• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • 1
  • 1
  • Tagged with
  • 11
  • 11
  • 8
  • 6
  • 5
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 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.
1

Interactomics identifies novel regulators of the IRF-1 tumour suppressor protein

Narayan, Vikram January 2011 (has links)
The highly unstructured interferon regulated transcription factor IRF-1 is a tumour suppressor protein that plays vital roles in the antiviral and DNA damage response pathways. To identify interacting factors that regulate IRF-1 function and expand on the available limited information on its interactome, an in vitro screen was developed using peptide-aptamer affinity chromatography coupled with mass spectrometry. Discrete identified which bind to a number of potential transcriptional regulators including NPM1, YB-1 and TRIM28. The screen also proved useful in identifying binding proteins to the C-terminal Mf1 domain, which is vital for IRF-1-mediated growth suppression and Cdk2 repression, and additionally regulates IRF-1 stability. Thus, an LXXLL motif in the MF1 domain was found to be required for the binding of Hsp70 family members and cooperation with Hsp90 to regulated IRF-1 turnover and activity. These conclusions were supproted by the finding the Hsp90 inhibitors suppressed IRF-1-dependent transcription shortly after treatment, whilst at later time points inhibition of Hsp90 led to an Hsp70-dependent depletion of nuclear IRF-1. Conversely, the half-life of IRF-1 was increase by Hsp90 in an ATPase-dependent manner leading to the accumulation of nuclear, but not cytoplasmic, IRF-1. Additionally, a stress specific interaction between IRF-1 and the Hsp70-associated ubiquitin E3 ligase CHIP, that targets Hsp70/Hsp90 clients for proteasomal degradation, was demonstrated. Consequently, decreases in IRF-1 protein levels in cells exposed to heat stress or heavy metal ions were accompanied by the formation of IRF-1:CHIP complexes. Based on observations that CHIP ubiquitination of IRF-1 occurred both in the presence and absence of Hsp70, a model was proposed wherein Hsp70 serves as a factor that recruits CHIP to its substrates and its dissociation from the complex allosterically activates CHIP-dependent substrate ubiquitination. In support of this model, in vitro and biophysical evidence is presented, showing that CHIP in complex with Hsp70 is less flexible and less effective as an E3 ligase that CHIP alone. Thus, in agreement with recent studies, the work done in this thesis highlights the importance of conformational flexibility and of direct binding or 'docking' of CHIP to its substrate(s) in its mechanism of action.
2

Development of an intrabody capable of activating interferon regulatory factor-1 (IRF-1) and identification of IRF-1-binding peptide motifs

Möller, Angeli January 2011 (has links)
Interferon regulatory factor 1 (IRF-1) is a tumour suppressor protein and transcription factor. It has been shown to modulate target gene expression in response to stimuli, which include viral infection and DNA damage, and to be down-regulated in several forms of cancer. This thesis details the development of an intrabody, an intracellular antibody, that binds specifically to endogenous IRF-1. The binding of the intrabody to IRF-1 enhanced transcription from IRF-1-responsive reporter gene constructs and endogenous promoters, thus it was shown to activate IRF-1. Intrabody binding also increased the rate at which IRF-1 was degraded, suggesting that the intrabody epitope may be regulating both IRF-1 activity and turnover. These results were supported point mutation within the intrabody epitope (P325 to A) as the resultant mutant also displayed both a higher transcriptional activity and increased rate of degradation. In an effort to understand the mechanisms which regulate IRF-1 activity a search for novel IRF-1-interacting proteins was carried out using phage peptide display. This in vitro technique enables the identification of peptides able to bind a specific target protein. The sequence of these peptides can then be used to search protein databases for homologous, full-length proteins that could also bind the target protein. This led to the identification of an IRF-1-binding peptide that held sequence similar to a region of Zinc Finger 350 (ZNF350), a transcription factor involved in regulating the DNA damage response. Subsequently, endogenous ZNF350 and IRF-1 were co-immunoprecipitated from a human cancer cell line. The extreme C-terminus of IRF-1 was shown to be sufficient for an interaction with ZNF350, although a second, more N-terminal site was also shown to be essential for a stable intracellular interaction. This data sheds new light on the role of the extreme C-terminus of IRF-1 in modulating the protein‟s activity. This study also provides new and IRF-1-specific molecular tools, in the form of intrabodies and IRF-1-binding peptides, which could be used in the future to further characterise the activity and regulation of this tumour suppressor protein.
3

