression. These studies predict that Tup1-hypoacetyalted histone interaction is important to the repression mechanism, and in vivo hyperacetylation might compromise the corepressors ability to repress target genes. ^ One way to alter histone acetylation levels in vivo is to alter the balance between histone acetyltransferases and histone deacetylases. To date five histone deacetylases (HDACs) have been identified in yeast Rpd3, Hos1, Hos2, Hos3 and Hda1. Deletion of single or double HDAC genes had little to no effect on Tup1-Ssn6 repression, but simultaneous deletion of three specific activities Rpd3, Hos1, and Hos2 abolished repression in vivo. Promoter regions of Tup1-Ssn6 target genes in these triple deacetylase mutant cells are dramatically hyperacetylated in both H3 and H4. Examination of bulk histone acetylation levels showed that this specific HDAC triple mutant combination (rpd3 hos1 hos2) caused a dramatic and concomitant hyperacetylation of both H3 and H4. The loss of repression in the rpd3 hos1 hos2 cells, but not in other mutants, is consistent with previous observations, which indicate that histones provide redundant functions in the repression mechanism and that high levels of acetylation are required to prevent Tup1 binding. Investigation into a potential direct interaction between the Tup1-Ssn6 corepressor complex and one or more HDAC activities showed that both Rpd3 and Hos2 interact with the corepressor complex in vivo. These findings indicate that Tup1-Ssn6 repression involves the recruitment of histone deacetylase activities to target promoters, where they locally deacetylate histone residues promoting Tup1-histone tail interaction to initiate and/or maintain the repressed state. ^

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Bortezomib (VELCADE™, formerly known as PS-341) is a selective and potent inhibitor of the proteasome that was recently FDA-approved for the treatment of multiple myeloma. Despite its success in multiple myeloma and progression into clinical trials for other malignancies, bortezomib's exact mechanism of action remains undefined. The major objective of this study was to evaluate the anticancer activity of this drug using in vitro and in vivo pancreatic cancer models and determine whether bortezomib-induced apoptosis occurs via induction of endoplasmic reticular (ER) stress. The investigation revealed that bortezomib inhibited tumor cell proliferation via abrogation of cdk activity and induced apoptosis in pancreatic cancer cell lines. I hypothesized that bortezomib-induced apoptosis was triggered by a large accumulation ubiquitin-conjugated proteins that resulted in ER stress. My data demonstrated that bortezomib induced a unique type of ER stress in that it inhibited PKR-like ER kinase (PERK) and subsequent phosphorylation of eukaryotic initiation factor 2α (eif2α), a key event in translational suppression. The combined effects of proteasome inhibition and the failure to attenuate translation resulted in an accumulation of aggregated proteins (proteotoxicity), JNK activation, cytochrome c release, caspase-3 activation, and DNA fragmentation. Bortezomib also enhanced apoptosis induced by other agents that stimulated the unfolded protein response (UPR), demonstrating that translational suppression is a critical cytoprotective mechanism during ER stress. Tumor cells attempt to survive bortezomib-induced ER stress by sequestering aggregated proteins into large structures, termed aggresomes. Since histone deacetylase 6 (HDAC6) is essential for aggresome formation, tumor cells may be sensitized to bortezomib-induced apoptosis by blocking HDAC function. My results demonstrated that HDAC inhibitors disrupted aggresome formation and synergized with bortezomib to induce apoptosis in pancreatic cancer or multiple myeloma cells in vitro and in orthotopic pancreatic tumors in vivo. Taken together, my data establish a mechanistic link between bortezomib-induced aggresome formation, ER stress, and apoptosis and identify a novel therapeutic strategy for the treatment of pancreatic cancer and other hematologic and solid malignancies. ^

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Gastrointestinal Stromal Tumors (GIST) are sarcomas driven by gain-of-function mutations of KIT or PDGFRA. Although, the introduction of tyrosine kinase inhibitors has dramatically changed the history of this disease, evidences emerge that inhibition of KIT or PDGFRA are not sufficient to cure patients. The developmental pathway Notch has a critical role in the cell fate, regulating cell proliferation and differentiation. Dysregulation of Notch pathway has been implicated in a wide variety of cancers functioning as a tumor promoter or a tumor suppressor in a cell context dependent manner. Given that Notch activation deregulates the morphogenesis of mesenchymal cells in the GI track, that Notch acts as a tumor suppressor in neuroendocrine tumors, and finally that the cell of origin of GIST are the Interstitial Cell of Cajal that arise from a mesenchymal origin with some neuroendocrine features, we hypothesized that Notch pathway signaling may play a role in growth, survival and differentiation of GIST cells. To test this hypothesis, we genetically and pharmacologically manipulated the Notch pathway in human GIST cells. In this study, we