Chemical genetic manipulation of interferon regulatory factor 1 (IRF-1) using synthetic biology

Al Samman, Khaldoon Mohammed A. January 2012 (has links)
Interferon regulatory factor 1 (IRF-1), the founding member of IRF family, is a nuclear transcription factor first described as a transcription factor that binds to the upstream region of interferon induced genes following viral infection. In addition, IRF-1 has been reported to be involved in cell growth regulation, induction of apoptosis, immune responses, post-transcriptional modification, and cell transformation by oncogenes. Thus, IRF-1 shows accumulative evidence supporting the theory that IRF-1 functions as a tumour suppressor. However, we still lack the knowledge in the regulation and function behind IRF-1 and many other tumour suppressors due to the lack of synthetic tools that can aid in understanding the mechanism of cancer biology. Here we described the creation of synthetic tools that can be applied to study the role of a transcription factor(s) in cancer biology. Firstly, we described the creation, using recombineering technology, of universal bacterial artificial chromosome (BAC) targeting vector. This targeting vector, carry a cre-conditioned STOP cassette that can be targeted at a desired specific area. The resulted targeting vector can aid the generation of mice models with a conditioned knock-in subtle mutation(s). The resulted cre-conditioned mice models are an essential tool for any outstanding research project in cancer biology. Secondly, we described the development of Flp-In System™ from Invitrogen; the system can ease the generation of isogenic stable mammalian expression cell lines. Using this system, we created two isogenic stable cell lines expressing wild-type IRF-1 and a mutant that abolish IRF-1 DNA binding ability (W11R). Both cell lines were investigated using microarray analysis revealing new IRF-1 target genes. We reported the up-regulation of expected standard interferon regulatory genes such as, interleukin-24 (IL-24) and interferon regulatory factor-2 binding protein-2 (IRF2BP2) and the up-regulation of standard apoptotic genes such as, early growth response-1 (EGR-1) and prostate transmembrane protein, androgen induced-1 (PMEPA1) confirming the role of IRF-1 as a tumour suppressor. However, we also reported the up-regulation of secreted phosphoprotein-1 (SPP1) and SH3 and PX domains-2A (SH3PXD2A) which are matricellular protein produced by cancer cells playing a role in cellular adhesion, invasion, tumour growth progression and metastasis. Thus, we proposed a new biological role of IRF-1 in cellular movement. Thirdly, we described the development of a synthetic stable reporter cell line which can report IRF-1 transcriptional activity; such reporter cell line can be used once large scale screening is needed. The created stable reporter cell line was used to screen a kinase inhibitor library which has revealed C3 as an IRF-1 modifier. The newly identified IRF-1 modifier regulates IRF-1 transcriptional activity by inhibiting platelet-derived growth factor receptor (PDGFR) and/or vascular endothelial growth factor receptor (VEGFR) tyrosine kinase. Finally, we validated the synthetic Flp-In System™ by testing the system using a novel oncoprotein model. We have developed a stable cell line that overexpresses an oncoprotein named Anterior Gradient 2 (AGR-2). We have found that AGR-2 can attenuate IRF-1 protein levels dependent of p53. In addition, AGR-2 has been identified as a cellular survivor factor during unfolding protein response. In conclusion, this study descried the creation and the validation of synthetic tools: synthetic cassette for cre-conditioned mice creation, the Flp-In System™ for isogenic stable cell line creation, and IRF-1 reporter cell line for high throughput screening. All synthetic tools were validated and used to investigate IRF-1, a transcription factor that plays a role in cancer and immune system.
4

Role of C-terminal phosphorylation in the regulation of the tumour suppressor IRF-1

Russell, Fiona Margaret M. January 2013 (has links)
The transcription factor Interferon Regulatory Factor-1 (IRF-1) has been demonstrated to suppress tumour growth through the regulation of many anti-oncogenic genes. Pro- and anti-apoptotic factors, cell cycle control genes, DNA damage response genes and prometastatic factors are all under the control of IRF-1, which effects both transcriptional activation and repression. In addition to these cell autonomous tumour suppressor activities, IRF-1 is also a key regulator of the immune system and, as such, mediates immune surveillance of tumours. Numerous studies have confirmed that loss or mis-regulation of IRF-1 is a key factor in several different types of cancer. Despite strong evidence for the crucial role of IRF-1 in cancer, and frequent assertions that this protein warrants further investigation as a drug target, very little is known about its regulation. Furthermore, since recent studies have linked upregulation of IRF-1 to the development of autoimmune diseases, it is particularly important that drugs be able to decouple autoimmune and anti-cancer functions of IRF-1 to avoid harmful side effects. This thesis describes how phosphorylation of IRF-1 in its regulatory C-terminal Mf1 domain modulates transactivatory and tumour suppressor activity. Phosphospecific antibodies were developed as tools to study the C-terminal phosphorylation. Using these, it was shown that treatment of cells with Interferon-γ(IFN-γ) not only causes accumulation of IRF-1 protein, but also results in phosphorylation of IRF-1 at two sites in the C-terminal Mf1 domain. Phosphomimetic mutants demonstrated that these phosphorylations enhanced the transactivatory activity of IRF-1 at various promoters, but did not affect repressor activity. Gel shift assays revealed that dual phosphorylation of IRF-1 (IRF-1 D/D) promoted DNAbinding and suggested this was through increased interaction with the cofactor/histone acetylase p300 which induces a conformational change in IRF-1, favouring DNA-binding. Acetylation by p300 appears to be important although it is not yet clear whether this directly or indirectly affects IRF-1 activity. Since the tumour suppressor activity of IRF-1 is of particular interest, the effect of phosphorylation was examined in clonogenic and invasion assays. IRF-1 D/D more efficiently suppressed colony formation in both anchorage dependent and independent assays, and may improve inhibition of invasion in Transwell assays. Thus, cell treatment with the therapeutic agent IFN-γ nduces phosphorylation of IRF-1, resulting in enhanced DNA binding of IRF-1 through improved p300 binding. In cells the outcome is more effective tumour suppression and inhibition of metastasis.
5

Epigenetické mechanismy v regulaci exprese molekul B7-H1 a IRF-1 v nádorových buňkách. / Epigenetic mechanisms in the regulation of the B7-H1 and IRF-1 expression in tumour cells.

Hrušková, Veronika January 2014 (has links)
Interferon γ is an important T-cell helper type 1 (Th1) cytokine involves in antimicrobial immunity. It is a part of the inflammatory immune response in the site of infection. However, for its proper function, the regulation of immunity is necessary to avoid injury of the tissue caused by long-term inflammation. While interferon γ triggers expression of proinflammatory genes, it also regulates genes which inactivate immune response. The B7-H1 molecule belongs among these inhibitory regulators. Furthermore, antitumour effect of interferon γ is well-known as well. After extensive experiments, interferon γ was tested as an immunotherapeutic drug against melanomas in clinical trials. However, the trials had to be terminated prematurely because of unsuccessful results. It started to be clear that interferon γ could have also a protumour effect. Interferon γ upregulates the expression of B7-H1 molecule which aids tumour in escape from immunity. The B7-H1 molecule possesses a binding site for interferon regulatory factor 1 (IRF-1) in its promoter region. This IRF-1 is induced by interferon γ - JAK/STAT signalling pathway. In our previous research, we observed interferon γ induced DNA demethylation of promoters in genes that are involved in antigen presenting machinery. Additionally, DNA methylation of...
6