demonstrated that constitutively active intracellular domain of Notch1 (ICN-1) expression potently induced growth arrest and downregulated KIT expression. We have performed a retrospective analysis of 15 primary GIST patients and found that high mRNA level of Hes1, a major target gene of Notch pathway, correlated with a significantly longer relapse-free survival. Therefore, we have established that treatment with the FDA approved histone deacetylase inhibitor SAHA (Vorinostat) caused dose-dependent upregulation of Notch1 expression and a parallel decrease in viability in these cells. Retroviral silencing of downstream targets of Notch with dominant negative Hes-1 as well as pharmacological inhibition of Notch pathway with a γ-secretase inhibitor partially rescued GIST cells from SAHA treatment. Taken together these results identify anti-tumor effect of Notch1 and a negative cross-talk between Notch1 and KIT pathways in GIST. Consequently, we propose that activation of this pathway with HDAC inhibitors may be a potential therapeutic strategy for GIST patients.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Targeting Histone deacetylases (HDAC) for the treatment of genetically complex soft tissue sarcoma Histone deactylase inhibitors (HDACi) are a new class of anticancer therapeutics; however, little is known about HDACi or the individual contribution of HDAC isoform activity in soft tissue sarcoma (STS). We investigated the potential efficacy of HDACi as monotherapy and in combination with chemotherapy in a panel of genetically complex STS. We found that HDACi combined with chemotherapy significantly induced anti-STS effects in vitro and in vivo. We then focused our study of HDACi in malignant peripheral nerve sheath tumor (MPNST), a subtype of highly aggressive, therapeutically resistant, and commonly fatal malignancies that occur in patients with neurofibromatosis type-1 (NF1) or sporadically. The therapeutic efficacy of HDACi was investigated in a panel of NF1-associated and sporadic MPNST cell lines. Our results demonstrate the NF1-assocaited cohort to be highly sensitive to HDACi while sporadic cell lines exhibited resistance. HDACi-induced productive autophagy was found to be a mode of resistance and inhibiting HDACi-induced autophagy significantly induced pro-apoptotic effects of HDACi in vitro and in vivo. HDACs are not a single enzyme consisting of 11 currently known isoforms. HDACis used in these studies inhibit a variety of these isoforms, namely class I HDACs which include HDAC1, 2, 3, and 8. Recently, HDAC8-specific inhibitors (HDAC8i) have been created and tested in various cancer cell lines. Lastly, the potential therapeutic efficacy of HDAC8i was investigated in human (NF1-associated and sporadic) and NF1-associated murine-derived MPNST. HDAC8i abrogated cell growth in human and murine-derived MPNST cells. Similar to the pattern noticed with pan-HDACis NF1-associated cells, especially murine-derived, were more sensitive to HDAC8i compared to human sporadic MPNST cell lines. S-phase arrest was observed in human and murine MPNST cells, independent of p53 mutational and NF1 status. HDAC8i induced apoptosis is all cell lines tested, with a more pronounced effects in human and murine-derived NF1-associated cells. Most importantly, HDAC8i abrogated murine-derived MPNST xenograft growth in vivo. Taken together, these findings support the evaluation of pan-HDACi and isoform-specific inhibitors as a novel therapy to treat MPNST, including in combination with autophagy blocking combination regimens in particular for patients with sporadic MPNST.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Amphibian metamorphosis is marked by dramatic, thyroid hormone (TH)-induced changes involving gene regulation by TH receptor (TR). It has been postulated that TR-mediated gene regulation involves chromatin remodeling. In the absence of ligand, TR can repress gene expression by recruiting a histone deacetylase complex, whereas liganded TR recruits a histone acetylase complex for gene activation. Earlier studies have led us to propose a dual function model for TR during development. In premetamorphic tadpoles, unliganded TR represses transcription involving histone deacetylation. During metamorphosis, endogenous TH allows TR to activate gene expression through histone acetylation. Here using chromatin immunoprecipitation assay, we directly demonstrate TR binding to TH response genes constitutively in vivo in premetamorphic tadpoles. We further show that TH treatment leads to histone deacetylase release from TH response gene promoters. Interestingly, in whole animals, changes in histone acetylation show little correlation with the expression of TH response genes. On the other hand, in the intestine and tail, where TH response genes are known to be up-regulated more dramatically by TH than in most other organs, we demonstrate that TH treatment induces gene activation and histone H4 acetylation. These data argue for a role of histone acetylation in transcriptional regulation by TRs during amphibian development in some tissues, whereas in others changes in histone acetylation levels may play no or only a minor role, supporting the existence of important alternative mechanisms in gene regulation by TR.