Understanding the relationship between IRF-1 and the transcriptional repressor ZNF350

Mallin, Lucy Janet January 2015 (has links)
Interferon regulatory factor-1 (IRF-1) is a transcription factor and tumour suppressor, involved in many diverse cellular processes including immune responses and growth regulation. An interesting feature of IRF-1 is that it can both activate and repress gene expression, possibly by acting with co-activator or co-repressor proteins. In a previous phage display assay, a homologous peptide to the known repressor protein, zinc finger 350 (ZNF350), was found to bind to the C-terminus of IRF-1. ZNF350, also known as ZBRK1 (Zinc finger and BRCA1-interacting protein with KRAB domain-1), is a member of the Krüppel-associated box (KRAB)-containing zinc finger (KZF) proteins, which is a group of the widely distributed transcriptional repression proteins in mammals. ZNF350 has previously been shown to repress the expression of a number of genes including ANG1 and GADD45A, often in complex with other proteins. This study confirms the direct interaction between IRF-1 and ZNF350 and identifies key residues, including the LXXLL repression motif within the C-terminus of IRF-1, necessary for the binding interface. The two proteins have additionally been shown to interact within a cellular environment, shown by using techniques including immunoprecipitation and a proximity ligation assay. In addition, the ZNF350/IRF-1 complex formation appears to occur in the basal state of the cell, as opposed to in response to cellular stress such as viral infection or DNA damage. On the basis of ZNF350 being a negative regulator of transcription, a novel technique was developed to identify putative targets of both ZNF350 and IRF-1. This involved an initial bioinformatics screen using candidate IRF-1 binding site data obtained from CENTIPEDE, an algorithm that combines genome sequence information, with cell-specific experimental data to map bound TF binding sites. This allowed for the identification of novel target genes that contained the ZNF350 consensus binding site, GGGxxCAGxxxTTT, within close proximity to an IRF-1 consensus site, such as the immune response gene IL-12A. Lastly, a peptide phage display screen was combined with high-throughput sequencing to identify other potential binding partners of ZNF350 and perhaps help to understand the mechanism by which transcriptional repression is controlled by complex formation.
7

Protein:protein interaction between Interferon Regulatory Factor (IRF-1) and necleophosmin (NPM1)

Neelagandan, Kalainanghi S. January 2016 (has links)
Interferon Regulatory Factor -1 (IRF-1) is a transcription factor that acts as a tumour suppressor in cancer cells. The inactivation or deletion of IRF-1 either in one or both allele has been frequently reported in leukaemia and myelodysplasia (MDS). On the other hand nucleophosmin (NPM), a nucleo-cytoplasmic shuttling phosphoprotein is also known to be aberrant in some form of leukaemia. NPM was first proposed as a binding partner of IRF-1 in 1997 and suggested to inactivate IRF-1 by inhibiting its DNA binding ability. No further researches on the interaction between IRF-1 and NPM1 was reported prior to the start of my PhD. In the research presented here the interaction and mechanism by which IRF-1 might be inactivated by NPM was studied. Under the context of both NPM and IRF-1 being frequently associated with leukaemia and MDS, the study was done to determine the role of NPM under its naïve condition and a most frequent mutated condition (NPMc+), where the C-terminal of NPM was frequently mutated to give rise to a cytoplasmic NPM in certain leukaemia. In this current research, the direct interaction between IRF1 and NPM was further confirmed both in vitro as well as within the cells. Following this, the effect of this interaction in respect to the leukaemic condition having NPMc+ mutation was done, by comparing the end results on AML2 (leukaemic cells with intact wild type NPM) and AML3 (leukaemic cells having a single NPM allele mutated to form NPMc+) cells. In this research, overexpression of wild type NPM (NPMwt) was found to increase IRF-1 transcriptional activity. On further analysis, the DNA binding activity of IRF-1 due to the presence of NPMwt or NPMc+ was not always inhibited, instead it shows a change in binding specificity, where NPMwt bound IRF-1, lacks DNA binding ability and DNA bound IRF-1 has a reduced binding towards NPM. This is being studied further in terms of NPM overexpression and increased IRF-1 transcriptional activity, as the order of addition (order of interaction in vivo) plays a major role in activating or deactivating IRF-1. This along with the increased transcriptional activity of IRF-1 suggests a novel function of NPM, where it could act in favour of IRF- 1 activity. Additionally, the NPM induced change in IRF-1 localisation was confirmed by the cytoplasmic localised IRF-1 in NPMc+ expressing cells and nucleolar sequestration in NPMwt overexpressing cells. This gives a novel mechanism by which NPM regulates IRF-1. Further, the NPMc+ specific colocalisation of IRF-1 urges to study the other proteins that may have been re-localised in AML cells due to the NPMc+ specific interaction. A mass spectrometric analysis on the cellular distribution of total proteins were analysed between AML2 (cells with NPMwt) and AML3 (cells containing NPMc+). Specific proteins related to cancer have been identified to be differentially distributed rather than being a random translocation. With this being said, a peptide phage display technology coupled with next generation sequencing was done to identify NPMwt binding peptides that can be used in drug discovery process or as small molecule inhibitors or activators. Three different peptides were selected at the end of the study that bind very effectively to NPMwt. These peptide can either aid or restrict NPM activity and need to be validated and studied in the future.
8