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Recent evidence suggests that the Myc and Mad1 proteins are implicated in the regulation of the gene encoding the human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase. We have analyzed the in vivo interaction between endogenous c-Myc and Mad1 proteins and the hTERT promoter in HL60 cells with the use of the chromatin immunoprecipitation assay. The E-boxes at the hTERT proximal promoter were occupied in vivo by c-Myc in exponentially proliferating HL60 cells but not in cells induced to differentiate by DMSO. In contrast, Mad1 protein was induced and bound to the hTERT promoter in differentiated HL60 cells. Concomitantly, the acetylation of the histones at the promoter was significantly reduced. These data suggest that the reciprocal E-box occupancy by c-Myc and Mad1 is responsible for activation and repression of the hTERT gene in proliferating and differentiated HL60 cells, respectively. Furthermore, the histone deacetylase inhibitor trichostatin A inhibited deacetylation of histones at the hTERT promoter and attenuated the repression of hTERT transcription during HL60 cell differentiation. In addition, trichostatin A treatment activated hTERT transcription in resting human lymphocytes and fibroblasts. Taken together, these results indicate that acetylation/deacetylation of histones is operative in the regulation of hTERT expression.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Resistance to radiotherapy due to insufficient cancer cell death is a significant cause of treatment failure in non-small cell lung cancer (NSCLC). The endogenous caspase-8 inhibitor, FLIP, is a critical regulator of cell death that is frequently overexpressed in NSCLC and is an established inhibitor of apoptotic cell death induced via the extrinsic death receptor pathway. Apoptosis induced by ionizing radiation (IR) has been considered to be mediated predominantly via the intrinsic apoptotic pathway; however, we found that IR-induced apoptosis was significantly attenuated in NSCLC cells when caspase-8 was depleted using RNA interference (RNAi), suggesting involvement of the extrinsic apoptosis pathway. Moreover, overexpression of wild-type FLIP, but not a mutant form that cannot bind the critical death receptor adaptor protein FADD, also attenuated IR-induced apoptosis, confirming the importance of the extrinsic apoptotic pathway as a determinant of response to IR in NSCLC. Importantly, when FLIP protein levels were down-regulated by RNAi, IR-induced cell death was significantly enhanced. The clinically relevant histone deacetylase (HDAC) inhibitors vorinostat and entinostat were subsequently found to sensitize a subset of NSCLC cell lines to IR in a manner that was dependent on their ability to suppress FLIP expression and promote activation of caspase-8. Entinostat also enhanced the anti-tumor activity of IR in vivo. Therefore, FLIP down-regulation induced by HDAC inhibitors is a potential clinical strategy to radio-sensitize NSCLC and thereby improve response to radiotherapy. Overall, this study provides the first evidence that pharmacological inhibition of FLIP may improve response of NCSLC to IR.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Protein phosphatase 2A (PP2A) plays a major role in maintaining cellular signaling homeostasis in human cells by reversibly affecting the phosphorylation of a variety of proteins. Protein phosphatase methylesterase-1 (PME-1) negatively regulates PP2A activity by reversible demethylation and active site binding. Thus far, it is known that overexpression of PME-1 in human gliomas contributes to ERK pathway signaling, cell proliferation, and malignant progression. Whether PME-1-mediated PP2A inhibition promotes therapy resistance in gliomas is unknown. Specific PP2A targets regulated by PME-1 in cancers also remain elusive. Additionally, whether oncogenic function of PME-1 can be generalized to various human cancers needs to be investigated. This study demonstrated that PME-1 expression promotes kinase inhibitor resistance in glioblastoma (GBM). PME-1 silencing sensitized GBM cells to a group of clinically used indolocarbazole multikinase inhibitors (MKIs). To facilitate the quantitative evaluation of MKIs by cancer-cell specific colony formation assay, Image-J software-plugin ‘ColonyArea’ was developed. PME-1-silencing was found to reactivate specific PP2A complexes and affect PP2A-target histone deacetylase HDAC4 activity. The HDAC4 inhibition induced synthetic lethality with MKIs similar to PME-1 depletion. However, synthetic lethality by both approaches required co-expression of a pro-apoptotic protein BAD. In gliomas, PME-1 and HDAC4 expression was associated with malignant progression. Using tumor PME-1, HDAC4 and BAD expression based stratification signatures this study defined patient subgroups that are likely to respond to MKI alone or in combination with HDAC4 inhibitor therapies. In contrast to the oncogenic role of PME-1 in certain cancer types, this study established that colorectal cancer (CRC) patients with high tumor PME-1 expression display favorable prognosis. Interestingly, PME-1 regulated survival signaling did not operate in CRC cells. Summarily, this study potentiates the candidacy of PME-1 as a therapy target in gliomas, but argues against generalization of these findings to other cancers, especially CRC.