Analyse der Regulationsmechanismen des humanen Tumorsuppressor Gens H-REV107-1

Reich, Steffen 03 July 2006 (has links)
H-REV107-1 wird in normalen Geweben ubiquitär exprimiert, während die Expression in humanen Mamma-, Ovarial-, und Lungentumoren unterdrückt ist. H-REV107-1 hemmt das Tumorwachstum in vitro und in vivo. Die Expression und Regulation des H-REV107-1 Gens wurde in verschiedenen, humanen Zelllinien untersucht. In Tumor Zelllinien wird die H-REV107-1 Expression durch IFNgamma induziert Eine Korrelation der H-REV107-1 und der IRF1 Expression nach Induktion mit IFNgamma wurde gezeigt. H-rev107-1 konnte nach konditionaler IRF1 Expression, Proteinsynthese-unabhängig, nachgewiesen werden und ist ein direktes Zielgen von IRF1. Die H-rev107-1 Expression ließ sich durch Unterdrückung des MEK/ERK Signalwegs mit dem MEK1 Inhibitor PD98059 aktivieren. Dies bedeutet, dass H-REV107-1 durch mindestens zwei verschiedene Signalwege, IFNgamma und MEK/ERK, reguliert wird. Der in vitro amplifizierte H-REV107-1 Promoter enthält keine TATA-Box, sondern ein Initiator Element sowie, in dem für eine TATA-Box definierten Abstand, eine ATF2 Bindungsstelle. Die Inkubation von transient transfizierten Zellen mit TNF alpha, cAMP und IFN gamma steigerte die Luciferase Aktivität. Mit Hilfe von Deletions- und Mutationskonstrukten wurden die regulatorischen Bereiche des Promoters bestimmt. Eine Mutation der cRel-Bindungsstelle, potentiell über NFkappaB reguliert, resultierte in einer Luciferase Aktivität von nur 9% des Wildtyp Promoters. Die Mutation der CREB/ATF2 Bindungsstelle reduzierte die Luciferase Aktivität auf 37%. Die Ko-Transfektion eines NFkappaB Suppressor reduzierte die Luciferase Aktivität um 53%. Diese Ergebnisse legen nahe, dass NFkappaB und ATF2 die H-REV107-1 Expression positiv regulieren. In einem EMSA wurde die Bindung von ATF2 an die CREB/ATF-2 Bindungsstelle gezeigt. Die Ergebnisse lassen vermuten, dass H-REV107-1 durch eine IFNgamma induzierte, möglicherweise PKR vermittelte, Signalkette von den Faktoren IRF1 und ATF2 direkt, sowie von NFkappaB indirekt, reguliert wird. / The H-REV107-1 class II tumor suppressor gene is ubiquitously expressed in normal tissues and down regulated in human breast, ovarian and lung tumors. H-REV107-1 has the capacity to suppress growth of tumor cells in vitro and in vivo. H-REV107-1 is up regulated after treatment with IFN gamma. A NIH3T3 cell line harboring an estrogen inducible IRF.1/hER fusion protein showed a protein synthesis independent up regulation of H-rev107-1 expression after induction of IRF-1. H-rev107-1 is a direct target of IRF-1. Inhibition of the MEK/ERK pathway, using the MEK1 inhibitor PD 98059, leads to a restored expression of H-rev107-1. Therefore, H-REV107-1 can be a target of the MEK/ERK-pathway. Thus, H-REV107-1 is regulated by at least two different pathways. To understand the regulatory mechanisms of the expression of the H-REV107-1 gene, the putative promoter region was analyzed in silico. The sequence was amplified and cloned. Induction of the promoter constructs with TNF alpha, cAMP and IFN gamma increased the luciferase activity. Several deletions constructs and constructs with putative transcription factor binding sites mutated were used to narrow down the important regulatory elements of the promoter. The mutations of a cRel binding site and a CREB/ATF-2 binding site decreased the luciferase activity by 91% and 63%, respectively. Co transfection of the full length promoter construct with a repressor of NFkappaB activation, reduced the luciferase activity to 47%. As a result of the investigation H-REV107-1 is directly regulated by IRF-1 and probably indirectly regulated by NFkappaB and the MEK/ERK signaling pathway. In an Electro Mobility Shift Assay (EMSA), the binding of ATF-2 to the CREB oligonucleotid was demonstrated by the use of a specific antibody. The ATF.2 binding site in the posititon –30 bp - 23 bp of the human, TATA-less H-REV107-1 promoter replaces the TATA-like element, which can be found in the H-rev107-1 promoter of rat and mouse.
9

MOLECULAR MECHANISMS OF SYNERGISTIC TRANSCRIPTIONAL REGULATION OF INDOLEAMINE 2,3-DIOXYGENASE

Robinson, Cory Michael 02 August 2004 (has links)
No description available.
10

Role de la protéine SOCS-1 dans la progression tumorale colique

Valentino, Lyne 30 September 2009 (has links) (PDF)
La protéine SOCS-1 (Suppressor Of Cytokine Signalling 1) a été historiquement caractérisée comme un régulateur négatif de la voie de signalisation JAK/STAT. Cette dernière, activée en réponse à de nombreuses cytokines, hormones et facteurs de croissance, aboutit à l'expression de nombreux gènes cibles, dont le gène codant pour la protéine SOCS-1. Dans un premier travail, nous avons étudié la régulation du gène Socs-1 après une stimulation par l'interféron-gamma. Nous avons ainsi mis en évidence l'implication des facteurs de transcription IRF-1 et Sp2 dans la régulation transcriptionnelle du gène Socs-1. Dans de nombreuses tumeurs humaines, l'expression du gène Socs-1 est inhibée par méthylation aberrante de l'ADN. Dans la lignée cellulaire métastatique colique, SW620, la réexpression de la protéine SOCS-1 provoque une inhibition des caractères invasifs. Cette transformation du phénotype cellulaire s'accompagne d'une réexpression de la E-cadherine à la membrane.

Page generated in 0.0272 seconds